1
|
Valle J. Biofilm-associated proteins: from the gut biofilms to neurodegeneration. Gut Microbes 2025; 17:2461721. [PMID: 39898557 PMCID: PMC11792866 DOI: 10.1080/19490976.2025.2461721] [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: 10/25/2024] [Revised: 12/07/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025] Open
Abstract
Human microbiota form a biofilm with substantial consequences for health and disease. Numerous studies have indicated that microbial communities produce functional amyloids as part of their biofilm extracellular scaffolds. The overlooked interplay between bacterial amyloids and the host may have detrimental consequences for the host, including neurodegeneration. This work gives an overview of the biofilm-associated amyloids expressed by the gut microbiota and their potential role in neurodegeneration. It discusses the biofilm-associated proteins (BAPs) of the gut microbiota, maps the amyloidogenic domains of these proteins, and analyzes the presence of bap genes within accessory genomes linked with transposable elements. Furthermore, the evidence supporting the existence of amyloids in the gut are presented. Finally, it explores the potential interactions between BAPs and α-synuclein, extending the literature on amyloid cross-kingdom interactions. Based on these findings, this study propose that BAP amyloids act as transmissible catalysts, facilitating the misfolding, accumulation, and spread of α-synuclein aggregates. This review contributes to the understanding of complex interactions among the microbiota, transmissible elements, and host, which is crucial for developing novel therapeutic approaches to combat microbiota-related diseases and improve overall health outcomes.
Collapse
Affiliation(s)
- Jaione Valle
- Microbial Biotechnology Department, Instituto de Agrobiotecnología, CSIC-Gobierno de Navarra, Mutilva, Navarra, Spain
| |
Collapse
|
2
|
Park SJ, Kim KW, Lee EJ. Gut-brain axis and environmental factors in Parkinson's disease: bidirectional link between disease onset and progression. Neural Regen Res 2025; 20:3416-3429. [PMID: 39688568 PMCID: PMC11974660 DOI: 10.4103/nrr.nrr-d-24-00994] [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: 08/28/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease has long been considered a disorder that primarily affects the brain, as it is defined by the dopaminergic neurodegeneration in the substantia nigra and the brain accumulation of Lewy bodies containing α-synuclein protein. In recent decades, however, accumulating research has revealed that Parkinson's disease also involves the gut and uncovered an intimate and important bidirectional link between the brain and the gut, called the "gut-brain axis." Numerous clinical studies demonstrate that gut dysfunction frequently precedes motor symptoms in Parkinson's disease patients, with findings including impaired intestinal permeability, heightened inflammation, and distinct gut microbiome profiles and metabolites. Furthermore, α-synuclein deposition has been consistently observed in the gut of Parkinson's disease patients, suggesting a potential role in disease initiation. Importantly, individuals with vagotomy have a reduced Parkinson's disease risk. From these observations, researchers have hypothesized that α-synuclein accumulation may initiate in the gut and subsequently propagate to the central dopaminergic neurons through the gut-brain axis, leading to Parkinson's disease. This review comprehensively examines the gut's involvement in Parkinson's disease, focusing on the concept of a gut-origin for the disease. We also examine the interplay between altered gut-related factors and the accumulation of pathological α-synuclein in the gut of Parkinson's disease patients. Given the accessibility of the gut to both dietary and pharmacological interventions, targeting gut-localized α-synuclein represents a promising avenue for developing effective Parkinson's disease therapies.
Collapse
Affiliation(s)
- Soo Jung Park
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Kyung Won Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon, South Korea
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
3
|
Li P, Li M, Chen WH. Best practices for developing microbiome-based disease diagnostic classifiers through machine learning. Gut Microbes 2025; 17:2489074. [PMID: 40186338 PMCID: PMC11980492 DOI: 10.1080/19490976.2025.2489074] [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: 12/05/2024] [Revised: 03/13/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025] Open
Abstract
The human gut microbiome, crucial in various diseases, can be utilized to develop diagnostic models through machine learning (ML). The specific tools and parameters used in model construction such as data preprocessing, batch effect removal and modeling algorithms can impact model performance and generalizability. To establish an generally applicable workflow, we divided the ML process into three above-mentioned steps and optimized each sequentially using 83 gut microbiome cohorts across 20 diseases. We tested a total of 156 tool-parameter-algorithm combinations and benchmarked them according to internal- and external- AUCs. At the data preprocessing step, we identified four data preprocessing methods that performed well for regression-type algorithms and one method that excelled for non-regression-type algorithms. At the batch effect removal step, we identified the "ComBat" function from the sva R package as an effective batch effect removal method and compared the performance of various algorithms. Finally, at the ML algorithm selection step, we found that Ridge and Random Forest ranked the best. Our optimized work flow performed similarly comparing with previous exhaustive methods for disease-specific optimizations, thus is generally applicable and can provide a comprehensive guideline for constructing diagnostic models for a range of diseases, potentially serving as a powerful tool for future medical diagnostics.
Collapse
Affiliation(s)
- Peikun Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- School of Biological Science, Jining Medical University, Rizhao, China
| |
Collapse
|
4
|
Zhang Y, Mo C, Ai P, He X, Xiao Q, Yang X. Pharmacomicrobiomics: a new field contributing to optimizing drug therapy in Parkinson's disease. Gut Microbes 2025; 17:2454937. [PMID: 39875349 PMCID: PMC11776486 DOI: 10.1080/19490976.2025.2454937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/19/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
Gut microbiota, which act as a determinant of pharmacokinetics, have long been overlooked. In recent years, a growing body of evidence indicates that the gut microbiota influence drug metabolism and efficacy. Conversely, drugs also exert a substantial influence on the function and composition of the gut microbiota. Pharmacomicrobiomics, an emerging field focusing on the interplay of drugs and gut microbiota, provides a potential foundation for making certain advances in personalized medicine. Understanding the communication between gut microbiota and antiparkinsonian drugs is critical for precise treatment of Parkinson's disease. Here, we provide a historical overview of the interplay between gut microbiota and antiparkinsonian drugs. Moreover, we discuss potential mechanistic insights into the complex associations between gut microbiota and drug metabolism. In addition, we also draw attention to microbiota-based biomarkers for predicting antiparkinsonian drug efficacy and examine current state-of-the-art knowledge of microbiota-based strategies to optimize drug therapy in Parkinson's disease.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengjun Mo
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Penghui Ai
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqin He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Xiao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Horn JA, Delgadillo DR, Mayer EA. Understanding Microbial Mediation of the Brain-Gut Axis. Gastroenterol Clin North Am 2025; 54:367-381. [PMID: 40348493 DOI: 10.1016/j.gtc.2024.12.004] [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: 05/14/2025]
Abstract
Bidirectional communications between the gut and the brain play an important role in the regulation of food intake, pain perception, mood, and cognitive function. The involved communication pathways are modulated by signals generated by the gut microbiome. Alterations in these communications have been implicated in several chronic brain and gut disorders, including food addiction, mood disorders, neurodevelopmental and neurodegenerative disorders, and functional and inflammatory bowel disorders. The gut microbiome holds great promise for the development of novel therapies normalizing altered brain-gut interactions.
Collapse
Affiliation(s)
- Jill A Horn
- Department of Population and Public Health Sciences, Keck School of Medicine at USC, 1845 N Soto Street, Los Angeles, CA 90032, USA
| | - Desiree R Delgadillo
- Goodman-Luskin Microbiome Center, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, CHS 42-210, MC737818, Los Angeles, CA 90095-73787, USA
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress & Resilience; UCLA Vatche & Tamar Manoukian Division of Digestive Diseases, Goodman Luskin Microbiome Center, UCLA.
| |
Collapse
|
6
|
Wirbel J, Andermann TM, Brooks EF, Evans L, Groth A, Dvorak M, Chakraborty M, Palushaj B, Reynolds GZM, Porter IE, Al Malki M, Rezvani A, Gooptu M, Elmariah H, Runaas L, Fei T, Martens MJ, Bolaños-Meade J, Hamadani M, Holtan S, Jenq R, Peled JU, Horowitz MM, Poston KL, Saber W, Kean LS, Perales MA, Bhatt AS. Accurate prediction of absolute prokaryotic abundance from DNA concentration. CELL REPORTS METHODS 2025; 5:101030. [PMID: 40300608 DOI: 10.1016/j.crmeth.2025.101030] [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: 11/02/2024] [Revised: 01/24/2025] [Accepted: 04/02/2025] [Indexed: 05/01/2025]
Abstract
Quantification of the absolute microbial abundance in a human stool sample is crucial for a comprehensive understanding of the microbial ecosystem, but this information is lost upon metagenomic sequencing. While several methods exist to measure absolute microbial abundance, they are technically challenging and costly, presenting an opportunity for machine learning. Here, we observe a strong correlation between DNA concentration and the absolute number of 16S ribosomal RNA copies as measured by digital droplet PCR in clinical stool samples from individuals undergoing hematopoietic cell transplantation (BMT CTN 1801). Based on this correlation and additional measurements, we trained an accurate yet simple machine learning model for the prediction of absolute prokaryotic load, which showed exceptional prediction accuracy on an external cohort that includes people living with Parkinson's disease and healthy controls. We propose that, with further validation, this model has the potential to enable accurate absolute abundance estimation based on readily available sample measurements.
Collapse
Affiliation(s)
- Jakob Wirbel
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA.
| | - Tessa M Andermann
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill, NC, USA
| | - Erin F Brooks
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | - Lanya Evans
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill, NC, USA
| | - Adam Groth
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill, NC, USA
| | - Mai Dvorak
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Bianca Palushaj
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA, USA
| | | | - Imani E Porter
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Monzr Al Malki
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Andrew Rezvani
- Blood and Marrow Transplantation and Cellular Therapy Unit, Stanford University School of Medicine, Stanford, CA, USA
| | - Mahasweta Gooptu
- Hematologic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hany Elmariah
- Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer and Research Center, Tampa, FL, USA
| | - Lyndsey Runaas
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Michael J Martens
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Javier Bolaños-Meade
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mehdi Hamadani
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shernan Holtan
- Blood and Marrow Transplantation Section, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Rob Jenq
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York City, NY, USA; Department of Medicine, Weill Cornell Medical College, New York City, NY, USA
| | - Mary M Horowitz
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kathleen L Poston
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Wael Saber
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Leslie S Kean
- Hematologic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Division of Pediatric Hematology and Oncology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Miguel-Angel Perales
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA; Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ami S Bhatt
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
7
|
Romano S, Wirbel J, Ansorge R, Schudoma C, Ducarmon QR, Narbad A, Zeller G. Machine learning-based meta-analysis reveals gut microbiome alterations associated with Parkinson's disease. Nat Commun 2025; 16:4227. [PMID: 40335465 PMCID: PMC12059030 DOI: 10.1038/s41467-025-56829-3] [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: 12/01/2023] [Accepted: 01/30/2025] [Indexed: 05/09/2025] Open
Abstract
There is strong interest in using the gut microbiome for Parkinson's disease (PD) diagnosis and treatment. However, a consensus on PD-associated microbiome features and a multi-study assessment of their diagnostic value is lacking. Here, we present a machine learning meta-analysis of PD microbiome studies of unprecedented scale (4489 samples). Within most studies, microbiome-based machine learning models accurately classify PD patients (average AUC 71.9%). However, these models are study-specific and do not generalise well across other studies (average AUC 61%). Training models on multiple datasets improves their generalizability (average LOSO AUC 68%) and disease specificity as assessed against microbiomes from other neurodegenerative diseases. Moreover, meta-analysis of shotgun metagenomes delineates PD-associated microbial pathways potentially contributing to gut health deterioration and favouring the translocation of pathogenic molecules along the gut-brain axis. Strikingly, microbial pathways for solvent and pesticide biotransformation are enriched in PD. These results align with epidemiological evidence that exposure to these molecules increases PD risk and raise the question of whether gut microbes modulate their toxicity. Here, we offer the most comprehensive overview to date about the PD gut microbiome and provide future reference for its diagnostic and functional potential.
Collapse
Affiliation(s)
- Stefano Romano
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Jakob Wirbel
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Rebecca Ansorge
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
| | - Christian Schudoma
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Quinten Raymond Ducarmon
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Arjan Narbad
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Georg Zeller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands.
- Center for Microbiome Analyses and Therapeutics (CMAT), Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
8
|
Lee JY, Jo S, Lee J, Choi M, Kim K, Lee S, Kim HS, Bae JW, Chung SJ. Distinct gut microbiome characteristics and dynamics in patients with Parkinson's disease based on the presence of premotor rapid-eye movement sleep behavior disorders. MICROBIOME 2025; 13:108. [PMID: 40307949 PMCID: PMC12042535 DOI: 10.1186/s40168-025-02095-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Alpha-synuclein aggregation, a hallmark of Parkinson's disease (PD), is hypothesized to often begin in the enteric or peripheral nervous system in "body-first" PD and progresses through the vagus nerve to the brain, therefore REM sleep behavior disorder (RBD) precedes the PD diagnosis. In contrast, "brain-first" PD begins in the central nervous system. Evidence that gut microbiome imbalances observed in PD and idiopathic RBD exhibit similar trends supports body-first and brain-first hypothesis and highlights the role of microbiota in PD pathogenesis. However, further investigation is needed to understand distinct microbiome changes in body-first versus brain-first PD over the disease progression. RESULTS Our investigation involved 104 patients with PD and 85 of their spouses as healthy controls (HC), with 57 patients (54.8%) categorized as PD-RBD(+) and 47 patients (45.2%) as PD-RBD(-) based on RBD presence before the PD diagnosis. We evaluated the microbiome differences between these groups over the disease progression through taxonomic and functional differential abundance analyses and carbohydrate-active enzyme (CAZyme) profiles based on metagenome-assembled genomes. The PD-RBD(+) gut microbiome showed a relatively stable microbiome composition irrespective of disease stage. In contrast, PD-RBD(-) microbiome exhibited a relatively dynamic microbiome change as the disease progressed. In early-stage PD-RBD(+), Escherichia and Akkermansia, associated with pathogenic biofilm formation and host mucin degradation, respectively, were enriched, which was supported by functional analysis. We discovered that genes of the UDP-GlcNAc synthesis/recycling pathway negatively correlated with biofilm formation; this finding was further validated in a separate cohort. Furthermore, fiber intake-associated taxa were decreased in early-stage PD-RBD(+) and the biased mucin-degrading capacity of CAZyme compared to fiber degradation. CONCLUSION We determined that the gut microbiome dynamics in patients with PD according to the disease progression depend on the presence of premotor RBD. Notably, early-stage PD-RBD(+) demonstrated distinct gut microbial characteristics, potentially contributing to exacerbation of PD pathophysiology. This outcome may contribute to the development of new therapeutic strategies targeting the gut microbiome in PD. Video Abstract.
Collapse
Affiliation(s)
- Jae-Yun Lee
- Department of Biology and Department of Life and Nanopharmaceutical Science, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sungyang Jo
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Jihyun Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Moongwan Choi
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Kijeong Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Sangjin Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Hyun Sik Kim
- Department of Biology and Department of Life and Nanopharmaceutical Science, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jin-Woo Bae
- Department of Biology and Department of Life and Nanopharmaceutical Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
9
|
Andreu-Sánchez S, Blanco-Míguez A, Wang D, Golzato D, Manghi P, Heidrich V, Fackelmann G, Zhernakova DV, Kurilshikov A, Valles-Colomer M, Weersma RK, Zhernakova A, Fu J, Segata N. Global genetic diversity of human gut microbiome species is related to geographic location and host health. Cell 2025:S0092-8674(25)00416-7. [PMID: 40311618 DOI: 10.1016/j.cell.2025.04.014] [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: 08/06/2024] [Revised: 01/23/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
The human gut harbors thousands of microbial species, each exhibiting significant inter-individual genetic variability. Although many studies have associated microbial relative abundances with human-health-related phenotypes, the substantial intraspecies genetic variability of gut microbes has not yet been comprehensively considered, limiting the potential of linking such genetic traits with host conditions. Here, we analyzed 32,152 metagenomes from 94 microbiome studies across the globe to investigate the human microbiome intraspecies genetic diversity. We reconstructed 583 species-specific phylogenies and linked them to geographic information and species' horizontal transmissibility. We identified 484 microbial-strain-level associations with 241 host phenotypes, encompassing human anthropometric factors, biochemical measurements, diseases, and lifestyle. We observed a higher prevalence of a Ruminococcus gnavus clade in nonagenarians correlated with distinct plasma bile acid profiles and a melanoma and prostate-cancer-associated Collinsella clade. Our large-scale intraspecies genetic analysis highlights the relevance of strain diversity as it relates to human health.
Collapse
Affiliation(s)
- Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Daoming Wang
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Davide Golzato
- Department of CIBIO, University of Trento, Trento, Italy
| | - Paolo Manghi
- Department of CIBIO, University of Trento, Trento, Italy
| | - Vitor Heidrich
- Department of CIBIO, University of Trento, Trento, Italy
| | | | - Daria V Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Mireia Valles-Colomer
- Department of CIBIO, University of Trento, Trento, Italy; MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.
| | - Nicola Segata
- Department of CIBIO, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy; Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|
10
|
Xia YM, Zhang MX, Ma XY, Tan LL, Li T, Wu J, Li MA, Zhao WJ, Qiao CM, Jia XB, Shen YQ, Cui C. Intranasal Transplantation of Microbiota Derived from Parkinson's Disease Mice Induced Astrocyte Activation and Neurodegenerative Pathology from Nose to Brain. Brain Sci 2025; 15:433. [PMID: 40426604 PMCID: PMC12109703 DOI: 10.3390/brainsci15050433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by early-onset olfactory dysfunction preceding motor symptoms, yet its mechanisms remain elusive. Based on the studies on microbiota-gut-brain axis, the microbiota-nose-brain axis might be involved in the pathogenesis of PD. However relative studies are rare. METHODS By consecutive 14-days intranasally transplanting bacteria, we established mice models exhibiting nasal microbiota dysbiosis (NMD), including animal group received intranasal drops of fecal bacterial suspension from normal mice (NB group) and animal group received intranasal drops of fecal bacterial suspension from PD mice (PB group), with animals that only received anesthesia used as the control group. Then we analyzed the nasal microbiota composition via 16S rRNA sequencing, evaluated the olfactory and motor functions through behavioral experiments, including buried food test, open field test, pole descent test, and traction test. The neuropathology in olfactory-related and PD-related brain regions, including olfactory bulb, pyriform cortex, hippocampus, substantia nigra and striatum, was also detected by western blotting, immunofluorescence and immunohistochemical experiments using the antibodies of NeuN, TH and GFAP. RESULTS 16S rRNA sequencing revealed that PB mice were primarily characterized by an increase in bacteria associated with inflammation and PD. Behavioral assessments revealed that mice with NMD demonstrated impairments in the buried food test and pole descent test, indicative of olfactory and motor dysfunction. By detecting NeuN and GFAP expression, we identified neuronal loss and astrocytes activation in olfactory-related brain regions and adjacent structures, including the olfactory bulb, pyriform cortex, hippocampus, substantia nigra and striatum of both NMD groups, which may contribute to the observed functional disorders. Notably, animals exposed to PD-derived bacteria exhibited more pronounced changes in nasal bacteria, with more severe neuropathology. CONCLUSIONS We present evidence supporting the microbiota-nose-brain axis, and the NMD-induced astrocyte activation and neurodegenerative pathology along the olfactory pathway may serve as a link between nose and brain.
Collapse
Affiliation(s)
- Yi-Meng Xia
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Mei-Xuan Zhang
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Xiao-Yu Ma
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Lu-Lu Tan
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ting Li
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Jian Wu
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Ming-An Li
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Wei-Jiang Zhao
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Chen-Meng Qiao
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Xue-Bing Jia
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yan-Qin Shen
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Chun Cui
- Laboratory of Neurodegenerative Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China; (Y.-M.X.); (M.-X.Z.); (X.-Y.M.); (L.-L.T.); (T.L.); (J.W.); (M.-A.L.); (W.-J.Z.); (C.-M.Q.); (X.-B.J.); (Y.-Q.S.)
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
11
|
Duru IC, Lecomte A, Laine P, Shishido TK, Suppula J, Paulin L, Scheperjans F, Pereira PAB, Auvinen P. Comparison of phage and plasmid populations in the gut microbiota between Parkinson's disease patients and controls. Sci Rep 2025; 15:13723. [PMID: 40258842 PMCID: PMC12012184 DOI: 10.1038/s41598-025-96924-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/01/2025] [Indexed: 04/23/2025] Open
Abstract
The aging population worldwide is on the rise, leading to a higher number of Parkinson's disease (PD) cases each year. PD is presently the second most prevalent neurodegenerative disease, affecting an estimated 7-10 million individuals globally. This research aimed to identify mobile genetic elements in human fecal samples using a shotgun metagenomics approach. We identified over 44,000 plasmid contigs and compared plasmid populations between PD patients (n = 68) and controls (n = 68). Significant associations emerged between groups (control vs PD) based on plasmid alpha and beta diversity. Moreover, the gene populations present on plasmids displayed marked differences in alpha and beta diversity between PD patients and controls. We identified a considerable number of phage contigs that were differentially abundant in the two groups. We also developed a predictive machine learning model based on phage abundance data, achieving a mean Area Under the Curve (AUC) of 0.74 with a standard deviation of 0.105 and a mean F1 score of 0.68 with a standard deviation of 0.14 across cross-validation folds, indicating moderate discriminatory power. Additionally, when tested on external data, the model yielded an AUC of 0.74 and an F1 score of 0.8, further demonstrating the predictive potential of phage populations in Parkinson's disease. Further, we improved the continuity and identification of the protein coding regions of the phage contigs by implementing alternative genetic codes.
Collapse
Affiliation(s)
- Ilhan Cem Duru
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
| | - Alexandre Lecomte
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Pia Laine
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | | - Joni Suppula
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Lars Paulin
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Filip Scheperjans
- Department of Neurology, Helsinki University Hospital and Clinicum, University of Helsinki, Helsinki, Finland
| | - Pedro A B Pereira
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
- Department of Neurology, Helsinki University Hospital and Clinicum, University of Helsinki, Helsinki, Finland.
| | - Petri Auvinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
12
|
Kumar R, Kumar R. Intestinal dysbiosis leads to the reduction in neurochemical production in Parkinson's disease (PD). INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:25-56. [PMID: 40414635 DOI: 10.1016/bs.irn.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Parkinson's Disease (PD) is a neurodegenerative disorder characterized by motor and non-motor symptoms, with emerging research suggesting a critical link between intestinal dysbiosis and PD progression. This review explores the pathophysiological mechanisms underlying PD, such as alpha-synuclein aggregation, mitochondrial dysfunction, neuroinflammation, and oxidative stress, while focusing on the impact of gut dysbiosis on intestinal barrier function and its role in reduced neurochemical production. The clinical features of PD, including dopamine, serotonin, and GABA deficiencies, are examined, with a focus on how dysbiosis contributes to neurotransmitter depletion. Current treatments of PD, such as levodopa and dopamine agonists, are discussed alongside gut health therapies such as probiotics, prebiotics, and Fecal Microbiota Transplantation (FMT). Future therapeutic directions, including synbiotics, engineered microbes, phage therapy, and the integration of machine learning (ML) and artificial intelligence (AI), are explored. The chapter also considers preventive strategies, such as lifestyle adjustments and early gut health monitoring using modern diagnostic tools and biosensors. Furthermore, a strong need for continued research into the gut-brain axis (GBA) to develop more effective, gut-targeted therapies for managing PD is discussed.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Life Sciences, GITAM School of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Rahul Kumar
- Department of Life Sciences, GITAM School of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India.
| |
Collapse
|
13
|
Bedarf JR, Romano S, Heinzmann SS, Duncan A, Traka MH, Ng D, Segovia-Lizano D, Simon MC, Narbad A, Wüllner U, Hildebrand F. A prebiotic dietary pilot intervention restores faecal metabolites and may be neuroprotective in Parkinson's Disease. NPJ Parkinsons Dis 2025; 11:66. [PMID: 40180909 PMCID: PMC11968880 DOI: 10.1038/s41531-025-00885-5] [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: 08/12/2024] [Accepted: 02/02/2025] [Indexed: 04/05/2025] Open
Abstract
Current treatment of Parkinson's Disease (PD) remains symptomatic, and disease-modifying approaches are urgently required. A promising approach is to modify intestinal microbiota and key metabolites of bacterial fermentation: short-chain fatty acids (SCFA), which are decreased in PD. A prospective, controlled pilot study (DRKS00034528) was conducted on 11 couples (PD patient plus healthy spouse as control (CO)). Participants followed a 4-week diet rich in dietary fibre, including intake of the prebiotic Lactulose. Gut metagenomes, faecal and urinary metabolites, and clinical characteristics were assessed. The dietary intervention significantly augmented faecal SCFA and increased Bifidobacteria spp., reducing PD-related gastrointestinal symptoms. The pre-existing bacterial dysbiosis in PD (depletion of Blautia, Dorea, Erysipelatoclostridium) persisted. Bacterial metabolite composition in faeces and urine positively changed with the intervention: Brain-relevant gut metabolic functions involved in neuroprotective and antioxidant pathways, including S-adenosyl methionine, glutathione, and inositol, improved in PD. These promising results warrant further investigation in larger cohorts.
Collapse
Affiliation(s)
- Janis Rebecca Bedarf
- Departent of Movement Disorders (PSB), Centre of Neurology, University Hospital Bonn, Bonn, Germany.
- German Centre for Neurodegenerative Diseases, DZNE Bonn, Bonn, Germany.
- Food, Microbiome, and Health, Quadram Institute Bioscience, Norfolk, UK.
| | - Stefano Romano
- Food, Microbiome, and Health, Quadram Institute Bioscience, Norfolk, UK
| | - Silke Sophie Heinzmann
- Research Unit Analytical BioGeoChemistry, Helmholtz Centre Munich, Neuherberg, Munich, Germany
| | - Anthony Duncan
- Food, Microbiome, and Health, Quadram Institute Bioscience, Norfolk, UK
- Decoding Biodiversity, Earlham Institute, Norfolk, UK
| | - Maria H Traka
- Food & Nutrition National Bioscience Research Infrastructure, Quadram Institute Bioscience, Norfolk, UK
| | - Duncan Ng
- Food & Nutrition National Bioscience Research Infrastructure, Quadram Institute Bioscience, Norfolk, UK
| | - Daniella Segovia-Lizano
- Food & Nutrition National Bioscience Research Infrastructure, Quadram Institute Bioscience, Norfolk, UK
| | - Marie-Christine Simon
- Institute of Nutritional and Food Sciences (IEL), Nutrition and Health, University of Bonn, Bonn, Germany
| | - Arjan Narbad
- Food, Microbiome, and Health, Quadram Institute Bioscience, Norfolk, UK
| | - Ullrich Wüllner
- Departent of Movement Disorders (PSB), Centre of Neurology, University Hospital Bonn, Bonn, Germany
- German Centre for Neurodegenerative Diseases, DZNE Bonn, Bonn, Germany
| | - Falk Hildebrand
- Food, Microbiome, and Health, Quadram Institute Bioscience, Norfolk, UK.
- Decoding Biodiversity, Earlham Institute, Norfolk, UK.
| |
Collapse
|
14
|
Sobral J, Empadinhas N, Esteves AR, Cardoso SM. Impact of Nutrition on the Gut Microbiota: Implications for Parkinson's Disease. Nutr Rev 2025; 83:713-727. [PMID: 39812804 DOI: 10.1093/nutrit/nuae208] [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] [Indexed: 01/16/2025] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disease that is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta and by the anomalous accumulation of α-synuclein aggregates into Lewy bodies and Lewy neurites. Research suggests 2 distinct subtypes of PD: the brain-first subtype if the pathology arises from the brain and then spreads to the peripheral nervous system (PNS) and the body-first subtype, where the pathological process begins in the PNS and then spreads to the central nervous system. This review primarily focuses on the body-first subtype. The influence of the gut microbiota on the development of PD has been the subject of growing interest among researchers. It has been suggested that gut inflammation may be closely associated with pathogenesis in PD, therefore leading to the hypothesis that gut microbiota modulation could play a significant role in this process. Nutrition can influence gut health and alter the risk and progression of PD by altering inflammatory markers. This review provides an overview of recent research that correlates variations in gut microbiota composition between patients with PD and healthy individuals with the impact of certain nutrients and dietary patterns, including the Mediterranean diet, the Western diet, and the ketogenic diet. It explores how these diets influence gut microbiota composition and, consequently, the risk of PD. Last, it examines fecal transplantation and the use of prebiotics, probiotics, or synbiotics as potential therapeutic strategies to balance the gut microbiome, aiming to reduce the risk or delay the progression of PD.
Collapse
Affiliation(s)
- Joana Sobral
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal
| | - Nuno Empadinhas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal
| | - Ana Raquel Esteves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal
| | - Sandra Morais Cardoso
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal
| |
Collapse
|
15
|
Ran Z, Mu BR, Wang DM, Xin-Huang, Ma QH, Lu MH. Parkinson's Disease and the Microbiota-Gut-Brain Axis: Metabolites, Mechanisms, and Innovative Therapeutic Strategies Targeting the Gut Microbiota. Mol Neurobiol 2025; 62:5273-5296. [PMID: 39531191 DOI: 10.1007/s12035-024-04584-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
The human gut microbiota is diverse and abundant and plays important roles in regulating health by participating in metabolism and controlling physiological activities. The gut microbiota and its metabolites have been shown to affect the functioning of the gut and central nervous system through the microbiota-gut-brain axis. It is well established that microbiota play significant roles in the pathogenesis and progression of Parkinson's disease (PD). Disorders of the intestinal microbiota and altered metabolite levels are closely associated with PD. Here, the changes in intestinal microbiota and effects of metabolites in patients with PD are reviewed. Potential mechanisms underlying intestinal microbiota disorders in the pathogenesis of PD are briefly discussed. Additionally, we outline the current strategies for the treatment of PD that target the gut microbiota, emphasizing the development of promising novel strategies.
Collapse
Affiliation(s)
- Zhao Ran
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ben-Rong Mu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dong-Mei Wang
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xin-Huang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215021, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215021, China.
| | - Mei-Hong Lu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
16
|
Namted S, Chailaor P, Bunchasak C. Effects of drinking water fructo-oligosaccharide supplementation on broiler chicken growth performance, blood glucose level, white blood cell count, carcass yield, meat quality, and cecal microbiota. Poult Sci 2025; 104:104901. [PMID: 40024010 PMCID: PMC11919399 DOI: 10.1016/j.psj.2025.104901] [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: 12/04/2024] [Revised: 02/09/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025] Open
Abstract
This study investigated the effects of fructo-oligosaccharides (FOS) supplementation on the growth performance, blood glucose level, white blood cell count, carcass yield, meat quality, and cecal microbiota of Ross 308 broiler chickens. A completely randomized design was employed; FOS was supplemented in the drinking water at concentrations of 0 %, 0.25 %, and 0.50 %. From 11 to 24 d of age, 0.25 % FOS supplementation significantly increased feed intake (FI), while feed cost per gain (FCG) was significantly reduced at 0.50 % FOS (P < 0.05). During the overall period (1-36 d of age), FOS supplementation significantly improved the European Production Efficiency Factor (EPEF) (P < 0.01), and slowed down the reduction in blood glucose levels after the re-feeding period (2, 3, 4, and 5 h) (P < 0.01). Furthermore, FOS supplementation decreased the heterophil/lymphocyte (H:L) ratio (P < 0.05). However, it had no significant effect on breast meat yield or abdominal fat, but 0.50 % FOS supplementation tended to increase the percentage of cecal weight (P = 0.08). Supplementation with FOS (0.25 % and 0.50 %) significantly reduced breast meat cooking loss (P < 0.05). Regarding cecal microbiota, the FOS-supplemented groups showed increased abundances of Lactobacillaceae and Acidaminococcaceae, whereas the abundances of Lachnospiraceae and Barnesiellaceae were reduced (P < 0.05). In conclusion, drinking water FOS supplementation had a beneficial effect on the overall productive performance and cooking loss of broiler chickens via stress reduction, which may involve an improvement in the gut microbiota.
Collapse
Affiliation(s)
- Siriporn Namted
- Department of Agriculture, Faculty of Agriculture Technology, Valaya Alongkorn Rajabhat University Under the Royal Patronage, Pathum Thani 13180, Thailand
| | | | - Chaiyapoom Bunchasak
- Department of Animal Science, Faculty of Agriculture, Kasetsart University, Bangkok, 10900, Thailand.
| |
Collapse
|
17
|
Hernández-García J, Muro-Reche P, Orenes-Piñero E. Gut microbiota and microRNAs as biomarkers in Parkinson's disease: early identification, diagnostic and potential treatments. Mol Cell Biochem 2025:10.1007/s11010-025-05271-6. [PMID: 40159518 DOI: 10.1007/s11010-025-05271-6] [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: 06/17/2024] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
The gut microbiota can affect both the enteric and the central nervous system, influencing individuals and their brain regulation. In this work, different pieces of scientific evidence are discussed, showing the relationship between changes in the microbiota and neurocognitive deterioration, focussing on Parkinson's disease (PD). Other factors that may cause or contribute to PD aetiology are the interactions between environmental factors and genetic susceptibility. According to the existing literature, there are several methods for the identification of neurocognitive impairment in different neurological diseases. However, such methods do not allow early identification, and therefore, the possibility of using other types of more effective diagnostic biomarkers in PD has also been investigated. Since this disease is characterised by specific microRNA (miRNA) expression, and the gut microbiota is an important factor in both PD and miRNA expression, the aim of this review is thoroughly analysing the role of microbiota and microRNAs in PD development. In addition, the relationship between these two factors and potential treatments will be also discussed.
Collapse
Affiliation(s)
- Javier Hernández-García
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Avda. de las Fuerzas Armadas, S/N, Lorca, 30800, Murcia, Spain
| | - Patricia Muro-Reche
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Avda. de las Fuerzas Armadas, S/N, Lorca, 30800, Murcia, Spain
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Avda. de las Fuerzas Armadas, S/N, Lorca, 30800, Murcia, Spain.
| |
Collapse
|
18
|
An JU, Mun SH, Kim WH, Seong JK, Park K, Cho S. Dynamics of the canine gut microbiota of a military dog birth cohort. Front Microbiol 2025; 16:1481567. [PMID: 40196028 PMCID: PMC11973337 DOI: 10.3389/fmicb.2025.1481567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction We systematically tracked early life stages in a military dog birth cohort to investigate canine gut microbiota dynamics related to environmental exposure during growth. This study utilized 16s rRNA amplicon sequencing-based analysis with molecular epidemiology of Enterococcus faecalis within a controlled environment at a military dog training center. Methods We examined shifts in gut microbiota diversity and taxonomic composition across four growth stages (lactation, weaning, starter, puppy) in three littermate groups. Additionally, E. faecalis dynamics was analyzed to confirm strain sharing among littermate groups. Results Gut microbiota changed rapidly during early growth, stabilizing at the puppy stage. This is supported by increased similarity in taxonomic composition among littermate groups, as they experienced an increased shared external environment and consumed the identical diets. E. faecalis strain sharing among littermate groups increased as dogs aged. Nine E. faecalis cluster types were identified; three specific types (type I, II, and IX) dominated in each littermate group during lactation. With greater exposure to the shared external environment, cluster type I gradually assumed dominance across all groups. Despite the dynamic shifts in microbiota, we found five genera within the core microbiota, Bacteroides, Peptoclostridium, Fusobacterium, Lactobacillus, and Blautia. Discussion This study is the first to explore the dynamic nature of early-life canine gut microbiota, illustrating its transition to stability and its resilience to environmental perturbations within the controlled training environment of a military dog birth cohort.
Collapse
Affiliation(s)
- Jae-Uk An
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Seung-Hyun Mun
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Woo-Hyun Kim
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, Republic of Korea
| | - Je Kyung Seong
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, Republic of Korea
| | - Kyoungwan Park
- Military Working Dogs Training Center, Gangwon, Republic of Korea
| | - Seongbeom Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, Republic of Korea
- Comparative Medicine Disease Research Center (CDRC), Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Krueger ME, Boles JS, Simon ZD, Alvarez SD, McFarland NR, Okun MS, Zimmermann EM, Forsmark CE, Tansey MG. Comparative analysis of Parkinson's and inflammatory bowel disease gut microbiomes reveals shared butyrate-producing bacteria depletion. NPJ Parkinsons Dis 2025; 11:50. [PMID: 40108151 PMCID: PMC11923181 DOI: 10.1038/s41531-025-00894-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Epidemiological studies reveal that inflammatory bowel disease (IBD) is associated with an increased risk of Parkinson's disease (PD). Gut dysbiosis has been documented in both PD and IBD, however it is currently unknown whether gut dysbiosis underlies the epidemiological association between both diseases. To identify shared and distinct features of the PD and IBD microbiome, we recruited 54 PD, 26 IBD, and 16 healthy control individuals and performed the first joint analysis of gut metagenomes. Larger, publicly available PD and IBD metagenomic datasets were also analyzed to validate and extend our findings. Depletions in short-chain fatty acid (SCFA)-producing bacteria, including Roseburia intestinalis, Faecalibacterium prausnitzii, Anaerostipes hadrus, and Eubacterium rectale, as well depletion in SCFA-synthesis pathways were detected across PD and IBD datasets, suggesting that depletion of these microbes in IBD may influence the risk for PD development.
Collapse
Affiliation(s)
- Maeve E Krueger
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jake Sondag Boles
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Zachary D Simon
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Stephan D Alvarez
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Nikolaus R McFarland
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Michael S Okun
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ellen M Zimmermann
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christopher E Forsmark
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
20
|
Cheng YK, Chiang HS. The interrelationship between intestinal immune cells and enteric α-synuclein in the progression of Parkinson's disease. Neurol Sci 2025:10.1007/s10072-025-08114-w. [PMID: 40085320 DOI: 10.1007/s10072-025-08114-w] [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: 01/29/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder primarily characterized by motor impairment, resulting from the accumulation of α-synuclein and neuronal cell death in the substantia nigra of the midbrain. Emerging evidence suggests that α-synuclein aggregation may originate in the enteric nervous system (ENS) and subsequently propagate to the brain via the vagus nerve. Clinical observations, such as prodromal gastrointestinal dysfunction in PD patients and the increased incidence of PD among individuals with inflammatory bowel disease, support the hypothesis that abnormal intestinal inflammation may contribute to the onset of motor dysfunction and neuropathology in PD. This review examines recent findings on the interplay between intestinal immune cells and α-synuclein aggregation within the framework of gut-originated PD pathogenesis. It begins by discussing evidence linking dysbiosis and intestinal inflammation to α-synuclein aggregation in the ENS. Additionally, it explores the potential role of intestinal immune cells in influencing enteric neurons and α-synuclein aggregation, furthering the understanding of PD development.
Collapse
Affiliation(s)
- Yuan-Kai Cheng
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hao-Sen Chiang
- Department of Life Science, National Taiwan University, Taipei, Taiwan.
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
21
|
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
|
22
|
Safarchi A, Al-Qadami G, Tran CD, Conlon M. Understanding dysbiosis and resilience in the human gut microbiome: biomarkers, interventions, and challenges. Front Microbiol 2025; 16:1559521. [PMID: 40104586 PMCID: PMC11913848 DOI: 10.3389/fmicb.2025.1559521] [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: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
The healthy gut microbiome is important in maintaining health and preventing various chronic and metabolic diseases through interactions with the host via different gut-organ axes, such as the gut-brain, gut-liver, gut-immune, and gut-lung axes. The human gut microbiome is relatively stable, yet can be influenced by numerous factors, such as diet, infections, chronic diseases, and medications which may disrupt its composition and function. Therefore, microbial resilience is suggested as one of the key characteristics of a healthy gut microbiome in humans. However, our understanding of its definition and indicators remains unclear due to insufficient experimental data. Here, we review the impact of key drivers including intrinsic and extrinsic factors such as diet and antibiotics on the human gut microbiome. Additionally, we discuss the concept of a resilient gut microbiome and highlight potential biomarkers including diversity indices and some bacterial taxa as recovery-associated bacteria, resistance genes, antimicrobial peptides, and functional flexibility. These biomarkers can facilitate the identification and prediction of healthy and resilient microbiomes, particularly in precision medicine, through diagnostic tools or machine learning approaches especially after antimicrobial medications that may cause stable dysbiosis. Furthermore, we review current nutrition intervention strategies to maximize microbial resilience, the challenges in investigating microbiome resilience, and future directions in this field of research.
Collapse
Affiliation(s)
- Azadeh Safarchi
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Ghanyah Al-Qadami
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Cuong D Tran
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Michael Conlon
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| |
Collapse
|
23
|
Liu H, Wang S, Yu X, Sun S, Su Y. Effect of Resveratrol on the intestinal microbiota in Type2 diabetes Mellitus mice. Braz J Microbiol 2025; 56:447-463. [PMID: 39621295 PMCID: PMC11885759 DOI: 10.1007/s42770-024-01573-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 11/04/2024] [Indexed: 03/09/2025] Open
Abstract
Some natural products have been found to have a positive effect on the treatment of Type 2 Diabetes Mellitus (T2DM). The effective mechanism by which these natural products work may be attributed to their ability to alter the gut microbiome. In this study, considering the beneficial properties of resveratrol for T2DM, the mechanism of resveratrol in releasing of T2DM, the correlation between the apparent traits of resveratrol in T2DM mice and the changes in intestinal flora structure were explored. The intervention of resveratrol in different dose did effectively improve the apparent T2DM symptoms in mice, including reduced fasting blood glucose, enhance to both the insulin tolerance and glucose tolerance. Simultaneously, resveratrol can effectively ameliorate the pathological condition of the colon and rectum in T2DM mice, leading to a reduction in intestinal villi swelling and fat vacuoles. The intervention of resveratrol led to changes in the gut flora structure of T2DM mice. Erysipelas and Ileibacterium are crucial taxa of resveratrol involved in maintaining the balance of the gut microbiota. The abundance of Erysipelotrichaceae and Ileibacterium was significantly elevated under the treatment of resveratrol, and the compose of the gut microbiota was more inclined to healthy mice. T2DM symptoms could be alleviated by resveratrol through different mechanisms compared to metformin intervention.
Collapse
Affiliation(s)
- Hao Liu
- Medical College, Kunming University, Kunming, 650214, Yunnan, P.R. China
| | - Shumin Wang
- Medical College, Kunming University, Kunming, 650214, Yunnan, P.R. China
| | - Xin Yu
- School of Basic Medicine, School of Clinical Medicine, Dali University, Dali, 671000, China
| | - Shuguang Sun
- Department of Endocrinology, School of Clinical Medicine, Dali University, Dali, 671000, China.
| | - Yingzhen Su
- Medical College, Kunming University, Kunming, 650214, Yunnan, P.R. China.
| |
Collapse
|
24
|
Almheiri RT, Hajjar B, Alkhaaldi SMI, Rabeh N, Aljoudi S, Abd-Elrahman KS, Hamdan H. Beyond weight loss: exploring the neurological ramifications of altered gut microbiota post-bariatric surgery. J Transl Med 2025; 23:223. [PMID: 39994634 PMCID: PMC11852891 DOI: 10.1186/s12967-025-06201-2] [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: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
This review discusses findings related to neurological disorders, gut microbiota, and bariatric surgery, focusing on neurotransmitters, neuroendocrine, the pathophysiology of bacteria contributing to disorders, and possible therapeutic interventions. Research on neurotransmitters suggests that their levels are heavily influenced by gut microbiota, which may link them to neurological disorders such as Alzheimer's disease, Parkinson's disease, Multiple sclerosis, Depression, and Autism spectrum disorder. The pathophysiology of bacteria that reach and influence the central nervous system has been documented. Trends in microbiota are often observed in specific neurological disorders, with a prominence of pro-inflammatory bacteria and a reduction in anti-inflammatory types. Furthermore, bariatric surgery has been shown to alter microbiota profiles similar to those observed in neurological disorders. Therapeutic interventions, including fecal microbiota transplants and probiotics, have shown potential to alleviate neurological symptoms. We suggest a framework for future studies that integrates knowledge from diverse research areas, employs rigorous methodologies, and includes long-trial clinical control groups.
Collapse
Affiliation(s)
- Rashed T Almheiri
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Baraa Hajjar
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Saif M I Alkhaaldi
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Nadia Rabeh
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Sara Aljoudi
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Khaled S Abd-Elrahman
- Department of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Department of Medical Sciences, College of Medicine and Health Science, Khalifa University, 127788, Abu Dhabi, United Arab Emirates.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Hamdan Hamdan
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates.
- Healthcare Engineering Innovation Group (HEIG), Khalifa University of Science and Technology, 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
25
|
Rout S, Dash R, Satish V, Venugopal G, Rao BN, Bandhyopadhyay D, Bhoi SK, Ramadass B. Exploring the role of acylated ghrelin and gut microbiome in delineating cognitive health in the elderly. Aging (Albany NY) 2025; 17:393-407. [PMID: 39927883 PMCID: PMC11892914 DOI: 10.18632/aging.206200] [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: 09/10/2024] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION With increased life expectancy, there is an increase in aging population and prevalence of dementia. Ghrelin is a key regulator of spatial memory and cognition. The gut microbiome may affect the circulating levels of unacylated ghrelin (UAG) and acylated ghrelin (AG). Thus, we explore the potential association of the gut microbiome, AG, and cognitive health in the aging dementia patient. METHODS 40 dementia patients and 40 controls were recruited. Fecal Microbiome analysis using 16S rRNA sequencing was performed on 18 samples. A mixed-method approach was employed for robust interpretation. RESULTS Dementia patients had an increased serum AG and AG/UAG ratio. With the increase in AG among dementia subjects, a significant decrease in species richness was observed. Bifidobacterium longum, Eubacterium biforme, Fecalibacterium prausnitzii, Lactobacillus ruminis, and Prevotella copri contributed to substantial differences in beta-diversity. Blautia obeum was associated with Mini-Mental State Examination (MMSE), and Fecalibacterium prausnitzii was associated with Montreal Cognitive Assessment (MoCA) Scale. DISCUSSION This pilot study indicates a complex interaction between AG, gut microbiome, and cognitive scores. Increased AG corresponds to both dementia and gut dysbiosis, intricately interconnecting the gut-brain axis. The circulating AG and associated gut microbiome might be a putative biomarker for dementia.
Collapse
Affiliation(s)
- Sudeshna Rout
- Department of Biochemistry, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Rishikesh Dash
- Centre of Excellence for Clinical Microbiome and Research (CCMR), All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Varsha Satish
- Centre of Excellence for Clinical Microbiome and Research (CCMR), All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Giriprasad Venugopal
- Centre of Excellence for Clinical Microbiome and Research (CCMR), All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Bodepudi Narasimha Rao
- Department of Physiology, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Debapriya Bandhyopadhyay
- Department of Biochemistry, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Sanjeev Kumar Bhoi
- Department of Neurology, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
| | - Balamurugan Ramadass
- Department of Biochemistry, All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
- Centre of Excellence for Clinical Microbiome and Research (CCMR), All India Institute of Medical Sciences Bhubaneswar, Odisha 751019, India
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
26
|
Wang L, Cui Y, Han B, Du Y, Salewala KS, Wang S, Zhao W, Zhang H, Wang S, Xu X, Ma J, Zhu Y, Tuo H. Gut microbiota and Parkinson's disease. Chin Med J (Engl) 2025; 138:289-297. [PMID: 39501822 PMCID: PMC11771718 DOI: 10.1097/cm9.0000000000003318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Indexed: 01/29/2025] Open
Abstract
ABSTRACT Emerging evidence suggests that dysbiosis of the gut microbiota is associated with the pathogenesis of Parkinson's disease (PD), a prevalent neurodegenerative disorder. The microbiota-gut-brain axis plays a crucial role in the development and progression of PD, and numerous studies have demonstrated the potential therapeutic benefits of modulations in the intestinal microbiota. This review provides insights into the characterization of the gut microbiota in patients with PD and highlights associations with clinical symptoms and underlying mechanisms. The discussion underscores the increased influence of the gut microbiota in the pathogenesis of PD. While the relationship is not fully elucidated, existing research demonstrates a strong correlation between changes in the composition of gut microbiota and disease development, and further investigation is warranted to explain the specific underlying mechanisms.
Collapse
Affiliation(s)
- Lin Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ying Cui
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bingyu Han
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yitong Du
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | | | - Shiya Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wenlu Zhao
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Hongxin Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Sichen Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xinran Xu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jianpeng Ma
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yan Zhu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Houzhen Tuo
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
27
|
Gorecki AM, Anyaegbu CC, Fitzgerald M, Fuller KA, Anderton RS. Imaging flow cytometry reveals LPS-induced changes to intracellular intensity and distribution of α-synuclein in a TLR4-dependent manner in STC-1 cells. Methods 2025; 234:93-111. [PMID: 39486562 DOI: 10.1016/j.ymeth.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Parkinson's disease is a chronic neurodegenerative disorder, where pathological protein aggregates largely composed of phosphorylated α-synuclein are implicated in disease pathogenesis and progression. Emerging evidence suggests that the interaction between pro-inflammatory microbial factors and the gut epithelium contributes to α-synuclein aggregation in the enteric nervous system. However, the cellular sources and mechanisms for α-synuclein pathology in the gut are still unclear. METHODS The STC-1 cell line, which models an enteroendocrine population capable of communicating with the gut microbiota, immune and nervous systems, was treated with a TLR4 inhibitor (TAK-242) prior to microbial lipopolysaccharide (LPS) exposure to investigate the role of TLR4 signalling in α-synuclein alterations. Antibodies targeting the full-length protein (α-synuclein) and the Serine-129 phosphorylated form (pS129) were used. Complex, multi-parametric image analysis was conducted through confocal microscopy (with Zen 3.8 analysis) and imaging flow cytometry (with IDEAS® analysis). RESULTS Confocal microscopy revealed heterogenous distribution of α-synuclein and pS129 in STC-1 cells, with prominent pS129 staining along cytoplasmic processes. Imaging flow cytometry further quantified the relationship between various α-synuclein morphometric features. Thereafter, imaging flow cytometry demonstrated a dose-specific effect of LPS, where the low (8 μg/mL), but not high dose (32 μg/mL), significantly altered measures related to α-synuclein intensity, distribution, and localisation. Pre-treatment with a TLR4 inhibitor TAK-242 alleviated some of these significant alterations. CONCLUSION This study demonstrates that LPS-TLR4 signalling alters the intracellular localisation of α-synuclein in enteroendocrine cells in vitro and showcases the utility of combining imaging flow cytometry to investigate subtle protein changes that may not be apparent through confocal microscopy alone. Further investigation is required to understand the apparent dose-dependent effects of LPS on α-synuclein in the gut epithelium in healthy states as well as conditions such as Parkinson's disease.
Collapse
Affiliation(s)
- Anastazja M Gorecki
- School of Health Sciences, University of Notre Dame Australia, Fremantle, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia; Perron Institute for Neurological and Translational Science, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia; School of Biological Sciences, University of Western Australia, Crawley, WA, Australia.
| | - Chidozie C Anyaegbu
- Curtin Health Innovation Research Institute, Curtin University, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia; Perron Institute for Neurological and Translational Science, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
| | - Melinda Fitzgerald
- Curtin Health Innovation Research Institute, Curtin University, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia; Perron Institute for Neurological and Translational Science, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
| | - Kathryn A Fuller
- Translational Cancer Pathology Laboratory, School of Biomedical Sciences (M504), The University of Western Australia, Crawley, WA, Australia
| | - Ryan S Anderton
- School of Health Sciences, University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
28
|
Sampson TR, Tansey MG, West AB, Liddle RA. Lewy body diseases and the gut. Mol Neurodegener 2025; 20:14. [PMID: 39885558 PMCID: PMC11783828 DOI: 10.1186/s13024-025-00804-5] [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: 10/17/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
Gastrointestinal (GI) involvement in Lewy body diseases (LBDs) has been observed since the initial descriptions of patients by James Parkinson. Recent experimental and human observational studies raise the possibility that pathogenic alpha-synuclein (⍺-syn) might develop in the GI tract and subsequently spread to susceptible brain regions. The cellular and mechanistic origins of ⍺-syn propagation in disease are under intense investigation. Experimental LBD models have implicated important contributions from the intrinsic gut microbiome, the intestinal immune system, and environmental toxicants, acting as triggers and modifiers to GI pathologies. Here, we review the primary clinical observations that link GI dysfunctions to LBDs. We first provide an overview of GI anatomy and the cellular repertoire relevant for disease, with a focus on luminal-sensing cells of the intestinal epithelium including enteroendocrine cells that express ⍺-syn and make direct contact with nerves. We describe interactions within the GI tract with resident microbes and exogenous toxicants, and how these may directly contribute to ⍺-syn pathology along with related metabolic and immunological responses. Finally, critical knowledge gaps in the field are highlighted, focusing on pivotal questions that remain some 200 years after the first descriptions of GI tract dysfunction in LBDs. We predict that a better understanding of how pathophysiologies in the gut influence disease risk and progression will accelerate discoveries that will lead to a deeper overall mechanistic understanding of disease and potential therapeutic strategies targeting the gut-brain axis to delay, arrest, or prevent disease progression.
Collapse
Affiliation(s)
- Timothy R Sampson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30329, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Malú Gámez Tansey
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
- Normal Fixel Institute of Neurological Diseases, Gainesville, FL, 32608, USA
| | - Andrew B West
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Duke Center for Neurodegeneration and Neurotherapeutic Research, Department of Pharmacology and Cancer Biology, Durham, NC, 27710, USA.
| | - Rodger A Liddle
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Duke Institute for Brain Sciences, Duke University, Durham, NC, 27710, USA.
- Department of Medicine, Duke University and Department of Veterans Affairs Health Care System, Durham, NC, 27710, USA.
| |
Collapse
|
29
|
Liu S, Tian M, Ma M, Qiu Y, Tang J, Hou J, Lu Q, Tian C, Ye G, Wang L, Gao K, Guo S, Jiang Z, Yang X. Effect of Gardeniae Fructus Powder on Growth Performance, Antioxidant Capacity, Intestinal Barrier Function, and Colonic Microbiota of Weaned Piglets. Animals (Basel) 2025; 15:221. [PMID: 39858221 PMCID: PMC11758313 DOI: 10.3390/ani15020221] [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: 12/10/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The present study aimed to explore the effect of GF powder on the growth performance, diarrhea rate, antioxidant and immune capacity, and intestinal health of weaned piglets. A total of 144 weaned piglets (8.29 ± 0.11 kg) at 21 d old were randomly assigned to four groups, with each treatment consisting of six replicate pens, with six piglets per pen, and each pen containing three barrows and three gilts. The piglets were fed a basal diet supplement with 0%, 0.4%, 0.6%, and 0.8% GF powder (n = 36). Our results indicated that compared with the basal diet, the F/G and diarrhea rate were remarkably decreased in the 0.8% GF group (p < 0.05). Serum biochemical parameters showed that supplementation with GF significantly increased the content of HDL-C (0.6 and 0.8% levels), IL-6 (0.8% level), IL-10 (0.4, 0.6, and 0.8% levels), Ig G (0.4% level), and Ig A (0.8% level) compared with the basal diet (p < 0.05). The index of antioxidant capacity showed that compared with a basal diet, supplementation with GF significantly decreased serum MDA content (0.4% and 0.8% levels) and jejunal and ileal MDA content (0.4%, 0.6%, and 0.8% levels) (p < 0.05). Additionally, compared with the basal diet, supplementation with GF significantly increased serum and ileal T-AOC content (0.4%, 0.6%, and 0.8% levels), serum T-SOD content (0.4% and 0.8% levels), ileal T-SOD content (0.4%, 0.6%, and 0.8% levels), CAT content (0.4%, 0.6%, and 0.8% levels), and jejunal GSH-Px content (0.8% level) (p < 0.05). The results of gene expression indicate that compared with the basal diet, supplementation with GF significantly increased Nrf 2 (0.4% level), NQO (0.4% level), SOD 1 (0.4% and 0.8% levels), and GCLC (0.4% level) and GCLM (0.8% level) abundance in jejunal mucosa; supplementation with GF significantly increased Nrf 2 (0.4%, 0.6%, and 0.8% levels), HO-1 (0.4% level), NQO (0.8% level), SOD 1 (0.4% and 0.8% levels), and GCLC (0.4% level) and GCLM (0.8% level) abundance in ileal mucosa (p < 0.05). Ulteriorly, the present results indicate that supplementation with GF at the 0.8% level significantly increased the villus height in the jejunum and ileum as well as the villus/crypt ratio in the ileum compared with the basal diet (p < 0.05). Compared with the basal diet, 0.4% GF significantly increased Occludin gene expression in ileal mucosa (p < 0.05), 0.6% GF significantly increased ZO-1, Claudin-1, and Occludin gene expression in jejunal mucosa (p < 0.05), and 0.8% GF significantly increased ZO-1 and Occludin gene expression in jejunal mucosa along with Occludin expression in ileal mucosa (p < 0.05). Furthermore, colonic microbiota composition showed that Shannon, observed species, and Chao 1 indices were significantly increased in the 0.8% GF group compared with the basal diet (p < 0.05). At the phylum level, in comparison with the basal diet, the relative abundance of Firmicutes significantly decreased in the 0.4%, 0.6%, and 0.8% GF groups, and Bacteroidetes increased in the 0.8% GF group (p < 0.05). At the genus level, compared with the basal diet, 0.6% and 0.8% GF significantly increased Prevotella abundance, and 0.6% GF significantly decreased Coprococcus abundance (p < 0.05). At the species level, compared with the basal diet, 0.8% GF significantly increased Prevotella copri abundance, and 0.4%, 0.6%, and 0.8% GF significantly decreased Blautia obeum abundance (p < 0.05). In summary, a dietary supplement with 0.8% Gardeniae Fructus powder significantly decreased the F/G and diarrhea rate and improved antioxidant capacity and intestinal barrier function, which may be associated with the improvement of the relative abundance of Prevotella copri. These findings indicate that Gardeniae Fructus powder may be used as a feed additive in swine weaning.
Collapse
Affiliation(s)
- Shilong Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (M.M.)
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Min Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Ming Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (M.M.)
| | - Yueqin Qiu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Jiaxi Tang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Jing Hou
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Qi Lu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Chaoyang Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Guohao Ye
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Li Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Kaiguo Gao
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Shining Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (S.L.); (M.M.)
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Xuefen Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (M.T.); (Y.Q.); (J.T.); (J.H.); (Q.L.); (C.T.); (G.Y.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| |
Collapse
|
30
|
Xia L, Li C, Zhao J, Sun Q, Mao X. Rebalancing immune homeostasis in combating disease: The impact of medicine food homology plants and gut microbiome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156150. [PMID: 39740376 DOI: 10.1016/j.phymed.2024.156150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Gut microbiota plays an important role in multiple human physiological processes and an imbalance in it, including the species, abundance, and metabolites can lead to diseases. These enteric microorganisms modulate immune homeostasis by presenting a myriad of antigenic determinants and microbial metabolites. Medicinal and food homologous (MFH) plants, edible herbal materials for both medicine and food, are important parts of Traditional Chinese Medicine (TCM). MFH plants have drawn much attention due to their strong biological activity and low toxicity. However, the interplay of MFH and gut microbiota in rebalancing the immune homeostasis in combating diseases needs systematic illumination. PURPOSE The review discusses the interaction between MFH and gut microbiota, including the effect of MFH on the major group of gut microbiota and the metabolic effect of gut microbiota on MFH. Moreover, how gut microbiota influences the immune system in terms of innate and adaptive immunity is addressed. Finally, the immunoregulatory mechanisms of MFH in regulation of host pathophysiology via gut microbiota are summarized. METHODS Literature was searched, analyzed, and collected using databases, including PubMed, Web of Science, and Google Scholar using relevant keywords. The obtained articles were screened and summarized by the research content of MFH and gut microbiota in immune regulation. RESULTS The review demonstrates the interaction between MFH and gut microbiota in disease prevention and treatment. Not only do the intestinal microorganisms and intestinal mucosa constitute an important immune barrier of the human body, but also lymphoid tissue and diffused immune cells within the mucosa participate in the response of innate immunity and adaptive immunity. MFH modulates immune regulation by affecting intestinal flora, helps maintain the balance of the immune system and interfere with the occurrence and development of a broad category of diseases. CONCLUSION Being absorbed from the gastrointestinal tract, MFH can have profound effects on gut microbiota. In turn, the gut microbiota also actively participate in the bioconversion of complex constituents from MFH, which could further influence their physiological and pharmacological properties. The review deepens the understanding of the relationship among MFH, gut microbiota, immune system, and human diseases and further promotes the progression of additional relevant research.
Collapse
Affiliation(s)
- Lu Xia
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Chuangen Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Jia Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, China
| | - Quancai Sun
- Department of Health, Nutrition, and Food sciences, Florida State University, Tallahassee, USA
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
31
|
Feng M, Zou Z, Shou P, Peng W, Liu M, Li X. Gut microbiota and Parkinson's disease: potential links and the role of fecal microbiota transplantation. Front Aging Neurosci 2024; 16:1479343. [PMID: 39679259 PMCID: PMC11638248 DOI: 10.3389/fnagi.2024.1479343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide and seriously affects the quality of life of elderly patients. PD is characterized by the loss of dopaminergic neurons in the substantia nigra as well as abnormal accumulation of α-synuclein in neurons. Recent research has deepened our understanding of the gut microbiota, revealing that it participates in the pathological process of PD through the gut-brain axis, suggesting that the gut may be the source of PD. Therefore, studying the relationship between gut microbiota and PD is crucial for improving our understanding of the disease's prevention, diagnosis, and treatment. In this review, we first describe the bidirectional regulation of the gut-brain axis by the gut microbiota and the mechanisms underlying the involvement of gut microbiota and their metabolites in PD. We then summarize the different species of gut microbiota found in patients with PD and their correlations with clinical symptoms. Finally, we review the most comprehensive animal and human studies on treating PD through fecal microbiota transplantation (FMT), discussing the challenges and considerations associated with this treatment approach.
Collapse
Affiliation(s)
- Maosen Feng
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Zhiyan Zou
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Pingping Shou
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Wei Peng
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Mingxue Liu
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Xiaoan Li
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
32
|
Ahmed HS. The Impact of Prevotella on Neurobiology in Aging: Deciphering Dendritic Cell Activity and Inflammatory Dynamics. Mol Neurobiol 2024; 61:9240-9251. [PMID: 38613648 DOI: 10.1007/s12035-024-04156-x] [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: 12/22/2023] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Prevotella species, notably Prevotella copri, significantly populate the human gut. In particular, P. copri is prevalent among non-Western populations with diets high in fiber. These species show complex relationships with diverse health aspects, associating with beneficial outcomes, including reduced visceral fat and improved glucose tolerance. Studies implicate various Prevotella species in specific diseases. Prevotella nigrescens and Porphyromonas gingivalis were linked to periodontal disease, promoting immune responses and influencing T helper type 17 (Th17) cells. Prevotella bivia was associated with bacterial vaginosis and a specific increase in activated cells in the vaginal mucosa. In contrast, they have shown substantial potential for inducing connective tissue degradation and alveolar bone resorption. Prevotella's role in neuroinflammatory disorders and autoinflammatory conditions such as Alzheimer's disease and Parkinson's disease has also been noted. The complex relationship between Prevotella and age-related conditions further extends to neurobiological changes in aging, with varying associations with Alzheimer's, Parkinson's, and other inflammatory conditions. Studies have also identified Prevotella to be implicated in cognitive decline in middle aged and the elderly. Future directions in this research area are anticipated to explore Prevotella-associated inflammatory mechanisms and therapeutic interventions. Investigating specific drug targets and immunomodulatory measures could lead to novel therapeutic strategies. Understanding how Prevotella-induced inflammation interacts with aging diseases would offer promising insights for treatments and interventions. This review urges ongoing research to discover therapeutic targets and mechanisms for moderating Prevotella-associated inflammation to further enhance our understanding and improve health outcomes.
Collapse
Affiliation(s)
- H Shafeeq Ahmed
- Bangalore Medical College and Research Institute, K.R Road, Bangalore, 560002, Karnataka, India.
| |
Collapse
|
33
|
Morais LH, Boktor JC, MahmoudianDehkordi S, Kaddurah-Daouk R, Mazmanian SK. α-synuclein overexpression and the microbiome shape the gut and brain metabolome in mice. NPJ Parkinsons Dis 2024; 10:208. [PMID: 39477976 PMCID: PMC11525669 DOI: 10.1038/s41531-024-00816-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Pathological forms of α-synuclein contribute to synucleinopathies, including Parkinson's disease (PD). Most cases of PD arise from gene-environment interactions. Microbiome composition is altered in PD, and gut bacteria are causal to symptoms in animal models. We quantitatively profiled nearly 630 metabolites in the gut, plasma, and brain of α-synuclein-overexpressing (ASO) mice, compared to wild-type (WT) animals, and comparing germ-free (GF) to specific pathogen-free (SPF) animals (n = 5 WT-SPF; n = 6 ASO-SPF; n = 6 WT-GF; n = 6 ASO-GF). Many differentially expressed metabolites in ASO mice are also dysregulated in human PD patients, including amine oxides, bile acids and indoles. The microbial metabolite trimethylamine N-oxide (TMAO) strongly correlates from the gut to the plasma to the brain in mice, notable since TMAO is elevated in the blood and cerebrospinal fluid of PD patients. These findings uncover broad metabolomic changes that are influenced by the intersection of host genetics and microbiome in a mouse model of PD.
Collapse
Affiliation(s)
- Livia H Morais
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Joseph C Boktor
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | | | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA.
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
| | - Sarkis K Mazmanian
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
34
|
Metcalfe-Roach A, Cirstea MS, Yu AC, Ramay HR, Coker O, Boroomand S, Kharazyan F, Martino D, Sycuro LK, Appel-Cresswell S, Finlay BB. Metagenomic Analysis Reveals Large-Scale Disruptions of the Gut Microbiome in Parkinson's Disease. Mov Disord 2024; 39:1740-1751. [PMID: 39192744 DOI: 10.1002/mds.29959] [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: 03/29/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) has been consistently linked to alterations within the gut microbiome. OBJECTIVE Our goal was to identify microbial features associated with PD incidence and progression. METHODS Metagenomic sequencing was used to characterize taxonomic and functional changes to the PD microbiome and to explore their relation to bacterial metabolites and disease progression. Motor and non-motor symptoms were tracked using Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS) and levodopa equivalent dose across ≤5 yearly study visits. Stool samples were collected at baseline for metagenomic sequencing (176 PD, 100 controls). RESULTS PD-derived stool samples had reduced intermicrobial connectivity and seven differentially abundant species compared to controls. A suite of bacterial functions differed between PD and controls, including depletion of carbohydrate degradation pathways and enrichment of ribosomal genes. Faecalibacterium prausnitzii-specific reads contributed significantly to more than half of all differentially abundant functional terms. A subset of disease-associated functional terms correlated with faster progression of MDS-UPDRS part IV and separated those with slow and fast progression with moderate accuracy within a random forest model (area under curve = 0.70). Most PD-associated microbial trends were stronger in those with symmetric motor symptoms. CONCLUSION We provide further evidence that the PD microbiome is characterized by reduced intermicrobial communication and a shift to proteolytic metabolism in lieu of short-chain fatty acid production, and suggest that these microbial alterations may be relevant to disease progression. We also describe how our results support the existence of gut-first versus brain-first PD subtypes. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Avril Metcalfe-Roach
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mihai S Cirstea
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Adam C Yu
- Pacific Parkinson's Research Centre, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hena R Ramay
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Olabisi Coker
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Seti Boroomand
- Borgland Family Brain Tissue and DNA Bank, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Faezeh Kharazyan
- Borgland Family Brain Tissue and DNA Bank, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Davide Martino
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Laura K Sycuro
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Silke Appel-Cresswell
- Pacific Parkinson's Research Centre, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Neurology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - B Brett Finlay
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
35
|
Zhang M, Gan X, Fang Y, Song X, Li Q, Huang B. Intestinal flora influences the progression of subarachnoid hemorrhage by affecting peripheral and central inflammatory pathways. Brain Res 2024; 1840:149032. [PMID: 38806092 DOI: 10.1016/j.brainres.2024.149032] [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: 03/20/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
Subarachnoid hemorrhage (SAH) is a debilitating condition that leaves survivors with neurological disability for the rest of their lives. No effective treatment for early brain injury (EBI) has been developed. Gut microbiome (GM) impact the host immune system and can influence disease processes in several organs, including the brain. However, it remains unclear whether the GM has an impact on the outcome of SAH brain injury. Here, we wondered whether microbiota could relieve the injury. We changed the microbiota of 8-week-old male rats by administering antibiotic-containing water for 2 weeks. Composition of the GM was profiled by using 16S rRNA. We induced SAH by puncture the internal carotid artery of control rats and rats with altered GM. Additionally, we studied inflammatory cells using HE stains, Intestinal lymphocyte flow cytometry, and Neuroinflammatory factor WB. SAH was significantly averted by alterations in GM using antibiotics. The altered GM significantly increased the intestinal and intracranial inflammation after SAH. This was manifested by Mosin (MSN) inflammatory cytokines. Our findings demonstrated that the brain injury following SAH is associated with GM.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, Jiangsu, China
| | - Xiaokui Gan
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, Jiangsu, China
| | - Yiming Fang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, Jiangsu, China
| | - Xiaowei Song
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, Jiangsu, China
| | - Qingquan Li
- Department of Neurosurgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, China
| | - Baosheng Huang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, Jiangsu, China.
| |
Collapse
|
36
|
Pal G, Bennett L, Roy J, Nyandege A, Mouradian MM, Gerhard T, Horton DB. Effects of antimicrobial exposure on the risk of Parkinson's disease. Parkinsonism Relat Disord 2024; 127:107081. [PMID: 39098264 DOI: 10.1016/j.parkreldis.2024.107081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/07/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND We aimed to assess how antimicrobial exposure affects Parkinson's disease (PD) risk. METHODS A nested case-control study was performed to examine the association between antimicrobial exposure and newly diagnosed PD using the Clinical Practice Research Datalink (CPRD). Each PD case was matched by age, sex, and year of diagnosis (index date) to up to 15 controls. Number of prescribed antimicrobial courses was assessed 1-5, 6-10, and 11-15 years prior to the index date. Logistic regression with generalized estimating equations (GEE) was used to estimate odds ratios (ORs) and false discovery rate-adjusted p-values between antimicrobial exposure and risk of PD. RESULTS We compared 12,557 PD cases with 80,804 matched controls. We found an inverse dose-response relationship between number of penicillin courses and PD risk across multiple time periods (5+ courses, 1-5 years prior: OR 0.85, 95 % CI 0.76-0.95, p = 0.043; 6-10 years prior: OR 0.84, 95 % CI: 0.73-0.95, p = 0.059; 11-15 years prior: OR 0.87, 95 % CI 0.74-1.02, p = 0.291). The number of macrolide courses was inversely but not significantly associated with PD risk 1-5 years prior to the index date (OR 0.89-0.91, 95 % CI: 0.79-0.99, adjusted p = 0.140-0.167). Exposure to ≥2 courses of antifungals 1-5 years prior was associated with an increased risk of PD (OR 1.16, 95 % CI: 1.06-1.27, p = 0.020). CONCLUSIONS In a large UK-representative population, the risk of PD was modestly lower among adults who had previously received multiple courses of penicillins in the last 15 years and modestly higher among those exposed to antifungal medicines in recent years.
Collapse
Affiliation(s)
- Gian Pal
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| | - Laura Bennett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA
| | - Jason Roy
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA; New Jersey Alliance for Clinical and Translational Science, New Brunswick, NJ, USA
| | - Abner Nyandege
- Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, USA
| | - M Maral Mouradian
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Robert Wood Johnson Medical School Institute for Neurological Therapeutics, Piscataway, NJ, USA
| | - Tobias Gerhard
- Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, USA; Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Piscataway, NJ, USA
| | - Daniel B Horton
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA; Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, USA; Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
37
|
Saadh MJ, Mustafa AN, Mustafa MA, S RJ, Dabis HK, Prasad GVS, Mohammad IJ, Adnan A, Idan AH. The role of gut-derived short-chain fatty acids in Parkinson's disease. Neurogenetics 2024; 25:307-336. [PMID: 39266892 DOI: 10.1007/s10048-024-00779-3] [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: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 09/14/2024]
Abstract
The emerging function of short-chain fatty acids (SCFAs) in Parkinson's disease (PD) has been investigated in this article. SCFAs, which are generated via the fermentation of dietary fiber by gut microbiota, have been associated with dysfunction of the gut-brain axis and, neuroinflammation. These processes are integral to the development of PD. This article examines the potential therapeutic implications of SCFAs in the management of PD, encompassing their capacity to modulate gastrointestinal permeability, neuroinflammation, and neuronal survival, by conducting an extensive literature review. As a whole, this article emphasizes the potential therapeutic utility of SCFAs as targets for the management and treatment of PD.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | - Mohammed Ahmed Mustafa
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Renuka Jyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | | | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra, Pradesh-531162, India
| | - Imad Jassim Mohammad
- College of Health and Medical Technology, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Ahmed Adnan
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
38
|
Walker AC, Bhargava R, Bucher MJ, Argote YM, Brust AS, Czyż DM. Identification of proteotoxic and proteoprotective bacteria that non-specifically affect proteins associated with neurodegenerative diseases. iScience 2024; 27:110828. [PMID: 39310761 PMCID: PMC11414702 DOI: 10.1016/j.isci.2024.110828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/05/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
There are no cures for neurodegenerative protein conformational diseases (PCDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Emerging evidence suggests the gut microbiota plays a role in their pathogenesis, though the influences of specific bacteria on disease-associated proteins remain elusive. Here, we reveal the effects of 229 human bacterial isolates on the aggregation and toxicity of Aβ1-42, α-synuclein, and polyglutamine tracts in Caenorhabditis elegans expressing these culprit proteins. Our findings demonstrate that bacterial effects on host protein aggregation are consistent across different culprit proteins, suggesting that microbes affect protein stability by modulating host proteostasis rather than selectively targeting disease-associated proteins. Furthermore, we found that feeding C. elegans proteoprotective Prevotella corporis activates the heat shock response, revealing an unexpected discovery of a microbial influence on host proteostasis. Insight into how individual bacteria affect PCD proteins could open new strategies for prevention and treatment by altering the abundance of microbes.
Collapse
Affiliation(s)
- Alyssa C. Walker
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Rohan Bhargava
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Michael J. Bucher
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Yoan M. Argote
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Amanda S. Brust
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Daniel M. Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
39
|
Dong W, Fan X, Guo Y, Wang S, Jia S, Lv N, Yuan T, Pan Y, Xue Y, Chen X, Xiong Q, Yang R, Zhao W, Zhu B. An expanded database and analytical toolkit for identifying bacterial virulence factors and their associations with chronic diseases. Nat Commun 2024; 15:8084. [PMID: 39278950 PMCID: PMC11402979 DOI: 10.1038/s41467-024-51864-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 08/16/2024] [Indexed: 09/18/2024] Open
Abstract
Virulence factor genes (VFGs) play pivotal roles in bacterial infections and have been identified within the human gut microbiota. However, their involvement in chronic diseases remains poorly understood. Here, we establish an expanded VFG database (VFDB 2.0) consisting of 62,332 nonredundant orthologues and alleles of VFGs using species-specific average nucleotide identity ( https://github.com/Wanting-Dong/MetaVF_toolkit/tree/main/databases ). We further develop the MetaVF toolkit, facilitating the precise identification of pathobiont-carried VFGs at the species level. A thorough characterization of VFGs for 5452 commensal isolates from healthy individuals reveals that only 11 of 301 species harbour these factors. Further analyses of VFGs within the gut microbiomes of nine chronic diseases reveal both common and disease-specific VFG features. Notably, in type 2 diabetes patients, long HiFi sequencing confirms that shared VF features are carried by pathobiont strains of Escherichia coli and Klebsiella pneumoniae. These findings underscore the critical importance of identifying and understanding VFGs in microbiome-associated diseases.
Collapse
Affiliation(s)
- Wanting Dong
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinyue Fan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yaqiong Guo
- Institute of Biotechnology and Health, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Siyi Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shulei Jia
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Na Lv
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Tao Yuan
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Yuanlong Pan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yong Xue
- National Engineering and Technology Research Center for Fruits and Vegetables, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Xi Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qian Xiong
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ruifu Yang
- Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| | - Weigang Zhao
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| | - Baoli Zhu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
- Beijing Key Laboratory of Antimicrobial Resistance and Pathogen Genomics, Beijing, 100101, China.
| |
Collapse
|
40
|
Rusch C, Beke M, Nieves C, Mai V, Stiep T, Tholanikunnel T, Ramirez-Zamora A, Hess CW, Langkamp-Henken B. Promotion of a Mediterranean Diet Alters Constipation Symptoms and Fecal Calprotectin in People with Parkinson's Disease: A Randomized Controlled Trial. Nutrients 2024; 16:2946. [PMID: 39275262 PMCID: PMC11396875 DOI: 10.3390/nu16172946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
Parkinson's disease is associated with gastrointestinal (GI) dysfunction, including constipation symptoms and abnormal intestinal permeability and inflammation. A Mediterranean diet (MediDiet) may aid in disease management. This parallel, randomized, controlled trial in people with Parkinson's (PwP) and constipation symptoms compared a MediDiet against standard of care on change in constipation symptoms, dietary intake, and fecal zonulin and calprotectin concentrations as markers of intestinal permeability and inflammation, respectively. Participants were randomized to either standard of care for constipation (control; n = 17, 65.1 ± 2.2 years) or a MediDiet plus standard of care (n = 19, 68.8 ± 1.4 years) for 8 weeks. Constipation scores decreased with both interventions (p < 0.01), but changes from baseline were not different between groups (MediDiet, -0.5 [-1.0, 0]; control, -0.8 [-1.0, 0.2]; median [25th, 75th]; p = 0.60). The MediDiet group had a higher intake of dietary fiber at week 4 than the control group (13.1 ± 0.7 g/1000 kcal vs. 9.8 ± 0.7 g/1000 kcal; p < 0.001). No differences in fecal zonulin were observed between groups (p = 0.33); however, fecal calprotectin tended to be lower in the MediDiet group at week 8 (45.8 ± 15.1 µg/g vs. 93.9 ± 26.8 µg/g; p = 0.05). The MediDiet and standard interventions reduced constipation symptoms; however, the MediDiet provided additional benefit of increased dietary fiber intake and less intestinal inflammation.
Collapse
Affiliation(s)
- Carley Rusch
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611-0370, USA; (C.R.); (M.B.)
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610-0236, USA; (T.S.); (T.T.); (A.R.-Z.); (C.W.H.)
| | - Matthew Beke
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611-0370, USA; (C.R.); (M.B.)
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610-0236, USA; (T.S.); (T.T.); (A.R.-Z.); (C.W.H.)
| | - Carmelo Nieves
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611-0370, USA; (C.R.); (M.B.)
| | - Volker Mai
- Department of Epidemiology, Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610-0009, USA;
| | - Tamara Stiep
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610-0236, USA; (T.S.); (T.T.); (A.R.-Z.); (C.W.H.)
| | - Tracy Tholanikunnel
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610-0236, USA; (T.S.); (T.T.); (A.R.-Z.); (C.W.H.)
| | - Adolfo Ramirez-Zamora
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610-0236, USA; (T.S.); (T.T.); (A.R.-Z.); (C.W.H.)
| | - Christopher W. Hess
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610-0236, USA; (T.S.); (T.T.); (A.R.-Z.); (C.W.H.)
| | - Bobbi Langkamp-Henken
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, University of Florida, Gainesville, FL 32611-0370, USA; (C.R.); (M.B.)
| |
Collapse
|
41
|
Bell J, Radial SL, Cuming RS, Trope G, Hughes KJ. Effects of fecal microbiota transplantation on clinical outcomes and fecal microbiota of foals with diarrhea. J Vet Intern Med 2024; 38:2718-2728. [PMID: 39266472 PMCID: PMC11423448 DOI: 10.1111/jvim.17185] [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: 03/16/2024] [Accepted: 08/12/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Diarrhea in foals can be associated with disruption of the intestinal microbiota (dysbiosis). Effective management of intestinal dysbiosis in foals has not been demonstrated. HYPOTHESIS/OBJECTIVES Fecal microbiota transplantation (FMT) in foals with diarrhea influences the intestinal microbiota and improves clinical and clinicopathological outcomes. ANIMALS Twenty-five foals <6 months of age with diarrhea and systemic inflammatory response syndrome at 3 veterinary hospitals. METHODS A prospective randomized placebo-controlled cohort study. Foals in the FMT group (n = 19) or control group (n = 9) received FMT or electrolyte solution once daily for 3 days. Fecal samples were obtained on Day 0 (D0), D1, D2, D3, and D7. Within group and between group data analyses were performed for clinical, clinicopathological, and microbiota variables. RESULTS Treatment had no effect on survival (FMT 79%; control 100%, P = .3) or resolution of diarrhea (FMT 68%; control 55%, P = .4). On D3, the white blood cell count of the FMT group was lower than the control group (D3 FMT group median 6.4 g/L [5-8.3 g/L]; D3 control group median 14.3 g/L [6.7-18.9 g/L] P = .04). Heart rate reduced over time in the FMT group (D0 median 80 bpm [60-150 bpm]; D2 median 70 bpm [52-110 bpm] [P = .005]; and D3 median 64, [54-102 bpm] [P < .001]). Phylum Verrucomicrobiota, genus Akkermansia, and family Prevotellaceae were enriched in the FMT group on D1 (linear discriminate analysis > 4). CONCLUSIONS AND CLINICAL IMPORTANCE In foals with diarrhea, FMT appears safe and can be associated with some clinical and microbiota changes suggestive of beneficial effect.
Collapse
Affiliation(s)
- Jillian Bell
- Charles Sturt University School of Agricultural, Environmental and Veterinary SciencesWagga WaggaNew South WalesAustralia
| | - Sharanne L. Radial
- Charles Sturt University School of Agricultural, Environmental and Veterinary SciencesWagga WaggaNew South WalesAustralia
| | | | - Gareth Trope
- South Eastern Equine HospitalNarre Warren NorthVictoriaAustralia
| | - Kristopher J. Hughes
- Charles Sturt University School of Agricultural, Environmental and Veterinary SciencesWagga WaggaNew South WalesAustralia
| |
Collapse
|
42
|
Borrego-Ruiz A, Borrego JJ. Influence of human gut microbiome on the healthy and the neurodegenerative aging. Exp Gerontol 2024; 194:112497. [PMID: 38909763 DOI: 10.1016/j.exger.2024.112497] [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: 03/04/2024] [Revised: 05/16/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
The gut microbiome plays a crucial role in host health throughout the lifespan by influencing brain function during aging. The microbial diversity of the human gut microbiome decreases during the aging process and, as a consequence, several mechanisms increase, such as oxidative stress, mitochondrial dysfunction, inflammatory response, and microbial gut dysbiosis. Moreover, evidence indicates that aging and neurodegeneration are closely related; consequently, the gut microbiome may serve as a novel marker of lifespan in the elderly. In this narrative study, we investigated how the changes in the composition of the gut microbiome that occur in aging influence to various neuropathological disorders, such as mild cognitive impairment (MCI), dementia, Alzheimer's disease (AD), and Parkinson's disease (PD); and which are the possible mechanisms that govern the relationship between the gut microbiome and cognitive impairment. In addition, several studies suggest that the gut microbiome may be a potential novel target to improve hallmarks of brain aging and to promote healthy cognition; therefore, current and future therapeutic interventions have been also reviewed.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Juan J Borrego
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Plataforma BIONAND, Málaga, Spain.
| |
Collapse
|
43
|
Duru IC, Lecomte A, Shishido TK, Laine P, Suppula J, Paulin L, Scheperjans F, Pereira PAB, Auvinen P. Metagenome-assembled microbial genomes from Parkinson's disease fecal samples. Sci Rep 2024; 14:18906. [PMID: 39143178 PMCID: PMC11324757 DOI: 10.1038/s41598-024-69742-4] [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: 03/13/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
The human gut microbiome composition has been linked to Parkinson's disease (PD). However, knowledge of the gut microbiota on the genome level is still limited. Here we performed deep metagenomic sequencing and binning to build metagenome-assembled genomes (MAGs) from 136 human fecal microbiomes (68 PD samples and 68 control samples). We constructed 952 non-redundant high-quality MAGs and compared them between PD and control groups. Among these MAGs, there were 22 different genomes of Collinsella and Prevotella, indicating high variability of those genera in the human gut environment. Microdiversity analysis indicated that Ruminococcus bromii was statistically significantly (p < 0.002) more diverse on the strain level in the control samples compared to the PD samples. In addition, by clustering all genes and performing presence-absence analysis between groups, we identified several control-specific (p < 0.05) related genes, such as speF and Fe-S oxidoreductase. We also report detailed annotation of MAGs, including Clusters of Orthologous Genes (COG), Cas operon type, antiviral gene, prophage, and secondary metabolites biosynthetic gene clusters, which can be useful for providing a reference for future studies.
Collapse
Affiliation(s)
- Ilhan Cem Duru
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
| | - Alexandre Lecomte
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | | - Pia Laine
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Joni Suppula
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Lars Paulin
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Filip Scheperjans
- Department of Neurology, Helsinki University Hospital and Clinicum, University of Helsinki, Helsinki, Finland
| | - Pedro A B Pereira
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
- Department of Neurology, Helsinki University Hospital and Clinicum, University of Helsinki, Helsinki, Finland.
| | - Petri Auvinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
44
|
Hu A, Zaongo SD, Harypursat V, Wang X, Ouyang J, Chen Y. HIV-associated neurocognitive disorder: key implications of the microbiota-gut-brain axis. Front Microbiol 2024; 15:1428239. [PMID: 39155987 PMCID: PMC11327151 DOI: 10.3389/fmicb.2024.1428239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
HIV-associated neurocognitive disorder (HAND) is now recognized to be relatively common in people living with HIV (PLWH), and remains a common cause of cognitive impairment. Unfortunately, the fundamental pathogenic processes underlying this specific outcome of HIV infection have not as yet been fully elucidated. With increased interest in research related to the microbiota-gut-brain axis, the gut-brain axis has been shown to play critical roles in regulating central nervous system disorders such as Alzheimer's disease and Parkinson's disease. PLWH are characterized by a particular affliction, referred to as gut-associated dysbiosis syndrome, which provokes an alteration in microbial composition and diversity, and of their associated metabolite composition within the gut. Interestingly, the gut microbiota has also been recognized as a key element, which both positively and negatively influences human brain health, including the functioning and development of the central nervous system (CNS). In this review, based on published evidence, we critically discuss the relevant interactions between the microbiota-gut-brain axis and the pathogenesis of HAND in the context of HIV infection. It is likely that HAND manifestation in PLWH mainly results from (i) gut-associated dysbiosis syndrome and a leaky gut on the one hand and (ii) inflammation on the other hand. In other words, the preceding features of HIV infection negatively alter the composition of the gut microbiota (microbes and their associated metabolites) and promote proinflammatory immune responses which singularly or in tandem damage neurons and/or induce inadequate neuronal signaling. Thus, HAND is fairly prevalent in PLWH. This work aims to demonstrate that in the quest to prevent and possibly treat HAND, the gut microbiota may ultimately represent a therapeutically targetable "host factor."
Collapse
Affiliation(s)
- Aizhen Hu
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Silvere D. Zaongo
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Vijay Harypursat
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Xin Wang
- Phase I Clinical Trial Center, Chonggang General Hospital, Chongqing, China
| | - Jing Ouyang
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Yaokai Chen
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
45
|
Kerstens R, Joyce P. The Gut Microbiome as a Catalyst and Emerging Therapeutic Target for Parkinson's Disease: A Comprehensive Update. Biomedicines 2024; 12:1738. [PMID: 39200203 PMCID: PMC11352163 DOI: 10.3390/biomedicines12081738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
Parkinson's Disease is the second most prevalent neurological disorder globally, and its cause is still largely unknown. Likewise, there is no cure, and existing treatments do little more than subdue symptoms before becoming ineffective. It is increasingly important to understand the factors contributing to Parkinson's Disease aetiology so that new and more effective pharmacotherapies can be established. In recent years, there has been an emergence of research linking gut dysbiosis to Parkinson's Disease via the gut-brain axis. Advancements in microbial profiling have led to characterisation of a Parkinson's-specific microbial signature, where novel treatments that leverage and correct gut dysbiosis are beginning to emerge for the safe and effective treatment of Parkinson's Disease. Preliminary clinical studies investigating microbiome-targeted therapeutics for Parkinson's Disease have revealed promising outcomes, and as such, the aim of this review is to provide a timely and comprehensive update of the most recent advances in this field. Faecal microbiota transplantation has emerged as a novel and potential frontrunner for microbial-based therapies due to their efficacy in alleviating Parkinson's Disease symptomology through modulation of the gut-brain axis. However, more rigorous clinical investigation, along with technological advancements in diagnostic and in vitro testing tools, are critically required to facilitate the widespread clinical translation of microbiome-targeting Parkinson's Disease therapeutics.
Collapse
Affiliation(s)
| | - Paul Joyce
- Centre for Pharmaceutical Innovation (CPI), UniSA Clinical & Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| |
Collapse
|
46
|
Zhao R. Can exercise benefits be harnessed with drugs? A new way to combat neurodegenerative diseases by boosting neurogenesis. Transl Neurodegener 2024; 13:36. [PMID: 39049102 PMCID: PMC11271207 DOI: 10.1186/s40035-024-00428-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Adult hippocampal neurogenesis (AHN) is affected by multiple factors, such as enriched environment, exercise, ageing, and neurodegenerative disorders. Neurodegenerative disorders can impair AHN, leading to progressive neuronal loss and cognitive decline. Compelling evidence suggests that individuals engaged in regular exercise exhibit higher production of proteins that are essential for AHN and memory. Interestingly, specific molecules that mediate the effects of exercise have shown effectiveness in promoting AHN and cognition in different transgenic animal models. Despite these advancements, the precise mechanisms by which exercise mimetics induce AHN remain partially understood. Recently, some novel exercise molecules have been tested and the underlying mechanisms have been proposed, involving intercommunications between multiple organs such as muscle-brain crosstalk, liver-brain crosstalk, and gut-brain crosstalk. In this review, we will discuss the current evidence regarding the effects and potential mechanisms of exercise mimetics on AHN and cognition in various neurological disorders. Opportunities, challenges, and future directions in this research field are also discussed.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education, Yangzhou University, 88 South Daxue Road, Yangzhou, 225009, China.
| |
Collapse
|
47
|
Yao Q, Chen L, Cai Y, Li C, Wen S, Yang C, Zhang Q, Zeng Y, Zheng S, Zou J, Huang G, Zeng Q. Exploring Causal Links Between Gut Microbiota and Geriatric Syndromes: A Two-Sample Mendelian Randomization Analysis. Int J Med Sci 2024; 21:1945-1963. [PMID: 39113894 PMCID: PMC11302557 DOI: 10.7150/ijms.94335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/21/2024] [Indexed: 08/10/2024] Open
Abstract
Background: Both observational studies and clinical trials have demonstrated a link between the gut microbiota and the geriatric syndrome. Nevertheless, the exact nature of this relationship, particularly concerning causality, remains elusive. Mendelian randomization (MR) is a method of inference based on genetic variation to assess the causal relationship between an exposure and an outcome. In this study, we conducted a two-sample Mendelian randomization (TSMR) study to fully reveal the potential genetic causal effects of gut microbiota on geriatric syndromes. Methods: This study used data from genome wide association studies (GWAS) to investigate causal relationships between the gut microbiota and geriatric syndromes, including frailty, Parkinson's disease (PD), delirium, insomnia, and depression. The primary causal relationships were evaluated using the inverse-variance weighted method, MR Egger, simple mode, weighted mode and weighted median. To assess the robustness of the results, horizontal pleiotropy was examined through MR-Egger intercept and MR-presso methods. Heterogeneity was assessed using Cochran's Q test, and sensitivity was evaluated via the leave-one-out method. Results: We identified 41 probable causal relationships between gut microbiota and five geriatric syndrome-associated illnesses using the inverse-variance weighted method. Frailty showed five positive and two negative causal relationships, while PD revealed three positive and four negative causal connections. Delirium showed three positive and two negative causal relationships. Similarly, insomnia demonstrated nine positive and two negative causal connections, while depression presented nine positive and two negative causal relationships. Conclusions: Using the TSMR method and data from the public GWAS database and, we observed associations between specific microbiota groups and geriatric syndromes. These findings suggest a potential role of gut microbiota in the development of geriatric syndromes, providing valuable insights for further research into the causal relationship between gut microbiota and these syndromes.
Collapse
Affiliation(s)
- Qiuru Yao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Ling Chen
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuxin Cai
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Changxi Li
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuyang Wen
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Chun Yang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, Guangdong, China
| | - Qi Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Yuting Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuqi Zheng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Jihua Zou
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
- Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Guozhi Huang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
48
|
Han N, Peng X, Zhang T, Qiang Y, Li X, Zhang W. Temporal dynamics and species-level complexity of Prevotella spp. in the human gut microbiota: implications for enterotypes and health. Front Microbiol 2024; 15:1414000. [PMID: 39044948 PMCID: PMC11265296 DOI: 10.3389/fmicb.2024.1414000] [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: 04/08/2024] [Accepted: 06/12/2024] [Indexed: 07/25/2024] Open
Abstract
The concept of "enterotypes" in microbiome research has attracted substantial interest, particularly focusing on the abundance of Prevotella spp. in the human gut. In this study, the intricate dynamics of Prevotella spp. in the human gut microbiota was investigated, based on the metagenomic method. First, 239 fecal samples from individuals across four regions of China revealed a bimodal distribution, highlighting the abundance and variability in Prevotella spp. within the Chinese population. Second, the longitudinal cohort study included 184 fecal samples from 52 time points collected from seven individuals who demonstrated either the outbreaks or disappearances of Prevotella spp., emphasizing the transient nature of Prevotella abundance levels and suggesting shifts in Prevotella "enterotypes." Furthermore, a turnover of the dominant Prevotella spp. was observed, indicating the potential presence of diverse subtypes of Prevotella enterotype. Notably, the genomic analysis demonstrated the persistence of specific Prevotella strains within individuals over extended periods, highlighting the enduring presence of Prevotella in the human gut. In conclusion, by integrating the temporal and geographical scales in our research, we gained deeper insights into the dynamics of Prevotella, emphasizing the importance of considering the dynamics at the time and species level in gut microbiota studies and their implications on human health.
Collapse
Affiliation(s)
- Na Han
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xianhui Peng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Tingting Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yujun Qiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuwen Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wen Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
49
|
Riegelman E, Xue KS, Wang JS, Tang L. Gut-Brain Axis in Focus: Polyphenols, Microbiota, and Their Influence on α-Synuclein in Parkinson's Disease. Nutrients 2024; 16:2041. [PMID: 38999791 PMCID: PMC11243524 DOI: 10.3390/nu16132041] [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: 06/11/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
With the recognition of the importance of the gut-brain axis in Parkinson's disease (PD) etiology, there is increased interest in developing therapeutic strategies that target α-synuclein, the hallmark abhorrent protein of PD pathogenesis, which may originate in the gut. Research has demonstrated that inhibiting the aggregation, oligomerization, and fibrillation of α-synuclein are key strategies for disease modification. Polyphenols, which are rich in fruits and vegetables, are drawing attention for their potential role in this context. In this paper, we reviewed how polyphenols influence the composition and functional capabilities of the gut microbiota and how the resulting microbial metabolites of polyphenols may potentially enhance the modulation of α-synuclein aggregation. Understanding the interaction between polyphenols and gut microbiota and identifying which specific microbes may enhance the efficacy of polyphenols is crucial for developing therapeutic strategies and precision nutrition based on the microbiome.
Collapse
Affiliation(s)
| | | | | | - Lili Tang
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602, USA; (E.R.); (K.S.X.); (J.-S.W.)
| |
Collapse
|
50
|
Bai F, You L, Lei H, Li X. Association between increased and decreased gut microbiota abundance and Parkinson's disease: A systematic review and subgroup meta-analysis. Exp Gerontol 2024; 191:112444. [PMID: 38679353 DOI: 10.1016/j.exger.2024.112444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
OBJECTIVE The objective of the study was to systematically investigate the association between gut microbiota (GM) abundance and Parkinson's disease (PD). METHODS PubMed, Medline, Cochrane Library and other literature datebase platforms were searched for eligible studies in the English-language from conception to March 1, 2024. Studies evaluating the association between GM and PD were included. The results of the included studies were analyzed using a random effects model with calculation of the mean difference (MD) with the 95 % confidence interval to quantify the incidence of differences in abundance of various bacterial families in PD patients. Continuous models were used to analyze the extracted data. RESULTS A total of 14 studies with 1045 PD cases and 821 healthy controls were included for data extraction and meta-analysis. All the included studies exhibited reasonable quality. The included studies reported the data on the ratios of 10 families of GM. Of these 10 microbiota families, Bifidobacteriaceae, Ruminococcaceae, Rikenellaceae, Lactobacillaceae, Verrucomicrobiaceae and Christensenellaceae were found to have increased ratios according to the pooled ratios, while Prevotellaceae, Lachnospiraceae, Erysipelotrichaceae and Faecalibacterium were decreased in PD cases. CONCLUSION Patients in the PD cohort exhibited distinctive microbiota compositions compared to healthy individuals, with unique differential patterns in gut microbiome abundance at the phylum, family, and genus levels that may be associated wtih PD pathogenesis.
Collapse
Affiliation(s)
- Fusheng Bai
- Department of Neurology, Jinqiu Hospital of Liaoning Province, Shenyang 110016, China
| | - Lin You
- Department of Neurology, Jinqiu Hospital of Liaoning Province, Shenyang 110016, China
| | - Hongyan Lei
- Department of Neurology, Jinqiu Hospital of Liaoning Province, Shenyang 110016, China
| | - Xinming Li
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Shenyang 110034, China.
| |
Collapse
|