1
|
Nunez H, Nieto PA, Mars RA, Ghavami M, Sew Hoy C, Sukhum K. Early life gut microbiome and its impact on childhood health and chronic conditions. Gut Microbes 2025; 17:2463567. [PMID: 39916516 PMCID: PMC11810090 DOI: 10.1080/19490976.2025.2463567] [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: 09/23/2024] [Revised: 12/20/2024] [Accepted: 02/02/2025] [Indexed: 02/12/2025] Open
Abstract
The development of the gut microbiome is crucial to human health, particularly during the first three years of life. Given its role in immune development, disturbances in the establishment process of the gut microbiome may have long term consequences. This review summarizes evidence for these claims, highlighting compositional changes of the gut microbiome during this critical period of life as well as factors that affect gut microbiome development. Based on human and animal data, we conclude that the early-life microbiome is a determinant of long-term health, impacting physiological, metabolic, and immune processes. The early-life gut microbiome field faces challenges. Some of these challenges are technical, such as lack of standardized stool collection protocols, inconsistent DNA extraction methods, and outdated sequencing technologies. Other challenges are methodological: small sample sizes, lack of longitudinal studies, and poor control of confounding variables. To address these limitations, we advocate for more robust research methodologies to better understand the microbiome's role in health and disease. Improved methods will lead to more reliable microbiome studies and a deeper understanding of its impact on health outcomes.
Collapse
Affiliation(s)
- Harold Nunez
- Seeding Inc, DBA Tiny Health, Austin, Texas, USA
| | | | - Ruben A. Mars
- Seeding Inc, DBA Tiny Health, Austin, Texas, USA
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
2
|
Prince N, Peralta Marzal LN, Roussin L, Monnoye M, Philippe C, Maximin E, Ahmed S, Salenius K, Lin J, Autio R, Adolfs Y, Pasterkamp RJ, Garssen J, Naudon L, Rabot S, Kraneveld AD, Perez-Pardo P. Mouse strain-specific responses along the gut-brain axis upon fecal microbiota transplantation from children with autism. Gut Microbes 2025; 17:2447822. [PMID: 39773319 PMCID: PMC11730631 DOI: 10.1080/19490976.2024.2447822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Several factors are linked to the pathophysiology of autism spectrum disorders (ASD); however, the molecular mechanisms of the condition remain unknown. As intestinal problems and gut microbiota dysbiosis are associated with ASD development and severity, recent studies have focused on elucidating the microbiota-gut-brain axis' involvement. This study aims to explore mechanisms through which gut microbiota might influence ASD. Briefly, we depleted the microbiota of conventional male BALB/cAnNCrl (Balb/c) and C57BL/6J (BL/6) mice prior to human fecal microbiota transplantation (hFMT) with samples from children with ASD or their neurotypical siblings. We found mouse strain-specific responses to ASD hFMT. Notably, Balb/c mice exhibit decreased exploratory and social behavior, and show evidence of intestinal, systemic, and central inflammation accompanied with metabolic shifts. BL/6 mice show less changes after hFMT. Our results reveal that gut microbiota alone induce changes in ASD-like behavior, and highlight the importance of mouse strain selection when investigating multifactorial conditions like ASD.
Collapse
Affiliation(s)
- Naika Prince
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lucia N. Peralta Marzal
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Léa Roussin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Magali Monnoye
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Catherine Philippe
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Elise Maximin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Sabbir Ahmed
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Karoliina Salenius
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Jake Lin
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Reija Autio
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Laurent Naudon
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Micalis Institute, Jouy-en-Josas, France
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Aletta D. Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Neuroscience, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
3
|
Wu H, Chen J, Guo S, Deng J, Zhou Z, Zhang X, Qi T, Yu F, Yang Q. Advances in the acting mechanism and treatment of gut microbiota in metabolic dysfunction-associated steatotic liver disease. Gut Microbes 2025; 17:2500099. [PMID: 40394806 PMCID: PMC12101596 DOI: 10.1080/19490976.2025.2500099] [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: 01/24/2025] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease(MASLD) is increasing in prevalence worldwide and has become the greatest potential risk for cirrhosis and hepatocellular liver cancer. Currently, the role of gut microbiota in the development of MASLD has become a research hotspot. The development of MASLD can affect the homeostasis of gut microbiota, and significant changes in the composition or abundance of gut microbiota and its metabolite abnormalities can influence disease progression. The regulation of gut microbiota is an important strategy and novel target for the treatment of MASLD with good prospects. In this paper, we summarize the role of gut microbiota and its metabolites in the pathogenesis of MASLD, and describe the potential preventive and therapeutic efficacy of gut microbiota as a noninvasive marker to regulate the pathogenesis of MASLD based on the "gut-hepatic axis", which will provide new therapeutic ideas for the clinic.
Collapse
Affiliation(s)
- Huaying Wu
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Jingjing Chen
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Shuyuan Guo
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jinhao Deng
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Zimeng Zhou
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Xuan Zhang
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - TianTian Qi
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- Department of Spine Surgery, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qi Yang
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
4
|
Lau RI, Su Q, Ng SC. Long COVID and gut microbiome: insights into pathogenesis and therapeutics. Gut Microbes 2025; 17:2457495. [PMID: 39854158 PMCID: PMC11776476 DOI: 10.1080/19490976.2025.2457495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Post-acute coronavirus disease 2019 syndrome (PACS), following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or coronavirus disease 2019 (COVID-19), is typically characterized by long-term debilitating symptoms affecting multiple organs and systems. Unfortunately, there is currently a lack of effective treatment strategies. Altered gut microbiome has been proposed as one of the plausible mechanisms involved in the pathogenesis of PACS; extensive studies have emerged to bridge the gap between the persistent symptoms and the dysbiosis of gut microbiome. Recent clinical trials have indicated that gut microbiome modulation using probiotics, prebiotics, and fecal microbiota transplantation (FMT) led to improvements in multiple symptoms related to PACS, including fatigue, memory loss, difficulty in concentration, gastrointestinal upset, and disturbances in sleep and mood. In this review, we highlight the latest evidence on the key microbial alterations observed in PACS, as well as the use of microbiome-based therapeutics in managing PACS symptoms. These novel findings altogether shed light on the treatment of PACS and other chronic conditions.
Collapse
Affiliation(s)
- Raphaela I. Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong KongSAR, China
- Microbiota I-Center (MagIC), Hong KongSAR, China
| | - Qi Su
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong KongSAR, China
- Microbiota I-Center (MagIC), Hong KongSAR, China
| | - Siew C. Ng
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong KongSAR, China
- Microbiota I-Center (MagIC), Hong KongSAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong KongSAR, China
| |
Collapse
|
5
|
Castagnoli R, Pala F, Subramanian P, Oguz C, Schwarz B, Lim AI, Burns AS, Fontana E, Bosticardo M, Corsino C, Angelova A, Delmonte OM, Kenney H, Riley D, Smith G, Ott de Bruin L, Oikonomou V, Dos Santos Dias L, Fink D, Bohrnsen E, Kimzey CD, Marseglia GL, Alva-Lozada G, Bergerson JR, Brett A, Brigatti KW, Dimitrova D, Dutmer CM, Freeman AF, Ale H, Holland SM, Licciardi F, Pasic S, Poskitt LE, Potts DE, Dasso JF, Sharapova SO, Strauss KA, Ward BR, Yilmaz M, Kuhns DB, Lionakis MS, Daley SR, Kong HH, Segre JA, Villa A, Pittaluga S, Walter JE, Vujkovic-Cvijin I, Belkaid Y, Notarangelo LD. Immunopathological and microbial signatures of inflammatory bowel disease in partial RAG deficiency. J Exp Med 2025; 222:e20241993. [PMID: 40314722 PMCID: PMC12047384 DOI: 10.1084/jem.20241993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
Partial RAG deficiency (pRD) can manifest with systemic and tissue-specific immune dysregulation, with inflammatory bowel disease (IBD) in 15% of the patients. We aimed at identifying the immunopathological and microbial signatures associated with IBD in patients with pRD and in a mouse model of pRD (Rag1w/w) with spontaneous development of colitis. pRD patients with IBD and Rag1w/w mice showed a systemic and colonic Th1/Th17 inflammatory signature. Restriction of fecal microbial diversity, abundance of pathogenic bacteria, and depletion of microbial species producing short-chain fatty acid were observed, which were associated with impaired induction of lamina propria peripheral Treg cells in Rag1w/w mice. The use of vedolizumab in Rag1w/w mice and of ustekinumab in a pRD patient were ineffective. Antibiotics ameliorated gut inflammation in Rag1w/w mice, but only bone marrow transplantation (BMT) rescued the immunopathological and microbial signatures. Our findings shed new light in the pathophysiology of gut inflammation in pRD and establish a curative role for BMT to resolve the disease phenotype.
Collapse
Affiliation(s)
- Riccardo Castagnoli
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ai Ing Lim
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew S. Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cristina Corsino
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Angelina Angelova
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heather Kenney
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Deanna Riley
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grace Smith
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Ott de Bruin
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Pediatric Stem Cell Transplantation Program, Leiden University Medical Center, Leiden, Netherlands
| | - Vasileios Oikonomou
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lucas Dos Santos Dias
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Danielle Fink
- Neutrophil Monitoring Lab, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Cole D. Kimzey
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Gian Luigi Marseglia
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Guisela Alva-Lozada
- Allergy and Immunology Division Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
| | - Jenna R.E. Bergerson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ana Brett
- Hospital Pediátrico, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
- Clínica Universitária de Pediatria, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| | | | - Dimana Dimitrova
- Experimental Transplantation and Immunotherapy Branch, National Cancer Institute of the National Institutes of Health, Bethesda, MD, USA
| | - Cullen M. Dutmer
- Allergy and Immunology, Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hanadys Ale
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children’s Hospital, Memorial Healthcare System, Hollywood, FL, USA
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Licciardi
- Immuno-reumatologia, Pediatria Specialistica Universitaria, Ospedale Infantile Regina Margherita, Torino, Italy
| | - Srdjan Pasic
- Department of Pediatric Immunology, Mother and Child Health Institute, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | | | - David E. Potts
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Joseph F. Dasso
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Svetlana O. Sharapova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | | | - Brant R. Ward
- Division of Allergy and Immunology, Children’s National Hospital, Washington, DC, USA
| | - Melis Yilmaz
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Douglas B. Kuhns
- Neutrophil Monitoring Lab, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Michail S. Lionakis
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephen R. Daley
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Heidi H. Kong
- Cutaneous Microbiome and Inflammation Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julia A. Segre
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anna Villa
- San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget), IRCSS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Stefania Pittaluga
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jolan E. Walter
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Ivan Vujkovic-Cvijin
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Immunology, Institut Pasteur, Paris, France
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Melby MK, Zent E, Ariste S, Shoukat R, Nichter M. "The Littlest Creatures that Live Inside Us": Public understandings influencing microbiome-related behaviors. Soc Sci Med 2025; 376:117864. [PMID: 40279787 DOI: 10.1016/j.socscimed.2025.117864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 04/29/2025]
Abstract
The impact of behaviors on the microbiome, and in turn its effects on health, are increasingly recognized by the scientific and medical communities. Yet little is known about public understandings of the microbiome and the relationships with health. We developed a semi-structured interview tool covering topics from microbiome composition to behavioral factors affecting microbiomes and related health throughout the life course. Drawing on semi-structured interviews conducted in 2021-22 with 32 US women purposefully recruited from 5 demographic categories (with pets, chronic illness, young children, complementary and alternative medicine use, and elders), we explore perspectives on topics including microbiome and diet, medicine use, cleaning practices, and travel. We conducted a thematic analysis, tallied response frequencies of elicited themes, and plotted emerging themes on semantic maps to provide visual representations of connections between concepts, and commonly mentioned metaphors to identify promising new lines of inquiry on public understandings of the microbiome. We observed interconnections between domains, as shown in semantic maps, and examples of ecological and militaristic analogies. Participants used analogies to estimate duration of microbiome disruption and recovery. Metaphors provide scaffolding for organizing new information about the microbiome and influence how people make choices about when and how to kill or promote microbes in their bodies and their environment. Characterizing these public understandings of microbiomes and linkages to health behaviors among different demographic groups is critical to designing and implementing health education and policy and informing future research inquiries in a participatory way through public-research collaborations.
Collapse
Affiliation(s)
- Melissa K Melby
- Department of Anthropology, University of Delaware, Newark, DE, USA; CIFAR Humans & the Microbiome, Canada.
| | - Eglee Zent
- Department of Anthropology, University of Delaware, Newark, DE, USA; Lab. Ecologia Humana, Instituto Venezolano de Investigaciones Cientificas, Altos de Pipe, VZ; Rubinstein School of Environment and Natural Resources, University of Vermont, Burlington, VT, USA
| | | | | | - Mark Nichter
- School of Anthropology, University of Arizona, Tucson, AZ, USA; CIFAR Humans & the Microbiome, Canada
| |
Collapse
|
7
|
Zhang N, Dong X. Causal relationship between gut microbiota, lipids, and neuropsychiatric disorders: A Mendelian randomization mediation study. J Affect Disord 2025; 379:19-35. [PMID: 40049531 DOI: 10.1016/j.jad.2025.02.091] [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: 09/30/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Numerous studies have shown an interconnection between the gut microbiota and the brain via the "gut-brain" axis. However, the causal relationships between gut microbiota, lipids, and neuropsychiatric disorders remain unclear. This study aimed to analyze potential associations among gut microbiota, lipids, and neuropsychiatric disorders-including AD, PD, ALS, MS, SCZ, MDD, and BD-using summary data from large-scale GWAS. METHODS Bidirectional Mendelian randomization (MR) with inverse variance weighting (IVW) was the primary method. Supplementary analyses included sensitivity analyses, Steiger tests, and Bayesian weighted MR (BWMR). Mediation analyses used two-step MR (TSMR) and multivariable MR (MVMR). RESULTS The analyses revealed 51 positive correlations (risk factors) (β > 0, P < 0.05) and 47 negative correlations (protective factors) (β < 0, P < 0.05) between gut microbiota and neuropsychiatric disorders. In addition, 35 positive correlations (β > 0, P < 0.05) and 22 negative correlations (β < 0, P < 0.05) between lipids and neuropsychiatric disorders were observed. Assessment of reverse causality with the seven neuropsychiatric disorders as exposures and the identified gut microbiota and lipids as outcomes revealed no evidence of reverse causality (P > 0.05). Mediation analysis indicated that the effect of the species Bacteroides plebeius on MDD is partially mediated through the regulation of phosphatidylcholine (16:0_20:4) levels (mediation proportion = 10.9 % [95 % CI = 0.0110-0.2073]). CONCLUSION This study provides evidence of a causal relationship between gut microbiota and neuropsychiatric disorders, suggesting lipids as mediators. These findings offer new insights into the mechanisms by which gut microbiota may influence neuropsychiatric disorders.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, the Seventh Clinical College of China Medical University, No. 24 Central Street, Xinfu District, Fushun 113000, Liaoning, China
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning, China.
| |
Collapse
|
8
|
Han Y, Zhou S, Yang Y, Hu S, Zhang W, Shen G, Peng C. Further negative effect of fibrous microplastics to the bioaccumulation and toxicity of decabromodiphenyl ethane on zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 980:179577. [PMID: 40319805 DOI: 10.1016/j.scitotenv.2025.179577] [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: 10/14/2024] [Revised: 04/06/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Co-pollution of microplastics (MPs) and novel brominated flame retardants (NBFRs) in aquatic environments is becoming increasingly common in aquatic environments, raising concerns about their comprehensive ecological impacts. This study investigated the effects of fibrous polyethylene terephthalate (PET) MPs on the bioaccumulation and toxicity of decabromodiphenyl ethane (DBDPE) in zebrafish (Danio rerio). In a 28-day water exposure experiment, co-exposure of fibrous MPs and DBDPE significantly increased the bioavailability of DBDPE in zebrafish and prolonged the half-life of DBDPE in vivo. The elimination rates of DBDPE concentration in muscles of single DBDPE exposure and co-exposure groups were 61.58 % and 56.63 %, respectively. Additionally, the co-exposure exacerbated intestinal damage, including structural deterioration and nutrients depletion, disrupted gut microbiota, promoted the enrichment of genes related to reproductive toxicity, and affected the gut-brain axis, indicating complex toxic interactions in zebrafish. Furthermore, genera of Aurantimicrobium, Cypionkella, and Gemmobacter were the gut microbes significantly associated with main differentially expressed genes(DEGs)in the brain. This study emphasized the exacerbating role of fibrous MPs in DBDPE toxicity, providing new insights into the ecological risks posed by the coexistence of MPs and NBFRs in aquatic ecosystems.
Collapse
Affiliation(s)
- Yanna Han
- School of Resource and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shanqi Zhou
- School of Resource and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, China; Xianghu Laboratory, Hangzhou 311231, China
| | - Yuhe Yang
- School of Resource and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shuangqing Hu
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai Academy of Environment Sciences, Shanghai 200233, China
| | - Wei Zhang
- School of Resource and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Genxiang Shen
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai Academy of Environment Sciences, Shanghai 200233, China
| | - Cheng Peng
- School of Resource and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, China; State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai Academy of Environment Sciences, Shanghai 200233, China.
| |
Collapse
|
9
|
Huang W, Jiang T, He J, Ruan J, Wu B, Tao R, Xu P, Wang Y, Chen R, Wang H, Yang Q, Zhang K, Jin L, Sun D, You J. Modulation of Intestinal Flora: a Novel Immunotherapeutic Approach for Enhancing Thyroid Cancer Treatment. Probiotics Antimicrob Proteins 2025; 17:1038-1063. [PMID: 39890752 DOI: 10.1007/s12602-025-10471-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Over the past 3 years, there has been a growing interest in clinical research regarding the potential involvement of intestinal flora in thyroid cancer (TC). This review delves into the intricate connection between intestinal flora and TC, focusing on the particular intestinal flora that is directly linked to the disease and identifying which may be able to predict potential microbial markers of TC. In order to shed light on the inflammatory pathways connected to the onset of TC, we investigated the impact of intestinal flora on immune modulation and the connection between chronic inflammation when investigating the role of intestinal flora in the pathogenesis of TC. Furthermore, the potential role of intestinal flora metabolites in the regulation of thyroid function was clarified by exploring the effects of short-chain fatty acids and lipopolysaccharide on thyroid hormone synthesis and metabolism. Based on these findings, we further explore the effects of probiotics, prebiotics, postbiotics, vitamins, and trace elements.
Collapse
Affiliation(s)
- Weiqiang Huang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Baihui Wu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Runchao Tao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Peiye Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Yongpan Wang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, SAR 999077, China
| | - Hanbing Wang
- The University of Hong Kong School of Biomedical Sciences, Hong Kong, 999077, SAR, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| |
Collapse
|
10
|
Lei Y, Deng Y, Xia R, Xie B, Yang Z, Xi S, Chen P, Tao R. Full-length 16S rRNA-based exploration of body site-specific bacterial signatures for origin determination and individual identification. Forensic Sci Int 2025; 371:112475. [PMID: 40286757 DOI: 10.1016/j.forsciint.2025.112475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
When the available human-derived information at a crime scene is limited, it poses challenges in determining the origin of the biological materials and identifying their donors. In this context, microorganisms have gradually emerged as a valuable complementary tool. Nowadays, the application of third-generation sequencing technology for full-length 16S rRNA sequencing to explore the specific bacterial biomarkers in various biological materials holds significant research and practical value. In this study, we performed full-length 16S rRNA gene sequencing on sterile swabs from palmar skin, oral mucosa, and nasal cavity using the PacBio single-molecule real-time sequencing (SMRT) platform. Alongside identifying specific bacterial biomarkers for these biological materials from different body sites, the study also preliminarily explored the specific bacterial taxa in 19 individuals at the phylum, genus, and species levels. The results showed that the palmar skin bacteria primarily consist of Cutibacterium, Staphylococcus, and Streptococcus, the oral mucosal bacteria are dominated by Streptococcus, Neisseria, and Haemophilus, while the dominant bacteria in nasal cavity are Staphylococcus and Cutibacterium. Beta diversity analysis revealed significant differences in the bacterial community composition across the three origins of biological materials. Furthermore, classification models based on the bacterial species were constructed using the Random Forest, XGBoost, and KNN algorithms. The results showed that both Random Forest and XGBoost models achieved an accuracy of 97 %, significantly outperforming the KNN model (79 %). The prediction accuracy at the OTU level was comparable to that at the species level. In addition, bacterial community differences between individuals were observed at both the genus and species levels. Overall, this study further explores the potential of classification prediction methods based on bacterial features for distinguishing the body site origins of different biological materials and enabling individual traceability, thereby providing valuable data to support the application of microbiological techniques in forensic practice.
Collapse
Affiliation(s)
- Yinlei Lei
- Key Laboratory of Cell Engineering of Guizhou Province, Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563099, China; Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Sciences, Key Laboratory of Forensic Science, Ministry of Justice, Shanghai 200063, China
| | - Yu Deng
- Key Laboratory of Cell Engineering of Guizhou Province, Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563099, China; Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Sciences, Key Laboratory of Forensic Science, Ministry of Justice, Shanghai 200063, China
| | - Ruocheng Xia
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Sciences, Key Laboratory of Forensic Science, Ministry of Justice, Shanghai 200063, China
| | - Baoyan Xie
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Sciences, Key Laboratory of Forensic Science, Ministry of Justice, Shanghai 200063, China
| | - Zhenchen Yang
- Criminal Science and Technology Research Institute, Fengxian Branch of Shanghai Municipal Public Security Bureau, Shanghai 201499, China
| | - Shuangyun Xi
- School of Forensic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Pengyu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563099, China.
| | - Ruiyang Tao
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Sciences, Key Laboratory of Forensic Science, Ministry of Justice, Shanghai 200063, China.
| |
Collapse
|
11
|
Park C, Park J, Chang D, Kim S. Development of reference-based model for improved analysis of bacterial community. Food Res Int 2025; 211:116380. [PMID: 40356165 DOI: 10.1016/j.foodres.2025.116380] [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: 02/26/2025] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Probiotic bacteria play a vital role in maintaining gut microbial homeostasis and are widely used in various commercial products. Although 16S rRNA amplicon-based next-generation sequencing (NGS) is commonly used to analyze probiotic products, biases can arise from various 16S rRNA amplification regions, sequencing platforms, and library kits. In this study, a reference-based bias correction model was developed to correct sequencing biases. The model was validated using eight mock communities and 12 commercial products, which were analyzed across multiple NGS platforms and various 16S rRNA regions. Specific primer-probe assays were developed for accurate bacterial quantification, and their specificity was validated and used in conjunction with droplet digital PCR (ddPCR) to establish initial bacterial ratios within communities. Analysis of the mock communities revealed platform- and region-specific biases, with specific species consistently over- or under-represented. Similarly, commercial product analyses have shown biased outcomes owing to varying sequencing protocols. The correction model, based on PCR efficiencies from the reference communities, successfully corrected biased ratios across different amplification regions and platforms to achieve results that closely matched the proportions predicted by ddPCR. The model effectively corrected the biases arising from the different polymerases. Notably, partial references containing approximately 40 % of the species achieved correction results that were comparable to those of the complete references. This approach demonstrates the potential for improving microbiome analysis accuracy within predictable ranges, and could serve as a model for addressing sequencing bias in metagenomic research.
Collapse
Affiliation(s)
- Changwoo Park
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea.
| | - Jinyoung Park
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea; Department of Precision Measurement, University of Science & Technology, Daejeon 34113, Republic of Korea.
| | - Dongho Chang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon 34141, Republic of Korea.
| | - Seil Kim
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea; Department of Precision Measurement, University of Science & Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
12
|
Silveira MAD, Rodrigues RR, Trinchieri G. Intestinal Microbiome Modulation of Therapeutic Efficacy of Cancer Immunotherapy. Gastroenterol Clin North Am 2025; 54:295-315. [PMID: 40348489 PMCID: PMC12066836 DOI: 10.1016/j.gtc.2024.10.005] [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: 05/14/2025]
Abstract
Bacteria are associated with certain cancers and may induce genetic instability and cancer progression. The gut microbiome modulates the response to cancer therapy. Training machine learning models with response associated taxa or bacterial genes predict patients' response to immunotherapies with moderate accuracy. Clinical trials targeting the gut microbiome to improve immunotherapy efficacy have been conducted. While single bacterial strains or small consortia have not be reported yet to be successful, encouraging results have been reported in small single arm and randomized studies using transplant of fecal microbiome from cancer patients who successfully responded to therapy or from healthy volunteers.
Collapse
Affiliation(s)
- Maruhen A D Silveira
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4146, Bethesda, MD 20852, USA
| | - Richard R Rodrigues
- Microbiome and Genetics Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4140B, Bethesda, MD 20852, USA; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Giorgio Trinchieri
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4146, Bethesda, MD 20852, USA.
| |
Collapse
|
13
|
Yang H, Tao H, Xu C, Song J, Teng C, Pan C, Wei S. Selenium-enriched green tea extracts: chemical constituents and effects on antioxidant and anti-inflammatory factors and four major intestinal flora in mice with intestinal disorders. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:4472-4482. [PMID: 40231393 DOI: 10.1002/jsfa.14242] [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: 04/17/2023] [Revised: 01/06/2025] [Accepted: 02/02/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Selenium-enriched tea represents an ideal source of selenium supplements, yet its effects on mice with intestinal disorders remain under-documented. Therefore, we have carried out relevant research. METHODS We determined the main chemical components of selenium-enriched green tea and evaluated the antioxidant and anti-inflammatory effects of selenium-enriched green tea extract and changes in intestinal flora of antibiotic-induced intestinal disease mice through BALB/c mouse experiments. RESULTS The main chemical components of selenium-enriched green tea and green tea are significantly different. Selenium-enriched green tea is characterized by a high selenium content, with tea polyphenols, flavonoids, tea polysaccharides and catechins being the primary constituents. The results of animal experiments indicate that the extract of green tea rich in selenium increased the content of antioxidant factors in the intestines of mice and reduced the levels of intestinal inflammatory factors. This was also confirmed by mRNA gene expression determination. In addition, selenium-enriched green tea extracts can reduce the weight loss and intestinal pathological damage induced by antibiotics, promote the colonization of Bifidobacterium and Lactobacillus in the intestinal tract of mice and inhibit the growth of Escherichia coli and Enterococcus in the intestinal tract. CONCLUSION Selenium-enriched green tea has high nutritional content. It demonstrates superior potential in alleviating oxidative stress and inflammatory responses caused by intestinal diseases, and plays a role in regulating the intestinal flora. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Hongbo Yang
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Hui Tao
- First Affiliated Hospital of Guizhou Medical University (Guian Hospital), Guiyang, China
| | - Chan Xu
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Jieyu Song
- School of Public Health, Guizhou Medical University, Guiyang, China
| | - Chunli Teng
- Guizhou Jianande Technology Co. Ltd, Guiyang, China
| | - Canping Pan
- College of Science, China Agricultural University, Beijing, China
| | - Shaofeng Wei
- School of Public Health, Guizhou Medical University, Guiyang, China
| |
Collapse
|
14
|
Cao T, Sun K, He E, Cao X, Zhao L, Xu X, Qiu H. Diverse Perspectives Illuminate the Intestinal Toxicity of Traditional and Biodegradable Agricultural Film Microplastics to Eisenia fetida under Varying Exposure Sequences. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:9943-9954. [PMID: 40371808 DOI: 10.1021/acs.est.5c01932] [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: 05/16/2025]
Abstract
The widespread use of plastic agricultural films necessitates a thorough evaluation of environmental risks posed by soil microplastics (MPs). While the intestinal tract is a critical site for MP interactions in soil organisms, current research predominantly focuses on overall physiological responses, overlooking organ-specific toxic mechanisms. To address this gap, we exposed earthworms (Eisenia fetida) to polyethylene (PE) and biodegradable polylactic acid (PLA) MPs sourced from agricultural films at an environmentally realistic concentration of 1.0 g/kg. Incorporating natural earthworm mobility, we designed two exposure scenarios: migration from clean to contaminated soil (scenario A) and vice versa (scenario B). Machine learning-driven image analysis and phenotypic profiling revealed that PE induced more severe intestinal lesions than PLA, adversely affecting intestinal immune functions. Furthermore, PE resulted in greater oxidative damage and significantly activated immune proteins such as melanin and antimicrobial peptides through reprograming immune-related gene and protein pathways. Conversely, PLA predominantly disrupted intestinal digestive and absorptive functions, though the gut microbial community partially mitigated damage through structural and compositional adaptation. Compared with scenario A, earthworms in scenario B exhibited reduced tissue damage, enhanced digestive enzyme activity, and upregulated energy-related metabolites and cell proliferation genes, indicating partial recovery from MP-induced intestinal dysfunction. These findings elucidate the distinct toxicity mechanisms of conventional and biodegradable agricultural MPs on soil organisms, while the scenario-based approach advances risk assessment by aligning experimental design with real-world ecological behaviors.
Collapse
Affiliation(s)
- Tianyi Cao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kailun Sun
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Erkai He
- School of Geographic Sciences, East China Normal University, Shanghai 200241, China
| | - Xinde Cao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ling Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoyun Xu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Qiu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
15
|
Yau R, Pavloudi C, Zeng Y, Saw J, Eleftherianos I. Infection with the entomopathogenic nematodes Steinernema alters the Drosophila melanogaster larval microbiome. PLoS One 2025; 20:e0323657. [PMID: 40378358 DOI: 10.1371/journal.pone.0323657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/11/2025] [Indexed: 05/18/2025] Open
Abstract
The fruit fly Drosophila melanogaster is a vital model for studying the microbiome due to the availability of genetic resources and procedures. To understand better the importance of microbial composition in shaping immune modulation, we can investigate the role of the microbiota through parasitic infection. For this, we use entomopathogenic nematodes (EPN) of the genus Steinernema which exhibit remarkable ability to efficiently infect a diverse array of insect species, facilitated by the mutualistic bacteria Xenorhabdus found within their gut. To examine the microbiome changes in D. melanogaster larvae in response to Steinernema nematode infection, D. melanogaster late second to early third instar larvae were exposed separately to S. carpocapsae and S. hermaphroditum infective juveniles. We have found that S. carpocapsae infective juveniles are more pathogenic to D. melanogaster larvae compared to the closely related S. hermaphroditum. Our microbiome analysis also indicates substantial changes in the size and composition of the D. melanogaster larval microbiome during infection with either nematode species compared to the uninfected controls. Our results serve as a foundation for future studies to elucidate the entomopathogenic-specific effector molecules that alter the D. melanogaster microbiome and understand the role of the microbiome in regulating insect anti-nematode immune processes.
Collapse
Affiliation(s)
- Raymond Yau
- Department of Biological Sciences, The George Washington University, Washington, DC, United States of America
| | - Christina Pavloudi
- Department of Biological Sciences, The George Washington University, Washington, DC, United States of America
- European Marine Biological Resource Centre-European Research Infrastructure Consortium (EMBRC-ERIC), Paris, France
| | - Yingying Zeng
- Department of Biological Sciences, The George Washington University, Washington, DC, United States of America
| | - Jimmy Saw
- Department of Biological Sciences, The George Washington University, Washington, DC, United States of America
| | - Ioannis Eleftherianos
- Department of Biological Sciences, The George Washington University, Washington, DC, United States of America
| |
Collapse
|
16
|
Rampelli S, Pomstra D, Barone M, Fabbrini M, Turroni S, Candela M, Henry AG. Consumption of only wild foods induces large scale, partially persistent alterations to the gut microbiome. Sci Rep 2025; 15:16593. [PMID: 40360545 PMCID: PMC12075472 DOI: 10.1038/s41598-025-00319-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The gut microbiome (GM) is implicated in human health and varies among lifestyles. So-called "traditional" diets have been suggested to promote health-associated taxa. However, most studies focused only on diets including domesticated foods. Historically, humans consumed only wild foods, which might uniquely shape GM composition. We explored the impact of a wild-food-only diet on GM, particularly whether it increases the presence of health-associated and/or "old friend" taxa, and if the alterations to GM are persistent or transient. One participant collected daily fecal samples and recorded daily food consumption over an eight-week period, the middle four weeks of which he consumed only wild foods. Samples were profiled by 16S rRNA sequencing, and oligotyping and network analysis were conducted to assess microbial co-occurrence patterns. A wild-food-only diet considerably alters the composition of the GM, and the magnitude of the changes is larger than that observed in other diet interventions. No new taxa, including "old friends" appeared; instead, the proportions of already-present taxa shifted. Network analysis revealed distinct microbial co-abundance groups restructuring across dietary phases. There is a clear successional shift from the pre-, during- and post-wild-food-only diet. This analysis highlighted structural and functional shifts in microbial interactions, underscoring diet's role in shaping the gut ecosystem.
Collapse
Affiliation(s)
- Simone Rampelli
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy.
| | - Diederik Pomstra
- Department of Archaeological Sciences, Faculty of Archaeology, Leiden University, Leiden, The Netherlands
| | - Monica Barone
- Department of Medical and Surgical Sciences (DiMeC), Microbiomics Unit, Alma Mater Studiorum - University of Bologna, 40138, Bologna, Italy
| | - Marco Fabbrini
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy
- Department of Medical and Surgical Sciences (DiMeC), Microbiomics Unit, Alma Mater Studiorum - University of Bologna, 40138, Bologna, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy
| | - Marco Candela
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy
| | - Amanda G Henry
- Department of Archaeological Sciences, Faculty of Archaeology, Leiden University, Leiden, The Netherlands.
- Naturalis Biodiversity Center, Leiden, The Netherlands.
| |
Collapse
|
17
|
Jung YS, Song NE, Oh SY, Park YK, Kim YJ, Seong H, You SM, Jung DH, Shin D, Lee MG, Lim MC, Han NS. Advances in in vitro cultivation techniques for comprehensive analysis of human gut microbiome. Biotechnol Adv 2025; 82:108595. [PMID: 40374084 DOI: 10.1016/j.biotechadv.2025.108595] [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: 12/12/2024] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/17/2025]
Abstract
The role of gut microbiota in human health and disease is becoming increasingly recognized. Historically, the impact of human gut microbiota on health has been studied using clinical trials and animal models. However, clinical studies often struggle with controlling variables and pinpointing disease-causing factors, while animal models fall short of accurately replicating the human gut environment. Additionally, continuous sample collection for gut microbiota analysis in vivo presents significant ethical and technical challenges. To address these limitations, in vitro fermentation models have emerged as promising alternatives. These models aim to simulate the structural and functional characteristics of the human gut in a controlled setting, offering valuable insights into microbial behavior. This review highlights current knowledge and technological advances in in vitro cultivation systems for human gut microbiota, focusing on key elements such as three-dimensional scaffolds, culture media, fermentation systems, and analytical techniques. By examining these components, the review establishes a framework for improving methods to cultivate and study human gut microbiota, enhancing research methodologies for better understanding microbial interactions, behavior, and adaptation in diverse environments.
Collapse
Affiliation(s)
- Young Sung Jung
- Korea Food Research Institute, Wanju, Jeollabuk-do 55365, Republic of Korea; Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Nho-Eul Song
- Korea Food Research Institute, Wanju, Jeollabuk-do 55365, Republic of Korea
| | - Seo Yeong Oh
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Young Kyoung Park
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea
| | - Ye-Jin Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hyunbin Seong
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sang-Mook You
- Center for Bio-Based Chemistry, Korea Research Institute of Chemical Technology, Ulsan 44429, Republic of Korea
| | - Dong-Hyun Jung
- Division of Food and Nutrition, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Donghyun Shin
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Mi-Gi Lee
- Bio-Center, Gyeonggi-do Business and Science Accelerator, Suwon 16229, Republic of Korea
| | - Min-Cheol Lim
- Korea Food Research Institute, Wanju, Jeollabuk-do 55365, Republic of Korea.
| | - Nam Soo Han
- Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea.
| |
Collapse
|
18
|
Fernandez E, Wargo JA, Helmink BA. The Microbiome and Cancer: A Translational Science Review. JAMA 2025:2833859. [PMID: 40354071 DOI: 10.1001/jama.2025.2191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Importance Growing evidence suggests that microbes located within the gastrointestinal tract and other anatomical locations influence the development and progression of diseases such as cancer. Observations Clinical and preclinical evidence suggests that microbes in the gastrointestinal tract and other anatomical locations, such as the respiratory tract, may affect carcinogenesis, development of metastases, cancer treatment response, and cancer treatment-related adverse effects. Within tumors of patients with cancer, microbes may affect response to treatment, and therapies that reduce or eliminate these microbes may improve outcomes in patients with cancer. Modulating gastrointestinal tract (gut) microbes through fecal microbiota transplant and other strategies such as dietary intervention (eg, high-fiber diet intervention) has improved outcomes in small studies of patients treated with cancer immunotherapy. In contrast, disruption of the gut microbiota by receipt of broad-spectrum antibiotics prior to treatment with cancer immunotherapy has been associated with poorer overall survival and higher rates of adverse effects in patients treated with immune checkpoint blockade for solid tumors and also with chimeric antigen receptor T-cell therapy for hematologic malignancies. Conclusions and Relevance Microbes in the gut and other locations in the body may influence the development and progression of cancer and may affect the response to adverse effects from cancer therapy. Future therapies targeting microbes in the gut and other locations in the body could potentially improve outcomes in patients with cancer.
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Beth A Helmink
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
19
|
Barak N, Brekhman V, Aharonovich D, Lotan T, Sher D. Jellyfish blooms through the microbial lens: temporal changes, cross-species and Jellyfish-water comparisons. ENVIRONMENTAL MICROBIOME 2025; 20:49. [PMID: 40346699 PMCID: PMC12063254 DOI: 10.1186/s40793-025-00714-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Jellyfish blooms have significant ecological and economic impacts, yet the microbial communities associated with these blooms remain poorly understood, despite their potential influence on host fitness and microbial communities in the surrounding water. In this study, we explored temporal and tissue-specific variations in the microbiota of Rhopilema nomadica, the dominant jellyfish species in the Eastern Mediterranean Sea, across winter and summer blooms. During late summer blooms, microbial richness declined, coinciding with an increase in Endozoicomonas and unclassified Rickettsiales, while Tenacibaculum predominantly characterized winter blooms. Tissue-specific analyses revealed bacterial groups that were more consistently associated with different jellyfish tissues (e.g., Bacteroides in the bell and Simkaniaceae in the gonads), suggesting different microbial niches within the host. Furthermore, some key bacteria associated with R. nomadica, including Endozoicomonas, unclassified Rickettsiales, and Bacteroides were detected in the surrounding bloom water but absent from remote seawater, suggesting potential localized transmission dynamics between jellyfish and their immediate marine environment. Finally, a comparative analysis with nine additional jellyfish species identified recurring microbial taxa, including Endozoicomonas, Mycoplasma, and Spiroplasma, though no universal core microbiota was observed. This study represents the first exploration of microbial dynamics within R. nomadica blooms and the most comprehensive analysis of jellyfish-associated microbiomes across bloom stages and tissues to date. Our findings reveal complex relationships between jellyfish species, bloom progression, their microbial communities, and the surrounding seawater.
Collapse
Affiliation(s)
- Noga Barak
- Department of Marine Biology, The Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, 3103301, Israel
| | - Vera Brekhman
- Department of Marine Biology, The Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, 3103301, Israel
| | - Dikla Aharonovich
- Department of Marine Biology, The Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, 3103301, Israel
| | - Tamar Lotan
- Department of Marine Biology, The Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, 3103301, Israel.
| | - Daniel Sher
- Department of Marine Biology, The Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, 3103301, Israel.
| |
Collapse
|
20
|
Song CH, Kim N, Choi Y, Kim S, Kim KS, Park MH, Lee SH, Lee DH. Beneficial effect of consuming milk containing only A2 beta-casein on gut microbiota: A single-center, randomized, double-blind, cross-over study. PLoS One 2025; 20:e0323016. [PMID: 40338897 PMCID: PMC12061139 DOI: 10.1371/journal.pone.0323016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/01/2025] [Indexed: 05/10/2025] Open
Abstract
Cow milk contains essential nutrients, with β-casein existing in A1 and A2 forms. Studies suggest that A2 milk (containing only A2 β-casein) may offer gastrointestinal (GI) benefits compared to A1/A2 milk (containing both forms). This study investigated the effects of A2 milk consumption on the gut microbiota of South Korean cohort experiencing GI discomfort after consuming A1/A2 milk. Thirty-five participants with GI discomfort after milk consumption were included. Stool DNA was analyzed using 16S rRNA gene sequencing before and after consuming either A1/A2 or A2 milk. Beta diversity analysis using the generalized UniFrac distance method revealed a significant shift in gut microbiota composition after A2 milk consumption (p = 0.04), but no significant change after consuming A1/A2 milk. Significant differences in gut microbiota composition were found between A1/A2 and A2 milk drinkers after milk consumption (p = 0.031). Alpha diversity indices remained unchanged. Notable increases in beneficial microbes, including Bifidobacterium and Blautia, were observed after A2 milk intake. Linear discriminant analysis Effect Size (LEfSe) analysis identified significant enrichment of Actinobacteria, particularly Bifidobacterium longum and Blautia wexlerae, in the A2 group. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) analysis highlighted enriched transport systems related to energy, peptides, sugars, and raffinose family oligosaccharides in the A2 group. Spearman correlation showed significant associations between Bifidobacterium, Blautia, and enhanced transport systems exclusively in the A2 group. Two weeks of A2 milk consumption led to significant alterations in gut microbiota, promoting beneficial microbes and related functions. A2 milk could be a suitable alternative for subjects who experience milk-intake-related GI discomfort.
Collapse
Affiliation(s)
- Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Seulgi Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kyung Su Kim
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Min Hee Park
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Sang Hee Lee
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Origüela V, Lopez-Zaplana A. Gut Microbiota: An Immersion in Dysbiosis, Associated Pathologies, and Probiotics. Microorganisms 2025; 13:1084. [PMID: 40431257 PMCID: PMC12113704 DOI: 10.3390/microorganisms13051084] [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: 03/17/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
The importance of the microbiome, particularly the gut microbiota and its implications for health, is well established. However, an increasing number of studies further strengthen the link between an imbalanced gut microbiota and a greater predisposition to different diseases. The gut microbiota constitutes a fundamental ecosystem for maintaining human health. Its alteration, known as dysbiosis, is associated with a wide range of conditions, including intestinal, metabolic, immunological, or neurological pathologies, among others. In recent years, there has been a substantial increase in knowledge about probiotics-bacterial species that enhance health or address various diseases-with numerous studies reporting their benefits in preventing or improving these conditions. This review aims to analyze the most common pathologies resulting from an imbalance in the gut microbiota, as well as detail the most important and known gut probiotics, their functions, and mechanisms of action in relation to these conditions.
Collapse
Affiliation(s)
- Valentina Origüela
- Department of Physiology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain;
| | | |
Collapse
|
22
|
Singh S, Saini V, Jha HC. The role of secondary genomes in neurodevelopment and co-evolutionary dynamics. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:245-297. [PMID: 40414634 DOI: 10.1016/bs.irn.2025.03.008] [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
This chapter examines how human biology and microbial "secondary genomes" have co-evolved to shape neurodevelopment through the gut-brain axis. Microbial communities generate metabolites that cross blood-brain and placental barriers, influencing synaptogenesis, immune responses, and neural circuit formation. Simultaneously, Human Accelerated Regions (HARs) and Endogenous Retroviruses (ERVs) modulate gene expression and immune pathways, determining which microbes thrive in the gut and impacting brain maturation. These factors converge to form a dynamic host-microbe dialogue with significant consequences for neurodevelopmental disorders (NDD), including autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and schizophrenia. Building on evolutionary perspectives, the chapter elucidates how genetic and immune mechanisms orchestrate beneficial and pathological host-microbe interactions in early brain development. It then explores therapeutic strategies, such as probiotics, prebiotics, fecal microbiota transplantation, and CRISPR-driven microbial engineering, targeting gut dysbiosis to mitigate or prevent neurodevelopmental dysfunctions. Furthermore, innovative organ-on-chip models reveal mechanistic insights under physiologically relevant conditions, offering a translational bridge between in vitro experiments and clinical applications. As the field continues to evolve, this work underscores the translational potential of manipulating the microbiome to optimize neurological outcomes. It enriches our understanding of the intricate evolutionary interplay between host genomes and the microbial world.
Collapse
Affiliation(s)
- Siddharth Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India.
| | - Vaishali Saini
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India.
| |
Collapse
|
23
|
D’Urso F, Paladini F, Miraglia A, D’Amuri A, Chieppa M, Pollini M, Broccolo F. Translating Patent Innovation into Clinical Practice: Two Decades of Therapeutic Advancements in Dysbiosis Management. Microorganisms 2025; 13:1064. [PMID: 40431238 PMCID: PMC12114573 DOI: 10.3390/microorganisms13051064] [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: 02/10/2025] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Dysbiosis, characterized by a microbial imbalance, particularly within the gut microbiota, has emerged as a significant health concern linked to various diseases. This study analyzed 8097 patent documents from The Lens database (2005-2024) to examine global innovation trends in dysbiosis management. The patent filings showed exponential growth, peaking at 1222 documents in 2022, with the United States leading in publications (4361 documents). The analysis revealed three primary innovation clusters: bacterial-based therapeutics (44.8% of patents), specific therapeutic applications (27.6%), and diagnostic methods (15.9%). The disease associations predominantly included inflammatory conditions, infections, and cancer. The patent classifications highlighted a significant focus on probiotic development and microbiota modulation. The surge in patent activity since 2014 correlates with advances in DNA sequencing technology and the growing recognition of dysbiosis's role in human health. This analysis provides valuable insights into the evolving landscape of microbiome therapeutics and future directions for dysbiosis management.
Collapse
Affiliation(s)
- Fabiana D’Urso
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy; (F.P.); (A.M.); (A.D.); (M.C.); (M.P.)
| | | | | | | | | | | | - Francesco Broccolo
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy; (F.P.); (A.M.); (A.D.); (M.C.); (M.P.)
| |
Collapse
|
24
|
Gao F, Cheng W, Ma Y, Yu B, Lang X, Jin X, Wang J, Liu X, Di C, Wang H, Ye F, Zhao T, Chen W, Li Q. Electron-beam FLASH whole brain irradiation induced a unique changes of intestinal flora. Mol Med 2025; 31:165. [PMID: 40316930 PMCID: PMC12049017 DOI: 10.1186/s10020-024-01053-w] [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: 07/15/2024] [Accepted: 12/18/2024] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Whole-brain radiotherapy (WBRT) is an important way to treat multiple metastases. Ultra-high dose rate (FLASH) can avoid neurotoxicity caused by conventional irradiation, it has attracted much attention. This study aims to study the difference of irradiation-induced intestinal flora between conventional dose rate and FLASH WBRT. METHODS WBRT with 10 Gy was performed with electron-beam conventional irradiation (2 Gy/s) and electron-beam FLASH (eFLASH) irradiation (230 Gy/s). The intestinal feces and whole brain of mice were isolated after behavioral evaluation at 1st, 3rd and 10th weeks post-irradiation. HE staining and immunofluorescence were used to access the level of brain damage. The differences in intestinal microbes and transcription levels were detected by 16S rRNA gene sequencing and transcriptome sequencing, respectively. RESULTS eFLASH irradiation significantly reduced radiation neurotoxicity and had a long-term protective effect on cognitive function and learning and memory ability. Compared with conventional irradiation, eFLASH irradiation not only up-regulated the expression of genes related to neuronal regeneration and digestive system, but also induced more abundant intestinal microflora, especially the "probiotics" such as Lachnospiraceae and others, which were proved to play a role in radiation protection, increased significantly after eFLASH irradiation. The up-regulated microbiota after eFLASH irradiation was significantly positively correlated with genes related to neuronal development and regeneration, while significantly negatively correlated with genes related to inhibitory synapses. Additionally, conventional irradiation down-regulated microbial metabolism-related pathways, while FLASH did not. CONCLUSIONS In summary, we explored the unique gut microbiota changes induced by eFLASH WBRT for the first time, providing a theoretical basis for exploring the mechanism of action of FLASH.
Collapse
Affiliation(s)
- Feifei Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Cheng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanxi Ma
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
| | - Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinle Lang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianxin Wang
- Institute of Applied Electronics, China Academy of Engineering Physics, Mianyang, 621900, China
| | - Xianhong Liu
- Zhongjiu Flash Medical Technology Co., Ltd., Mianyang, 621000, China
| | - Cuixia Di
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Wang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
| | - Fei Ye
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, Gansu Province, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
25
|
Metwaly A, Kriaa A, Hassani Z, Carraturo F, Druart C, Arnauts K, Wilmes P, Walter J, Rosshart S, Desai MS, Dore J, Fasano A, Blottiere HM, Maguin E, Haller D. A Consensus Statement on establishing causality, therapeutic applications and the use of preclinical models in microbiome research. Nat Rev Gastroenterol Hepatol 2025; 22:343-356. [PMID: 40033063 DOI: 10.1038/s41575-025-01041-3] [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] [Accepted: 01/17/2025] [Indexed: 03/05/2025]
Abstract
The gut microbiome comprises trillions of microorganisms and profoundly influences human health by modulating metabolism, immune responses and neuronal functions. Disruption in gut microbiome composition is implicated in various inflammatory conditions, metabolic disorders and neurodegenerative diseases. However, determining the underlying mechanisms and establishing cause and effect is extremely difficult. Preclinical models offer crucial insights into the role of the gut microbiome in diseases and help identify potential therapeutic interventions. The Human Microbiome Action Consortium initiated a Delphi survey to assess the utility of preclinical models, including animal and cell-based models, in elucidating the causal role of the gut microbiome in these diseases. The Delphi survey aimed to address the complexity of selecting appropriate preclinical models to investigate disease causality and to study host-microbiome interactions effectively. We adopted a structured approach encompassing a literature review, expert workshops and the Delphi questionnaire to gather insights from a diverse range of stakeholders. Experts were requested to evaluate the strengths, limitations, and suitability of these models in addressing the causal relationship between the gut microbiome and disease pathogenesis. The resulting consensus statements and recommendations provide valuable insights for selecting preclinical models in future studies of gut microbiome-related diseases.
Collapse
Affiliation(s)
- Amira Metwaly
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University Munich, Freising, Germany
- ZIEL Institute for Food & Health, Technical University Munich, Freising, Germany
| | - Aicha Kriaa
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Federica Carraturo
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | | | - Kaline Arnauts
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jens Walter
- APC Microbiome Ireland, School of Microbiology, and Department of Medicine, University College Cork, Cork, Ireland
| | - Stephan Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Joel Dore
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, France
| | - Alessio Fasano
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
- Department of Paediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center,Massachusetts General Hospital Brigham, Harvard Medical School, Boston, MA, USA
| | - Hervé M Blottiere
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, France
- Nantes Université, INRAE, UMR1280, PhAN, Nantes, France
| | - Emmanuelle Maguin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| | - Dirk Haller
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University Munich, Freising, Germany.
- ZIEL Institute for Food & Health, Technical University Munich, Freising, Germany.
| |
Collapse
|
26
|
Jing T, Tang D. Intratumoral microbiota: a new force in the development and treatment of esophageal cancer. Clin Transl Oncol 2025; 27:1921-1932. [PMID: 39455494 DOI: 10.1007/s12094-024-03757-1] [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: 04/14/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Esophageal cancer (EC) ranks among the most prevalent cancers worldwide, with a particularly high incidence in the Asian population. Due to the inconspicuous nature of early symptoms, patients with esophageal cancer are typically diagnosed in the middle to late stages, resulting in suboptimal overall treatment outcomes. Consequently, there is an urgent need to explore and refine therapeutic strategies. Microorganisms have been identified in numerous tumor tissues, including EC, and these microorganisms are referred to as the intratumoral microbiome. Intratumoral microbiota and their metabolic byproducts can influence the progression and treatment of esophageal cancer through various mechanisms, such as modulating tumor cell metabolism and local immune responses. Therefore, the intratumoral microbiota may potentially serve as a target for the treatment of esophageal cancer. This review delineates the composition, origin, and diagnostic significance of intratumoral microbiota in esophageal cancer tissue, and discusses the mechanisms by which intratumoral microbiota contribute to the onset of esophageal cancer. In addition, the impact of intratumoral microbiota on the treatment of esophageal cancer and its intervention measures are also addressed.
Collapse
Affiliation(s)
- Tianyang Jing
- Clinical Medical College, Yangzhou University, Yangzhou, 22500, Jiangsu Province, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
27
|
Pope Q, Varma R, Tataru C, David MM, Fern X. Learning a deep language model for microbiomes: The power of large scale unlabeled microbiome data. PLoS Comput Biol 2025; 21:e1011353. [PMID: 40334224 PMCID: PMC12058177 DOI: 10.1371/journal.pcbi.1011353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/24/2025] [Indexed: 05/09/2025] Open
Abstract
We use open source human gut microbiome data to learn a microbial "language" model by adapting techniques from Natural Language Processing (NLP). Our microbial "language" model is trained in a self-supervised fashion (i.e., without additional external labels) to capture the interactions among different microbial taxa and the common compositional patterns in microbial communities. The learned model produces contextualized taxon representations that allow a single microbial taxon to be represented differently according to the specific microbial environment in which it appears. The model further provides a sample representation by collectively interpreting different microbial taxa in the sample and their interactions as a whole. We demonstrate that, while our sample representation performs comparably to baseline models in in-domain prediction tasks such as predicting Irritable Bowel Disease (IBD) and diet patterns, it significantly outperforms them when generalizing to test data from independent studies, even in the presence of substantial distribution shifts. Through a variety of analyses, we further show that the pre-trained, context-sensitive embedding captures meaningful biological information, including taxonomic relationships, correlations with biological pathways, and relevance to IBD expression, despite the model never being explicitly exposed to such signals.
Collapse
Affiliation(s)
- Quintin Pope
- School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, United States of America
| | - Rohan Varma
- School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, United States of America
| | - Christine Tataru
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Maude M David
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Xiaoli Fern
- School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
28
|
Muttiah B, Hanafiah A. Gut Microbiota and Cardiovascular Diseases: Unraveling the Role of Dysbiosis and Microbial Metabolites. Int J Mol Sci 2025; 26:4264. [PMID: 40362500 PMCID: PMC12072866 DOI: 10.3390/ijms26094264] [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: 04/09/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular diseases (CVDs), including heart failure (HF), hypertension, myocardial infarction (MI), and atherosclerosis, are increasingly linked to gut microbiota dysbiosis and its metabolic byproducts. HF, affecting over 64 million individuals globally, is associated with systemic inflammation and gut barrier dysfunction, exacerbating disease progression. Similarly, hypertension and MI correlate with reduced microbial diversity and an abundance of pro-inflammatory bacteria, contributing to vascular inflammation and increased cardiovascular risk. Atherosclerosis is also influenced by gut dysbiosis, with key microbial metabolites such as trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs) playing crucial roles in disease pathogenesis. Emerging evidence highlights the therapeutic potential of natural compounds, including flavonoids, omega-3 fatty acids, resveratrol, curcumin, and marine-derived bioactives, which modulate the gut microbiota and confer cardioprotective effects. These insights underscore the gut microbiota as a critical regulator of cardiovascular health, suggesting that targeting dysbiosis may offer novel preventive and therapeutic strategies. Further research is needed to elucidate underlying mechanisms and optimize microbiome-based interventions for improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- GUT Research Group, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
29
|
Yi K, Huang Y, Jiang Y, Zhou L. Causal relationship between gut microbiota and laryngeal cancer: a mendelian randomization analysis. Braz J Otorhinolaryngol 2025; 91:101634. [PMID: 40305979 PMCID: PMC12118545 DOI: 10.1016/j.bjorl.2025.101634] [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: 10/23/2024] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
OBJECTIVE Laryngeal cancer incidence is rising globally; the role of gut microbiota remains underexplored. This study aimed to establish a causal link between gut microbiota and laryngeal cancer to inform preventive and therapeutic strategies. METHODS Gut microbiota data from GWAS conducted by the MiBioGen consortium served as the exposure variable, with laryngeal cancer as the outcome variable. the exposure variable and the outcome variable were analyzed using Mendelian Randomization. The primary method was Inverse Variance Weighted analysis, with heterogeneity and pleiotropy assessed through Cochran's Q test, MR-Egger regression, and MR-PRESSO. RESULTS In the study, we identified five bacterial taxa with potential causal relationships with laryngeal cancer risk: Higher levels of Clostridiaceae1 (OR = 0.9993, 95% CI 0.9986-0.9999, p = 0.0463) and Turicibacter (OR = 0.9995, 95% CI 0.9989-0.9999, p = 0.0384) were linked to reduced cancer risk, while Mollicutes RF9 (OR = 1.0010, 95% CI 1.0003-1.0016, p = 0.0027), Euryarchaeota (OR = 1.0004, 95% CI 1.0001-1.0007, p = 0.0234), and Cyanobacteria (OR = 1.0005, 95% CI 1.0000-1.0009, p = 0.0464) were associated with increased risk. CONCLUSION Our findings suggest a causal relationship between gut microbiota composition and laryngeal cancer risk. Clostridiaceae1 and Turicibacter may play a protective role, while Mollicutes RF9, Euryarchaeota, and Cyanobacteria could contribute to increased cancer susceptibility. These insights highlight potential microbiome-based strategies for early detection, prevention, and therapeutic intervention in laryngeal cancer. LEVEL OF EVIDENCE Level 5.
Collapse
Affiliation(s)
- Kaiyan Yi
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yu Huang
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yun Jiang
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Lingling Zhou
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Campisciano G, Rupel K, D'Amico F, Tettamanti M, Vella F, Cason C, Comar M, Turroni S, Marcon G. Oral microbiota profile is related to cognitive status in centenarians: a clinical and biological study. Maturitas 2025; 198:108593. [PMID: 40315555 DOI: 10.1016/j.maturitas.2025.108593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
OBJECTIVES A growing body of evidence supports the potential role of the oral microbiota in influencing cognitive function. Centenarians, at the extreme end of the lifespan, are the ideal cohort to study the long-term effects of inflammaging. STUDY DESIGN Twenty-three centenarians were examined by a neurologist, neuropsychologist and dentist to assess cognitive status and oral health. They were also profiled for oral microbiota and inflammasome. RESULTS We found less alpha diversity in the oral microbiota of participants with dementia and an overall depletion of typical oral commensals, including Alloprevotella, Prevotella, Veillonella, Fusobacterium and Leptotrichia. The latter two were also underrepresented in edentulous compared with dentate subjects. Moreover, levels of pro-inflammatory cytokines and chemokines tended to be higher in participants with dementia. CONCLUSIONS Our data support a relationship between oral microbiota, cognitive status and inflammation, which deserves further exploration to counteract cognitive decline while promoting healthy aging.
Collapse
Affiliation(s)
| | - Katia Rupel
- Department of Medical Science, University of Trieste, Trieste, Italy
| | - Federica D'Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Mauro Tettamanti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS - Milano, Italy
| | - Filomena Vella
- ASUGI, Azienda Sanitaria Universitaria Giuliano Isontina -Trieste, Italy
| | - Carolina Cason
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - Trieste, Italy; Department of Medical Science, University of Trieste, Trieste, Italy
| | - Manola Comar
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - Trieste, Italy; Department of Medical Science, University of Trieste, Trieste, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| | - Gabriella Marcon
- Department of Medical Science, University of Trieste, Trieste, Italy; DMED - University of Udine, Udine, Italy
| |
Collapse
|
31
|
Thapa HB, Passegger CA, Fleischhacker D, Kohl P, Chen YC, Kalawong R, Tam-Amersdorfer C, Gerstorfer MR, Strahlhofer J, Schild-Prüfert K, Zechner EL, Blesl A, Binder L, Busslinger GA, Eberl L, Gorkiewicz G, Strobl H, Högenauer C, Schild S. Enrichment of human IgA-coated bacterial vesicles in ulcerative colitis as a driver of inflammation. Nat Commun 2025; 16:3995. [PMID: 40301356 PMCID: PMC12041585 DOI: 10.1038/s41467-025-59354-5] [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: 03/01/2024] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
The gut microbiome contributes to chronic inflammatory responses in ulcerative colitis (UC), but molecular mechanisms and disease-relevant effectors remain unclear. Here we analyze the pro-inflammatory properties of colonic fluid obtained during colonoscopy from UC and control patients. In patients with UC, we find that the pelletable effector fraction is composed mostly of bacterial extracellular vesicles (BEVs) that exhibit high IgA-levels and incite strong pro-inflammatory responses in IgA receptor-positive (CD89+) immune cells. Biopsy analyses reveal higher infiltration of CD89+ immune cells in the colonic mucosa from patients with UC than control individuals. Further studies show that IgA-coated BEVs, but not host-derived vesicles nor soluble IgA, are potent activators of pro-inflammatory responses in CD89+ cells. IgA-coated BEVs also exacerbate intestinal inflammation in a dextran sodium sulfate colitis model using transgenic mice expressing human CD89. Our data thus implicate a link between IgA-coated BEVs and intestinal inflammation via CD89+ immune cells, and also hint a potential new therapeutic target for UC.
Collapse
Affiliation(s)
- Himadri B Thapa
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Christina A Passegger
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | | | - Paul Kohl
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Yi-Chi Chen
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Ratchara Kalawong
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Carmen Tam-Amersdorfer
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Michael R Gerstorfer
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Jana Strahlhofer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Ellen L Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
- Field of Excellence Biohealth - University of Graz, Graz, Austria
| | - Andreas Blesl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Lukas Binder
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Georg A Busslinger
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Gregor Gorkiewicz
- BioTechMed, Graz, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Herbert Strobl
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
| | - Christoph Högenauer
- BioTechMed, Graz, Austria.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed, Graz, Austria.
- Field of Excellence Biohealth - University of Graz, Graz, Austria.
- Austrian Agency for Health and Food Safety (AGES), Institute for Medical Microbiology and Hygiene, Graz, Austria.
| |
Collapse
|
32
|
Li J, Lv Y, Shao M, Lv D, Fu Z, Guo P, Li Q, Shang Q. Fermentation of Alginate and Its Oligosaccharides by the Human Gut Microbiota: Structure-Property Relationships and New Findings Focusing on Bacteroides xylanisolvens. Nutrients 2025; 17:1424. [PMID: 40362733 PMCID: PMC12074081 DOI: 10.3390/nu17091424] [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: 03/30/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Alginate and its oligosaccharides (AOS) are widely used in the food industry all over the world. However, how they are fermented by the human gut microbiota has not been fully elucidated. Here, we aim to explore the structure-property relationships of the fermentation of these carbohydrates by the human gut microbiota. Methods: High-performance liquid chromatography, 16S rRNA gene amplicon high-throughput sequencing, whole genome sequencing, and metabolome analysis were used to study the fermentation of alginate and AOS by the human gut microbiota. Results and Conclusions: Low-molecular-weight alginate and AOS were more fermentable than alginate. Moreover, fermentation of AOS with a molecular weight (Mw) of 0.8 kDa produced higher amounts of acetate and butyrate than that with a Mw of 0.3 kDa. B. xylanisolvens was a keystone species responsible for the fermentation. Additionally, each B. xylanisolvens strain was characterized with a unique capability for AOS fermentation. Specifically, B. xylanisolvens P19-10, a bacterium isolated from healthy human colon, exhibited the best fermentation capacity. Genomic analysis suggested that B. xylanisolvens P19-10 was armed with a plethora of carbohydrate-active enzymes. Additionally, the polysaccharide lyase family 6_1 was identified as a candidate enzyme responsible for the utilization of AOS. Moreover, fermentation of AOS by B. xylanisolvens P19-10 was associated with significant changes in bacterial metabolites and metabolic pathways. Future perspectives: Our study provides novel mechanistic insights into the fermentation of alginate and AOS by human gut microbiota, which has applications for the development of new carbohydrate-based nutraceuticals and foods.
Collapse
Affiliation(s)
- Jiayi Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (J.L.); (Y.L.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Youjing Lv
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (J.L.); (Y.L.)
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China; (M.S.); (D.L.); (Z.F.); (P.G.)
| | - Meng Shao
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China; (M.S.); (D.L.); (Z.F.); (P.G.)
| | - Depeng Lv
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China; (M.S.); (D.L.); (Z.F.); (P.G.)
| | - Zhiliang Fu
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China; (M.S.); (D.L.); (Z.F.); (P.G.)
| | - Peng Guo
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China; (M.S.); (D.L.); (Z.F.); (P.G.)
| | - Quancai Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (J.L.); (Y.L.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China; (M.S.); (D.L.); (Z.F.); (P.G.)
| | - Qingsen Shang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Key Laboratory of Glycoscience and Glycotherapeutics, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (J.L.); (Y.L.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China; (M.S.); (D.L.); (Z.F.); (P.G.)
| |
Collapse
|
33
|
Su X, Tian Z, Fang Y, Zhou S, Ma S. Effects of high-dose glucocorticoids on gut microbiota in the treatment of Graves' ophthalmopathy. Microbiol Spectr 2025:e0246724. [PMID: 40261021 DOI: 10.1128/spectrum.02467-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
Many studies indicate the gut microbiome is associated with diseases caused by administering high-dose glucocorticoids (GCs), such as hypertension, hyperglycemia, and osteoporosis. However, the association between intestinal flora and the use of high-dose GCs remains elusive. We aimed to characterize gut microbiome in Graves' ophthalmopathy (GO) patients after administering high-dose GCs. In this study, 20 primary GO patients were recruited. The differences in gut microbiota of GO patients before and after administering high-dose GCs were analyzed by 16S rDNA sequencing technology. Untargeted metabolomic analysis was used to examine the differences in gut metabolites between two groups. There were significant differences in α and β diversities of gut microbiota in GO patients before and after administering high-dose GCs. The random forest analysis indicated that three intestinal bacteria (Faecalibacterium, Streptococcus, and Prevotella) could distinguish the two groups with the highest accuracy, which was proven by receiver operator characteristic curve and linear discriminant analysis effect size analysis. The short-chain fatty acid-producing ability in GO patients' gut after high-dose GC administration was significantly decreased. The 5-hydroxytryptamine levels significantly increased in the gut of GO patients after administering high-dose GCs. Our study suggests that high-dose GC administration causes the changes in gut microbiome and metabolites. Moreover, the altered flora and metabolites are related to hypertension, hyperglycemia, and osteoporosis. These findings can help understand the development of side effects caused by high-dose GCs and can be further used to develop potential probiotics to facilitate the prevention for those side effects.IMPORTANCEFor the first time, we revealed that gut microbiome and metabolome in Graves' ophthalmopathy patients after high-dose glucocorticoid (GC) administration significantly changed, and the altered flora and metabolites are related to hypertension, hyperglycemia, and osteoporosis. These findings can help understand the development of side effects caused by high-dose GCs and can be further used to develop potential probiotics to facilitate the prevention for those side effects.
Collapse
Affiliation(s)
- Xinhuan Su
- Department of Endocrinology, Department of Geriatrics, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhenyu Tian
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yalun Fang
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Shengnan Zhou
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shizhan Ma
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
34
|
Samira R, Monsur M, Trina NA. How the Built Environment Shapes Children's Microbiome: A Systematic Review. Microorganisms 2025; 13:950. [PMID: 40284786 PMCID: PMC12029762 DOI: 10.3390/microorganisms13040950] [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: 04/01/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
This systematic review aims to synthesize key empirical findings to understand how various elements of the built environment influence the microbiome concerning children's health and well-being. A comprehensive literature search was conducted across multiple databases, focusing on studies that examined the relationship between built environment factors and the microbiome aspects of childhood. A total of 42 studies were included in the final systematic review. We analyzed these studies from a range of different lenses, starting with basic research questions and variables to types of built environments, age groups of children, sampling strategy, bioinformatics, and the biological methods utilized. This review highlights a growing emphasis on children's exposure to nature within built environments and its potential to beneficially alter the microbiome, with 38% of studies addressing this link. It also identifies a significant research gap in connecting built environment design features (landscape and/or architectural) to microbiome outcomes and associated health, behavioral, and mental health impacts on children. The findings indicate that interventions aimed at improving the built environment quality via design could foster healthier microbiomes in children's environments. This review underscores the need for interdisciplinary research and policy initiatives that integrate microbiome science with built environment design to promote children's health and well-being.
Collapse
Affiliation(s)
- Rozalynne Samira
- Department of Plant and Soil Science, Institute of Genomics for Crop Abiotic Tolerance (IGCAST), Texas Tech University, 1006 Canton Ave, Lubbock, TX 79409, USA
| | - Muntazar Monsur
- Department of Landscape Architecture (DoLA), Davis College of Agricultural Sciences and Natural, Texas Tech University, 2904 15th St., Lubbock, TX 79409, USA;
| | - Nazia Afrin Trina
- Department of Landscape Architecture (DoLA), Davis College of Agricultural Sciences and Natural, Texas Tech University, 2904 15th St., Lubbock, TX 79409, USA;
| |
Collapse
|
35
|
Cai W, Haddad M, Haddad R, Kesten I, Hoffman T, Laan R, Westfall S, Defaye M, Abdullah NS, Wong C, Brown N, Tansley S, Lister KC, Hooshmandi M, Wang F, Lorenzo LE, Hovhannisyan V, Ho-Tieng D, Kumar V, Sharif B, Thurairajah B, Fan J, Sahar T, Clayton C, Wu N, Zhang J, Bar-Yoseph H, Pitashny M, Krock E, Mogil JS, Prager-Khoutorsky M, Séguéla P, Altier C, King IL, De Koninck Y, Brereton NJB, Gonzalez E, Shir Y, Minerbi A, Khoutorsky A. The gut microbiota promotes pain in fibromyalgia. Neuron 2025:S0896-6273(25)00252-1. [PMID: 40280127 DOI: 10.1016/j.neuron.2025.03.032] [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: 02/27/2024] [Revised: 02/09/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025]
Abstract
Fibromyalgia is a prevalent syndrome characterized by widespread pain in the absence of evident tissue injury or pathology, making it one of the most mysterious chronic pain conditions. The composition of the gut microbiota in individuals with fibromyalgia differs from that of healthy controls, but its functional role in the syndrome is unknown. Here, we show that fecal microbiota transplantation from fibromyalgia patients, but not from healthy controls, into germ-free mice induces pain and numerous molecular phenotypes that parallel known changes in fibromyalgia patients, including immune activation and metabolomic profile alterations. Replacing the fibromyalgia microbiota with a healthy microbiota substantially alleviated pain in mice. An open-label trial in women with fibromyalgia (Registry MOH_2021-11-04_010374) showed that transplantation of a healthy microbiota is associated with reduced pain and improved quality of life. We conclude that altered gut microbiota has a role in fibromyalgia pain, highlighting it as a promising target for therapeutic interventions.
Collapse
Affiliation(s)
- Weihua Cai
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - May Haddad
- Rambam Health Campus, Haifa, Israel; Ruth and Bruce Rapaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | | | - Inbar Kesten
- Rambam Health Campus, Haifa, Israel; Clinical Research Institute at Rambam (CRiR), Haifa, Israel
| | | | - Reut Laan
- Ruth and Bruce Rapaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Susan Westfall
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; McGill Centre for Microbiome Research, McGill University, Montreal, QC, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nasser S Abdullah
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Nicole Brown
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Shannon Tansley
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Kevin C Lister
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Mehdi Hooshmandi
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Feng Wang
- Faculty of Dentistry, CERVO Brain Research Center, University Laval, Quebec City, QC, Canada
| | - Louis-Etienne Lorenzo
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, University Laval, Quebec City, QC, Canada
| | | | - David Ho-Tieng
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Vibhu Kumar
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Behrang Sharif
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada
| | - Bavanitha Thurairajah
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; McGill Centre for Microbiome Research, McGill University, Montreal, QC, Canada
| | - Jonathan Fan
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Tali Sahar
- Alan Edwards Pain Management Unit, McGill University Health Centre, Montreal, QC, Canada
| | | | - Neil Wu
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Ji Zhang
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Haggai Bar-Yoseph
- Rambam Health Campus, Haifa, Israel; Ruth and Bruce Rapaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel; Clinical Research Institute at Rambam (CRiR), Haifa, Israel
| | - Milena Pitashny
- Rambam Health Campus, Haifa, Israel; Ruth and Bruce Rapaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel; Clinical Research Institute at Rambam (CRiR), Haifa, Israel
| | - Emerson Krock
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Jeffrey S Mogil
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada; Departments of Psychology and Anesthesia, McGill University, Montreal, QC, Canada
| | | | - Philippe Séguéla
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Irah L King
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; McGill Centre for Microbiome Research, McGill University, Montreal, QC, Canada
| | - Yves De Koninck
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, University Laval, Quebec City, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Nicholas J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Emmanuel Gonzalez
- McGill Centre for Microbiome Research, McGill University, Montreal, QC, Canada; Canadian Center for Computational Genomics, McGill University and Genome Quebec Innovation Center, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Yoram Shir
- Department of Anesthesia, McGill University, Montreal, QC, Canada; Alan Edwards Pain Management Unit, McGill University Health Centre, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada.
| | - Amir Minerbi
- Rambam Health Campus, Haifa, Israel; Ruth and Bruce Rapaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada.
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC, Canada; McGill Centre for Microbiome Research, McGill University, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada.
| |
Collapse
|
36
|
Kim D, El Khoury S, Pérez-Carrascal OM, DeSousa C, Jung DK, Bohley S, Wijaya L, Trang K, Shapira M. Gut microbiome remodeling provides protection from an environmental toxin. iScience 2025; 28:112209. [PMID: 40230520 PMCID: PMC11995125 DOI: 10.1016/j.isci.2025.112209] [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: 08/22/2024] [Revised: 01/09/2025] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
Gut microbiomes contribute to animal health and fitness. The immense biochemical diversity of bacteria holds particular potential for neutralizing environmental toxins and thus helping hosts deal with new toxic challenges. To explore this potential, we used Caenorhabditis elegans harboring a defined microbiome, and the antibiotic neomycin as a model toxin, differentially affecting microbiome strains, and also toxic to worms. Worms exposed to neomycin showed delayed development and reduced survival but were protected when colonized with neomycin-resistant Stenotrophomonas. 16S rRNA sequencing, bacterial load quantification, genetic manipulation, and behavioral assays showed that protection was linked to enrichment of Stenotrophomonas carrying a neomycin-modifying enzyme. Enrichment was facilitated by altered bacterial competition in the gut, as well as by KGB-1/JNK-dependent behavioral changes. While microbiome remodeling conferred toxin resistance, it was associated with reduced infection resistance and metabolic changes. These findings suggest that microbiome adaptation can help animals cope with stressors but may have long-term consequences that add to effects of direct intoxication.
Collapse
Affiliation(s)
- Dan Kim
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sarah El Khoury
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Catherin DeSousa
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Da Kyung Jung
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Seneca Bohley
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lila Wijaya
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kenneth Trang
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael Shapira
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
37
|
Wang X, Yuan X, Lin Y, Lan Q, Mei S, Cai M, Lei F, Dong B, Zhao M, Zhu B. Exploratory study on source identification of saliva stain and its TsD inference based on the microbial relative and absolute abundance. Int J Legal Med 2025:10.1007/s00414-025-03456-8. [PMID: 40240552 DOI: 10.1007/s00414-025-03456-8] [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: 06/27/2024] [Accepted: 02/16/2025] [Indexed: 04/18/2025]
Abstract
In recent years, it has become a major research trend to obtain the microbial relative abundance in common body fluid stains at the crime scenes through 16S rRNA next generation sequencing to explore the effectiveness in forensic application. However, few scholars have combined the determination of tissue sources of body fluid stains with the inference of time since deposition (TsD) based on the relative and absolute abundance of microorganism in the same sample in a single study. Therefore, we preliminarily used the four abundant saliva-related bacteria to distinguish fresh saliva, saliva stains (exposure ≤60 days) from the four kinds of fresh body fluids and epidermal tissue, simultaneously assessed the temporal variation regularities in both microbial relative and absolute abundance in these saliva stains. Quantitative real-time PCR results demonstrated that fresh saliva samples and saliva stains exposed for up to 60 days still retained two or more abundant saliva-related bacteria, demonstrating sufficient discriminative power to identify saliva stain from other four kinds of body fluids and tissue. Microbial compositions and temporal analyses of 56 saliva samples revealed that many phyla and genera with abundance higher than 1% had different temporal variation regularities in relative and absolute abundance data, except for some genera such as Neisseria, etc. Beta diversity analysis indicated greater differences in absolute quantitative data among fresh saliva samples and saliva stains at different time points compared with relative quantitative data. The support vector machine (svm) model based on microbial relative or absolute abundance both have the prediction accuracy higher than 0.8 in classifying saliva stains deposited at 1 h, 1 day, and 7 to 60 days. This study combined the tissue origin identification and TsD inference of saliva stains, and the absolute quantitative technology was applied for the first time to the TsD inference of saliva stains. And the results indicated that using the absolute quantitative technology might be more suitable for early TsD inference (within 14 days) of saliva stains in this study, which helped to accurately infer the TsD of saliva stains, providing an important clue for forensic investigation.
Collapse
Affiliation(s)
- Xi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Yuan
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Yifeng Lin
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Qiong Lan
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Shuyan Mei
- School of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Meiming Cai
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Fanzhang Lei
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Bonan Dong
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Ming Zhao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Bofeng Zhu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China.
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Upadhyay R, Mani S, Sevanan M. Microbiome-based dietary supplements for better development and healthy brain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:329-368. [PMID: 40414637 DOI: 10.1016/bs.irn.2025.03.009] [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
Microbiome-based dietary supplements have gained attention for their role in enhancing brain development and cognitive health. The gut microbiome influences neurological functions through the gut-brain axis, impacting neurotransmitter production, immune regulation, and metabolic pathways. Dysbiosis is linked to neurological disorders such as Alzheimer's, Parkinson's, and autism spectrum disorders. This chapter explores dietary interventions targeting the microbiome, emphasising probiotics, prebiotics, and postbiotics. Additionally, AI and machine learning are transforming microbiome research by enabling personalised supplementation strategies tailored to individual gut profiles. Ethical challenges, including data privacy and algorithmic bias, are also discussed. Advances in big data analytics and predictive modelling are paving the way for precision-targeted interventions to optimise brain health. While microbiome-based therapies hold great promise, further clinical validation and regulatory frameworks are needed to ensure their efficacy and accessibility. This chapter highlights the future potential of microbiome-targeted strategies in neuroprotection and cognitive well-being.
Collapse
Affiliation(s)
- Riddhi Upadhyay
- Division of Biotechnology, Karunya Institute of Technology and Sciences (Deemed University), Coimbatore, Tamil Nadu, India
| | - Sugumar Mani
- Palamur Biosciences Private Limited, Mahabubnagar, Telangana, India
| | - Murugan Sevanan
- Division of Biotechnology, Karunya Institute of Technology and Sciences (Deemed University), Coimbatore, Tamil Nadu, India.
| |
Collapse
|
39
|
Yu M, Lu Y, Zhang W, Gong X, Hao Z, Xu L, Wen Y, Dong X, Han F, Gao X. Preliminary analysis of salivary microbiota in catathrenia (nocturnal groaning) using machine learning algorithms. J Oral Microbiol 2025; 17:2489613. [PMID: 40247863 PMCID: PMC12004722 DOI: 10.1080/20002297.2025.2489613] [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: 10/29/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025] Open
Abstract
Objectives The present study aimed to characterize the salivary microbiota in patients with catathrenia and to longitudinally validate potential biomarkers after treatment with mandibular advancement devices (MAD). Materials and methods Twenty-two patients with catathrenia (12 M/10 F, median age 28 y) and 22 age-matched control volunteers (8 M/14 F, median age 30 y) were included in the cross-sectional study. Video/audio polysomnography was conducted for diagnosis. All patients received treatment with custom-fit MAD and were followed for one month. Ten patients (6 M/4 F) underwent post-treatment PSG. Salivary samples were collected, and microbial characteristics were analyzed using 16S rRNA gene sequencing. The 10-fold cross-validated XGBoost and nested Random Forest Classifier machine learning algorithms were utilized to identify potential biomarkers. Results In the cross-sectional study, patients with catathrenia had lower α-diversity represented by Chao 1, Faith's phylogenetic diversity (pd), and observed species. Beta-diversity based on the Bray-Curtis dissimilarities revealed a significant inter-group separation (p = 0.001). The inter-group microbiota distribution was significantly different on the phylum and family levels. The treatment of MAD did not alter salivary microbiota distribution significantly. Among the most important genera in catathrenia and control classification identified by machine learning algorithms, four genera, Alloprevotella, Peptostreptococcaceae_XI_G1, Actinomyces and Rothia, changed significantly with MAD treatment. Correlation analysis revealed that Alloprevotella was negatively related to the severity of catathrenia (r2= -0.63, p < 0.001). Conclusions High-throughput sequencing revealed that the salivary microbiota composition was significantly altered in patients with catathrenia. Some characteristic genera (Alloprevotella, Peptostreptococcaceae_XI_G1, Actinomyces, and Rothia) could be potential biomarkers sensitive to treatment. Future studies are needed to confirm and determine the mechanisms underlying these findings.
Collapse
Affiliation(s)
- Min Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P.R. China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, P.R. China
- National Center for Stomatology, Beijing, P.R. China
| | - Yujia Lu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P.R. China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, P.R. China
- National Center for Stomatology, Beijing, P.R. China
| | - Wanxin Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P.R. China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, P.R. China
- National Center for Stomatology, Beijing, P.R. China
| | - Xu Gong
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P.R. China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, P.R. China
- National Center for Stomatology, Beijing, P.R. China
| | - Zeliang Hao
- Department of Stomatology, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| | - Liyue Xu
- Sleep Division, Peking University People’s Hospital, Beijing, P.R. China
| | - Yongfei Wen
- Sleep Division, Peking University People’s Hospital, Beijing, P.R. China
| | - Xiaosong Dong
- Sleep Division, Peking University People’s Hospital, Beijing, P.R. China
| | - Fang Han
- Sleep Division, Peking University People’s Hospital, Beijing, P.R. China
| | - Xuemei Gao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P.R. China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, P.R. China
- National Center for Stomatology, Beijing, P.R. China
| |
Collapse
|
40
|
Parthasarathy S, Giridharan B, Panigrahi J, Konyak LM, Jamir N, Tharumasivam SV. Abnormal microbiota due to prenatal antibiotic as a possible risk factor for Attention-Deficit / Hyperactivity Disorder (ADHD). INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:299-328. [PMID: 40414636 DOI: 10.1016/bs.irn.2025.03.007] [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
One of the major issues modern medicine faces is the increasing use of antibiotics in reaction to the increased incidence of infectious agents. The current trend of antibiotic overuse contributes to microbial dysbiosis. Recent studies have hypothesized that antibiotic exposure during pregnancy, which alters the composition of the microbiome, might increase the likelihood of attention deficit hyperactivity disorder (ADHD). In addition to the ongoing discussion about the potential links between antibiotic usage, microbiome dysbiosis, and ADHD, there is a rising interest in integrating AI and ML into healthcare practices. Diagnosis, treatment plans, and prognoses are all enhanced by these technological advancements. Remote monitors or telemedicine monitoring are among the management techniques described in this chapter for effectively managing illnesses. Also discussed are ways to halt the progression of diseases by preventative measures that use biosensor technology and dietary approaches. Personalized treatment programs, disease progression stages, and prognosis evaluations are all made possible with the use of artificial intelligence and machine learning. By using these technologies to provide individualized therapy, healthcare practitioners may get a better understanding of ADHD and perhaps improve patient outcomes.
Collapse
Affiliation(s)
| | - Bupesh Giridharan
- Department of Forestry, Nagaland University (Hqrs.), Lumami, Nagaland, India; Department of Biotechnology, Berhampur University, Bhanja Bihar, Ganjam, Odisha, India.
| | - Jogeswar Panigrahi
- Department of Biotechnology, Berhampur University, Bhanja Bihar, Ganjam, Odisha, India
| | - Longnyu M Konyak
- Department of Forestry, Nagaland University (Hqrs.), Lumami, Nagaland, India
| | - Nokenketla Jamir
- Department of Forestry, Nagaland University (Hqrs.), Lumami, Nagaland, India
| | | |
Collapse
|
41
|
Han B, Zhang Y, Feng X, Yang J, Wang B, Fang J, Wang Z, Zhu J, Niu G, Guo Y. The power of microbes: the key role of gut microbiota in the initiation and progression of colorectal cancer. Front Oncol 2025; 15:1563886. [PMID: 40297806 PMCID: PMC12034544 DOI: 10.3389/fonc.2025.1563886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer (CRC) is ranked as the third most prevalent malignancy and is a leading cause of cancer-related mortality globally, significantly affecting the health and longevity of middle-aged individuals and the elderly. The primary risk factors for CRC are mainly due to unhealthy dietary habits and lifestyle choices, and they have been shown to profoundly influence the composition of the gut microbiota. Given that dietary patterns are critical determinants of gut microbial diversity, a compelling association exists between gut microbiota and the pathogenesis of CRC. Recent research has increasingly focused on the intricate interplay between gut microbiota and CRC, exploring its role in disease initiation, progression, and the modulation of host immune responses. Investigations have demonstrated that certain specific microbial communities can promote inflammation, disrupt metabolic pathways, and produce carcinogenic compounds, thereby contributing to the development of CRC. Conversely, a diverse and balanced gut microbiome may confer protective effects against cancer through mechanisms such as the production of short-chain fatty acids and the enhancement of intestinal barrier integrity. This article provides a comprehensive overview of the characteristics of the gut microbial community and its complex relationship with CRC. It highlights potential mechanisms through which gut microbiota may influence CRC pathogenesis, including chronic inflammation, toxins, metabolites, epigenetic dysregulation, and immune regulatory dysfunction. Additionally, this review summarizes innovative strategies for CRC prevention and treatment, emphasizing the therapeutic potential of probiotics and natural plant extracts. By elucidating these connections, this work aims to enhance the understanding of the gut microbiome's role in CRC.
Collapse
Affiliation(s)
- Bo Han
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Yongfeng Zhang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Xue Feng
- Department of Cardiology, 63650 Military Hospital, Urumqi, China
| | - Jun Yang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Baolin Wang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Jiang Fang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Zhigang Wang
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Jun Zhu
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Ge Niu
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| | - Youxiang Guo
- Department of General Surgery, 63650 Military Hospital, Urumqi, China
| |
Collapse
|
42
|
Jagadesan S, Guda C. MetaDAVis: An R shiny application for metagenomic data analysis and visualization. PLoS One 2025; 20:e0319949. [PMID: 40193328 PMCID: PMC11975103 DOI: 10.1371/journal.pone.0319949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/11/2025] [Indexed: 04/09/2025] Open
Abstract
The human microbiome exerts tremendous influence on maintaining a balance between human health and disease. High-throughput sequencing has enabled the study of microbial communities at an unprecedented resolution. Generation of massive amounts of sequencing data has also presented novel challenges to analyzing and visualizing data to make biologically relevant interpretations. We have developed an interactive Metagenome Data Analysis and Visualization (MetaDAVis) tool for 16S rRNA as well as the whole genome sequencing data analysis and visualization to address these challenges using an R Shiny application. MetaDAVis can perform six different types of analyses that include: i) Taxonomic abundance distribution; ii) Alpha and beta diversity analyses; iii) Dimension reduction tasks using PCA, t-SNE, and UMAP; iv) Correlation analysis using taxa- or sample-based data; v) Heatmap generation; and vi) Differential abundance analysis. MetaDAVis creates interactive and dynamic figures and tables from multiple methods enabling users to easily understand their data using different variables. Our program is user-friendly and easily customizable allowing those without any programming background to perform comprehensive data analyses using a standalone or web-based interface.
Collapse
Affiliation(s)
- Sankarasubramanian Jagadesan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
43
|
Leigh J, Skidmore B, Wong A, Maleki Vareki S, Ng TL. Exploring the Microbiome's Impact on Glioma and Brain Metastases: Insights into Development, Progression, and Treatment Response-A Scoping Review. Cancers (Basel) 2025; 17:1228. [PMID: 40227812 PMCID: PMC11988003 DOI: 10.3390/cancers17071228] [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: 03/02/2025] [Revised: 03/29/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
Background: The human microbiome plays a crucial role in health and disease. Dysbiosis, an imbalance of microorganisms, has been implicated in cancer development and treatment response, including in primary brain tumors and brain metastases, through interactions mediated by the gut-brain axis. This scoping review synthesizes current evidence on the relationship between the human microbiome and brain tumors. Methods: A systematic search of five electronic databases was conducted by an expert librarian, using controlled vocabulary and keywords. A targeted grey literature search in Google Scholar and clinical trial registries was also undertaken. Eligible studies included primary research involving human patients, or in vivo, or in vitro models of glioma or brain metastasis, with a focus on the microbiome's role in tumor development, treatment response, and outcomes. Results: Out of 584 citations screened, 40 studies met inclusion criteria, comprising 24 articles and 16 conference abstracts. These included 12 human studies, 16 using mouse models, 7 combining both, and 5 employing large datasets or next-generation sequencing of tumor samples. Thirty-one studies focused on primary brain tumors, six on brain metastases, and three on both. Of the 20 studies examining dysbiosis in tumor development, 95% (n = 19) found an association with tumor growth. Additionally, 71.4% (n = 5/7) of studies reported that microbiome alterations influenced treatment efficacy. Conclusions: Although the role of the gut-brain axis in brain tumors is still emerging and is characterized by heterogeneity across studies, existing evidence consistently supports a relationship between the gut microbiome and both brain tumor development and treatment outcomes.
Collapse
Affiliation(s)
- Jennifer Leigh
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, ON K1Y 4E9, Canada;
- Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Becky Skidmore
- Skidmore Research & Information Consulting Inc., Ottawa, ON, Canada;
| | - Adrian Wong
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Saman Maleki Vareki
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada;
- Verspeeten Family Cancer Centre, London Health Sciences Research Institute, London, ON N6A 5W9, Canada
- Department of Oncology, Western University, London, ON N6A 3K7, Canada
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
| | - Terry L. Ng
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital Cancer Centre, Ottawa, ON K1Y 4E9, Canada;
| |
Collapse
|
44
|
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
|
45
|
Zouiouich S, Wan Y, Vogtmann E, Porras C, Abnet CC, Shi J, Sinha R. Sample Size Estimations Based on Human Microbiome Temporal Stability Over 6 Months: A Shallow Shotgun Metagenome Sequencing Analysis. Cancer Epidemiol Biomarkers Prev 2025; 34:588-597. [PMID: 39927868 DOI: 10.1158/1055-9965.epi-24-0839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/04/2024] [Accepted: 02/06/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Biological factors affect the human microbiome, highlighting the need for reasonably estimating sample sizes in future population studies. METHODS We assessed the temporal stability of fecal microbiome diversity, species composition, and genes and functional pathways through shallow shotgun metagenome sequencing. Using intraclass correlation coefficients (ICC), we measured biological variability over 6 months. We estimated case numbers for 1:1 or 1:3 matched case-control studies, considering significance levels of 0.05 and 0.001 with 80% power, based on the collected fecal specimens per participant. RESULTS The fecal microbiome's temporal stability over 6 months varied (ICC < 0.6) for most alpha and beta diversity metrics. Heterogeneity was seen in species, genes, and pathways stability (ICC, 0.0-0.9). Detecting an OR of 1.5 per SD required 1,000 to 5,000 cases (0.05 significance for alpha and beta; 0.001 for species, genes, and pathways) with equal cases and controls. Low-prevalence species needed 15,102 cases, and high-prevalence species required 3,527. Similar needs applied to genes and pathways. In a 1:3 matched case-control study with one fecal specimen, 10,068 cases were needed for low-prevalence species and 2,351 for high-prevalence species. For ORs of 1.5 with multiple specimens, cases needed for low-prevalence species were 15,102 (one specimen), 8,267 (two specimens), and 5,989 (three specimens). CONCLUSIONS Detecting disease associations requires a large number of cases. Repeating prediagnostic samples and matching cases to more controls could decrease the needed number of cases for such detections. IMPACT Our results will help future epidemiologic study designs and implement well-powered microbiome studies.
Collapse
Affiliation(s)
- Semi Zouiouich
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Yunhu Wan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Emily Vogtmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Carolina Porras
- Costa Rican Agency for Biomedical Research-INCIENSA Foundation, San José, Costa Rica
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
46
|
Gilbert JA, Azad MB, Bäckhed F, Blaser MJ, Byndloss M, Chiu CY, Chu H, Dugas LR, Elinav E, Gibbons SM, Gilbert KE, Henn MR, Ishaq SL, Ley RE, Lynch SV, Segal E, Spector TD, Strandwitz P, Suez J, Tropini C, Whiteson K, Knight R. Clinical translation of microbiome research. Nat Med 2025; 31:1099-1113. [PMID: 40217076 DOI: 10.1038/s41591-025-03615-9] [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: 10/27/2024] [Accepted: 02/26/2025] [Indexed: 04/18/2025]
Abstract
The landscape of clinical microbiome research has dramatically evolved over the past decade. By leveraging in vivo and in vitro experimentation, multiomic approaches and computational biology, we have uncovered mechanisms of action and microbial metrics of association and identified effective ways to modify the microbiome in many diseases and treatment modalities. This Review explores recent advances in the clinical application of microbiome research over the past 5 years, while acknowledging existing barriers and highlighting opportunities. We focus on the translation of microbiome research into clinical practice, spearheaded by Food and Drug Administration (FDA)-approved microbiome therapies for recurrent Clostridioides difficile infections and the emerging fields of microbiome-based diagnostics and therapeutics. We highlight key examples of studies demonstrating how microbiome mechanisms, metrics and modifiers can advance clinical practice. We also discuss forward-looking perspectives on key challenges and opportunities toward integrating microbiome data into routine clinical practice, precision medicine and personalized healthcare and nutrition.
Collapse
Affiliation(s)
- Jack A Gilbert
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
- Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Martin J Blaser
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Mariana Byndloss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Fransisco, San Francisco, CA, USA
- Department of Medicine, Division of Infectious Diseases, University of California, San Fransisco, San Francisco, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Hiutung Chu
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines, La Jolla, CA, USA
| | - Lara R Dugas
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- Microbiome and Cancer Division, DKFZ, Heidelberg, Germany
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | - Katharine E Gilbert
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | | | - Suzanne L Ishaq
- School of Food and Agriculture, University of Maine, Orono, ME, USA
- Microbes and Social Equity working group, Orono, ME, USA
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Biology, Tübingen, Germany
| | - Susan V Lynch
- Benioff Center for Microbiome Medicine, Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- ZOE Ltd, London, UK
| | | | - Jotham Suez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Carolina Tropini
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
47
|
Tahiri M, Gilbert JA. Examining the potential prebiotic effect of almonds. J Appl Microbiol 2025; 136:lxaf078. [PMID: 40156575 DOI: 10.1093/jambio/lxaf078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/01/2025]
Abstract
Almonds, a nutrient-dense food rich in dietary fiber, polyphenols, and unsaturated fatty acids, exhibit significant potential as a functional food with prebiotic effects. Prebiotics selectively stimulate the growth and activity of beneficial gut microbiota, leading to improved gut and systemic health. This review synthesizes evidence from in vitro studies, clinical trials, and systematic reviews to elucidate the prebiotic effects of almond consumption. Almonds enhance gut microbiota diversity and composition, particularly increasing beneficial bacteria such as Bifidobacterium and Roseburia, while promoting the production of short-chain fatty acids (SCFAs), such as butyrate, which are critical for gut barrier integrity and inflammation modulation. The presence of polyphenols, such as proanthocyanidins, contributes to their antioxidative and antimicrobial properties, further supporting microbiome health. Despite variability in study outcomes, likely due to differences in population health status, study design, and almond preparation methods, the cumulative findings underscore almonds' role as a potential prebiotic food with the potential to improve cardiovascular health. Continued research focusing on individualized responses and standardized methodologies is essential to fully harness the health benefits of almond consumption.
Collapse
Affiliation(s)
- Maha Tahiri
- Nutrition Sustainability Strategies, St Petersburg, FL 33702, United States
- Tufts University, Friedmann School of Nutrition Science and Policy, Boston, MA 02111, United States
| | - Jack A Gilbert
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92023, United States
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92023, United States
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92023, United States
| |
Collapse
|
48
|
Melby MK, Watanabe K, Haraoui LP. Addressing Antimicrobial Resistance by Changing Our Relationships with Microbes: Lessons from Japan. Dela J Public Health 2025; 11:28-31. [PMID: 40331181 PMCID: PMC12051887 DOI: 10.32481/djph.2025.04.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Antimicrobial resistance (AMR) is a global health problem, but it is only the 'tip of the iceberg' of microbial disruption caused by antibiotics. Under the surface, cultural factors such as understandings of and attitudes toward microbes may play a significant role influencing relationships between humans and microbes. Western strategies to address pathogenic microbes and AMR often overlook the symbiotic relationship humans share with beneficial microbes (our microbiota), viewing humans as separate from nature and focusing on control. Given the increasing prevalence of novel pathogens, antimicrobial resistance, and chronic illnesses associated with disturbed microbiota (dysbiosis), alternative approaches are needed. Cross-cultural studies may provide ways forward. An exploration of Japanese perspectives on microbes through the lens of food and health reveals practices where microbes are often regarded as partners and friends rather than foes.
Collapse
Affiliation(s)
- Melissa K Melby
- University of Delaware, Department of Anthropology, USA
- CIFAR Humans & the Microbiome Program
| | | | - Louis-Patrick Haraoui
- CIFAR Humans & the Microbiome Program
- Department of Microbiology and Infectious Disease, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Centre de Recherche Charles-Le Moyne, CISSS de la Montérégie -Centre - Hôpital Charles-Le Moyne, Greenfield Park, Québec, Canada
| |
Collapse
|
49
|
Abolhasani FS, Vaghefinanekaran N, Yarahmadi A, Akrami S, Mirmahdavi S, Yousefi MH, Afkhami H, Shafiei M. Outer membrane vesicles in gram-negative bacteria and its correlation with pathogenesis. Front Immunol 2025; 16:1541636. [PMID: 40236702 PMCID: PMC11996793 DOI: 10.3389/fimmu.2025.1541636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/11/2025] [Indexed: 04/17/2025] Open
Abstract
There is a widespread distribution of gram-negative bacteria worldwide, which are responsible for the deaths of numerous patients each year. The illnesses they cause can be localized and systemic, and these bacteria possess several key virulence factors that contribute to their pathogenicity. In recent years, several distinct mechanisms of pathogenesis have evolved that remain largely unknown to scientists and medical experts. Among these, outer membrane vesicles (OMVs) are undoubtedly one of the most significant factors influencing virulence. OMVs contain various bacterial compounds and can have diverse effects on host organisms and the immune system, potentially exacerbating disease and inflammation while evading immune responses. This review comprehensively examines the role of OMVs in bacterial pathogenesis, their interaction with host cells, and their potential biomedical applications. Understanding the molecular mechanisms governing OMV biogenesis and function could pave the way for novel antimicrobial strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Fatemeh Sadat Abolhasani
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Sousan Akrami
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Mirmahdavi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Hasan Yousefi
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Hamed Afkhami
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, School of Medicine, Shahed University, Tehran, Iran
| | - Morvarid Shafiei
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
50
|
Best L, Dost T, Esser D, Flor S, Gamarra AM, Haase M, Kadibalban AS, Marinos G, Walker A, Zimmermann J, Simon R, Schmidt S, Taubenheim J, Künzel S, Häsler R, Franzenburg S, Groth M, Waschina S, Rosenstiel P, Sommer F, Witte OW, Schmitt-Kopplin P, Baines JF, Frahm C, Kaleta C. Metabolic modelling reveals the aging-associated decline of host-microbiome metabolic interactions in mice. Nat Microbiol 2025; 10:973-991. [PMID: 40140706 PMCID: PMC11964932 DOI: 10.1038/s41564-025-01959-z] [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: 05/03/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025]
Abstract
Aging is accompanied by considerable changes in the gut microbiome, yet the molecular mechanisms driving aging and the role of the microbiome remain unclear. Here we combined metagenomics, transcriptomics and metabolomics from aging mice with metabolic modelling to characterize host-microbiome interactions during aging. Reconstructing integrated metabolic models of host and 181 mouse gut microorganisms, we show a complex dependency of host metabolism on known and previously undescribed microbial interactions. We observed a pronounced reduction in metabolic activity within the aging microbiome accompanied by reduced beneficial interactions between bacterial species. These changes coincided with increased systemic inflammation and the downregulation of essential host pathways, particularly in nucleotide metabolism, predicted to rely on the microbiota and critical for preserving intestinal barrier function, cellular replication and homeostasis. Our results elucidate microbiome-host interactions that potentially influence host aging processes. These pathways could serve as future targets for the development of microbiome-based anti-aging therapies.
Collapse
Affiliation(s)
- Lena Best
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Dost
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Daniela Esser
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel/Lübeck, Germany
| | - Stefano Flor
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Andy Mercado Gamarra
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Madlen Haase
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - A Samer Kadibalban
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Georgios Marinos
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- CAU Innovation GmbH, Kiel University, Kiel, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, Helmholtz Munich, Neuherberg, Germany
| | - Johannes Zimmermann
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- Evolutionary Ecology and Genetics, Zoological Institute, Kiel University, Kiel, Germany
- Antibiotic resistance group, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Rowena Simon
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Silvio Schmidt
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Jan Taubenheim
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sven Künzel
- Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Marco Groth
- Core Facility Next-Generation Sequencing, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Silvio Waschina
- Nutriinformatics, Institute of Human Nutrition and Food Science, Kiel University, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Felix Sommer
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Otto W Witte
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Munich, Neuherberg, Germany
- Institute of Analytical Food Chemistry, Technical University München, Freising, Germany
| | - John F Baines
- Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Christiane Frahm
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|