1
|
Zheng XQ, Wang DB, Jiang YR, Song CL. Gut microbiota and microbial metabolites for osteoporosis. Gut Microbes 2025; 17:2437247. [PMID: 39690861 DOI: 10.1080/19490976.2024.2437247] [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: 07/11/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Osteoporosis is an age-related bone metabolic disease. As an essential endocrine organ, the skeletal system is intricately connected with extraosseous organs. The crosstalk between bones and other organs supports this view. In recent years, the link between the gut microecology and bone metabolism has become an important research topic, both in preclinical studies and in clinical trials. Many studies have shown that skeletal changes are accompanied by changes in the composition and structure of the gut microbiota (GM). At the same time, natural or artificial interventions targeting the GM can subsequently affect bone metabolism. Moreover, microbiome-related metabolites may have important effects on bone metabolism. We aim to review the relationships among the GM, microbial metabolites, and bone metabolism and to summarize the potential mechanisms involved and the theory of the gut‒bone axis. We also describe existing bottlenecks in laboratory studies, as well as existing challenges in clinical settings, and propose possible future research directions.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Ding-Ben Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Yi-Rong Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
2
|
Prince N, Peralta Marzal LN, Roussin L, Monnoye M, Philippe C, Maximin E, Ahmed S, Salenius K, Lin J, Autio R, Adolfs Y, Pasterkamp RJ, Garssen J, Naudon L, Rabot S, Kraneveld AD, Perez-Pardo P. Mouse strain-specific responses along the gut-brain axis upon fecal microbiota transplantation from children with autism. Gut Microbes 2025; 17:2447822. [PMID: 39773319 PMCID: PMC11730631 DOI: 10.1080/19490976.2024.2447822] [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: 07/18/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Several factors are linked to the pathophysiology of autism spectrum disorders (ASD); however, the molecular mechanisms of the condition remain unknown. As intestinal problems and gut microbiota dysbiosis are associated with ASD development and severity, recent studies have focused on elucidating the microbiota-gut-brain axis' involvement. This study aims to explore mechanisms through which gut microbiota might influence ASD. Briefly, we depleted the microbiota of conventional male BALB/cAnNCrl (Balb/c) and C57BL/6J (BL/6) mice prior to human fecal microbiota transplantation (hFMT) with samples from children with ASD or their neurotypical siblings. We found mouse strain-specific responses to ASD hFMT. Notably, Balb/c mice exhibit decreased exploratory and social behavior, and show evidence of intestinal, systemic, and central inflammation accompanied with metabolic shifts. BL/6 mice show less changes after hFMT. Our results reveal that gut microbiota alone induce changes in ASD-like behavior, and highlight the importance of mouse strain selection when investigating multifactorial conditions like ASD.
Collapse
Affiliation(s)
- Naika Prince
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lucia N. Peralta Marzal
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Léa Roussin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Magali Monnoye
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Catherine Philippe
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Elise Maximin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Sabbir Ahmed
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Karoliina Salenius
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Jake Lin
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Reija Autio
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Laurent Naudon
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Micalis Institute, Jouy-en-Josas, France
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Aletta D. Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Neuroscience, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
3
|
Wang X, Hu M, Wu W, Lou X, Gao R, Ma T, Dheen ST, Cheng J, Xiong J, Chen X, Wang J. Indole derivatives ameliorated the methamphetamine-induced depression and anxiety via aryl hydrocarbon receptor along "microbiota-brain" axis. Gut Microbes 2025; 17:2470386. [PMID: 39996473 PMCID: PMC11864316 DOI: 10.1080/19490976.2025.2470386] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/24/2025] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
In addition to the high neurotoxicity, depression, and anxiety are the most prominent characteristics of methamphetamine (Meth) withdrawal. Studies to date on the issue of Meth-associated depression and anxiety are focused on the brain, however, whether peripheral homeostasis, especially the "microbiota-gut" axis participates in these adverse outcomes, remains poorly understood. In the current study, with the fecal microbiota transplantation (FMT) assay, the mice received microbiota from Meth withdrawal mice displayed marked depression and anxiety behaviors. The 16S rRNA sequencing results showed that Meth withdrawal contributed to a striking reduction of Akkermansia, Bacteroides, Faecalibaculum, Desulfovibrio, and Anaerostipes, which are known to be associated with tryptophan (TRP) metabolism. Noteworthily, the substantial decreases of the indole derivatives from the TRP metabolic pathway, including IAA, IPA, ILA, IET, IArA, IAld, and TRM were observed in the serum of both Meth abusing humans and mice during Meth withdrawal with the UHPLC-MS/MS analysis. Combining the high and low TRP diet mouse model, the mice with high TRP diet obviously impeded Meth-associated depression and anxiety behaviors, and these results were further strengthened by the evidence that administration of IPA, IAA, and indole dramatically ameliorated the Meth induced aberrant behaviors. Importantly, these protective effects were remarkably counteracted in aryl hydrocarbon receptor knockout (AhR KO) mice, underlining the key roles of microbiota-indoles-AhR signaling in Meth-associated depression and anxiety. Collectively, the important contribution of the present work is that we provide the first evidence that peripheral gut homeostasis disturbance but not limited to the brain, plays a key role in driving the Meth-induced depression and anxiety in the periods of withdrawal, especially the microbiota and the indole metabolic disturbance. Therefore, targeting AhR may provide novel insight into the therapeutic strategies for Meth-associated psychological disorders.
Collapse
Affiliation(s)
- Xi Wang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Miaoyang Hu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Weilan Wu
- Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinyu Lou
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rong Gao
- Department of Hygienic Analysis and Detection, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Tengfei Ma
- Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - S Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jie Cheng
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jianping Xiong
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xufeng Chen
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Wang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Jin T, Li SY, Zheng HL, Liu XD, Huang Y, Ma G, Zhao YX, Zhao XT, Yang L, Wang QH, Wang HJ, Gu C, Pan Z, Lin F. Gut microbes-spinal connection is required for itch sensation. Gut Microbes 2025; 17:2495859. [PMID: 40289281 PMCID: PMC12036491 DOI: 10.1080/19490976.2025.2495859] [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: 08/26/2024] [Revised: 03/23/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
The gut microbiota has been linked to a number of neurological disorders. However, it is unclear whether the gut microbiota is involved in the genesis of chronic itch, a refractory condition that afflicts patients both physically and mentally. Here, we report that depletion of gut microbiota enhances tolerance to itch in mice orally administered with antibiotics (ABX) and mice free of germ. Of note, oral gavage with Bacteroides fragilis (B. fragilis), a prominent species of the genus Bacteroides with most differential change, corrected the ABX-induced itch dysfunction through its driven metabolite acetyl-l-carnitine (ALC). Mechanistically, gut microbiota or B. fragilis depletion caused a decrease in RNA N6-methyladenosine (m6A) demethylase FTO expression in the dorsal horn and a consequent increase in RNA m6A sites in Mas-related G protein-coupled receptor F (MrgprF) mRNA, leading to decreased MRGPRF protein. The downregulation of FTO was triggered by inactivation of ETS proto-oncogene 1 (ETS1), a transcription factor that binds to the Fto promoter. These findings support a gut microbe - spinal connection in modulation of itch sensation in RNA m6A epigenetic-dependent manner and highlight a critical role of ALC in linking the altered B. fragilis and itch dysfunction.
Collapse
Affiliation(s)
- Tong Jin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Department of Pain, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Si-Yuan Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hong-Li Zheng
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Department of Pain, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- Anesthesiology Department, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Xiao-Dan Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Department of Anesthesiology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Yue Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Gan Ma
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Ya-Xuan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xiao-Tian Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Li Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qi-Hui Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hong-Jun Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Chengyong Gu
- Anesthesiology Department, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Zhiqiang Pan
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Fuqing Lin
- Department of Pain, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
5
|
Du W, Zou ZP, Ye BC, Zhou Y. Gut microbiota and associated metabolites: key players in high-fat diet-induced chronic diseases. Gut Microbes 2025; 17:2494703. [PMID: 40260760 PMCID: PMC12026090 DOI: 10.1080/19490976.2025.2494703] [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: 01/03/2025] [Revised: 02/26/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Excessive intake of dietary fats is strongly associated with an increased risk of various chronic diseases, such as obesity, diabetes, hepatic metabolic disorders, cardiovascular disease, chronic intestinal inflammation, and certain cancers. A significant portion of the adverse effects of high-fat diet on disease risk is mediated through modifications in the gut microbiota. Specifically, high-fat diets are linked to reduced microbial diversity, an overgrowth of gram-negative bacteria, an elevated Firmicutes-to-Bacteroidetes ratio, and alterations at various taxonomic levels. These microbial alterations influence the intestinal metabolism of small molecules, which subsequently increases intestinal permeability, exacerbates inflammatory responses, disrupts metabolic functions, and impairs signal transduction pathways in the host. Consequently, diet-induced changes in the gut microbiota play a crucial role in the initiation and progression of chronic diseases. This review explores the relationship between high-fat diets and gut microbiota, highlighting their roles and underlying mechanisms in the development of chronic metabolic diseases. Additionally, we propose probiotic interventions may serve as a promising adjunctive therapy to counteract the negative effects of high-fat diet-induced alterations in gut microbiota composition.
Collapse
Affiliation(s)
- Wei Du
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhen-Ping Zou
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Bang-Ce Ye
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ying Zhou
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
6
|
Gustafson KL, Rodriguez TR, McAdams ZL, Coghill LM, Ericsson AC, Franklin CL. Failure of colonization following gut microbiota transfer exacerbates DSS-induced colitis. Gut Microbes 2025; 17:2447815. [PMID: 39812347 PMCID: PMC11740679 DOI: 10.1080/19490976.2024.2447815] [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: 09/25/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
To study the impact of differing specific pathogen-free gut microbiomes (GMs) on a murine model of inflammatory bowel disease, selected GMs were transferred using embryo transfer (ET), cross-fostering (CF), and co-housing (CH). Prior work showed that the GM transfer method and the microbial composition of donor and recipient GMs can influence microbial colonization and disease phenotypes in dextran sodium sulfate-induced colitis. When a low richness GM was transferred to a recipient with a high richness GM via CH, the donor GM failed to successfully colonize, and a more severe disease phenotype resulted when compared to ET or CF, where colonization was successful. By comparing CH and gastric gavage for fecal material transfer, we isolated the microbial component of this effect and determined that differences in disease severity and survival were associated with microbial factors rather than the transfer method itself. Mice receiving a low richness GM via CH and gastric gavage exhibited greater disease severity and higher expression of pro-inflammatory immune mediators compared to those receiving a high richness GM. This study provides valuable insights into the role of GM composition and colonization in disease modulation.
Collapse
Affiliation(s)
- Kevin L. Gustafson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Trevor R. Rodriguez
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
| | - Zachary L. McAdams
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- Molecular Pathogenesis and Therapeutics Program, University of Missouri, Columbia, MO, USA
| | - Lyndon M. Coghill
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- University of Missouri Bioinformatics and Analytics Core, University of Missouri, Columbia, MO, USA
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- University of Missouri College of Veterinary Medicine, Columbia, MO, USA
- University of Missouri Metagenomics Center, Columbia, MO, USA
| | - Craig L. Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| |
Collapse
|
7
|
Wang Z, Wang Z, Lu T, Yuan G, Chen W, Jin J, Jiang X, Yan W, Yuan K, Zou G, Bao Y, Shi J, Liu X, Wei H, Han Y, Lu L. Gut microbiota regulate insomnia-like behaviors via gut-brain metabolic axis. Mol Psychiatry 2025; 30:2597-2611. [PMID: 39658705 DOI: 10.1038/s41380-024-02867-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Abstract
Sleep interacts reciprocally with the gut microbiota. However, mechanisms of the gut microbe-brain metabolic axis that are responsible for sleep behavior have remained largely unknown. Here, we showed that the absence of the gut microbiota can alter sleep behavior. Sleep deprivation reduced butyrate levels in fecal content and the hypothalamus in specific pathogen-free mice but not in germ-free mice. The microbial metabolite butyrate can promote sleep by modulating orexin neuronal activity in the lateral hypothalamic area in mice. Insomnia patients had lower serum butyrate levels and a deficiency in butyrate-producing species within the gut microbiota. Transplantation of the gut microbiota from insomnia patients to germ-free mice conferred insomnia-like behaviors, accompanied by a decrease in serum butyrate levels. The oral administration of butyrate rescued sleep disturbances in recipient mice. Overall, these findings reveal the causal role of microbial metabolic pathways in modulating insomnia-like behaviors, suggesting potential therapeutic strategies for treating sleep disorders.
Collapse
Affiliation(s)
- Zhe Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Zhong Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China
| | - Guohao Yuan
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wenhao Chen
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jin Jin
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xianhong Jiang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400030, China
| | - Wei Yan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Guichang Zou
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China.
| | - Hong Wei
- Yu-Yue Pathology Scientific Research Center, Chongqing, 401329, China.
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China.
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, 100191, Beijing, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China.
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorders, Chinese Academy of Medical Sciences, 100730, Beijing, China.
| |
Collapse
|
8
|
Yang B, Rutkowski N, Ruta A, Gray-Gaillard E, Maestas DR, Kelly SH, Krishnan K, Wu X, Wu S, Chen A, Mejías JC, Hooks JST, Vanderzee I, Mensah P, Celik N, Eric M, Abraham P, Tam A, Housseau F, Pardoll DM, Sears CL, Elisseeff JH. Murine gut microbiota dysbiosis via enteric infection modulates the foreign body response to a distal biomaterial implant. Proc Natl Acad Sci U S A 2025; 122:e2422169122. [PMID: 40354538 DOI: 10.1073/pnas.2422169122] [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: 11/26/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
The gut microbiota influences systemic immunity and the function of distal tissues, including the brain, liver, skin, lung, and muscle. However, the role of the gut microbiota in the foreign body response and fibrosis is largely unexplored. To investigate this connection, we perturbed the homeostasis of the murine gut microbiota via infection with the pathogenic bacterial species enterotoxigenic Bacteroides fragilis (ETBF) and implanted particulate material (mean particle size <600 μm) of the synthetic polymer polycaprolactone (PCL) into a distal muscle injury. ETBF infection in mice led to increased neutrophil and γδ T cell infiltration into the PCL implant site. ETBF infection alone promoted systemic inflammation, increased levels of neutrophils in lymphoid tissues, and altered skeletal muscle gene expression. At the PCL implant site, we found significant changes in the transcriptome of sorted stromal cells between infected and control mice, including differences related to ECM components such as proteoglycans and glycosaminoglycans. However, we did not observe ETBF-induced differences in fibrosis levels. These results demonstrate the ability of the gut microbiota to mediate long-distance effects such as immune and stromal responses to a distal biomaterial implant.
Collapse
Affiliation(s)
- Brenda Yang
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Natalie Rutkowski
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Anna Ruta
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Elise Gray-Gaillard
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - David R Maestas
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Sean H Kelly
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Kavita Krishnan
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Xinqun Wu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Shaoguang Wu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Allen Chen
- Department of Biomedical Engineering, Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218
| | - Joscelyn C Mejías
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Joshua S T Hooks
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Isabel Vanderzee
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Patricia Mensah
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Nazmiye Celik
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Marie Eric
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Peter Abraham
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
| | - Ada Tam
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Franck Housseau
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Drew M Pardoll
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Cynthia L Sears
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287
| | - Jennifer H Elisseeff
- Department of Biomedical Engineering, Cellular and Molecular Medicine, or Ophthalmology, Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231
- Sidney Kimmel Comprehensive Cancer Center, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| |
Collapse
|
9
|
Hasegawa R, Poulin R, Salloum PM. Testing for Consistency in Co-occurrence Patterns Among Bacterial Taxa Across the Microbiomes of Four Different Trematode Parasites. MICROBIAL ECOLOGY 2025; 88:45. [PMID: 40382531 DOI: 10.1007/s00248-025-02545-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
Elucidating the specific processes and drivers of community assembly in the host microbiome is essential to fully understand host biology. Toward this goal, an important first step is to describe co-occurrence patterns among different microbial taxa, which can be driven by numerous factors, such as host identity. While host identity can be an important influential factor on co-occurrence patterns, a limited number of studies have explored the relative importance of host identity after controlling for other environmental factors. Here, we examined microbial co-occurrence patterns in four phylogenetically distinct trematode species living within the same snail species, collected concomitantly from the same habitat. Our previous study determined that all these trematodes shared some bacterial taxa, and the relative abundance of microbial taxa differed among trematodes, possibly due to differences in their eco-physiological traits. Here, we specifically predict that pairwise microbial co-occurrence patterns also vary among trematode host species. Our results showed that co-occurrence patterns among eight microbial families varied greatly among the four trematode hosts, with some microbial families co-occurring in some trematode species, whereas no such patterns were observed in other trematodes. Our study suggests that the habitat identity (trematode species) and its associated biotic characteristics, such as physiological and ecological traits, can determine co-occurrence patterns among microbial taxa, with substantial effects on local community composition.
Collapse
Affiliation(s)
- Ryota Hasegawa
- Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand.
| | - Robert Poulin
- Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Priscila M Salloum
- Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| |
Collapse
|
10
|
Zhang X, Wang Y, E Q, Naveed M, Wang X, Liu Y, Li M. The biological activity and potential of probiotics-derived extracellular vesicles as postbiotics in modulating microbiota-host communication. J Nanobiotechnology 2025; 23:349. [PMID: 40380331 PMCID: PMC12082936 DOI: 10.1186/s12951-025-03435-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/01/2025] [Indexed: 05/19/2025] Open
Abstract
Probiotics such as Lactobacillus and Bifidobacterium spp. have been shown to be critical for maintaining host homeostasis. In recent years, key compounds of postbiotics derived from probiotic metabolism and cellular secretion have been identified for their role in maintaining organ immunity and regulating intestinal inflammation. In particular, probiotic-derived extracellular vesicles (PEVs) can act as postbiotics, maintaining almost the same functional activity as probiotics. They also have strong biocompatibility and loading capacity to carry exogenous or parental active molecules to reach distal organs to play their roles. This provides a new direction for understanding the intrinsic microbiota-host communication mechanism. However, most current studies on PEVs are limited to their functional effects/benefits, and their specific physicochemical properties, composition, intrinsic mechanisms for maintaining host homeostasis, and possible threats remain to be explored. Here, we review and summarize the unique physicochemical properties of PEVs and their bioactivities and mechanisms in mediating microbiota-host communication, and elucidate the limitations of the current research on PEVs and their potential application as postbiotics.
Collapse
Affiliation(s)
- Xiaoming Zhang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ye Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qiyu E
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Muhammad Naveed
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiuli Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| |
Collapse
|
11
|
Schmitt C, Gasparini J, Moullec H, Walch L, Leroux-Coyau M, Leloup J. Local, environmental and trace metal effects on gut microbiota diversity in urban feral pigeons. ENVIRONMENTAL RESEARCH 2025; 273:121263. [PMID: 40024502 DOI: 10.1016/j.envres.2025.121263] [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: 11/18/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Nowadays, understanding the biotic responses to the enhanced urbanization need to encompass not the only the physiological and phenotypic features but also the related microbiota of wildlife animals. One of main threats in urban ecosystems is the chemical pollution. Thus, we have explored whether the cloacal microbiota of feral pigeons (Columba livia) is impacted by both their geographical foraging area, and metal exposure in an urban context. First, pigeons were captured in 4 specific areas of Paris (France) and placed in captivity. By applying a 16SrRNA metabarcoding approach, we observed that the gut microbiota diversity was structured according to the capture sites, with strong variation of Actinobacteria, Bacilli and Clostridia, that could be linked to the granivorous or low-protein diets. Subsequently, we experimentally exposed these pigeons to zinc and/or lead (two-factor cross design) during 90 days in a non-urban environment, but no impact on the composition nor diversity of pigeon gut microbiota, has been observed after 45 and 90 days of metal exposures. However, the composition and diversity significantly differed from the microbiota at the capture period, with the emergence of taxa belonging to Corynebacterium and Bifidobacterium in captive conditions. These data highlight a strong impact of the lifestyles (captivity in non-urban environment) on the gut microbiota composition. In parallel, we hypothesized that the diet and the local environment might have smoothed the impact of the metal exposure for pigeons that could quickly change the structure of their gut microbiota. Our findings shed light on the effects of urban pollution and environment on bird communities, that can be extended to their gut microbiota causing potential additive or synergic negative effects to host organisms and populations.
Collapse
Affiliation(s)
- Clarence Schmitt
- Sorbonne Université, Univ Paris-Cité, Univ Paris-Est, CNRS, IRD, INRAE, Institut d'écologie et des sciences de l'environnement de Paris, IEES Paris, F-75005, Paris, France
| | - Julien Gasparini
- Sorbonne Université, Univ Paris-Cité, Univ Paris-Est, CNRS, IRD, INRAE, Institut d'écologie et des sciences de l'environnement de Paris, IEES Paris, F-75005, Paris, France
| | - Héloïse Moullec
- Sorbonne Université, Univ Paris-Cité, Univ Paris-Est, CNRS, IRD, INRAE, Institut d'écologie et des sciences de l'environnement de Paris, IEES Paris, F-75005, Paris, France; Department of Biology, University of Turku, Turku, Finland
| | - Laurence Walch
- Sorbonne Université, Univ Paris-Cité, Univ Paris-Est, CNRS, IRD, INRAE, Institut d'écologie et des sciences de l'environnement de Paris, IEES Paris, F-75005, Paris, France
| | - Mathieu Leroux-Coyau
- Sorbonne Université, Univ Paris-Cité, Univ Paris-Est, CNRS, IRD, INRAE, Institut d'écologie et des sciences de l'environnement de Paris, IEES Paris, F-75005, Paris, France
| | - Julie Leloup
- Sorbonne Université, Univ Paris-Cité, Univ Paris-Est, CNRS, IRD, INRAE, Institut d'écologie et des sciences de l'environnement de Paris, IEES Paris, F-75005, Paris, France.
| |
Collapse
|
12
|
Ismeurt-Walmsley C, Giannoni P, Servant F, Mekki LN, Baranger K, Rivera S, Marin P, Lelouvier B, Claeysen S. The same but different: impact of animal facility sanitary status on a transgenic mouse model of Alzheimer's disease. mBio 2025; 16:e0400124. [PMID: 40243365 PMCID: PMC12077201 DOI: 10.1128/mbio.04001-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The gut-brain axis has emerged as a key player in the regulation of brain function and cognitive health. Gut microbiota dysbiosis has been observed in preclinical models of Alzheimer's disease and patients. Manipulating the composition of the gut microbiota enhances or delays neuropathology and cognitive deficits in mouse models. Accordingly, the health status of the animal facility may strongly influence these outcomes. In the present study, we longitudinally analyzed the fecal microbiota composition and amyloid pathology of 5XFAD mice housed in a specific opportunistic pathogen-free (SOPF) and a conventional facility. The composition of the microbiota of 5XFAD mice after aging in conventional facility showed marked differences compared to WT littermates that were not observed when the mice were bred in SOPF facility. The development of amyloid pathology was also enhanced by conventional housing. We then transplanted fecal microbiota (FMT) from both sources into wild-type (WT) mice and measured memory performance, assessed in the novel object recognition test, in transplanted animals. Mice transplanted with microbiota from conventionally bred 5XFAD mice showed impaired memory performance, whereas FMT from mice housed in SOPF facility did not induce memory deficits in transplanted mice. Finally, 18 weeks of housing SOPF-born animals in a conventional facility resulted in the reappearance of specific microbiota compositions in 5XFAD vs WT mice. In conclusion, these results show a strong impact of housing conditions on microbiota-associated phenotypes and question the relevance of breeding preclinical models in specific pathogen-free (SPF) facilities. IMPORTANCE Housing conditions affect the composition of the gut microbiota. Gut microbiota of 6-month-old conventionally bred Alzheimer's mice is dysbiotic. Gut dysbiosis is absent in Alzheimer's mice housed in highly sanitized facilities. Transfer of fecal microbiota from conventionally bred mice affects cognition. Microbiota of mice housed in highly sanitized facilities has no effect on cognition.
Collapse
Affiliation(s)
| | - Patrizia Giannoni
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, Occitanie, France
| | | | - Linda-Nora Mekki
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, Occitanie, France
| | - Kevin Baranger
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Santiago Rivera
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Philippe Marin
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, Occitanie, France
| | | | - Sylvie Claeysen
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, Occitanie, France
| |
Collapse
|
13
|
He Y, Xie K, Yang K, Wang N, Zhang L. Unraveling the Interplay Between Metabolism and Neurodevelopment in Health and Disease. CNS Neurosci Ther 2025; 31:e70427. [PMID: 40365712 PMCID: PMC12076066 DOI: 10.1111/cns.70427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Neurodevelopment is a multifaceted and tightly regulated process essential for the formation, maturation, and functional specialization of the nervous system. It spans critical stages, including cellular proliferation, differentiation, migration, synaptogenesis, and synaptic pruning, which collectively establish the foundation for cognitive, behavioral, and emotional functions. Metabolism serves as a cornerstone for neurodevelopment, providing the energy and substrates necessary for biosynthesis, signaling, and cellular activities. RESULTS Key metabolic pathways, including glycolysis, lipid metabolism, and amino acid metabolism, support processes such as cell proliferation, myelination, and neurotransmitter synthesis. Crucial signaling pathways, such as insulin, mTOR, and AMPK, further regulate neuronal growth, synaptic plasticity, and energy homeostasis. Dysregulation of these metabolic processes is linked to a spectrum of neurodevelopmental disorders, including autism spectrum disorders (ASDs), intellectual disabilities, and epilepsy. CONCLUSIONS This review investigates the intricate interplay between metabolic processes and neurodevelopment, elucidating the molecular mechanisms that govern brain development and the pathogenesis of neurodevelopmental disorders. Additionally, it highlights potential avenues for the development of innovative strategies aimed at enhancing brain health and function.
Collapse
Affiliation(s)
- Yanqing He
- Department of Neurosurgery, and National Clinical Research Center of Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of NeurosurgeryXiangya Hospital, Central South University, Jiangxi (National Regional Center for Neurological Diseases)NanchangChina
| | - Kang Xie
- Department of Neurosurgery, and National Clinical Research Center of Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of NeurosurgeryXiangya Hospital, Central South University, Jiangxi (National Regional Center for Neurological Diseases)NanchangChina
| | - Kang Yang
- Department of Neurosurgery, and National Clinical Research Center of Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of NeurosurgeryXiangya Hospital, Central South University, Jiangxi (National Regional Center for Neurological Diseases)NanchangChina
| | - Nianhua Wang
- Department of NeurosurgeryChangde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City)ChangdeHunanChina
| | - Longbo Zhang
- Department of Neurosurgery, and National Clinical Research Center of Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of NeurosurgeryXiangya Hospital, Central South University, Jiangxi (National Regional Center for Neurological Diseases)NanchangChina
| |
Collapse
|
14
|
Dai J, Yang J, Han S, Li N, Wang S, Xia S, Kim HH, Jun Y, Lee S, Kitagawa Y, Xie F, Yang L, Shen S, Chen L, Turner DP, Hodin RA, Martyn JAJ, Mao J, You Z. Deficiency of intestinal alkaline phosphatase affects behavior and microglia activity in mice. Brain Behav Immun 2025; 126:297-310. [PMID: 39984137 PMCID: PMC12051184 DOI: 10.1016/j.bbi.2025.02.006] [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: 08/17/2024] [Revised: 12/26/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
The gut microbiota plays crucial roles in the development and functions of the central nervous system (CNS) as well as in modulation of neurobehavior in heath and disease. The gut brush border enzyme intestinal alkaline phosphatase (IAP) is an important positive regulator of gut microbial homeostasis. In mice, IAP is encoded by Akp3 gene, which is specifically expressed in the duodenum of the small intestine. IAP deficiency alters gut bacterial composition and gut barrier function. Decreased IAP activity has been observed in aging, gut inflammatory diseases, and metabolic disorders. We hypothesized that this enzyme could also play an important role in modulating neurobehavior. We performed deep sequencing of gut bacterial 16S rRNA and found that IAP deficiency changed gut microbiota composition at various taxonomic levels. Using targeted metabolomic analysis, we also found that IAP deficiency resulted in changes of gut bacteria-derived metabolites in serum and brain metabolism. Neurobehavioral analyses revealed that Akp3-/- (IAP knockout) mice had decreased basal nociception thresholds, increased anxiety-like behavior, and reduced locomotor activity. Furthermore, Akp3-/- mice had more pronounced brain microglial phagocytic activity, together with an increase in the activated microglia population. Fecal microbiota transplantation from wildtype to Akp3-/- mice partially improved neurobehavior and reduced brain microglial phagocytic activity in Akp3-/- mice. This study demonstrates that deficiency of the endogenous gut-derived host factor IAP induces behavioral phenotype changes (nociception; motor activity, and anxiety) and affects brain microglia activity. Changes in the gut microbiota induced by knocking down Akp3 contribute to behavioral changes, which is probably mediated by microglia activity modulated by the gut bacteria-derived metabolites.
Collapse
Affiliation(s)
- Jiajia Dai
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospital for Children, Boston, MA, the United States of America
| | - Jinsheng Yang
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America
| | - Sen Han
- Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115, the United States of America
| | - Na Li
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America
| | - Shiyu Wang
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America
| | - Suyun Xia
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America
| | - Hyung-Hwan Kim
- Department of Radiology, Neurovascular Research Laboratory, Massachusetts General Hospital, Charlestown, MA, the United States of America
| | - Yonghyun Jun
- Department of Radiology, Neurovascular Research Laboratory, Massachusetts General Hospital, Charlestown, MA, the United States of America; Department of Anatomy, School of Medicine, Chosun University, Dong-gu, Dong-gu, Gwangju, South Korea
| | - Seeun Lee
- Department of Radiology, Neurovascular Research Laboratory, Massachusetts General Hospital, Charlestown, MA, the United States of America
| | - Yoshinori Kitagawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospital for Children, Boston, MA, the United States of America; Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Fei Xie
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospital for Children, Boston, MA, the United States of America
| | - Liuyue Yang
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America
| | - Shiqian Shen
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America
| | - Lucy Chen
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America
| | - Dana P Turner
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America
| | - Richard A Hodin
- Department of Surgery, Massachusetts General Hospital, Boston, MA, the United States of America
| | - J A Jeevendra Martyn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospital for Children, Boston, MA, the United States of America
| | - Jianren Mao
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America.
| | - Zerong You
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, the United States of America; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospital for Children, Boston, MA, the United States of America.
| |
Collapse
|
15
|
Lee SH, Han C, Shin C. IUPHAR Review: Microbiota-Gut-Brain Axis and its role in Neuropsychiatric Disorders. Pharmacol Res 2025; 216:107749. [PMID: 40306604 DOI: 10.1016/j.phrs.2025.107749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
The human gut microbiome, composed of a vast array of microorganisms that have co-evolved with humans, is crucial for the development and function of brain systems. Research has consistently shown bidirectional communication between the gut and the brain through neuronal, endocrine, and immunological, and chemical pathways. Recent neuroscience studies have linked changes in the microbiome and microbial metabolites to various neuropsychiatric disorders such as autism, depression, anxiety, schizophrenia, eating disorders, and neurocognitive disorders. Novel metagenome-wide association studies have confirmed these microbiome variations in large samples and expanded our understanding of the interactions between human genes and the gut microbiome. The causal relationship between gut microbiota and neuropsychiatric disorders is being elucidated through the establishment of large cohort studies incorporating microbiome data and advanced statistical techniques. Ongoing animal and human studies focused on the microbiota-gut-brain axis are promising for developing new prevention and treatment strategies for neuropsychiatric conditions. The scope of these studies has broadened from microbiome-modulating therapies including prebiotics, probiotics, synbiotics and postbiotics to more extensive approaches such as fecal microbiota transplantation. Recent systematic reviews and meta-analyses have strengthened the evidence base for these innovative treatments. Despite extensive research over the past decade, many intriguing aspects still need to be elucidated regarding the role and therapeutic interventions of the microbiota-gut-brain axis in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Changsu Han
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Cheolmin Shin
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Milligan YC, Peters NV, West G, Cortes LR, Chassaing B, de Vries GJ, Castillo-Ruiz A. The microbiota shapes the development of the mouse hypothalamic paraventricular nucleus. Horm Behav 2025; 172:105742. [PMID: 40262424 DOI: 10.1016/j.yhbeh.2025.105742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
Microbes massively colonize the mammalian newborn at birth. We previously reported that the microbiota influences key neurodevelopmental events, e.g., when compared to their conventionally colonized (CC) counterparts, sterile newborn mice ("germ-free" or GF) show higher cell death in the hypothalamic paraventricular nucleus (PVN). Here, we tested the hypothesis that the microbiota, perhaps via cell death mechanisms, shapes PVN development. To this aim, we used a cross-fostering approach that also allowed us to test whether any potential effects are influenced by microbial colonization at birth or programmed prenatally via the maternal microbiota. Specifically, we cross-fostered GF pups to CC dams (GF → CC) immediately after birth and compared them to control groups cross-fostered within microbial status (CC → CC, GF → GF). At postnatal day 7, GF → GF and GF → CC newborns had fewer PVN cells than did CC → CC newborns, without affecting PVN volume. In a follow-up experiment, we confirmed a reduction in PVN cell number with no change in PVN volume in adult GF mice. Thus, the greater cell death previously observed in the PVN of newborn GF mice is associated with a permanent reduction in cell number. Because the deficit is not altered by introducing a microbiota at birth, our findings also suggest that the maternal microbiota shapes development of the PVN starting in utero.
Collapse
Affiliation(s)
- Yvonne C Milligan
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - Nicole V Peters
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - Gabby West
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - Benoit Chassaing
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - Geert J de Vries
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA; Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | |
Collapse
|
17
|
Hajjeh O, Rajab I, Bdair M, Saife S, Zahran A, Nazzal I, AbuZahra MI, Jallad H, Abukhalil MM, Hallak M, Al-Said OS, Al-Braik R, Sawaftah Z, Milhem F, Almur O, Saife S, Aburemaileh M, Abuhilal A. Enteric nervous system dysfunction as a driver of central nervous system disorders: The Forgotten brain in neurological disease. Neuroscience 2025; 572:232-247. [PMID: 40088964 DOI: 10.1016/j.neuroscience.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
The Enteric Nervous System (ENS), often called the "second brain," is a complex network of neurons and glial cells within the gastrointestinal (GI) tract. It functions autonomously while maintaining close communication with the central nervous system (CNS) via the gut-brain axis (GBA). ENS dysfunction plays a crucial role in neurodegenerative and neurodevelopmental disorders, including Parkinson's disease, Alzheimer's disease, and autism spectrum disorder. Disruptions such as altered neurotransmission, gut microbiota imbalance, and neuroinflammation contribute to disease pathogenesis. The GBA enables bidirectional communication through the vagus nerve, gut hormones, immune signaling, and microbial metabolites, linking gut health to neurological function. ENS dysregulation is implicated in conditions like irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD), influencing systemic and CNS pathology through neuroinflammation and impaired barrier integrity. This review highlights emerging therapeutic strategies targeting ENS dysfunction, including prebiotics, probiotics, fecal microbiota transplantation (FMT), and vagus nerve stimulation, which offer novel ways to modulate gut-brain interactions. Unlike previous perspectives that view the ENS as a passive disease marker, this review repositions it as an active driver of neurological disorders. By integrating advances in ENS biomarkers, therapeutic targets, and GBA modulation, this article presents a paradigm shift-emphasizing ENS dysfunction as a fundamental mechanism in neurodegeneration and neurodevelopmental disorders. This perspective paves the way for innovative diagnostics, personalized gut-targeted therapies, and a deeper understanding of the ENS's role in brain health and disease.
Collapse
Affiliation(s)
- Orabi Hajjeh
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Islam Rajab
- Internal Medicine Department, St. Joseph's University Medical Center, 703 Main St, Paterson, NJ 07503, USA
| | - Mohammad Bdair
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Sarah Saife
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Anwar Zahran
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Iyad Nazzal
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammad Ibrahem AbuZahra
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Hammam Jallad
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Maram M Abukhalil
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mira Hallak
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Osama S Al-Said
- Department Of Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Rama Al-Braik
- Department Of Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Zaid Sawaftah
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Fathi Milhem
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Omar Almur
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Sakeena Saife
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammed Aburemaileh
- Department Of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Anfal Abuhilal
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| |
Collapse
|
18
|
Gui M, Lv L, Hu S, Qin L, Wang C. Sarcopenia in Parkinson's disease: from pathogenesis to interventions. Metabolism 2025; 169:156272. [PMID: 40258411 DOI: 10.1016/j.metabol.2025.156272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025]
Abstract
Parkinson's disease (PD) and sarcopenia are prevalent age-related conditions that often coexist in affected individuals. Sarcopenia is particularly common among PD patients, with severe cases affecting approximately one in five individuals with the disease. Furthermore, sarcopenia is closely linked to the accelerated progression of PD, diminished quality of life, greater susceptibility to falls and fractures, and increased mortality risk. Although the precise mechanisms remain unclear, numerous studies suggest that factors such as the accumulation of α-Synuclein in skeletal muscle, loss of motor neurons, inflammation, phosphate toxicity, hormonal dysregulation, vitamin D deficiency, intestinal flora imbalances, and dysfunction of the gut-muscle-brain axis contribute to sarcopenia in PD. Understanding these mechanisms provides valuable insights into the relationship between PD and sarcopenia and establishes a foundation for future research and therapeutic strategies. This review examines the mechanisms underlying sarcopenia in PD, methods for its screening and assessment, and potential avenues for future research, including strategies for risk reduction and treatment.
Collapse
Affiliation(s)
- Meilin Gui
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lingling Lv
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Shenglan Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lixia Qin
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China; Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Chunyu Wang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China; Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China; Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha 410011, China.
| |
Collapse
|
19
|
Zhao Y, Zhao W, Chai X, Sun P, Huang J, Guo X, Zhang L, Ren D, Yi C, Zhu X, Zhao S. Reshaping the gut microbiota: A novel oppinion of Eucommiae cortex polysaccharide alleviate learning and memory impairments in Alzheimer's disease. J Adv Res 2025:S2090-1232(25)00269-3. [PMID: 40252828 DOI: 10.1016/j.jare.2025.04.025] [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: 11/21/2024] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD), which is a chronic neurodegenerative disorder, is marked by the progressive deteriorations in learning and memory capabilities. The microbiota-gut-brain axis has come to be regarded as a crucial element in relation to the pathogenesis as well as the treatment of AD. Eucommiae cortex polysaccharides (EPs), being among the most plentiful substances present in the Eucommiae cortex, show the potential to exert immunomodulatory and neuroprotective function. However, whether EPs are protective against AD and their mechanism of action remain to be investigated OBJECTIVES: We hypothesize that EPs can regulate brain glutamine metabolism through gut microbiota and the butyric acid metabolized by them, improve oxidative stress and autophagy in the brain, and thus alleviate AD. METHODS In the present study, we used EPs (0.25 % w/w in food) and fecal microbiota transplantation, as well as butyrate supplementation (0.1 M in water), to intervene in AD mice. Multi-omics were used to determine the mechanism by which EPs improve AD-related learning and memory impairments. RESULTS Our results suggest that EPs, functioning as a prebiotic, alleviated learning and memory impairments in AD mice. Mechanistically, EPs are able to reshape the gut microbiota, promote the growth of gut microbiota involved in short-chain fatty acid metabolism, particularly butyrate-producing microbes. The butyrate produced by these microbes improves the brain microenvironment by modulating oxidative stress and autophagy mediated by brain glutamate metabolism, improving learning and memory impairments in AD mice, and inhibiting the formation and deposition of beta-amyloid proteins. Fecal microbiota transplantation (FMT) and butyrate supplementation further confirm this conclusion. CONCLUSIONS Our results highlighted that EPs can alleviate learning and memory impairments in AD with a gut microbiota-dependent manner and that butyric acid metabolized by butyric acid-metabolizing bacteria in the gut plays a central role in regulating brain glutamine metabolism to improve brain microenvironmental homeostasis. Meanwhile, the present study provides new insights into the treatment of AD with natural products.
Collapse
Affiliation(s)
- Yongkang Zhao
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China
| | - Wenxing Zhao
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China
| | - Xuejun Chai
- College of Basic Medicine, Xi'an Medical University, 710000 Xi'an, China.
| | - Penghao Sun
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China
| | - Junlang Huang
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China
| | - Xinrui Guo
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China
| | - Lulu Zhang
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China
| | - Duoduo Ren
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, 528000 Shenzhen, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China.
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, China.
| |
Collapse
|
20
|
Ben-Ari Y, Danchin ÉÉ. Limitations of genomics to predict and treat autism: a disorder born in the womb. J Med Genet 2025; 62:303-310. [PMID: 40081874 PMCID: PMC12015019 DOI: 10.1136/jmg-2024-110224] [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: 07/24/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Brain development involves the sequential expression of vulnerable biological processes including cell proliferation, programmed cell death, neuronal migration, synapse and functional unit formation. All these processes involve gene and activity-dependent events that can be distorted by many extrinsic and intrinsic environmental factors, including stress, microbiota, inflammatory signals, hormonal signals and epigenetic factors, hence leading to disorders born in the womb that are manifested later in autism spectrum disorders (ASDs) and other neurodevelopmental disorders. Predicting and treating such disorders call for a conceptual framework that includes all aspects of developmental biology. Here, taking the high incidence of ASDs as an example, we first discuss the intrinsic limitations of the genetic approach, notably the widely used twin studies and SNPs. We then review the long list of in utero events that can deviate developmental sequences, leading to persistent aberrant activity generated by immature misplaced and misconnected neuronal ensembles that are the direct cause of ASD. In a clinical perspective, we suggest analysing non-genetic maternity data to enable an early prediction of babies who will develop ASD years later, thereby facilitating early psycho-educative techniques. Subsequently, agents capable of selectively silencing malformed immature networks offer promising therapeutic perspectives. In summary, understanding developmental processes is critical to predicting, understanding and treating ASD, as well as most other disorders that arise in the womb.
Collapse
Affiliation(s)
| | - Étienne É Danchin
- Centre de biologie integrative, Centre de recherches sur la cognition animale, Toulouse University, Toulouse, France
| |
Collapse
|
21
|
Ramadan YN, Alqifari SF, Alshehri K, Alhowiti A, Mirghani H, Alrasheed T, Aljohani F, Alghamdi A, Hetta HF. Microbiome Gut-Brain-Axis: Impact on Brain Development and Mental Health. Mol Neurobiol 2025:10.1007/s12035-025-04846-0. [PMID: 40234288 DOI: 10.1007/s12035-025-04846-0] [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: 07/12/2024] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
The current discovery that the gut microbiome, which contains roughly 100 trillion microbes, affects health and disease has catalyzed a boom in multidisciplinary research efforts focused on understanding this relationship. Also, it is commonly demonstrated that the gut and the CNS are closely related in a bidirectional pathway. A balanced gut microbiome is essential for regular brain activities and emotional responses. On the other hand, the CNS regulates the majority of GI physiology. Any disruption in this bidirectional pathway led to a progression of health problems in both directions, neurological and gastrointestinal diseases. In this review, we hope to shed light on the complicated connections of the microbiome-gut-brain axis and the critical roles of gut microbiome in the early development of the brain in order to get a deeper knowledge of microbiome-mediated pathological conditions and management options through rebalancing of gut microbiome.
Collapse
Affiliation(s)
- Yasmin N Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt.
| | - Saleh F Alqifari
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Khaled Alshehri
- Department of Internal Medicine (Neurology), Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Amirah Alhowiti
- Department of Family and Community Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Hyder Mirghani
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Tariq Alrasheed
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Faisal Aljohani
- Division of Medicine and Gastroenterology, Department of Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdulaziz Alghamdi
- Department of Medicine, Division of Psychiatry, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Helal F Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, 71491, Tabuk, Saudi Arabia
| |
Collapse
|
22
|
Budin AJ, Brown WA, MacCormick AD, Caterson I, Sumithran P. Depressive symptoms at short-, medium-, and long-term follow-up after bariatric surgical procedures: A systematic review and meta-analysis. Obes Rev 2025:e13927. [PMID: 40222815 DOI: 10.1111/obr.13927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
IMPORTANCE Patients experience both positive and negative changes in mood following bariatric surgery and mental health outcomes have been reported to differ between procedure types. Understanding changes in symptoms over time and between surgical procedures is vital to providing meaningful, long-term, patient-centered care. OBJECTIVE To examine the nature and time course of changes in depressive symptoms after different bariatric procedures. EVIDENCE REVIEW Medline, Embase, Emcare, PsycINFO, CINAHL, and CENTRAL databases were systematically searched from inception to January 18, 2024. Ninety publications describing patient-reported depressive symptoms in 13,146 individuals undergoing bariatric procedures were included. FINDINGS Qualitative analysis indicated a reduction of depressive symptoms at all time points following all bariatric procedure types. However, a subset of patients experienced worsening symptoms post-surgery. Meta-analyses indicated depressive symptoms improve following bariatric surgery by an SMD of -0.6 (95% CI: -0.8, -0.4) in the short term (0-4 months post-surgery), -0.9 (95% CI: -1.0, -0.8) in the medium term (5-12 months), and -0.7 (95% CI: -0.9, -0.5) in the long term (> 12 months). There was no evidence that surgery type was associated with the change in depressive symptoms at any time point post-surgery. CONCLUSIONS AND RELEVANCE Patient-reported depressive symptoms improve following bariatric surgery with improvements peaking in the medium term and diminishing over time. Significant heterogeneity in the results cannot be explained by surgery type, baseline depression, or depression instrument used across studies. Long-term management of post-bariatric surgery patients must consider the potential for adverse psychological effects of surgery.
Collapse
Affiliation(s)
- Alyssa J Budin
- Department of Surgery, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| | - Wendy A Brown
- Department of Surgery, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
- Alfred Health, The Alfred Centre, Melbourne, Victoria, Australia
| | - Andrew D MacCormick
- Department of Surgery, The University of Auckland, Auckland, New Zealand
- Middlemore Hospital, Te Whatu Ora Counties Manukau Otahuhu, Auckland, New Zealand
| | - Ian Caterson
- The Boden Initiative, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Priya Sumithran
- Department of Surgery, School of Translational Medicine, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
- Department of Endocrinology and Diabetes, Alfred Health Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Crain E, Minaya DM, de La Serre CB. Microbiota-induced inflammation mediates the impacts of a Western diet on hippocampal-dependent memory. Nutr Res 2025; 138:89-106. [PMID: 40339190 DOI: 10.1016/j.nutres.2025.04.002] [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: 05/13/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 05/10/2025]
Abstract
Obesity is associated with impaired hippocampal-dependent memory, but the mechanisms driving this pathology are not fully understood. Western diets (WD) contribute to obesity, and previous reviews have described a role for WD in impaired hippocampal-dependent memory. However, there is need for a more detailed description of the pathways by which WD may impair memory. The short vs long-term effect of specific dietary components on brain structure and functions as well as the precise mechanism and molecular pathways involved are still not fully understood. This review focuses on the mechanisms and effects of gut microbiota-driven neuroinflammation. WD leads to changes and imbalance in bacterial taxa abundances that are deleterious to the host health (gut dysbiosis) and studies in rodent models show these changes are sufficient to impair hippocampal-dependent memory. Here, we discuss a variety of proposed mechanisms linking microbiota composition to hippocampal function, with a focus on neuroinflammation. Gut microbiota impacts gastrointestinal barrier function, leading to increased circulating proinflammatory bacterial products, increased blood-brain barrier permeability, and neuroinflammation.
Collapse
Affiliation(s)
- Eden Crain
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| | - Dulce M Minaya
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
24
|
Li N, Fang X, Li H, Liu J, Chen N, Zhao X, Yang Q, Chen X. Ginsenoside CK modulates glucose metabolism via PPARγ to ameliorate SCOP-induced cognitive dysfunction. Metab Brain Dis 2025; 40:168. [PMID: 40178645 DOI: 10.1007/s11011-025-01596-9] [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: 09/27/2024] [Accepted: 03/23/2025] [Indexed: 04/05/2025]
Abstract
Ginsenoside compound K (CK) exhibits neuroprotective properties; however, the underlying mechanisms behind these effects have not been investigated thoroughly. CK is the primary active compound derived from ginseng and is metabolized in the gut. It enhances neuronal function by modulating the gut microflora. Therefore, the present study aimed to elucidate the mechanism through which CK enhances cognitive function, employing gut microbiome and microarray analyses. The results revealed that CK upregulated the expression of peroxisome proliferator-activated receptor gamma (PPARγ), suppressed amyloid-β (Aβ) aggregation in hippocampal neurons, and influenced the expression of cyclin-dependent kinase-5 (CDK5), (including insulin receptor substrate 2) IRS2, insulin-degrading enzyme (IDE), glycogen synthase kinase-3 beta (GSK-3β), glucose transporter type 1 (GLUT1), and glucose transporter type 3 (GLUT3) proteins. These proteins play crucial roles in regulating brain glucose metabolism, increasing neuronal energy, and reducing neuronal apoptosis, thereby ameliorating cognitive impairment in mice.
Collapse
Affiliation(s)
- Na Li
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Xingyu Fang
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
| | - Hui Li
- Qian Wei Hospital of Jilin Province, Changchun, 130117, Jilin, P.R. China
| | - Jian Liu
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
| | - Nan Chen
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
| | - Xiaohui Zhao
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China
| | - Qing Yang
- Jinlin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, P.R. China.
| | - Xijun Chen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P.R. China.
| |
Collapse
|
25
|
Lam LY, Liang TR, Wu WJ, Lam HYP. Intestinal Lactobacillus johnsonii protects against neuroangiostrongyliasis in BALB/c mice through modulation of immune response. PLoS Negl Trop Dis 2025; 19:e0012977. [PMID: 40198714 PMCID: PMC11978024 DOI: 10.1371/journal.pntd.0012977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025] Open
Abstract
Neuroangiostrongyliasis is characterized by eosinophilic meningoencephalitis with a robust onset of severe neurological symptoms, by which immunological factors and peripheral metabolites have been postulated to affect the course of the disease. The gut-brain axis provides a bidirectional communication between the gut and the central nervous system, and therefore, understanding the gut microbiome may provide us with a deeper insight into the pathogenesis of angiostrongyliasis. Using 16S rRNA sequencing, we identified an increase in the abundance of different Lactobacillus species in Angiostrongylus cantonensis-infected mice, which was correlated to the disease severity. However, attempts to inoculate L. johnsonii into A. cantonensis-infected mice surprisingly revealed an improvement in neuroinflammation and prolonged survival. RNA sequencing suggested an immune-modulatory effect of L. johnsonii, which was confirmed by ELISA, showing increased levels of IL-10 and reduced levels of IL-2, IL-4, IL-5, and MCP-1 in the brain. Nevertheless, L. johnsonii-associated improvements were not associated with microbiome-related metabolites, as UHPLC-MS/MS analysis revealed no change in short-chain fatty acids, tryptophan metabolites, and bile acids. Our results suggest that while intestinal L. johnsonii appears to be linked to the progression of neuroangiostrongyliasis, these bacteria are likely attempting to modulate the dysregulated immune response to combat the disease. This is one of the first studies to investigate the gut microbiome in mice with A. cantonensis infection, which extends our knowledge from the microbiome-point-of-view of the pathogenesis of angiostrongyliasis and how the body defends against A. cantonensis. This work also extends to possible treatment approaches using L. johnsonii as probiotics.
Collapse
Affiliation(s)
- Long Yin Lam
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Ting-Ruei Liang
- PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien, Taiwan
| | - Wen-Jui Wu
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
| | - Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
26
|
Anka IZ, Uren Webster T, McLaughlin S, Overland B, Hitchings M, Garcia de Leaniz C, Consuegra S. Gut microbiota diversity affects fish behaviour and is influenced by host genetics and early rearing conditions. Open Biol 2025; 15:240232. [PMID: 40237041 PMCID: PMC12001083 DOI: 10.1098/rsob.240232] [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: 08/14/2024] [Revised: 11/12/2024] [Accepted: 03/06/2025] [Indexed: 04/17/2025] Open
Abstract
The gut microbiota influences human and animal cognition and behaviour through its effects on the endocrine and immune systems. The microbiome-behaviour relationship may be especially relevant for fish, due to their diverse evolutionary history and potential implications for farming and conservation. Yet, there is limited research on the interaction between gut microbiome and behaviour in non-model fish. We manipulated the rearing environment and diet of fish from two inbred strains of the self-fertilizing mangrove killifish (Kryptolebias marmoratus) and assessed the effects on the gut microbiome and its interactions with anxiety-like behaviours. We found that microbiota composition and alpha diversity were significantly influenced by host genetics (strain), hatching mode (naturally or artificial dechorionation) and diet, but not by environmental enrichment. Fish activity level and inspections of a novel object were strongly associated with microbiota community composition and alpha diversity. The microbial taxa associated with differences in behaviour were dominated by Bacteroidales, potentially related to the production of metabolites affecting neural development. We suggest that the association between microbiome and fish behaviour could be an indirect effect of the modulation of the gut microbiota by host genetics and early rearing conditions, which could be affecting the production of microbial metabolites that interact with the fish physiology.
Collapse
Affiliation(s)
- Ishrat Z. Anka
- Department of Biosciences, Swansea University, Swansea, UK
- Department of Aquaculture, Chattogram Veterinary and Animal Sciences University, Chittagong, Bangladesh
| | | | - Sam McLaughlin
- Department of Biosciences, Swansea University, Swansea, UK
| | | | | | - Carlos Garcia de Leaniz
- Department of Biosciences, Swansea University, Swansea, UK
- Centro de Investigaciones Marinas, Universidade de Vigo, Vigo, Spain
| | - Sofia Consuegra
- Department of Biosciences, Swansea University, Swansea, UK
- Departamento de Biotecnologia y Acuicultura, Instituto de Investigaciones Marinas (IIM-CSIC), Vigo, Spain
| |
Collapse
|
27
|
Jeong S, Davis CK, Chokkalla AK, Kim B, Park S, Vemuganti R. Fecal microbiota transplantation fails to impart the benefits of circadian-dependent intermittent fasting following ischemic stroke. J Cereb Blood Flow Metab 2025; 45:779-789. [PMID: 39917846 PMCID: PMC11806450 DOI: 10.1177/0271678x251319636] [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: 06/04/2024] [Revised: 10/28/2024] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
Intermittent fasting (IF) is known to induce significant ischemic tolerance. Diet is a major proponent of gut microbiota, and gut microbial dysbiosis plays a role in post-stroke brain damage. Hence, we currently evaluated whether IF-mediated ischemic tolerance is mediated by gut microbiota. Additionally, circadian cycle is known to modulate post-ischemic outcomes, and thus we further evaluated if gut microbiota would be influenced by prophylactic IF during the inactive phase (fasting during daytime; IIF) or active phase (fasting during nighttime; AIF). The AIF, but not IIF, cohort showed a significantly decreased fecal Firmicutes/Bacteroidetes ratio compared with the ad libitum (AL) cohort. Moreover, the levels of gut microbiota-derived metabolites butyrate and propionate decreased in AL cohort following focal ischemia, whereas they increased in AIF cohort. However, fecal microbiota transplantation (FMT) from IIF or AIF cohort had no significant effects on post-ischemic motor and cognitive function recovery, anxiety-, and depression-like behaviors compared with FMT from AL cohort. Furthermore, FMT from IIF or AIF cohort did not influence the post-ischemic infarct volume, atrophy volume or white matter damage. Overall, the current findings indicate that the beneficial effects of IF after focal ischemia are not mediated by the gut microbiota.
Collapse
Affiliation(s)
- Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Bori Kim
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Sena Park
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
- William S. Middleton Veterans Hospital, Madison, WI, USA
| |
Collapse
|
28
|
Gupta M, Cilkiz M, Ibrahim MMA, Athrey G. Gut Microbiome-Brain Crosstalk in the Early Life of Chicken Reveals the Circadian Regulation of Key Metabolic and Immune Signaling Processes. Microorganisms 2025; 13:789. [PMID: 40284627 PMCID: PMC12029235 DOI: 10.3390/microorganisms13040789] [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: 02/17/2025] [Revised: 03/13/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Circadian rhythms are innate biological systems that control everyday behavior and physiology. Furthermore, bilateral interaction between the host's circadian rhythm and the gut microbes influences a variety of health ramifications, including metabolic diseases, obesity, and mental health including GALT physiology and the microbiome population. Therefore, we are studying the correlation between differential gene expression in the chicken brain and microbiota abundance during circadian rhythms. To understand this, we raised freshly hatched chicks under two photoperiod treatments: normal photoperiod (NP = 12/12 LD) and extended photoperiod (EP 23/1 LD). The chicks were randomly assigned to one of two treatments. After 21 days of circadian entrainment, the chicks were euthanized at nine time points spaced six hours apart over 48 h to characterize the brain transcriptomes. Each sample's RNA was extracted, and 36 mRNA libraries were generated and sequenced using Illumina technology, followed by data processing, count data generation, and differential gene expression analysis. We generated an average of 17.5 million reads per library for 237.9 M reads. When aligned to the Galgal6 reference genome, 11,867 genes had detectable expression levels, with a common dispersion value of 0.105. To identify the genes that follow 24 h rhythms, counts per million data were performed in DiscoRhythm. We discovered 577 genes with Cosinor and 417 with the JTK cycle algorithm that exhibit substantial rhythms. We used weighted gene co-expression network analysis (WGCNA) to analyze the correlation between differentially expressed genes and microbiota abundance. The most enriched pathways included aldosterone-regulated sodium reabsorption, endocrine and other factor-regulated calcium reabsorption, GABAergic synapse, oxidative phosphorylation, serotonergic synapse, dopaminergic synapse and circadian entrainment. This study builds on our previous study, and adds new findings about the specific interactions and co-regulation of the brain transcriptome and the gut microbiota. The interaction between gut microbiota and host gene expression highlights the potential benefits of microbiome-modulation approaches to improve gut health and performance in poultry.
Collapse
Affiliation(s)
- Mridula Gupta
- Department of Poultry Science, Texas A&M University, 2472 TAMU, College Station, TX 77843, USA;
| | - Mustafa Cilkiz
- Soil and Crop Sciences, Texas A&M University, College Station, TX 77843, USA;
| | - Mohamed M. A. Ibrahim
- Department of Laser Applications in Metrology, Photochemistry and Agriculture, National Institute of Laser Enhanced Sciences, Cairo University, Giza 12613, Egypt;
| | - Giridhar Athrey
- Department of Poultry Science, Texas A&M University, 2472 TAMU, College Station, TX 77843, USA;
- Faculty of Ecology & Evolutionary Biology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
29
|
Schwerdtfeger LA, Lanser TB, Montini F, Moreira T, LeServe DS, Cox LM, Weiner HL. Akkermansia mono-colonization modulates microglia and astrocytes in a strain specific manner. J Neuroinflammation 2025; 22:94. [PMID: 40148962 PMCID: PMC11951737 DOI: 10.1186/s12974-025-03417-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Microglia and astrocytes are the primary glial cells in the central nervous system (CNS) and their function is shaped by multiple factors. Regulation of CNS glia by the microbiota have been reported, although the role of specific bacteria has not been identified. We colonized germ-free mice with the type strain Akkermansia muciniphila (AmT) and a novel A. muciniphila strain BWH-H3 (Am-H3) isolated from a subject with multiple sclerosis and compared to mice colonized with Bacteroides cellulosilyticus strain BWH-E5 (Bc) isolated from a healthy control subject. We then investigated the effect of these bacteria on microglia and astrocyte gene expression by RNA sequencing. We found altered gene expression profiles in brain microglia, with Akkermansia downregulating genes related to antigen presentation and cell migration. Furthermore, we observed strain specific effects, with Akkermansia H3 upregulating histone and protein binding associated genes and downregulating channel and ion transport genes. Astrocyte pathways that were altered by Akkermansia H3 mono-colonization included upregulation of proliferation pathways and downregulation in cytoskeletal associated genes. Furthermore, animals colonized with type strain Akkermansia and strain H3 had effects on the immune system including elevated splenic γδ-T cells and increased IFNγ production in CD4 + T cells. We also measured intestinal short chain fatty acids and found that both A. muciniphila strains produced proprionate while B. cellulosilyticus produced acetate, proprionate, and isovalerate. Taken together, our study shows that specific members of the intestinal microbiota influence both microglial and astroyctes which may be mediated by changes in short chain fatty acids and peripheral immune signaling.
Collapse
Affiliation(s)
- Luke A Schwerdtfeger
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Toby B Lanser
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Federico Montini
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Thais Moreira
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Danielle S LeServe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Laura M Cox
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA.
| |
Collapse
|
30
|
Lewis N, Villani A, Lagopoulos J. Gut dysbiosis as a driver of neuroinflammation in attention-deficit/hyperactivity disorder: A review of current evidence. Neuroscience 2025; 569:298-321. [PMID: 39848564 DOI: 10.1016/j.neuroscience.2025.01.031] [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: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
There is mounting evidence for the involvement of the immune system, neuroinflammation and disturbed gut microbiota, or dysbiosis, in attention-deficit/hyperactivity disorder (ADHD). Gut dysbiosis is strongly implicated in many physical, autoimmune, neurological, and neuropsychiatric conditions, however knowledge of its particular pathogenic role in ADHD is sparse. As such, this narrative review examines and synthesizes the available evidence related to inflammation, dysbiosis, and neural processes in ADHD. Minimal differences in microbiota diversity measures between cases and controls were found, however many relative abundance differences were observed at all classification levels (phylum to strain). Compositional differences of taxa important to key gut-brain axis pathways, in particular Bacteroides species and Faecalibacterium, may contribute to inflammation, brain functioning differences, and symptoms, in ADHD. We have identified one possible model of ADHD etiopathogenesis involving systemic inflammation, an impaired blood-brain barrier, and neural disturbances as downstream consequences of gut dysbiosis. Nevertheless, studies conducted to date have varied degrees of methodological rigour and involve diverse participant characteristics and analytical techniques, highlighting a need for additional research.
Collapse
Affiliation(s)
- Naomi Lewis
- School of Health, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs, QLD 4556, Australia; Thompson Institute, University of the Sunshine Coast, 12 Innovation Pkwy, Birtinya, QLD 4575, Australia.
| | - Anthony Villani
- School of Health, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs, QLD 4556, Australia.
| | - Jim Lagopoulos
- Thompson Brain and Mind Healthcare, Eccles Blvd, Birtinya, QLD 4575, Australia.
| |
Collapse
|
31
|
Menozzi E, Schapira AHV, Borghammer P. The Gut-Brain Axis in Parkinson disease: Emerging Concepts and Therapeutic Implications. Mov Disord Clin Pract 2025. [PMID: 40079755 DOI: 10.1002/mdc3.70029] [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: 09/10/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The gut-brain axis, i.e. the bidirectional communication system between the gut and the brain, has become of central importance in Parkinson disease (PD) research over the past 20 years. AIMS We aimed to describe the milestones of the gut-brain axis research in PD and the development of theories proposing the involvement of the gastrointestinal tract in PD pathogenesis. METHODS We searched PubMed using the terms 'gut-brain axis' AND 'Parkinson disease', and selected relevant articles to provide the foundation for reconstructing an historical overview of the gut-brain axis research in PD. RESULTS Mounting evidence from preclinical, clinical and post-mortem studies suggests that a subgroup of PD patients present with a range of prodromal symptoms (e.g., autonomic dysfunction, rapid eye movement sleep behaviour disorder) which reflect initial accumulation and later spread of pathological α-synuclein rostrally from the gastrointestinal tract ("body-first" PD). Through neural connections along the gut-brain axis, pathological α-synuclein may spread to the brain, producing clinically manifest disease. Recently, two mechanisms involving the gut-brain axis have attracted increasing attention for their role in PD pathogenesis and progression, namely the perturbation of the composition of the microorganisms living in the gut (the gut microbiome), and the dysfunction of enteroendocrine cells. CONCLUSION Treatments targeting the gut-brain axis, especially the gut microbiome and the enteroendocrine cells pathway, could potentially slow disease progression or even prevent disease onset. Among these, pre/probiotics, faecal microbiota transplantation, and glucagon-like peptide-1 receptor agonists, have entered advanced stages of clinical trials in humans and shown potential symptomatic and disease-modifying effects.
Collapse
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Per Borghammer
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
32
|
Gu T, Guo R, Chen L, Zong Y, Tian Y, Xu W, Zeng T, Lu L. Multi-omics uncover acute stress vulnerability through gut-hypothalamic communication in ducks. Br Poult Sci 2025:1-10. [PMID: 40072466 DOI: 10.1080/00071668.2025.2454960] [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: 10/29/2024] [Accepted: 01/01/2025] [Indexed: 03/14/2025]
Abstract
1. The avian gut hosts a complex and dynamic microbial ecosystem, which is essential for regulating host organ function. However, the relationship between the gut microbiota and the hypothalamic axis in acute stress vulnerability in ducks remains unclear.2. This study investigated how the gut microbiota affects microbial metabolism and the host stress response by comparing hypothalamic neurotransmitter availability, microbial composition and co-metabolites generated by both the microbiota and hypothalamus in ducks exhibiting the lowest active avoidance (LAA) and highest active avoidance (HAA) behaviour.3. The HAA group experienced a significant increase in the availability of arginine, histidine, glutamine, norepinephrine, L-tyrosine and melatonin during acute stress in the hypothalamus, compared to that in the LAA group. The 16S rRNA sequencing revealed significant differences in the gut microbiota composition based on acute stress vulnerabilities.4. Both caecal and hypothalamic metabolomic analyses identified 71 metabolites altered in caecal content and 95 in the hypothalamus. There was significant enrichment in pathways such as the cGMP-PKG signalling, dopaminergic synapse and endocrine resistance.5. Correlation analyses demonstrated that certain co-metabolites, including 1,3-dicyclohexylurea, 1-deoxyvaleric acid, 2-amino-2-methyl-1,3-propanediol, 3-chloroaniline, methenamine, N4-acetylcytidine-triphosphate and traumatin, may play a role in the gut microbiota-hypothalamic axis.6. The results suggested that the gut microbiome influenced acute stress responses. This provided a basis for understanding gut-hypothalamic communication and its impact on behaviour in ducks.
Collapse
Affiliation(s)
- T Gu
- State Key Laboratory for Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - R Guo
- State Key Laboratory for Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - L Chen
- State Key Laboratory for Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Y Zong
- State Key Laboratory for Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Y Tian
- State Key Laboratory for Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - W Xu
- State Key Laboratory for Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - T Zeng
- State Key Laboratory for Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - L Lu
- State Key Laboratory for Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs of China, Hangzhou, China
| |
Collapse
|
33
|
Xialu S, Faqiang M. Mechanisms of action of intestinal microorganisms and advances in head and neck tumors. Discov Oncol 2025; 16:303. [PMID: 40072772 PMCID: PMC11903988 DOI: 10.1007/s12672-025-02035-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
In the last decade, it has been discovered that intestinal flora can affect various organ-specific cancers by altering the body's energy balance, synthesizing genetic toxins and small signaling molecules, and initiating and modulating immune responses. In this review, we will focus on elucidating the role of intestinal flora based on its molecular mechanisms and its possible impact on head and neck cancers in the near future, and explore how it may be a novel approach to treating head and neck cancers in the future.
Collapse
Affiliation(s)
- Su Xialu
- Graduate School of Guizhou Medical University, Guiyang, 550000, China
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, 556000, China
| | - Ma Faqiang
- Graduate School of Guizhou Medical University, Guiyang, 550000, China.
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, 556000, China.
| |
Collapse
|
34
|
Ghimire S, Lehman PC, Aguilar Meza LS, Shahi SK, Hoang J, Olalde H, Paullus M, Cherwin C, Wang K, Gill C, Cho T, Mangalam AK. Specific microbial ratio in the gut microbiome is associated with multiple sclerosis. Proc Natl Acad Sci U S A 2025; 122:e2413953122. [PMID: 40030030 PMCID: PMC11912405 DOI: 10.1073/pnas.2413953122] [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: 07/11/2024] [Accepted: 01/02/2025] [Indexed: 03/19/2025] Open
Abstract
Gut microbiota dysbiosis is associated with multiple sclerosis (MS), but the causal relationship between specific gut bacteria and MS pathogenesis remains poorly understood. Therefore, we profiled the stool microbiome of people with MS (PwMS) and healthy controls (HC) using shotgun metagenomic sequencing. PwMS showed a distinct microbiome compared to HC, with Prevotella copri (PC) and Blautia species as drivers of microbial communities in HC and PwMS, respectively. Administration of MS-driving Blautia species (Blautia wexlerae; BW) to mice resulted in increased levels of gut inflammatory markers and altered microbiota with increased capacity to induce proinflammatory cytokines. Utilizing experimental autoimmune encephalomyelitis (EAE), an animal model of MS, we identified a lower gut Bifidobacterium to Akkermansia ratio as a hallmark of the disease. BW-administered mice also showed a lower Bifidobacterium to Akkermansia ratio pre-EAE induction which correlated with increased disease severity post-EAE induction. The importance of the Bifidobacterium to Akkermansia ratio at the species level, lower Bifidobacterium adolescentis to Akkermansia muciniphila (BA:AM), was validated in our MS cohort and a large International Multiple Sclerosis Microbiome Study. Thus, our findings highlight the BA:AM ratio as a potential gut microbial marker in PwMS, opening avenues for microbiome-based diagnosis, prognosis, and therapy in MS.
Collapse
Affiliation(s)
- Sudeep Ghimire
- Department of Pathology, University of Iowa, Iowa City, IA52242
- Clinician Scientist, Iowa City Veterans Affairs Health Care System, Iowa City, IA52242
| | - Peter C. Lehman
- Department of Pathology, University of Iowa, Iowa City, IA52242
- Graduate Program in Experimental Pathology, University of Iowa, Iowa City, IA52242
| | | | - Shailesh K. Shahi
- Department of Pathology, University of Iowa, Iowa City, IA52242
- Clinician Scientist, Iowa City Veterans Affairs Health Care System, Iowa City, IA52242
| | - Jemmie Hoang
- College of Nursing, University of Iowa, Iowa City, IA52242
| | - Heena Olalde
- Department of Neurology, University of Iowa Hospital and Clinics, Iowa City, IA52242
| | - Mishelle Paullus
- Department of Neurology, University of Iowa Hospital and Clinics, Iowa City, IA52242
| | | | - Kai Wang
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA52242
| | - Christine Gill
- Department of Neurology, University of Iowa Hospital and Clinics, Iowa City, IA52242
| | - Tracey Cho
- Department of Neurology, University of Iowa Hospital and Clinics, Iowa City, IA52242
| | - Ashutosh K. Mangalam
- Department of Pathology, University of Iowa, Iowa City, IA52242
- Clinician Scientist, Iowa City Veterans Affairs Health Care System, Iowa City, IA52242
- Graduate Program in Experimental Pathology, University of Iowa, Iowa City, IA52242
- Microbiome Core, University of Iowa, Iowa City, IA52242
| |
Collapse
|
35
|
Philip V, Kraimi N, Zhang H, Lu J, Palma GD, Shimbori C, McCoy KD, Hapfelmeier S, Schären OP, Macpherson AJ, Chirdo F, Surette MG, Verdu EF, Liu F, Collins SM, Bercik P. Innate immune system signaling and intestinal dendritic cells migration to the brain underlie behavioral changes after microbial colonization in adult mice. Brain Behav Immun 2025; 127:238-250. [PMID: 40068794 DOI: 10.1016/j.bbi.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/19/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND AIMS Accumulating evidence suggests the microbiota is a key factor in Disorders of Gut-Brain Interaction (DGBI), by affecting host immune and neural systems. However, the underlying mechanisms remain elusive due to their complexity and clinical heterogeneity of patients with DGBIs. We aimed to identify neuroimmune pathways that are critical in microbiota-gut-brain communication during de novo gut colonization. METHODS We employed a combination of gnotobiotic and state-of-the-art microbial tools, behavioral analysis, immune and pharmacological approaches. Germ-free wild type, TLR signaling-deficient MyD88-/- Ticam1-/- and lymphocyte-deficient SCID mice were studied before and after colonization with specific pathogen-free microbiota, Altered Schaedler Flora, E. coli or S. typhimurium (permanent or transient colonizers). TLR agonists and antagonists, CCR7 antagonist or immunomodulators were used to study immune pathways. We assessed brain c-Fos, brain-derived neurotrophic factor, and dendritic and glial cells by immunofluorescence, expression of neuroimmune genes by NanoString and performed brain proteomics. RESULTS Bacterial monocolonization, conventionalization or administration of microbial products to germ-free mice altered mouse behavior similarly, acting through Toll-like receptor or nucleotide-binding oligomerization domain signaling. The process required CD11b+CD11c+CD103+ dendritic cell activation and migration into the brain. The change in behavior did not require the continued presence of bacteria and was associated with activation of multiple neuro-immune networks in the gut and the brain. CONCLUSIONS Changes in neural plasticity occur rapidly upon initial gut microbial colonization and involve innate immune signaling to the brain, mediated by CD11b+CD11c+CD103+ dendritic cell migration. The results identify a new target with therapeutic potential for DGBIs developing in context of increased gut and blood-brain barrier permeability.
Collapse
Affiliation(s)
- Vivek Philip
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada; Campbell Family Mental Health Research Institute, the Centre for Addiction and Mental Health, Toronto, Canada
| | - Narjis Kraimi
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Hailong Zhang
- Campbell Family Mental Health Research Institute, the Centre for Addiction and Mental Health, Toronto, Canada
| | - Jun Lu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Giada De Palma
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Chiko Shimbori
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Kathy D McCoy
- Department of Biomedical Research, University Hospital, Bern, Switzerland; Dept. of Physiology and Pharmacology, Snyder Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Olivier P Schären
- University of Bern, Institute for Infectious Diseases, Bern, Switzerland
| | | | - Fernando Chirdo
- Instituto de Estudios Inmunologicos y Fisiopatologicos - IIFP (UNLP-CONICET), La Plata, Argentina
| | - Michael G Surette
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, the Centre for Addiction and Mental Health, Toronto, Canada
| | - Stephen M Collins
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada.
| |
Collapse
|
36
|
Nakhal MM, Mydeen AB, Yassin LK, Almazrouei R, Alkamali R, Alsulaimi M, Elsaleh RI, BaniYas S, Al Houqani S, Al-Marzooq F, Hassane M, Voitetskii R, Statsenko Y, Allam M, Akour A, Hamad MIK. Antibiotics-induced dysbiosis impacts dendritic morphology of adult mouse cortical interneurons. Front Neuroanat 2025; 19:1557961. [PMID: 40124111 PMCID: PMC11925899 DOI: 10.3389/fnana.2025.1557961] [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: 01/09/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction A growing body of evidence suggests that the gut microbiome may contribute to changes in brain morphology. The microbiota-gut-brain axis (MGBA) has been shown to influence neurogenesis, axon myelination, and synapse structure. However, it remains unclear whether the MGBA can influence the morphology and density of inhibitory GABAergic interneurons. The aim of this study was to determine whether antibiotic-induced dysbiosis (AID) is associated with alterations in dendritic morphology of GABAergic inhibitory interneurons in the medial entorhinal cortex (mEC), somatosensory cortex (SSC), motor cortex (MC), and hippocampus (Hp). Methods A cohort of six-month-old GAD-67-EGFP transgenic mice was treated with an antibiotic cocktail for two weeks, resulting in gut dysbiosis as validated by collecting stool samples at baseline and after treatment, then using next-generation sequencing of 16S ribosomal RNA. Results The results demonstrate that the proposed model effectively exhibited the defining features of gut dysbiosis, including a significant reduction in microbiome diversity, expansion of pathobionts, and loss of beneficial microbes. The AID group showed alterations in density and morphology of GABAergic interneurons in different brain areas. The mean dendritic length and mean dendritic segments of the SSC and Hp were found to be significantly decreased, while no such decrease was observed in the mEC or MC. Furthermore, the density of interneurons was decreased in the mEC, Hp, and SSC areas, while no change was observed in the MC area. Discussion The interneuron dysfunction plays a role in the pathogenesis of neurological disease. The findings of this study suggest that AID potentially influences the density and morphology of the interneurons, which may contribute to the development of neurological disorders.
Collapse
Affiliation(s)
- Mohammed M. Nakhal
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ayishal B. Mydeen
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Lydia K. Yassin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Reem Almazrouei
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rasha Alkamali
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mahra Alsulaimi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rawan I. Elsaleh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Shamsa BaniYas
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Shaikha Al Houqani
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Farah Al-Marzooq
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Maya Hassane
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Roman Voitetskii
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Yauhen Statsenko
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mushal Allam
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
37
|
Xu M, Zhou EY, Shi H. Tryptophan and Its Metabolite Serotonin Impact Metabolic and Mental Disorders via the Brain-Gut-Microbiome Axis: A Focus on Sex Differences. Cells 2025; 14:384. [PMID: 40072112 PMCID: PMC11899299 DOI: 10.3390/cells14050384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
The crisis of metabolic and mental disorders continues to escalate worldwide. A growing body of research highlights the influence of tryptophan and its metabolites, such as serotonin, beyond their traditional roles in neural signaling. Serotonin acts as a key neurotransmitter within the brain-gut-microbiome axis, a critical bidirectional communication network affecting both metabolism and behavior. Emerging evidence suggests that the gut microbiome regulates brain function and behavior, particularly through microbial influences on tryptophan metabolism and the serotonergic system, both of which are essential for normal functioning. Additionally, sex differences exist in multiple aspects of serotonin-mediated modulation within the brain-gut-microbiome axis, affecting feeding and affective behaviors. This review summarizes the current knowledge from human and animal studies on the influence of tryptophan and its metabolite serotonin on metabolic and behavioral regulation involving the brain and gut microbiome, with a focus on sex differences and the role of sex hormones. We speculate that gut-derived tryptophan and serotonin play essential roles in the pathophysiology that modifies neural circuits, potentially contributing to eating and affective disorders. We propose the gut microbiome as an appealing therapeutic target for metabolic and affective disorders, emphasizing the importance of understanding sex differences in metabolic and behavioral regulation influenced by the brain-gut-microbiome axis. The therapeutic targeting of the gut microbiota and its metabolites may offer a viable strategy for treating serotonin-related disorders, such as eating and affective disorders, with potential differences in treatment efficacy between men and women. This review would promote research on sex differences in metabolic and behavioral regulation impacted by the brain-gut-microbiome axis.
Collapse
Affiliation(s)
- Mengyang Xu
- Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, USA
| | - Ethan Y. Zhou
- Institute for the Environment and Sustainability, Miami University, Oxford, OH 45056, USA
| | - Haifei Shi
- Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, USA
| |
Collapse
|
38
|
Mustafa N, Afroz R, Batool Z, Salman T, Nawaz S, Haleem DJ. Exploring Serotonin-1A receptor function in the effects of buspirone on cognition by molecular receptor expression and EEG analytical studies. Eur J Pharmacol 2025; 990:177275. [PMID: 39855288 DOI: 10.1016/j.ejphar.2025.177275] [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: 08/28/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Buspirone, a commonly prescribed medication for generalized anxiety disorder (GAD), is gaining attention for its narrow window of side effects such as lack of physical dependence, non-sedative properties as compared to other anxiolytic drugs. Its dose-specific therapeutic effects beyond anxiety highlights its clinical significance. Pharmacologically, buspirone activates serotonin-1A pre-synaptic autoreceptors and post-synaptic heteroreceptors which modulate serotonergic neurotransmission induced behavioral changes such as anxiolytic and nootropic effects. This study explored change in neural activity associated serotonin-1A receptors, induced by repeated administration of buspirone at specific doses (0.1 mg/kg and 3 mg/kg). Buspirone induced behavioral changes were assessed by Morris Water Maze (MWM) for cognitive functions, Elevated Plus Maze (EPM) for anxiety, RT-PCR (Reverse transcriptase-polymerase chain reaction) for 5-HT1A receptor expression levels, and EEG (electroencephalography) analysis of neuronal electrical activity in the frontal cortex. Our findings revealed that a low dose of buspirone (0.1 mg/kg) significantly enhanced spatial learning and memory compared to high dose (3 mg/kg). Low-dose treatment elevated mRNA expression levels of serotonin-1A receptors in hippocampus and decreased in midbrain raphe nuclei, with the opposite patterns observed in the high dose. In addition, EEG spectral analysis have revealed dose specific cross coupling frequency of theta-gamma and delta-beta brain waves. At low dose (0.1 mg/kg) positive correlation of theta-gamma coupling effect and negative correlation of delta beta as decoupling effect were observed. Conversely, at high dose (3 mg/kg), results showed opposite pattern with weak correlation of theta gamma coupling effect and positive correlation of delta-beta as coupling effect. These results suggest that buspirone enhances learning and memory with differential activation of pre and postsynaptic serotonin-1A receptors, altering its expression levels which influence neural activity associated with theta-gamma and delta-beta coupling effects. It provides valuable molecular insights on clinical significance of buspirone in mitigating neuropathological disorders such as behavioral disorders and neurocognitive decline associated with disrupted regulation of serotonin-1A neurotransmission at specific doses. Our findings provide molecular insights of dose dependent therapeutic potential of buspirone against neuropathological symptoms of behavioral disorders, neurocognitive decline associated with dysregulated serotonin-1A neurotransmission.
Collapse
Affiliation(s)
- Nazish Mustafa
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Rushda Afroz
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Tabinda Salman
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Shazia Nawaz
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Darakhshan Jabeen Haleem
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| |
Collapse
|
39
|
Lee JW, Lee KA, Jang IH, Nam K, Kim SH, Kyung M, Cho KC, Lee JH, You H, Kim EK, Koh YH, Lee H, Park J, Hwang SY, Chung YW, Ryu CM, Kwon Y, Roh SH, Ryu JH, Lee WJ. Microbiome-emitted scents activate olfactory neuron-independent airway-gut-brain axis to promote host growth in Drosophila. Nat Commun 2025; 16:2199. [PMID: 40038269 PMCID: PMC11880416 DOI: 10.1038/s41467-025-57484-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 02/13/2025] [Indexed: 03/06/2025] Open
Abstract
While it is now accepted that the microbiome has strong impacts on animal growth promotion, the exact mechanism has remained elusive. Here we show that microbiome-emitted scents contain volatile somatotrophic factors (VSFs), which promote host growth in an olfaction-independent manner in Drosophila. We found that inhaled VSFs are readily sensed by olfactory receptor 42b non-neuronally expressed in subsets of tracheal airway cells, enteroendocrine cells, and enterocytes. Olfaction-independent sensing of VSFs activates the airway-gut-brain axis by regulating Hippo, FGF and insulin-like growth factor signaling pathways, which are required for airway branching, organ oxygenation and body growth. We found that a mutant microbiome that did not produce (2R,3R)-2,3-butanediol failed to activate the airway-gut-brain axis for host growth. Importantly, forced inhalation of (2R,3R)-2,3-butanediol completely reversed these defects. Our discovery of contact-independent and olfaction-independent airborne interactions between host and microbiome provides a novel perspective on the role of the airway-gut-brain axis in microbiome-controlled host development.
Collapse
Affiliation(s)
- Jin-Woo Lee
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Kyung-Ah Lee
- School of Biological Sciences, Seoul National University, Seoul, South Korea
- Saeloun Bio Inc., Seoul, South Korea
| | - In-Hwan Jang
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Kibum Nam
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sung-Hee Kim
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Minsoo Kyung
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Kyu-Chan Cho
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Ji-Hoon Lee
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Hyejin You
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Eun-Kyoung Kim
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Young Hoon Koh
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Hansol Lee
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Junsun Park
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Soo-Yeon Hwang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Youn Wook Chung
- Severance Biomedical Science Institute, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Choong-Min Ryu
- Molecular Phytobacteriology Laboratory, Infectious Disease Research Center, KRIBB, Daejeon, South Korea
| | - Youngjoo Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Soung-Hun Roh
- School of Biological Sciences, Seoul National University, Seoul, South Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Ji-Hwan Ryu
- Severance Biomedical Science Institute, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Won-Jae Lee
- School of Biological Sciences, Seoul National University, Seoul, South Korea.
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.
- Wide River Institute of Immunology, Seoul National University, Hongcheon, South Korea.
| |
Collapse
|
40
|
Huwart SJP, Morales-Puerto N, Everard A. Gut microbiota-related neuroinflammation at the crossroad of food reward alterations: implications for eating disorders. Gut 2025:gutjnl-2024-333397. [PMID: 39961644 DOI: 10.1136/gutjnl-2024-333397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
The link between gut microbiome and eating behaviours, especially palatable food intake, is a growing focus of scientific investigation. The complex ecosystem of microorganisms in the gut influences host metabolism, immune function and neurobehavioural signalling. This review explores the role of neuroinflammation in dysregulations of food-induced reward signalling and the potential causal role of the gut microbiota on these proinflammatory processes. Particular attention is given to eating disorders (ED, specifically anorexia nervosa, binge eating disorder and bulimia nervosa) and potential links with the gut microbiota, food reward alterations and neuroinflammation. Finally, we propose gut microbiota modulation as a promising therapeutic strategy in food reward alterations and ED.
Collapse
Affiliation(s)
- Sabrina J P Huwart
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life BIOtechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| | - Nuria Morales-Puerto
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life BIOtechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life BIOtechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
41
|
Puglisi CH, Kim M, Aldhafeeri M, Lewandowski M, Vuong HE. Interactions of the maternal microbiome with diet, stress, and infection influence fetal development. FEBS J 2025; 292:1437-1453. [PMID: 39988792 PMCID: PMC11927046 DOI: 10.1111/febs.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/07/2024] [Accepted: 01/14/2025] [Indexed: 02/25/2025]
Abstract
Humans and other animals contain multitudes of microorganisms including bacteria, fungi, and viruses, which make up a diverse microbiome. Across body sites including skin, gastrointestinal tract, and oral cavity there are distinct microbial niches that are made up of trillions of microorganisms that have co-evolved to inhabit and interact with the host. The microbiome also interacts with the changing environment. This tripartite interaction between the host, microbiome, and environment suggests microbial communities play a key role in the biological processes of the host, such as development and behaviors. Over the past two decades, emerging research continues to reveal how host and microbe interactions impact nervous system signaling and behaviors, and influence neurodevelopmental, neurological, and neurodegenerative disorders. In this review, we will describe the unique features of the maternal microbiome that exist during the perinatal period and discuss evidence for the function of the maternal microbiome in offspring development. Finally, we will discuss how the maternal environment interacts with the microbiome and nervous system development and then postulate how the maternal microbiome can modify early offspring development to have lasting influence on brain health.
Collapse
Affiliation(s)
- Chloe H Puglisi
- Division of Neonatology, Department of PediatricsUniversity of MinnesotaMinneapolisMNUSA
| | - Minjeong Kim
- Division of Neonatology, Department of PediatricsUniversity of MinnesotaMinneapolisMNUSA
| | - Modi Aldhafeeri
- Division of Neonatology, Department of PediatricsUniversity of MinnesotaMinneapolisMNUSA
| | - Megan Lewandowski
- Division of Neonatology, Department of PediatricsUniversity of MinnesotaMinneapolisMNUSA
| | - Helen E. Vuong
- Division of Neonatology, Department of PediatricsUniversity of MinnesotaMinneapolisMNUSA
| |
Collapse
|
42
|
Moraes MA, Arabe LB, Resende BL, Codo BC, Reis ALAL, Souza BR. The gold standard control groups in physiological and pharmacological research are not that shiny: Intraperitoneal saline injection and needle pricking affect prepubescent mice's behavior in a sex-specific manner. Horm Behav 2025; 169:105707. [PMID: 39965530 DOI: 10.1016/j.yhbeh.2025.105707] [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: 10/24/2024] [Revised: 01/12/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
Study design and experimental tools are crucial for good quality science, and an essential part of it is the choice of control groups to best test the hypothesis. Two of the standard control groups in physiological and pharmacological research are needle pricking without substance injection (Sham) and/or vehicle injection (Saline). However, both needle pricking and saline injection can act as stressors, potentially influencing the analyzed outcome. This raises the question of whether the dependent variable remains unaffected by the stress induced by these procedures. Despite the significance of this issue, very few studies have investigated the behavioral effects of a single intraperitoneal (I.P.) Sham and/or single I.P. Saline injection in mice, and those that have used mostly adult males. In this study, we investigated if a single I.P. Sham and/or I.P. Saline injection affects female and male prepubertal (4-weeks-old) mice behavior. After Sham or Saline injection, we examined exploratory/motor behavior (open field test - OFT), anxiety-like behavior (elevated plus-maze - EPM), and behavioral despair/depressive-like behavior (forced swimming test - FST). We observed that both Sham prepubertal females and males showed behavioral alterations in OFT and EPM, and Saline males showed behavioral alterations in OFT and FST. On the other hand, prepubertal Saline females showed an increase in exploratory behavior, risk assessment/anxiety-like behavior, and behavioral despair/depressive-like behavior. Thus, our findings indicate that control procedures commonly used in physiological and pharmacological experimental designs affect the behavior of prepubescent mice, with more pronounced effects in females than in males. This study suggests considering Naïve animals together with Sham and/or Vehicle for a better and more honest interpretation of the data.
Collapse
Affiliation(s)
- Muiara Aparecida Moraes
- Laboratory of Neurodevelopment and Evolution - Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Laila Blanc Arabe
- Laboratory of Neurodevelopment and Evolution - Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Bruna Lopes Resende
- Laboratory of Neurodevelopment and Evolution - Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Beatriz Campos Codo
- Laboratory of Neurodevelopment and Evolution - Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ana Luiza Araújo Lima Reis
- Laboratory of Neurodevelopment and Evolution - Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Bruno Rezende Souza
- Laboratory of Neurodevelopment and Evolution - Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
43
|
Yin M, Peng J, Chen M, Zhan Q, Zhong H. Rosa Roxburghii Tratt Polysaccharides Prevent Alzheimer's Disease-Like Cognitive Dysfunctions and Pathology in Rats by Regulating the Microbiota-Gut-Brain Axis and Oxidative Stress. J Med Food 2025; 28:232-242. [PMID: 40042065 DOI: 10.1089/jmf.2024.k.0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
The microbial-gut-brain axis and oxidative stress may be important to the pathogenesis of Alzheimer's disease (AD). Rosa roxburghii Tratt polysaccharides (RRTP) have a strong antioxidant effect and can affect the gut microbiota, and whether it can affect AD is unknown. So, AlCl3 and d-galactose were used to establish AD model rats, and RRTP was used as an intervention treatment. Morris water maze test was used to detect cognitive functions. The hippocampus was used to observe the pathological changes, and the cortex was used to measure antioxidant markers. The stool was collected for 16S rDNA sequencing. Morris water maze test showed that the learning ability and memory level of AD group rats were decreased, and RRTP intervention could mitigate the injury to a certain extent. In the AD group, hematoxylin-eosin staining revealed changes in the morphology of neurons, silver glycine staining revealed neurofibrillary tangles and Congo red staining revealed β-amyloid. RRTP could ameliorate the above changes to some extent. The results of superoxide dismutase, malondialdehyde, and glutathione peroxidase showed that the antioxidant capacity in the RRTP intervention group was significantly higher than that in the AD group. 16S rDNA sequencing results showed that there were differences in the species composition of gut microbiota, and the ratio of Firmicutes to Bacteroidetes in the AD group was decreased. After RRTP intervention, the proportion of Lactobacillus increased. In conclusion, RRTP may prevent AD pathology and cognitive functions in rats to a certain extent through the microbiota-gut-brain axis and oxidative stress.
Collapse
Affiliation(s)
- Mingyue Yin
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| | - Jiangjiang Peng
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| | - Ming Chen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| | - Qingqing Zhan
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| | - Hui Zhong
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| |
Collapse
|
44
|
Mehta I, Juneja K, Nimmakayala T, Bansal L, Pulekar S, Duggineni D, Ghori HK, Modi N, Younas S. Gut Microbiota and Mental Health: A Comprehensive Review of Gut-Brain Interactions in Mood Disorders. Cureus 2025; 17:e81447. [PMID: 40303511 PMCID: PMC12038870 DOI: 10.7759/cureus.81447] [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] [Accepted: 03/30/2025] [Indexed: 05/02/2025] Open
Abstract
The human gut flora of trillions of bacteria is vital for general health and greatly influences digestion, immune system function, and brain development. Through neuronal, hormonal, and immunological channels, the gut-brain axis (GBA), a bidirectional communication network, links the gut microbiota to the central nervous system (CNS). This relationship has been linked to affective diseases, including depression and anxiety, as well as mental health issues. This review explores the intricate relationship between gut bacteria and mood disorders, focusing on how gut microbiota-host interactions, immune system modulation, and neurotransmitter control support mental health. The function of important microbial metabolites, including short-chain fatty acids (SCFAs), in preserving blood-brain barrier integrity and modulating neuroinflammation is covered in this review. It also examines the bidirectional impact between gut health and mental health, including how dysbiosis could aggravate mood disorders and how depressed states might change the composition of gut bacteria. Furthermore, we discuss how psychotropic drugs affect gut flora and consider other elements such as nutrition and lifestyle that affect gut microbiome composition. Potential paths for treating mood disorders through gut microbiota modification are presented as emerging treatment techniques, including probiotics, nutritional therapies, and precision medicine. The development of new therapeutic approaches for mood disorders depends on the awareness of the GBA. Gut bacteria significantly affect mental health through immune modulation, neurotransmitter generation, and other intricate processes. Future studies should concentrate on large, varied populations to better understand these interactions and to create customized treatments that combine gut microbiota modulation with conventional mental health therapies.
Collapse
Affiliation(s)
- Ishani Mehta
- Psychiatry and Behavioral Sciences, Maharaja Agrasen Institute of Medical Research and Education, Hisar, IND
| | | | - Tharun Nimmakayala
- Medicine and Surgery, Apollo Institute of Medical Sciences and Research, Chittoor, IND
| | - Lajpat Bansal
- Psychiatry and Behavioral Sciences, Maharaja Agrasen Institute of Medical Research and Education, Hisar, IND
| | - Shivani Pulekar
- General Practice, Davao Medical School Foundation, Davao, PHL
| | | | | | - Nishi Modi
- Medicine, Government Medical College, Surat, Surat, IND
| | - Salma Younas
- Pharmacy, Punjab University College of Pharmacy, Lahore, PAK
| |
Collapse
|
45
|
Green JE, Wrobel A, Todd E, Marx W, Berk M, Lotfaliany M, Castle D, Cryan JF, Athan E, Hair C, Nierenberg AA, Jacka FN, Dawson S. Early antibiotic exposure and risk of psychiatric and neurocognitive outcomes: systematic review and meta-analysis. Br J Psychiatry 2025; 226:171-183. [PMID: 39658347 DOI: 10.1192/bjp.2024.121] [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] [Indexed: 12/12/2024]
Abstract
BACKGROUND The prenatal and early-life periods pose a crucial neurodevelopmental window whereby disruptions to the intestinal microbiota and the developing brain may have adverse impacts. As antibiotics affect the human intestinal microbiome, it follows that early-life antibiotic exposure may be associated with later-life psychiatric or neurocognitive outcomes. AIMS To explore the association between early-life (in utero and early childhood (age 0-2 years)) antibiotic exposure and the subsequent risk of psychiatric and neurocognitive outcomes. METHOD A search was conducted using Medline, PsychINFO and Excerpta Medica databases on 20 November 2023. Risk of bias was assessed using the Newcastle-Ottawa scale, and certainty was assessed using the grading of recommendations, assessment, development and evaluation (GRADE) certainty assessment. RESULTS Thirty studies were included (n = 7 047 853 participants). Associations were observed between in utero antibiotic exposure and later development of autism spectrum disorder (ASD) (odds ratio 1.09, 95% CI: 1.02-1.16) and attention-deficit hyperactivity disorder (ADHD) (odds ratio 1.19, 95% CI: 1.11-1.27) and early-childhood exposure and later development of ASD (odds ratio 1.19, 95% CI: 1.01-1.40), ADHD (odds ratio 1.33, 95% CI: 1.20-1.48) and major depressive disorder (MDD) (odds ratio 1.29, 95% CI: 1.04-1.60). However, studies that used sibling control groups showed no significant association between early-life exposure and ASD or ADHD. No studies in MDD used sibling controls. Using the GRADE certainty assessment, all meta-analyses but one were rated very low certainty, largely owing to methodological and statistical heterogeneity. CONCLUSIONS While there was weak evidence for associations between antibiotic use in early-life and later neurodevelopmental outcomes, these were attenuated in sibling-controlled subgroup analyses. Thus, associations may be explained by genetic and familial confounding, and studies failing to utilise sibling-control groups must be interpreted with caution. PROSPERO ID: CRD42022304128.
Collapse
Affiliation(s)
- Jessica Emily Green
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Monash Alfred Psychiatry Research Centre (MAPrc), Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Prahran, Australia
- Department of Psychiatry, Peninsula Health, Frankston, Australia
| | - Anna Wrobel
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Emma Todd
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Wolfgang Marx
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Michael Berk
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Melbourne, Parkville, Australia
- Orygen Youth Health Research Centre and the Centre of Youth Mental Health, Melbourne, Australia
- The Florey Institute for Neuroscience and Mental Health, Parkville, Australia
- Department of Mental Health Drug and Alcohol Services, Barwon Health, Geelong, Australia
| | - Mojtaba Lotfaliany
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - David Castle
- School of Psychological Sciences, University of Tasmania, Hobart, Tasmania
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork and APC Microbiome, Cork, Ireland
| | - Eugene Athan
- Department of Mental Health Drug and Alcohol Services, Barwon Health, Geelong, Australia
| | - Christopher Hair
- Department of Mental Health Drug and Alcohol Services, Barwon Health, Geelong, Australia
| | - Andrew A Nierenberg
- Dauten Family Center for Bipolar Treatment Innovation, Department of Psychiatry, Massachusetts General Hospital, Boston, USA
- Department of Psychiatry, Harvard Medical School, Boston, USA
| | - Felice N Jacka
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Department of Immunology, Therapeutics, and Vaccines, James Cook University, Townsville, Australia
| | - Samantha Dawson
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| |
Collapse
|
46
|
Zhou X, Ganz AB, Rayner A, Cheng TY, Oba H, Rolnik B, Lancaster S, Lu X, Li Y, Johnson JS, Hoyd R, Spakowicz DJ, Slavich GM, Snyder MP. Dynamic human gut microbiome and immune shifts during an immersive psychosocial intervention program. Brain Behav Immun 2025; 125:428-443. [PMID: 39701328 PMCID: PMC11903166 DOI: 10.1016/j.bbi.2024.12.027] [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: 05/08/2024] [Revised: 10/24/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Although depression is a leading cause of disability worldwide, the pathophysiological mechanisms underlying this disorder-particularly those involving the gut microbiome-are poorly understood. METHOD To investigate, we conducted a community-based observational study to explore complex associations between changes in the gut microbiome, cytokine levels, and depression symptoms in 51 participants (Mage = 49.56, SD = 13.31) receiving an immersive psychosocial intervention. A total of 142 multi-omics samples were collected from participants before, during, and three months after the nine-day inquiry-based stress reduction program. RESULTS Results revealed that depression was associated with both an increased presence of putatively pathogenic bacteria and reduced microbial beta-diversity. Following the intervention, we observed reductions in neuroinflammatory cytokines and improvements in several mental health indicators. Interestingly, participants with a Prevotella-dominant microbiome showed milder symptoms when depressed, along with a more resilient microbiome and more favorable inflammatory cytokine profile, including reduced levels of CXCL-1. CONCLUSIONS These findings reveal a potentially protective link between the Prevotella-dominant microbiome and depression, as evidenced by a reduced pro-inflammatory environment and fewer depressive symptoms. These insights, coupled with observed improvements in neuroinflammatory markers and mental health from the intervention, may highlight potential avenues for microbiome-targeted therapies for managing depression.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Genetics, Stanford University School of Medicine, CA, USA; Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, CA, USA
| | - Ariel B Ganz
- Department of Genetics, Stanford University School of Medicine, CA, USA; Stanford Healthcare Innovation Lab, Stanford University, CA, USA
| | - Andre Rayner
- Department of Genetics, Stanford University School of Medicine, CA, USA
| | - Tess Yan Cheng
- Department of Genetics, Stanford University School of Medicine, CA, USA; Department of Microbiology, College of Arts and Sciences, University of Washington, WA, USA
| | - Haley Oba
- Department of Genetics, Stanford University School of Medicine, CA, USA
| | - Benjamin Rolnik
- Department of Genetics, Stanford University School of Medicine, CA, USA; Stanford Healthcare Innovation Lab, Stanford University, CA, USA
| | - Samuel Lancaster
- Department of Genetics, Stanford University School of Medicine, CA, USA
| | - Xinrui Lu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Sichuan, China
| | - Yizhou Li
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Sichuan, China
| | - Jethro S Johnson
- Oxford Centre for Microbiome Studies, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Rebecca Hoyd
- The Ohio State University Comprehensive Cancer Center, OH, USA
| | | | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, CA, USA; Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, CA, USA; Stanford Healthcare Innovation Lab, Stanford University, CA, USA.
| |
Collapse
|
47
|
Petrut SM, Bragaru AM, Munteanu AE, Moldovan AD, Moldovan CA, Rusu E. Gut over Mind: Exploring the Powerful Gut-Brain Axis. Nutrients 2025; 17:842. [PMID: 40077713 PMCID: PMC11901622 DOI: 10.3390/nu17050842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Background: The human gastrointestinal tract is home to a wide variety of microorganisms. For some decades now, bacteria known as probiotics have been added to various foods because of their beneficial effects for human health. Evidence indicates that probiotics significantly regulate gut microbiota, which is vital for digestion, metabolism, immune function, and mental health. Methods: We conducted a narrative review of available original research published in PubMed for the past ten years focusing on recent advancements that provide a thorough understanding of the relationship between the gastrointestinal system and the brain. Results: Recent advances in research have focused on the importance of gut microbiota in influencing mental health. The microbiota-gut-brain axis is a complex, bidirectional communication network linking the central nervous system and the gastrointestinal tract, which highlights how the gut and brain are deeply interconnected and influence each other in ways that affect our overall health, emotions, and behavior. This powerful link is a major area of research as scientists discover more about how gut health can impact mental well-being. Conclusions: A comprehensive understanding of microbiota composition and mechanisms involved in these interactions between the gut and the brain could shape future medical and therapeutic approaches. It would balance scientific explanation with clinical relevance, offering insights into how understanding the brain-gut axis can revolutionize our approach to treating mental health and gastrointestinal disorders.
Collapse
Affiliation(s)
- Stefana-Maria Petrut
- Department of Preclinical Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania; (S.-M.P.); (E.R.)
| | - Alexandra Maria Bragaru
- Doctoral School of Medicine, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (A.M.B.); (A.-D.M.)
| | - Alice Elena Munteanu
- Department of Medico-Surgical and Prophylactic Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
- Department of Cardiology, “Dr. Carol Davila” Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Adina-Diana Moldovan
- Doctoral School of Medicine, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (A.M.B.); (A.-D.M.)
- MedLife SA, 010719 Bucharest, Romania
| | - Cosmin-Alec Moldovan
- Department of Medico-Surgical and Prophylactic Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
- Department of General Surgery, Witting Clinical Hospital, 010243 Bucharest, Romania
| | - Elena Rusu
- Department of Preclinical Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania; (S.-M.P.); (E.R.)
| |
Collapse
|
48
|
Lan G, Ma R, Zhou Y, Lu Z, Zhu B, Liu J, Wu W, Zhang Y, Liu J, Gu H, Lin J, Wei W, Qi D. Gut microbiota alterations in golden snub-nosed monkeys during food shortage and parturition-nursing periods. Front Microbiol 2025; 16:1556648. [PMID: 40083783 PMCID: PMC11903488 DOI: 10.3389/fmicb.2025.1556648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
Adopting unique survival strategies during spring food shortages and simultaneous parturition and nursing is crucial for golden snub-nosed monkeys. Social behaviors, such as altruism within one-male units (OMUs), are decisive for family health, but the role of microbiota in regulating these behaviors remains unknown. We conducted the gut microbiota from members of 10 OMUs using 16S RNA sequencing technology. We found that in adult males, gut microbiota diversity significantly decreased in food shortages and parturition-nursing period. Meanwhile, there was a notable reduction in 12 metabolism-related pathways, including those related to carbohydrates, amino acids, and lipid. The gut microbiota of adult male monkeys shifts from being enriched with the genera Akkermansia in winter to the genera norank Muribaculaceae in spring. This transition alters the pathways for nutrient acquisition, thereby reducing the consumption of stored energy. In contrast, other OMU members (adult females and subadults) did not experience adverse effects on the metabolic functions of their gut microbiota during the food-scarce spring, which is also a critical period for parturition and lactation in adult females. This study elucidates the co-evolution of altruistic behavior and gut microbiota in Sichuan snub-nosed monkeys, insights into the interaction mechanisms between mammalian microbiota and survival strategies.
Collapse
Affiliation(s)
- Guanwei Lan
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan, China
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Rui Ma
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan, China
| | - Yanshan Zhou
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan, China
| | - Zhantao Lu
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan, China
- College of Life and Environmental Sciences, Central South University of Forestry and Technology, Changsha, China
| | - Biqing Zhu
- Administrative Bureau of Baihe National Nature Reserve, Aba, China
| | - Juan Liu
- Administrative Bureau of Baihe National Nature Reserve, Aba, China
| | - Wei Wu
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan, China
| | - Yue Zhang
- Administrative Bureau of Baihe National Nature Reserve, Aba, China
| | - Jiabin Liu
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan, China
- College of Life and Environmental Sciences, Central South University of Forestry and Technology, Changsha, China
| | - Haijun Gu
- Sichuan Provincial Bureau of Forestry and Grassland, Chengdu, Sichuan, China
| | - Jie Lin
- Administrative Bureau of Baihe National Nature Reserve, Aba, China
| | - Wei Wei
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Dunwu Qi
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, Chengdu, Sichuan, China
| |
Collapse
|
49
|
Krawczyk A, Sladowska GE, Strzalka-Mrozik B. The Role of the Gut Microbiota in Modulating Signaling Pathways and Oxidative Stress in Glioma Therapies. Cancers (Basel) 2025; 17:719. [PMID: 40075568 PMCID: PMC11899293 DOI: 10.3390/cancers17050719] [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: 01/12/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Tumors of the central nervous system (CNS), especially gliomas, pose a significant clinical challenge due to their aggressive nature and limited therapeutic options. Emerging research highlights the critical role of the gut microbiota in regulating CNS health and disease. The composition of the gut microbiota is essential for maintaining CNS homeostasis, as it modulates immune responses, oxidative status, and neuroinflammation. The microbiota-gut-brain axis, a bidirectional communication network, plays a pivotal role in cancer and CNS disease treatment, exerting its influence through neural, endocrine, immunological, and metabolic pathways. Recent studies suggest that the gut microbiota influences the solidification of the tumor microenvironment and that dysbiosis may promote glioma development by modulating systemic inflammation and oxidative stress, which contributes to tumorigenesis and CNS tumor progression. This review interrogates the impact of the gut microbiota on glioma, focusing on critical pathways such as NF-κB, MAPK, PI3K/Akt/mTOR, and Kynurenine/AhR that drive tumor proliferation, immune evasion, and therapy resistance. Furthermore, we explore emerging therapeutic strategies, including probiotics and microbiota-based interventions, which show potential in modulating these pathways and enhancing immunotherapies such as checkpoint inhibitors. By focusing on the multifaceted interactions between the gut microbiota, oxidative stress, and CNS tumors, this review highlights the potential of microbiota-targeted therapies and their manipulation to complement and enhance current treatments.
Collapse
Affiliation(s)
| | | | - Barbara Strzalka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.K.); (G.E.S.)
| |
Collapse
|
50
|
Takahashi K, Kurokawa K, Miyagawa K, Mochida-Saito A, Takeda H, Tsuji M. Repeated exposure to antibiotics exhibits anxiety-like behaviors with a reduction in neurogenesis in the ventral hippocampus of dentate gyrus. Neurosci Lett 2025; 849:138131. [PMID: 39842766 DOI: 10.1016/j.neulet.2025.138131] [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: 11/28/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Disruption of gut microbiota balance is known to contribute to the development of anxiety; however, it remains unclear whether dysbiosis-induced anxiety involves the glycogen synthase kinase-3β (GSK-3β)/cAMP response element binding protein (CREB)/brain-derived neurotrophic factor (BDNF) pathway and neurogenesis in the ventral hippocampal dentate gyrus (DG). In this study, Male ddY mice were administered an antibacterial cocktail to induce dysbiosis. The dysbiosis model displayed anxiety-like behaviors in the hole-board and elevated plus-maze tests, decreased the phosphorylation levels of GSK-3β (Ser9) and CREB, decreased the expression level of BDNF in the ventral hippocampus, and reduced neurogenesis in the ventral hippocampal DG. This suggests that dysbiosis-induced anxiety-like behaviors are associated with reduced neurogenesis in the ventral hippocampal DG via the GSK-3β/CREB/BDNF pathway.
Collapse
Affiliation(s)
- Kohei Takahashi
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Kazuhiro Kurokawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Kazuya Miyagawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Atsumi Mochida-Saito
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Hiroshi Takeda
- Research Administration Office, International University of Health and Welfare, 4-1-26 Akasaka, Minato-ku, Tokyo 107-8402, Japan
| | - Minoru Tsuji
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan.
| |
Collapse
|