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are technical, such as lack of standardized stool collection protocols, inconsistent DNA extraction methods, and outdated sequencing technologies.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
|
Boulund U, Thorsen J, Trivedi U, Tranæs K, Jiang J, Shah SA, Stokholm J. The role of the early-life gut microbiome in childhood asthma. Gut Microbes 2025; 17:2457489. [PMID: 39882630 PMCID: PMC11784655 DOI: 10.1080/19490976.2025.2457489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/05/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Asthma is a chronic disease affecting millions of children worldwide, and in severe cases requires hospitalization. The etiology of asthma is multifactorial, caused by both genetic and environmental factors. In recent years, the role of the early-life gut microbiome in relation to asthma has become apparent, supported by an increasing number of population studies, in vivo research, and intervention trials. Numerous early-life factors, which for decades have been associated with the risk of developing childhood asthma, are now being linked to the disease through alterations of the gut microbiome. These factors include cesarean birth, antibiotic use, breastfeeding, and having siblings or pets, among others. Association studies have highlighted several specific microbes that are altered in children developing asthma, but these can vary between studies and disease phenotype. This demonstrates the importance of the gut microbial ecosystem in asthma, and the necessity of well-designed studies to validate the underlying mechanisms and guide future clinical applications. In this review, we examine the current literature on the role of the gut microbiome in childhood asthma and identify research gaps to allow for future microbial-focused therapeutic applications in asthma.
Collapse
Affiliation(s)
- Ulrika Boulund
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Jonathan Thorsen
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Urvish Trivedi
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Section of Microbiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kaare Tranæs
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Jie Jiang
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Shiraz A. Shah
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Takyi E, Nirmalkar K, Adams J, Krajmalnik-Brown R. Interventions targeting the gut microbiota and their possible effect on gastrointestinal and neurobehavioral symptoms in autism spectrum disorder. Gut Microbes 2025; 17:2499580. [PMID: 40376856 PMCID: PMC12087657 DOI: 10.1080/19490976.2025.2499580] [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: 08/25/2024] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/18/2025] Open
Abstract
Autism spectrum disorder (ASD) is a developmental disorder that is characterized by deficits in social communication and restricted, repetitive, and stereotyped behaviors. In addition to neurobehavioral symptoms, children with ASD often have gastrointestinal symptoms (e.g. constipation, diarrhea, gas, abdominal pain, reflux). Several studies have proposed the role of gut microbiota and metabolic disorders in gastrointestinal symptoms and neurodevelopmental dysfunction in ASD patients; these results offer promising avenues for novel treatments of this disorder. Interventions targeting the gut microbiota - such as fecal microbiota transplant (FMT), microbiota transplant therapy (MTT), probiotics, prebiotics, synbiotics, antibiotics, antifungals, and diet - promise to improve gut health and can potentially improve neurological symptoms. The modulation of the gut microbiota using MTT in ASD has shown beneficial and long-term effects on GI symptoms and core symptoms of autism. Also, the modulation of the gut microbiota to resemble that of typically developing individuals seems to be the most promising intervention. As most of the studies carried out with MTT are open-label studies, more extensive double-blinded randomized control trials are needed to confirm the efficacy of MTT as a therapeutic option for ASD. This review examines the current clinical research evidence for the use of interventions that target the microbiome - such as antibiotics, antifungals, probiotics/prebiotics, synbiotics, and MTT - and their effectiveness in changing the gut microbiota and improving gastrointestinal and neurobehavioral symptoms in ASD.
Collapse
Affiliation(s)
- Evelyn Takyi
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Khemlal Nirmalkar
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - James Adams
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
4
|
Schoultz I, Claesson MJ, Dominguez‐Bello MG, Fåk Hållenius F, Konturek P, Korpela K, Laursen MF, Penders J, Roager H, Vatanen T, Öhman L, Jenmalm MC. Gut microbiota development across the lifespan: Disease links and health-promoting interventions. J Intern Med 2025; 297:560-583. [PMID: 40270478 PMCID: PMC12087861 DOI: 10.1111/joim.20089] [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] [Indexed: 04/25/2025]
Abstract
The gut microbiota plays a pivotal role in human life and undergoes dynamic changes throughout the human lifespan, from infancy to old age. During our life, the gut microbiota influences health and disease across life stages. This review summarizes the discussions and presentations from the symposium "Gut microbiota development from infancy to old age" held in collaboration with the Journal of Internal Medicine. In early infancy, microbial colonization is shaped by factors such as mode of delivery, antibiotic exposure, and milk-feeding practices, laying the foundation for subsequent increased microbial diversity and maturation. Throughout childhood and adolescence, microbial maturation continues, influencing immune development and metabolic health. In adulthood, the gut microbiota reaches a relatively stable state, influenced by genetics, diet, and lifestyle. Notably, disruptions in gut microbiota composition have been implicated in various inflammatory diseases-including inflammatory bowel disease, Type 1 diabetes, and allergies. Furthermore, emerging evidence suggests a connection between gut dysbiosis and neurodegenerative disorders such as Alzheimer's disease. Understanding the role of the gut microbiota in disease pathogenesis across life stages provides insights into potential therapeutic interventions. Probiotics, prebiotics, and dietary modifications, as well as fecal microbiota transplantation, are being explored as promising strategies to promote a healthy gut microbiota and mitigate disease risks. This review focuses on the gut microbiota's role in infancy, adulthood, and aging, addressing its development, stability, and alterations linked to health and disease across these critical life stages. It outlines future research directions aimed at optimizing the gut microbiota composition to improve health.
Collapse
Affiliation(s)
- Ida Schoultz
- School of Medical SciencesFaculty of Medicine and Health Örebro UniversityOrebroSweden
| | | | - Maria Gloria Dominguez‐Bello
- Department of Biochemistry & Microbiology and of AnthropologyRutgers University–New BrunswickNew BrunswickNew JerseyUSA
| | - Frida Fåk Hållenius
- Department of Food Technology, Engineering and NutritionLund UniversityLundSweden
| | - Peter Konturek
- Department of Medicine, Thuringia Clinic SaalfeldTeaching Hospital of the University JenaJenaGermany
| | - Katri Korpela
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | | | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtthe Netherlands
| | - H. Roager
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenFrederiksbergDenmark
| | - Tommi Vatanen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFinland
- Department of Microbiology, Faculty of Agriculture and ForestryUniversity of HelsinkiHelsinkiFinland
- Research Program for Clinical and Molecular Metabolism, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Liggins InstituteUniversity of AucklandAucklandNew Zealand
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maria C. Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden
| |
Collapse
|
5
|
Fernández-Pato A, Sinha T, Garmaeva S, Gulyaeva A, Kuzub N, Roux S, Fu J, Kurilshikov A, Zhernakova A. Early-life development of the gut virome and plasmidome: A longitudinal study in cesarean-born infants. Cell Rep 2025; 44:115731. [PMID: 40413742 DOI: 10.1016/j.celrep.2025.115731] [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: 09/20/2024] [Revised: 03/14/2025] [Accepted: 04/30/2025] [Indexed: 05/27/2025] Open
Abstract
Mobile genetic elements (MGE) are critical yet understudied determinants of gut microbiome composition. In this secondary analysis of a randomized controlled trial (NCT06030713), we characterized the gut virome and plasmidome in 195 samples from 28 mother-infant dyads delivered by cesarean section. Infant mobilome increases in richness over the first 6 postnatal weeks, demonstrating high individual-specificity and temporal stability, establishing a personal persistent mobilome. Formula-fed infants exhibit greater mobilome richness than breastfed infants, with plasmid composition being influenced by antibiotic exposure and birth weight. Plasmids constitute a reservoir of antibiotic resistance genes (ARG), with around 5% of infant gut plasmid taxonomic units carrying ARG. Notably, ARG profiles do not differ with antibiotic exposure at birth. Mother-infant sharing of viral and plasmid strains primarily occurs after 6 months of age. Overall, our integrative analysis offers insights into the dynamics, modulation, and origin of MGE in the developing gut microbiome.
Collapse
Affiliation(s)
- Asier Fernández-Pato
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands
| | - Trishla Sinha
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands
| | - Sanzhima Garmaeva
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands
| | - Anastasia Gulyaeva
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands
| | - Nataliia Kuzub
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands
| | - Simon Roux
- US Department of Energy, Joint Genome Institute, Berkeley, CA 94720, USA
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands.
| |
Collapse
|
6
|
Feng J, Xu B, Magnuson JT, Wang J, Gao Y, Qiu W, Xuan R. Exposure of infants to antibiotics via cord blood, breast milk, and formula: A review on exposure level, temporal variation, and risk assessment. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138665. [PMID: 40403372 DOI: 10.1016/j.jhazmat.2025.138665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/26/2025] [Accepted: 05/17/2025] [Indexed: 05/24/2025]
Abstract
The pervasive use of antibiotics across various sectors, including agriculture, medicine, and aquaculture, has led to a notable increase in environmental antibiotic residues. This phenomenon has raised significant public concern regarding the potential health risks antibiotics may pose, particularly to vulnerable populations such as infants. However, the conceptualization of exposure routes of antibiotics to infants and the associated health risks has not been conducted. This review summarized three main pathways infants are exposed to antibiotics, including umbilical cord blood, breast milk, and infant formula. Antibiotic exposure levels in infants were synthesized, examining spatial and temporal trends in antibiotic concentrations across different media through clinical testing. We also analyzed the doses of antibiotics consumed by infants over time through breast milk and formula, evaluating the associated risks. Furthermore, we explored the potential adverse effects of early-life antibiotic exposure on the infant gut microbiota, physical development, and multiple organ systems. Given the global significance of antibiotic distribution, it is pertinent to comprehensively monitor antibiotic concentrations in infants and conduct longitudinal follow-up studies on their growth and development, accurately quantifying and assessing the impacts on fetal and infant health.
Collapse
Affiliation(s)
- Jiating Feng
- Gynaecology and obstetrics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315020, China; Health Science Center, Ningbo University, Ningbo, Zhejiang Province 315211, China
| | - Bentuo Xu
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution, School of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Jason T Magnuson
- US Geological Survey, Columbia Environmental Research Center, Columbia, MO 65201, USA
| | - Jiayi Wang
- Gynaecology and obstetrics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315020, China
| | - Yajie Gao
- Gynaecology and obstetrics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315020, China
| | - Wenhui Qiu
- Guangdong-Hong Kong Joint Laboratory for Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Rongrong Xuan
- Gynaecology and obstetrics, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province 315020, China.
| |
Collapse
|
7
|
Zhao X, Lei C, Wang Y, Zhang L, Tang W, Li Z, Jiang L, Li X. Impact of TLR5 overexpression on gut microbiota and metabolites: Insights into Salmonella infection outcomes. Microb Pathog 2025; 205:107708. [PMID: 40381961 DOI: 10.1016/j.micpath.2025.107708] [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: 12/05/2024] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND The gut microbiome and the host immune system work together to maintain intestinal health and protect against infections. Toll-like receptor 5 (TLR5) recognizes bacterial flagellin and plays a crucial role in this network. However, the precise role of TLR5 in regulating gut microbiota and resistance to infection remains unclear. This study utilized a TLR5 intestine-specific overexpression mouse model to explore these interactions and their impact on Salmonella infection. METHODS TLR5 intestine-specific overexpression mice (TLR5+/+) and wild-type (WT) mice were infected with Salmonella to assess TLR5's protective role. Survival time, fecal Salmonella load, and intestinal tissue integrity were evaluated. Subsequently, 16S rRNA sequencing and LC-MS-based metabolomics were performed to analyze gut microbiota composition and fecal metabolites. Fecal microbiota transplantation (FMT) and metabolite transplantation experiments were conducted to evaluate the functional impact of microbiota and metabolites on resistance to infection. RESULTS TLR5 overexpression significantly improved survival time and reduced fecal Salmonella load, demonstrating its protective role against infection. 16S rRNA sequencing revealed enrichment of beneficial taxa, while metabolomic analysis identified altered metabolites in TLR5+/+ mice. Although fecal microbiota and metabolite transplantation did not fully replicate the protective effects, these experiments highlighted the important roles of microbiota and metabolites in infection outcomes, with limitations in transplantation likely affecting the results. These findings underscore the significance of microbiota and metabolites in TLR5-mediated gut immunity. CONCLUSION TLR5 overexpression significantly alters gut microbiota and metabolite profiles, contributing to improved infection outcomes. These findings highlight the critical roles of microbiota and metabolites in TLR5-mediated immunity and provide a foundation for exploring targeted strategies to enhance resistance against enteric pathogens.
Collapse
Affiliation(s)
- Xu Zhao
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Lei
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Yue Wang
- Department of Biological Medicines, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
| | - Lu Zhang
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Weiyi Tang
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Zhi Li
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Lijing Jiang
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| | - Xiang Li
- Department of Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| |
Collapse
|
8
|
Laue HE, Willis AD, Wang F, MacDougall MC, Xu Y, Karagas MR, Madan JC, Fleisch AF, Lanphear BP, Cecil KM, Yolton K, Chen A, Buckley JP, Braun JM. Early-life and concurrent predictors of the healthy adolescent microbiome in a cohort study. Genome Med 2025; 17:50. [PMID: 40340756 PMCID: PMC12060534 DOI: 10.1186/s13073-025-01481-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/25/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND The microbiome of adolescents is poorly understood, as are factors influencing its composition. We aimed to describe the healthy adolescent microbiome and identify early-life and concurrent predictors of its composition. METHODS We performed metagenomic sequencing of 247 fecal specimens from 167 adolescents aged 11-14 years participating in the Health Outcomes and Measures of the Environment (HOME) Study, a longitudinal pregnancy and birth cohort (Cincinnati, OH). We described common features of the adolescent gut microbiome and applied self-organizing maps (SOMs)-a machine-learning approach-to identify distinct microbial profiles (n = 4). Using prospectively collected data on sociodemographic characteristics, lifestyle, diet, and sexual maturation, we identified early-life and concurrent factors associated with microbial diversity and phylum relative abundance with linear regression models and composition with Kruskal-Wallis and Fisher's exact tests. RESULTS We found that household income and other sociodemographic factors were consistent predictors of the microbiome, with higher income associated with lower diversity and differential relative abundances of Firmicutes (increased) and Actinobacteria (decreased). Sexual maturation, distinct from chronological age, was related to higher diversity in females and differences in phylum relative abundances and compositional profiles in both males and females. CONCLUSIONS Our study suggests that adolescence is a unique window for gut microbial composition and that it may be shaped by both early-life and concurrent exposures, highlighting its potential in future epidemiologic research.
Collapse
Grants
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- R35GM133420 NIGMS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- National Institute of General Medical Sciences
- National Center for Advancing Translational Sciences
Collapse
Affiliation(s)
- Hannah E Laue
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst School of Public Health and Health Sciences, 715 N. Pleasant Street, Arnold House 429, Amherst, MA, 01003, USA.
| | - Amy D Willis
- Department of Biostatistics, University of Washington Hans Rosling Center for Population Health, 3980 15 Avenue NE, Box 351617, Seattle, WA, 98195-1617, USA
| | - Fang Wang
- Department of Environmental Health Sciences, Columbia Mailman School of Public Health, 630 W 168th St, P&S 16-416, New York, NY, 10032, USA
| | - Melinda C MacDougall
- Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Yingying Xu
- Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Margaret R Karagas
- Department of Epidemiology, Dartmouth Geisel School of Medicine, One Medical Center Dr Lebanon, Lebanon, NH, 03756, USA
| | - Juliette C Madan
- Department of Epidemiology, Dartmouth Geisel School of Medicine, One Medical Center Dr Lebanon, Lebanon, NH, 03756, USA
- Department of Psychiatry, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Abby F Fleisch
- Center for Interdisciplinary and Population Health Research, Maine Institute for Research, Westbrook, ME, USA
- Pediatric Endocrinology and Diabetes, Maine Medical Center, 887 Congress St, Portland, ME, USA
| | - Bruce P Lanphear
- Faculty of Health Sciences, Simon Fraser University, Blusson Hall, 8888 University Dr, Burnaby, BC, Canada
| | - Kim M Cecil
- Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Radiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Environmental and Public Health Sciences, University of Cincinnati, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Kimberly Yolton
- Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Environmental and Public Health Sciences, University of Cincinnati, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Jessie P Buckley
- Department of Epidemiology, University of North Carolina at Chapel Hill, 2106-B McGavran-Greenberg Hall CB#7435, Chapel Hill, NC, 27599, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University, 121 S Main St, Providence, RI, USA
| |
Collapse
|
9
|
Farooq S, Talat A, Dhariwal A, Petersen FC, Khan AU. Transgenerational gut dysbiosis: Unveiling the dynamics of antibiotic resistance through mobile genetic elements from mothers to infants. Int J Antimicrob Agents 2025; 65:107458. [PMID: 39921114 DOI: 10.1016/j.ijantimicag.2025.107458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/04/2025] [Accepted: 01/29/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVES The initial microbial colonization of the gut is seeded by microbes transmitted from the mother's gut, skin, and vaginal tract. As the gut microbiome evolves, a few transmitted microbes persist throughout life. Understanding the impact of mother-to-neonate gut microbiome and antibiotic resistance genes (ARGs) transmission is crucial for establishing its role in infants' immunity against pathogens. METHODS This study primarily explores mother-neonate ARG transmission through 125 publicly available fecal metagenomes, isolated from eighteen mother-neonate pairs. RESULTS The core ARGs, detected in both mothers and their respective infants at all stages (birth, 1st, 2nd, 3rd, 4th, 8th and 12th months) included aminoglycosidases APH(3')-IIIa, Bifidobacterium adolescentis rpoB mutants conferring resistance to rifampicin, β-lactamases CblA-1, CfxA2, multidrug resistance gene CRP, diaminopyrimidine resistance gene dfrF, fluoroquinolone-resistance gene emrR, macrolide; lincosamide; streptogramin resistance gene ErmB, ErmG, macrolide resistance gene Mef(En2), nucleosidase SAT-4, and tetracycline-resistance genes tet(O), tet(Q), and tet(W). Most of these infants and mothers were not administered any antibiotics. In infants, ARGs were predominantly carried by Bacillota, Pseudomonadota, and Actinomycetota, similar to the mothers. The dominant ARG-carrying opportunistic pathogens were Escherichia coli, Klebsiella, and Streptococcus, found across all infant cohorts. All the core ARGs were associated with mobile genetic elements, signifying the role of horizontal gene transfer(HGT). We detected 132 virulence determinants, mostly E. coli-specific, including pilus chaperones, general secretion pathway proteins, type III secretion system effectors, and heme-binding proteins. CONCLUSIONS Maternal-neonate transmission of ARGs along with possible nosocomial infections, mode of delivery, breastfeeding versus formula feeding, and gestation period, must be considered for mother-neonate health.
Collapse
Affiliation(s)
- Samiya Farooq
- Antimicrobial Resistance Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India; Bioinformatics and Computational Biology Centre of DBT Government of India, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Absar Talat
- Antimicrobial Resistance Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India; Bioinformatics and Computational Biology Centre of DBT Government of India, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Achal Dhariwal
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | | | - Asad U Khan
- Antimicrobial Resistance Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India; Bioinformatics and Computational Biology Centre of DBT Government of India, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India.
| |
Collapse
|
10
|
Husain N, Kumar A, Anbazhagan AN, Gill RK, Dudeja PK. Intestinal luminal anion transporters and their interplay with gut microbiome and inflammation. Am J Physiol Cell Physiol 2025; 328:C1455-C1472. [PMID: 40047092 PMCID: PMC12023768 DOI: 10.1152/ajpcell.00026.2025] [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: 01/10/2025] [Revised: 01/29/2025] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
The intestine, as a critical interface between the external environment and the internal body, plays a central role in nutrient absorption, immune regulation, and maintaining homeostasis. The intestinal epithelium, composed of specialized epithelial cells, harbors apical anion transporters that primarily mediate the transport of chloride and bicarbonate ions, essential for maintaining electrolyte balance, pH homeostasis, and fluid absorption/secretion. In addition, the intestine hosts a diverse population of gut microbiota that plays a pivotal role in various physiological processes including nutrient metabolism, immune regulation, and maintenance of intestinal barrier integrity, all of which are critical for host gut homeostasis and health. The anion transporters and gut microbiome are intricately interconnected, where alterations in one can trigger changes in the other, leading to compromised barrier integrity and increasing susceptibility to pathophysiological states including gut inflammation. This review focuses on the interplay of key apical anion transporters including Down-Regulated in Adenoma (DRA, SLC26A3), Putative Anion Transporter-1 (PAT1, SLC26A6), and Cystic Fibrosis Transmembrane Conductance Regulator [CFTR, ATP-binding cassette subfamily C member 7 (ABCC7)] with the gut microbiome, barrier integrity, and their relationship to gut inflammation.
Collapse
Affiliation(s)
- Nazim Husain
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Arivarasu N. Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pradeep. K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
11
|
Zeng S, Wang S, Mu D. Seeding microbes in defined early-life windows to fight diabetes. Trends Endocrinol Metab 2025:S1043-2760(25)00080-3. [PMID: 40316483 DOI: 10.1016/j.tem.2025.04.005] [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] [Received: 03/31/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/04/2025]
Abstract
Establishment of the early-life gut microbiome, coinciding with the host development, predisposes to long-term disease risk if disrupted. Recently, Hill et al. precisely delineated a critical early-life window in humans and mice during which specific fungi and bacteria play indispensable roles in β-cell development, thereby regulating lifelong metabolic homeostasis.
Collapse
Affiliation(s)
- Shuqin Zeng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Shaopu Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
Andreu-Sánchez S, Blanco-Míguez A, Wang D, Golzato D, Manghi P, Heidrich V, Fackelmann G, Zhernakova DV, Kurilshikov A, Valles-Colomer M, Weersma RK, Zhernakova A, Fu J, Segata N. Global genetic diversity of human gut microbiome species is related to geographic location and host health. Cell 2025:S0092-8674(25)00416-7. [PMID: 40311618 DOI: 10.1016/j.cell.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/23/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
The human gut harbors thousands of microbial species, each exhibiting significant inter-individual genetic variability. Although many studies have associated microbial relative abundances with human-health-related phenotypes, the substantial intraspecies genetic variability of gut microbes has not yet been comprehensively considered, limiting the potential of linking such genetic traits with host conditions. Here, we analyzed 32,152 metagenomes from 94 microbiome studies across the globe to investigate the human microbiome intraspecies genetic diversity. We reconstructed 583 species-specific phylogenies and linked them to geographic information and species' horizontal transmissibility. We identified 484 microbial-strain-level associations with 241 host phenotypes, encompassing human anthropometric factors, biochemical measurements, diseases, and lifestyle. We observed a higher prevalence of a Ruminococcus gnavus clade in nonagenarians correlated with distinct plasma bile acid profiles and a melanoma and prostate-cancer-associated Collinsella clade. Our large-scale intraspecies genetic analysis highlights the relevance of strain diversity as it relates to human health.
Collapse
Affiliation(s)
- Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Daoming Wang
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Davide Golzato
- Department of CIBIO, University of Trento, Trento, Italy
| | - Paolo Manghi
- Department of CIBIO, University of Trento, Trento, Italy
| | - Vitor Heidrich
- Department of CIBIO, University of Trento, Trento, Italy
| | | | - Daria V Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Mireia Valles-Colomer
- Department of CIBIO, University of Trento, Trento, Italy; MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.
| | - Nicola Segata
- Department of CIBIO, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy; Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|
13
|
Coates N, John DA, Jordan S, Storey M, Thornton CA, Garaiova I, Wang D, Allen SJ, Michael DR, Plummer SF, Facey PD. The Impact of Probiotic Supplementation on the Development of the Infant Gut Microbiota: An Exploratory Follow-Up of a Randomised Controlled Trial. Microorganisms 2025; 13:984. [PMID: 40431157 DOI: 10.3390/microorganisms13050984] [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: 03/25/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/29/2025] Open
Abstract
Early-life establishment of the gut microbiota plays a role in lifelong health, with disruptions linked to heightened risks of metabolic and immune disorders. Probiotic supplementation may be used to modulate the infant gut microbiome to promote favourable development. Here, we evaluate how Lab4B probiotic supplementation shapes the development of the infant gut microbiome over the first 6 months. Faecal samples collected from infants enrolled in PROBAT (ISRCTN26287422), a randomised, double-blind, placebo-controlled trial, were analysed using culture-dependent and -independent (16S rDNA and metagenomic shotgun sequencing) techniques to examine the composition, diversity, and metabolic capabilities of the microbiome, as well as the abundance of antimicrobial resistance genes (ARGs). Probiotic supplementation encouraged the development of a microbiome with a distinct composition characterised by elevated abundances of Bifidobacteriaceae in the first 6 weeks (p = 0.006) and Lactobacillaceae throughout the first 6 months (p < 0.05 at every 6-week time point), accelerated microbial diversification, reduced abundance of beta-lactam- and cephalosporin-resistance genes, and differences in predicted metabolic capabilities at the start and end points. Supplementation of this neonatal population, which is at high risk of atopy, with the Lab4B probiotic significantly influenced the development of the infant gut microbiota during the first 6 months.
Collapse
Affiliation(s)
- Niall Coates
- Research & Development, Cultech Ltd., Port Talbot SA12 7BZ, UK
| | - Daniel A John
- Research & Development, Cultech Ltd., Port Talbot SA12 7BZ, UK
| | - Sue Jordan
- Faculty of Medicine, Health and Life Science, Swansea University, Swansea SA2 8PP, UK
| | - Melanie Storey
- Faculty of Medicine, Health and Life Science, Swansea University, Swansea SA2 8PP, UK
| | - Catherine A Thornton
- Faculty of Medicine, Health and Life Science, Swansea University, Swansea SA2 8PP, UK
| | - Iveta Garaiova
- Research & Development, Cultech Ltd., Port Talbot SA12 7BZ, UK
| | - Duolao Wang
- Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | | | - Daryn R Michael
- Research & Development, Cultech Ltd., Port Talbot SA12 7BZ, UK
| | - Susan F Plummer
- Research & Development, Cultech Ltd., Port Talbot SA12 7BZ, UK
| | - Paul D Facey
- Biomedical Sciences, Swansea University, Swansea SA2 8PP, UK
| |
Collapse
|
14
|
Keshet A, Hochwald O, Lavon A, Borenstein-Levin L, Shoer S, Godneva A, Glantz-Gashai Y, Cohen-Dolev N, Timstut F, Lotan-Pompan M, Solt I, Weinberger A, Segal E, Shilo S. Development of antibiotic resistome in premature infants. Cell Rep 2025; 44:115515. [PMID: 40198224 DOI: 10.1016/j.celrep.2025.115515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/17/2024] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
Preterm birth is a major concern in neonatal care, significantly impacting infant survival and long-term health. The gut microbiome, essential for infant development, often becomes imbalanced in preterm infants, making it crucial to understand the effects of antibiotics on its development. Our study analyzed weekly, 6-month, and 1-year stool samples from 100 preterm infants, correlating clinical data on antibiotic use and feeding patterns. Comparing infants who received no antibiotics with those given empirical post-birth treatment, we observed notable alterations in the gut microbiome's composition and an increase in antibiotic resistance gene abundance early in life. Although these effects diminished over time, their long-term clinical impacts remain unclear. Human milk feeding was associated with beneficial microbiota like Actinobacteriota and reduced antibiotic resistance genes, underscoring its protective role. This highlights the importance of judicious antibiotic use and promoting human milk to foster a healthy gut microbiome in preterm infants.
Collapse
Affiliation(s)
- Ayya Keshet
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ori Hochwald
- Neonatal Intensive Care Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel; Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amit Lavon
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Liron Borenstein-Levin
- Neonatal Intensive Care Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel; Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Saar Shoer
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anastasia Godneva
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yitav Glantz-Gashai
- Neonatal Intensive Care Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Noa Cohen-Dolev
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Fanny Timstut
- Neonatal Intensive Care Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Maya Lotan-Pompan
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Solt
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel
| | - Adina Weinberger
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Smadar Shilo
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
15
|
Dasari MR, Roche KE, Jansen D, Anderson J, Alberts SC, Tung J, Gilbert JA, Blekhman R, Mukherjee S, Archie EA. Social and environmental predictors of gut microbiome age in wild baboons. eLife 2025; 13:RP102166. [PMID: 40244653 PMCID: PMC12005720 DOI: 10.7554/elife.102166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Mammalian gut microbiomes are highly dynamic communities that shape and are shaped by host aging, including age-related changes to host immunity, metabolism, and behavior. As such, gut microbial composition may provide valuable information on host biological age. Here, we test this idea by creating a microbiome-based age predictor using 13,563 gut microbial profiles from 479 wild baboons collected over 14 years. The resulting 'microbiome clock' predicts host chronological age. Deviations from the clock's predictions are linked to some demographic and socio-environmental factors that predict baboon health and survival: animals who appear old-for-age tend to be male, sampled in the dry season (for females), and have high social status (both sexes). However, an individual's 'microbiome age' does not predict the attainment of developmental milestones or lifespan. Hence, in our host population, gut microbiome age largely reflects current, as opposed to past, social and environmental conditions, and does not predict the pace of host development or host mortality risk. We add to a growing understanding of how age is reflected in different host phenotypes and what forces modify biological age in primates.
Collapse
Affiliation(s)
- Mauna R Dasari
- Department of Biological Sciences, University of Notre DameNotre DameUnited States
- Department of Biological Sciences, University of PittsburghPittsburghUnited States
- California Academy of SciencesSan FranciscoUnited States
| | - Kimberly E Roche
- Program in Computational Biology and Bioinformatics, Duke UniversityDurhamUnited States
| | - David Jansen
- Department of Biological Sciences, University of Notre DameNotre DameUnited States
| | - Jordan Anderson
- Department of Evolutionary Anthropology, Duke UniversityDurhamUnited States
| | - Susan C Alberts
- Department of Evolutionary Anthropology, Duke UniversityDurhamUnited States
- Department of Biology, Duke UniversityDurhamUnited States
- Duke University Population Research Institute, Duke UniversityDurhamUnited States
| | - Jenny Tung
- Department of Evolutionary Anthropology, Duke UniversityDurhamUnited States
- Department of Biology, Duke UniversityDurhamUnited States
- Duke University Population Research Institute, Duke UniversityDurhamUnited States
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
- Canadian Institute for Advanced ResearchTorontoCanada
- Faculty of Life Sciences, Institute of Biology, Leipzig UniversityLeipzigGermany
| | - Jack A Gilbert
- Department of Pediatrics and the Scripps Institution of Oceanography, University of California, San DiegoSan DiegoUnited States
| | - Ran Blekhman
- Section of Genetic Medicine, Department of Medicine, University of ChicagoChicagoUnited States
| | - Sayan Mukherjee
- Departments of Statistical Science, Mathematics, Computer Science, and Bioinformatics and Biostatistics, Duke UniversityDurhamUnited States
- Center for Scalable Data Analytics and Artificial Intelligence, University of LeipzigLeipzigGermany
- Max Planck Institute for Mathematics in the Natural SciencesLeipzigGermany
| | - Elizabeth A Archie
- Department of Biological Sciences, University of Notre DameNotre DameUnited States
| |
Collapse
|
16
|
Ramadan YN, Alqifari SF, Alshehri K, Alhowiti A, Mirghani H, Alrasheed T, Aljohani F, Alghamdi A, Hetta HF. Microbiome Gut-Brain-Axis: Impact on Brain Development and Mental Health. Mol Neurobiol 2025:10.1007/s12035-025-04846-0. [PMID: 40234288 DOI: 10.1007/s12035-025-04846-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
The current discovery that the gut microbiome, which contains roughly 100 trillion microbes, affects health and disease has catalyzed a boom in multidisciplinary research efforts focused on understanding this relationship. Also, it is commonly demonstrated that the gut and the CNS are closely related in a bidirectional pathway. A balanced gut microbiome is essential for regular brain activities and emotional responses. On the other hand, the CNS regulates the majority of GI physiology. Any disruption in this bidirectional pathway led to a progression of health problems in both directions, neurological and gastrointestinal diseases. In this review, we hope to shed light on the complicated connections of the microbiome-gut-brain axis and the critical roles of gut microbiome in the early development of the brain in order to get a deeper knowledge of microbiome-mediated pathological conditions and management options through rebalancing of gut microbiome.
Collapse
Affiliation(s)
- Yasmin N Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt.
| | - Saleh F Alqifari
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Khaled Alshehri
- Department of Internal Medicine (Neurology), Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Amirah Alhowiti
- Department of Family and Community Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Hyder Mirghani
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Tariq Alrasheed
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Faisal Aljohani
- Division of Medicine and Gastroenterology, Department of Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdulaziz Alghamdi
- Department of Medicine, Division of Psychiatry, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Helal F Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, 71491, Tabuk, Saudi Arabia
| |
Collapse
|
17
|
Sawhney SS, Thänert R, Thänert A, Hall-Moore C, Ndao IM, Mahmud B, Warner BB, Tarr PI, Dantas G. Gut microbiome evolution from infancy to 8 years of age. Nat Med 2025:10.1038/s41591-025-03610-0. [PMID: 40175737 DOI: 10.1038/s41591-025-03610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 02/24/2025] [Indexed: 04/04/2025]
Abstract
The human gut microbiome is most dynamic in early life. Although sweeping changes in taxonomic architecture are well described, it remains unknown how, and to what extent, individual strains colonize and persist and how selective pressures define their genomic architecture. In this study, we combined shotgun sequencing of 1,203 stool samples from 26 mothers and their twins (52 infants), sampled from childbirth to 8 years after birth, with culture-enhanced, deep short-read and long-read stool sequencing from a subset of 10 twins (20 infants) to define transmission, persistence and evolutionary trajectories of gut species from infancy to middle childhood. We constructed 3,995 strain-resolved metagenome-assembled genomes across 399 taxa, and we found that 27.4% persist within individuals. We identified 726 strains shared within families, with Bacteroidales, Oscillospiraceae and Lachnospiraceae, but not Bifidobacteriaceae, vertically transferred. Lastly, we identified weaning as a critical inflection point that accelerates bacterial mutation rates and separates functional profiles of genes accruing mutations.
Collapse
Affiliation(s)
- Sanjam S Sawhney
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert Thänert
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna Thänert
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - I Malick Ndao
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Bejan Mahmud
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gautam Dantas
- Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
18
|
Bahat H, Paret M, Uzan A, Klainer H, Sharon E, Turjeman S, Koren O, Goldman M, Youngster I. Fecal microbiome composition in neonates with or without urinary tract infection. Pediatr Nephrol 2025; 40:1015-1021. [PMID: 39607509 PMCID: PMC11885367 DOI: 10.1007/s00467-024-06612-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Most infants with febrile urinary tract infection (UTI) do not have an underlying anatomical risk factor. Thus, other non-anatomical risk factors should be considered. Since the most common pathogens arise from the fecal microbiota, our aim was to investigate whether the gut microbiota composition differs between febrile infants younger than 2 months with or without UTI. METHODS In this prospective, case-control, pilot study, we performed 16S ribosomal ribonucleic acid amplicon sequencing to characterize gut microbiota of febrile neonates with and without UTI admitted to the pediatric ward at Shamir Medical Center between February 2019 and May 2021. RESULTS The study cohort included 42 febrile neonates: 17 with and 25 without febrile UTI. We found a significant difference in beta diversity (i.e. between-sample/study group similarity indices) between the UTI and non-UTI group (p = 0.016). There were also distinct differences in the relative abundance of the 20 most prevalent genera. Furthermore, several genera were significantly enriched in the UTI group, with others dominating the non-UTI group. Streptococci were underrepresented in the UTI group. There was no difference in alpha diversity (i.e. within-sample diversity/richness) between groups. CONCLUSION Febrile neonates with UTI have a different fecal microbiota composition (beta-diversity), but not alpha diversity, in comparison to febrile neonates without UTI. A larger study is warranted to confirm these findings and their potential applications.
Collapse
Affiliation(s)
- Hilla Bahat
- Department of Pediatrics, Shamir Medical Center, 70300, Zerifin, Israel.
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Michal Paret
- Department of Pediatrics, Shamir Medical Center, 70300, Zerifin, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Atara Uzan
- The Center for Microbiome Research, Shamir Medical Center, Zerifin, Israel
| | - Hodaya Klainer
- The Center for Microbiome Research, Shamir Medical Center, Zerifin, Israel
| | - Efrat Sharon
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Michael Goldman
- Department of Pediatrics, Shamir Medical Center, 70300, Zerifin, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Youngster
- Department of Pediatrics, Shamir Medical Center, 70300, Zerifin, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
- The Center for Microbiome Research, Shamir Medical Center, Zerifin, Israel
| |
Collapse
|
19
|
Canada K, Evans TM, Pelphrey KA. Microbiome's effect on white matter in autism. J Neurophysiol 2025; 133:1150-1158. [PMID: 39998297 DOI: 10.1152/jn.00607.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/21/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025] Open
Abstract
Autism spectrum disorder (ASD) is characterized by deficits in social communication and restricted, repetitive behavioral patterns. Although other physiological presentations in individuals with ASD are heterogeneous, neuroimaging studies have consistently revealed a developmental pattern of initial white matter hypermyelination followed by reduced myelination compared with typically developing peers. Multiple studies have demonstrated that core ASD symptoms, including impairments in social skills, language acquisition, learning capabilities, motor performance, and sensory processing, correlate significantly with white matter dysregulation measured through diffusion tensor imaging (DTI). Longitudinal studies have shown that decreased gut microbiome diversity, particularly reductions in beneficial bacteria such as Bifidobacterium and Lactobacillus, correlates with symptom severity. Emerging mechanistic evidence suggests bidirectional relationships between microbiome composition and white matter development, both directly through metabolites like short-chain fatty acids (SCFAs) that regulate oligodendrocyte function and subsequent myelination, and indirectly through modulation of neuroinflammatory pathways. By integrating molecular-level gut physiology findings with macro-level brain imaging data, we may identify novel therapeutic approaches targeting the gut-brain axis in ASD management.
Collapse
Affiliation(s)
- Katherine Canada
- Department of Neurology, University of Virginia, Gilmer Hall, Charlottesville, Virginia, United States
| | - Tanya M Evans
- School of Education and Human Development, University of Virginia, Ridley Hall 126, Charlottesville, Virginia, United States
| | - Kevin A Pelphrey
- Department of Neurology, University of Virginia, Gilmer Hall, Charlottesville, Virginia, United States
| |
Collapse
|
20
|
Kim S, Ndwandwe C, Devotta H, Kareem L, Yao L, O'Mahony L. Role of the microbiome in regulation of the immune system. Allergol Int 2025; 74:187-196. [PMID: 39955207 DOI: 10.1016/j.alit.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 02/17/2025] Open
Abstract
Immune health and metabolic functions are intimately connected via diet and the microbiota. Immune cells are continuously exposed to a wide range of microbes and microbial-derived compounds, with important mucosal and systemic ramifications. Microbial fermentation of dietary components in vivo generates thousands of molecules, some of which are integral components of the molecular circuitry that regulates immune and metabolic functions. These in turn protect against aberrant inflammatory or hyper-reactive processes and promote effector immune responses that quickly eliminate pathogens, such as SARS-CoV-2. Potent tolerance mechanisms should ensure that these immune cells do not over-react to non-pathogenic factors (e.g. food proteins), while maintaining the ability to respond to infectious challenges in a robust, effective and well controlled manner. In this review we examine the factors and mechanisms that shape microbiota composition and interactions with the host immune system, their associations with immune mediated disorders and strategies for intervention.
Collapse
Affiliation(s)
- Songhui Kim
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Cebile Ndwandwe
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hannah Devotta
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Lamiah Kareem
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Lu Yao
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Liam O'Mahony
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|
21
|
Wang X, Zhao D, Bi D, Li L, Tian H, Yin F, Zuo T, Ianiro G, Li N, Chen Q, Qin H. Fecal microbiota transplantation: transitioning from chaos and controversial realm to scientific precision era. Sci Bull (Beijing) 2025; 70:970-985. [PMID: 39855927 DOI: 10.1016/j.scib.2025.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/27/2025]
Abstract
With the popularization of modern lifestyles, the spectrum of intestinal diseases has become increasingly diverse, presenting significant challenges in its management. Traditional pharmaceutical interventions have struggled to keep pace with these changes, leaving many patients refractory to conventional pharmaceutical treatments. Fecal microbiota transplantation (FMT) has emerged as a promising therapeutic approach for enterogenic diseases. Still, controversies persist regarding its active constituents, mechanism of action, scheme of treatment evaluation, indications, and contraindications. In this review, we investigated the efficacy of FMT in addressing gastrointestinal and extraintestinal conditions, drawing from follow-up data on over 8000 patients. We systematically addressed the controversies surrounding FMT's clinical application. We delved into key issues such as its technical nature, evaluation methods, microbial restoration mechanisms, and impact on the host-microbiota interactions. Additionally, we explored the potential colonization patterns of FMT-engrafted new microbiota throughout the entire intestine and elucidated the specific pathways through which the new microbiota modulates host immunity, metabolism, and genome.
Collapse
Affiliation(s)
- Xinjun Wang
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China.
| | - Di Zhao
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Dexi Bi
- Department of Pathology, Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Long Li
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Hongliang Tian
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Fang Yin
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, 00168, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato, Rome, 00168, Italy
| | - Ning Li
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Qiyi Chen
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China.
| | - Huanlong Qin
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China.
| |
Collapse
|
22
|
Ni M, Fan Y, Liu Y, Li Y, Qiao W, Davey LE, Zhang XS, Ksiezarek M, Mead EA, Tourancheau A, Jiang W, Blaser MJ, Valdivia RH, Fang G. Epigenetic phase variation in the gut microbiome enhances bacterial adaptation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.11.632565. [PMID: 39829898 PMCID: PMC11741434 DOI: 10.1101/2025.01.11.632565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The human gut microbiome within the gastrointestinal tract continuously adapts to variations in diet, medications, and host physiology. A strategy for bacterial genetic adaptation is epigenetic phase variation (ePV) mediated by bacterial DNA methylation, which can regulate gene expression, enhance clonal heterogeneity, and enable a single bacterial strain to exhibit variable phenotypic states. Genome-wide and site-specific ePVs have been characterized in human pathogens' antigenic variation and virulence factor production. However, the role of ePV in facilitating adaptation within the human microbiome remains poorly understood. Here, we comprehensively cataloged genome-wide and site-specific ePV in human infant and adult gut microbiomes. First, using long-read metagenomic sequencing, we detected genome-wide ePV mediated by complex structural variations of DNA methyltransferases, highlighting those associated with antibiotics or fecal microbiota transplantation. Second, we analyzed a collection of public short-read metagenomic sequencing datasets, uncovering a great prevalence of genome-wide ePV in the human gut microbiome. Third, we quantitatively detected site-specific ePVs using single-molecule methylation analysis to identify dynamic variation associated with antibiotic treatment or probiotic engraftment. Finally, we performed an in-depth assessment of an Akkermansia muciniphila isolate from an infant, highlighting that ePVs can regulate gene expression and enhance the bacterial adaptive capacity by employing a bet-hedging strategy to increase tolerance to differing antibiotics. Our findings indicate that epigenetic modifications are a common strategy used by gut bacteria to adapt to the fluctuating environment.
Collapse
Affiliation(s)
- Mi Ni
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yu Fan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yujie Liu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yangmei Li
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wanjin Qiao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren E. Davey
- Department of Integrative Immunobiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Magdalena Ksiezarek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edward A. Mead
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alan Tourancheau
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenyan Jiang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Raphael H. Valdivia
- Department of Integrative Immunobiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, USA
| | - Gang Fang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
23
|
An JU, Mun SH, Kim WH, Seong JK, Park K, Cho S. Dynamics of the canine gut microbiota of a military dog birth cohort. Front Microbiol 2025; 16:1481567. [PMID: 40196028 PMCID: PMC11973337 DOI: 10.3389/fmicb.2025.1481567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction We systematically tracked early life stages in a military dog birth cohort to investigate canine gut microbiota dynamics related to environmental exposure during growth. This study utilized 16s rRNA amplicon sequencing-based analysis with molecular epidemiology of Enterococcus faecalis within a controlled environment at a military dog training center. Methods We examined shifts in gut microbiota diversity and taxonomic composition across four growth stages (lactation, weaning, starter, puppy) in three littermate groups. Additionally, E. faecalis dynamics was analyzed to confirm strain sharing among littermate groups. Results Gut microbiota changed rapidly during early growth, stabilizing at the puppy stage. This is supported by increased similarity in taxonomic composition among littermate groups, as they experienced an increased shared external environment and consumed the identical diets. E. faecalis strain sharing among littermate groups increased as dogs aged. Nine E. faecalis cluster types were identified; three specific types (type I, II, and IX) dominated in each littermate group during lactation. With greater exposure to the shared external environment, cluster type I gradually assumed dominance across all groups. Despite the dynamic shifts in microbiota, we found five genera within the core microbiota, Bacteroides, Peptoclostridium, Fusobacterium, Lactobacillus, and Blautia. Discussion This study is the first to explore the dynamic nature of early-life canine gut microbiota, illustrating its transition to stability and its resilience to environmental perturbations within the controlled training environment of a military dog birth cohort.
Collapse
Affiliation(s)
- Jae-Uk An
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Seung-Hyun Mun
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Woo-Hyun Kim
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, Republic of Korea
| | - Je Kyung Seong
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, Republic of Korea
| | - Kyoungwan Park
- Military Working Dogs Training Center, Gangwon, Republic of Korea
| | - Seongbeom Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, Republic of Korea
- Comparative Medicine Disease Research Center (CDRC), Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Morrison ML, Xue KS, Rosenberg NA. Quantifying compositional variability in microbial communities with FAVA. Proc Natl Acad Sci U S A 2025; 122:e2413211122. [PMID: 40063791 PMCID: PMC11929398 DOI: 10.1073/pnas.2413211122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/21/2025] [Indexed: 03/19/2025] Open
Abstract
Microbial communities vary across space, time, and individual hosts, generating a need for statistical methods capable of quantifying variability across multiple microbiome samples at once. To understand heterogeneity across microbiome samples from different host individuals, sampling times, spatial locations, or experimental replicates, we present FAVA (FST-based Assessment of Variability across vectors of relative Abundances), a framework for characterizing compositional variability across two or more microbiome samples. FAVA quantifies variability across many samples of taxonomic or functional relative abundances in a single index ranging between 0 and 1, equaling 0 when all samples are identical and 1 when each sample is entirely composed of a single taxon (and at least two distinct taxa are present across samples). Its definition relies on the population-genetic statistic FST, with samples playing the role of "populations" and taxa playing the role of "alleles." Its mathematical properties allow users to compare datasets with different numbers of samples and taxonomic categories. We introduce extensions that incorporate phylogenetic similarity among taxa and spatial or temporal distances between samples. We demonstrate FAVA in two examples. First, we use FAVA to measure how the taxonomic and functional variability of gastrointestinal microbiomes across individuals from seven ruminant species changes along the gastrointestinal tract. Second, we use FAVA to quantify the increase in temporal variability of gut microbiomes in healthy humans following an antibiotic course and to measure the duration of the antibiotic's influence on temporal microbiome variability. We have implemented this tool in an R package, FAVA, for use in pipelines for the analysis of microbial relative abundances.
Collapse
|
25
|
Verster AJ, Salerno P, Valls R, Barrack K, Price CE, McClure EA, Madan JC, O’Toole GA, Sanville JL, Ross BD. Persistent delay in maturation of the developing gut microbiota in infants with cystic fibrosis. mBio 2025; 16:e0342024. [PMID: 39945545 PMCID: PMC11898760 DOI: 10.1128/mbio.03420-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/23/2025] [Indexed: 02/19/2025] Open
Abstract
The healthy human infant gut microbiome undergoes stereotypical changes in taxonomic composition between birth and maturation to an adult-like stable state. During this time, extensive communication between microbiota and the host immune system contributes to health status later in life. Although there are many reported associations between microbiota compositional alterations and disease in adults, less is known about how microbiome development is altered in pediatric diseases. One pediatric disease linked to altered gut microbiota composition is cystic fibrosis (CF), a multi-organ genetic disease involving impaired chloride secretion across epithelia and heightened inflammation both in the gut and at other body sites. Here, we use shotgun metagenomics to profile the strain-level composition and developmental dynamics of the infant fecal microbiota from several CF and non-CF longitudinal cohorts spanning from birth to greater than 36 months of life. We identify a set of keystone species that define microbiota development in early life in non-CF infants but are missing or decreased in relative abundance in infants with CF, resulting in a delayed pattern of microbiota maturation, persistent entrenchment in a transitional developmental phase, and subsequent failure to attain an adult-like stable microbiota. Delayed maturation is strongly associated with cumulative antibiotic treatments, and we also detect the increased relative abundance of oral-derived bacteria and higher levels of fungi in infants with CF, features that are associated with decreased gut bacterial density. These findings suggest the potential for future directed therapies targeted at overcoming developmental delays in microbiota maturation for infants with CF.IMPORTANCEThe human gastrointestinal tract harbors a diversity of microbes that colonize upon birth and collectively contribute to host health throughout life. Infants with the disease cystic fibrosis (CF) harbor altered gut microbiota compared to non-CF counterparts, with lower levels of beneficial bacteria. How this altered population is established in infants with CF and how it develops over the first years of life is not well understood. By leveraging multiple large non-CF infant fecal metagenomic data sets and samples from a CF cohort collected prior to highly effective modulator therapy, we define microbiome maturation in infants up to 3 years of age. Our findings identify conserved age-diagnostic species in the non-CF infant microbiome that are diminished in abundance in CF counterparts that instead exhibit an enrichment of oral-derived bacteria and fungi associated with antibiotic exposure. Together, our study builds toward microbiota-targeted therapy to restore healthy microbiota dynamics in infants with CF.
Collapse
Affiliation(s)
- Adrian J. Verster
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Paige Salerno
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Rebecca Valls
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Kaitlyn Barrack
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Courtney E. Price
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Emily A. McClure
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Juliette C. Madan
- Department of Pediatrics, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - George A. O’Toole
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Julie L. Sanville
- Department of Pediatrics, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Benjamin D. Ross
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
26
|
Hill JH, Bell R, Barrios L, Baird H, Ost K, Greenewood M, Monts JK, Tracy E, Meili CH, Chiaro TR, Weis AM, Guillemin K, Beaudin AE, Murtaugh LC, Stephens WZ, Round JL. Neonatal fungi promote lifelong metabolic health through macrophage-dependent β cell development. Science 2025; 387:eadn0953. [PMID: 40048508 PMCID: PMC12036834 DOI: 10.1126/science.adn0953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 11/19/2024] [Indexed: 03/14/2025]
Abstract
Loss of early-life microbial diversity is correlated with diabetes, yet mechanisms by which microbes influence disease remain elusive. We report a critical neonatal window in mice when microbiota disruption results in lifelong metabolic consequences stemming from reduced β cell development. We show evidence for the existence of a similar program in humans and identify specific fungi and bacteria that are sufficient for β cell growth. The microbiota also plays an important role in seeding islet-resident macrophages, and macrophage depletion during development reduces β cells. Candida dubliniensis increases β cells in a macrophage-dependent manner through distinctive cell wall composition and reduces murine diabetes incidence. Provision of C. dubliniensis after β cell ablation or antibiotic treatment improves β cell function. These data identify fungi as critical early-life commensals that promote long-term metabolic health.
Collapse
Affiliation(s)
- Jennifer Hampton Hill
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Rickesha Bell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Logan Barrios
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Halli Baird
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Kyla Ost
- Department of Immunology and Microbiology, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Morgan Greenewood
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Josh K. Monts
- HSC Flow Cytometry Core, University of Utah, Salt Lake City, UT, USA
| | - Erin Tracy
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Casey H. Meili
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Tyson R. Chiaro
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Allison M. Weis
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, OR, USA
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Anna E. Beaudin
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Hematology and Hematologic Malignancies, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | | | - W. Zac Stephens
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| | - June L. Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
27
|
Safarchi A, Al-Qadami G, Tran CD, Conlon M. Understanding dysbiosis and resilience in the human gut microbiome: biomarkers, interventions, and challenges. Front Microbiol 2025; 16:1559521. [PMID: 40104586 PMCID: PMC11913848 DOI: 10.3389/fmicb.2025.1559521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
The healthy gut microbiome is important in maintaining health and preventing various chronic and metabolic diseases through interactions with the host via different gut-organ axes, such as the gut-brain, gut-liver, gut-immune, and gut-lung axes. The human gut microbiome is relatively stable, yet can be influenced by numerous factors, such as diet, infections, chronic diseases, and medications which may disrupt its composition and function. Therefore, microbial resilience is suggested as one of the key characteristics of a healthy gut microbiome in humans. However, our understanding of its definition and indicators remains unclear due to insufficient experimental data. Here, we review the impact of key drivers including intrinsic and extrinsic factors such as diet and antibiotics on the human gut microbiome. Additionally, we discuss the concept of a resilient gut microbiome and highlight potential biomarkers including diversity indices and some bacterial taxa as recovery-associated bacteria, resistance genes, antimicrobial peptides, and functional flexibility. These biomarkers can facilitate the identification and prediction of healthy and resilient microbiomes, particularly in precision medicine, through diagnostic tools or machine learning approaches especially after antimicrobial medications that may cause stable dysbiosis. Furthermore, we review current nutrition intervention strategies to maximize microbial resilience, the challenges in investigating microbiome resilience, and future directions in this field of research.
Collapse
Affiliation(s)
- Azadeh Safarchi
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Ghanyah Al-Qadami
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Cuong D Tran
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Michael Conlon
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| |
Collapse
|
28
|
Shukla A, Sharma C, Malik MZ, Singh AK, Aditya AK, Mago P, Shalimar, Ray AK. Deciphering the tripartite interaction of urbanized environment, gut microbiome and cardio-metabolic disease. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 377:124693. [PMID: 40022791 DOI: 10.1016/j.jenvman.2025.124693] [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: 12/02/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
The world is experiencing a sudden surge in urban population, especially in developing Asian and African countries. Consequently, the global burden of cardio-metabolic disease (CMD) is also rising owing to gut microbiome dysbiosis due to urbanization factors such as mode of birth, breastfeeding, diet, environmental pollutants, and soil exposure. Dysbiotic gut microbiome indicated by altered Firmicutes to Bacteroides ratio and loss of beneficial short-chain fatty acids-producing bacteria such as Prevotella, and Ruminococcus may disrupt host-intestinal homeostasis by altering host immune response, gut barrier integrity, and microbial metabolism through altered T-regulatory cells/T-helper cells balance, activation of pattern recognition receptors and toll-like receptors, decreased mucus production, elevated level of trimethylamine-oxide and primary bile acids. This leads to a pro-inflammatory gut characterized by increased pro-inflammatory cytokines such as tumour necrosis factor-α, interleukin-2, Interferon-ϒ and elevated levels of metabolites or metabolic endotoxemia due to leaky gut formation. These pathophysiological characteristics are associated with an increased risk of cardio-metabolic disease. This review aims to comprehensively elucidate the effect of urbanization on gut microbiome-driven cardio-metabolic disease. Additionally, it discusses targeting the gut microbiome and its associated pathways via strategies such as diet and lifestyle modulation, probiotics, prebiotics intake, etc., for the prevention and treatment of disease which can potentially be integrated into clinical and professional healthcare settings.
Collapse
Affiliation(s)
- Avaneesh Shukla
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Chanchal Sharma
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Md Zubbair Malik
- Department of Translational Medicine, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Abhishek Kumar Aditya
- Department of Medicine, K.D. Medical College, Hospital and Research Center, Mathura, India
| | - Payal Mago
- Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, New Delhi, India; Campus of Open Learning, University of Delhi, New Delhi, India
| | - Shalimar
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| |
Collapse
|
29
|
Hayden HS, Nelson MT, Ross SE, Verster AJ, Bouzek DC, Eng A, Waalkes A, Penewit K, Kopp BT, Siracusa C, Rock MJ, Salipante SJ, Hoffman LR, Sanders DB. Effects of Therapeutic Antibiotic Exposure on the Oropharyngeal and Fecal Microbiota in Infants With Cystic Fibrosis. Pediatr Pulmonol 2025; 60:e71024. [PMID: 40042126 PMCID: PMC11881217 DOI: 10.1002/ppul.71024] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 01/11/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Systemic antibiotics can impact all microbes inhabiting patients, regardless of the intended target organism(s). We studied the simultaneous effects on respiratory and fecal microbiomes of β-lactam antibiotics administered for respiratory symptoms in infants with cystic fibrosis (IWCF). OBJECTIVE To compare the magnitude and duration of intended (respiratory) and unintended (fecal) antimicrobial action by analyzing oropharyngeal (OP) and fecal microbiota in IWCF. DESIGN Shotgun metagenomic sequencing and qPCR were performed on OP and fecal samples collected longitudinally from 14 IWCF (ages 1-17 months) during ("On Antibiotics") and after ("Off Antibiotics") β-lactam therapy, and from 5 IWCF (3-16 months) never treated with antibiotics. RESULTS Total bacterial loads (TBL) for On Antibiotics samples were lower than for both Never (OP and fecal) and Off Antibiotics samples (fecal only). α-diversities (within-sample) for OP On Antibiotics samples were lower than for Never and Off Antibiotics samples but did not differ between fecal sample groups. β-diversity (between-sample) differed between all OP sample groups and between fecal On and Never Antibiotics and Off and Never antibiotics samples; however, fecal On and Off Antibiotics sample β-diversities did not differ. Patterns of change in antibiotic resistance gene abundances reflected shifts in microbial community composition. CONCLUSIONS β-lactam antibiotic exposure was followed by marked alterations in both OP and fecal microbiota. While microbiota appeared to rebound after treatment in both sample types, our results suggest that fecal microbiota recovered less than OP. The clinical consequences of these findings should be studied in IWCF and other populations frequently treated with antibiotics.
Collapse
Affiliation(s)
- Hillary S. Hayden
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Maria T. Nelson
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Sydney E. Ross
- Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Drake C. Bouzek
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Alex Eng
- Department of MicrobiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Adam Waalkes
- Department of MicrobiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Kelsi Penewit
- Department of Laboratory MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Benjamin T. Kopp
- Department of PediatricsEmory University School of MedicineAtlantaGeorgiaUSA
| | | | - Michael J. Rock
- Department of PediatricsUniversity of WisconsinMadisonWisconsinUSA
| | | | - Lucas R. Hoffman
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Don B. Sanders
- Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
30
|
Hollinger MK, Grayson EM, Ferreira CM, Sperling AI. Harnessing the Farm Effect: Microbial Products for the Treatment and Prevention of Asthma Throughout Life. Immunol Rev 2025; 330:e70012. [PMID: 40035333 PMCID: PMC11877632 DOI: 10.1111/imr.70012] [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: 09/19/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
It has long been appreciated that farm exposure early in life protects individuals from allergic asthma. Understanding what component(s) of this exposure is responsible for this protection is crucial to understanding allergic asthma pathogenesis and developing strategies to prevent or treat allergic asthma. In this review, we introduce the concept of Farm-Friends, or specific microbes associated with both a farm environment and protection from allergic asthma. We review the mechanism(s) by which these Farm-Friends suppress allergic inflammation, with a focus on the molecule(s) produced by these Farm-Friends. Finally, we discuss the relevance of Farm-Friend administration (oral vs. inhaled) for preventing the development and severity of allergic asthma throughout childhood and adulthood. By developing a fuller understanding of which Farm-Friends modulate host immunity, a greater wealth of prophylactic and therapeutic options becomes available to counter the current allergy epidemic.
Collapse
Affiliation(s)
- Maile K. Hollinger
- Beirne B. Carter Center for Immunology ResearchUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Medicine, Pulmonary and Critical CareUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Emily M. Grayson
- Beirne B. Carter Center for Immunology ResearchUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Medicine, Pulmonary and Critical CareUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Microbiology, Immunology, and Cancer BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Caroline M. Ferreira
- Department of Medicine, Pulmonary and Critical CareUniversity of VirginiaCharlottesvilleVirginiaUSA
- Institute of Environmental, Chemistry and Pharmaceutics Sciences, Department of Pharmaceutics SciencesFederal University of São PauloSão PauloBrazil
| | - Anne I. Sperling
- Beirne B. Carter Center for Immunology ResearchUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Medicine, Pulmonary and Critical CareUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Microbiology, Immunology, and Cancer BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
31
|
Green JE, Wrobel A, Todd E, Marx W, Berk M, Lotfaliany M, Castle D, Cryan JF, Athan E, Hair C, Nierenberg AA, Jacka FN, Dawson S. Early antibiotic exposure and risk of psychiatric and neurocognitive outcomes: systematic review and meta-analysis. Br J Psychiatry 2025; 226:171-183. [PMID: 39658347 DOI: 10.1192/bjp.2024.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
BACKGROUND The prenatal and early-life periods pose a crucial neurodevelopmental window whereby disruptions to the intestinal microbiota and the developing brain may have adverse impacts. As antibiotics affect the human intestinal microbiome, it follows that early-life antibiotic exposure may be associated with later-life psychiatric or neurocognitive outcomes. AIMS To explore the association between early-life (in utero and early childhood (age 0-2 years)) antibiotic exposure and the subsequent risk of psychiatric and neurocognitive outcomes. METHOD A search was conducted using Medline, PsychINFO and Excerpta Medica databases on 20 November 2023. Risk of bias was assessed using the Newcastle-Ottawa scale, and certainty was assessed using the grading of recommendations, assessment, development and evaluation (GRADE) certainty assessment. RESULTS Thirty studies were included (n = 7 047 853 participants). Associations were observed between in utero antibiotic exposure and later development of autism spectrum disorder (ASD) (odds ratio 1.09, 95% CI: 1.02-1.16) and attention-deficit hyperactivity disorder (ADHD) (odds ratio 1.19, 95% CI: 1.11-1.27) and early-childhood exposure and later development of ASD (odds ratio 1.19, 95% CI: 1.01-1.40), ADHD (odds ratio 1.33, 95% CI: 1.20-1.48) and major depressive disorder (MDD) (odds ratio 1.29, 95% CI: 1.04-1.60). However, studies that used sibling control groups showed no significant association between early-life exposure and ASD or ADHD. No studies in MDD used sibling controls. Using the GRADE certainty assessment, all meta-analyses but one were rated very low certainty, largely owing to methodological and statistical heterogeneity. CONCLUSIONS While there was weak evidence for associations between antibiotic use in early-life and later neurodevelopmental outcomes, these were attenuated in sibling-controlled subgroup analyses. Thus, associations may be explained by genetic and familial confounding, and studies failing to utilise sibling-control groups must be interpreted with caution. PROSPERO ID: CRD42022304128.
Collapse
Affiliation(s)
- Jessica Emily Green
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Monash Alfred Psychiatry Research Centre (MAPrc), Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Prahran, Australia
- Department of Psychiatry, Peninsula Health, Frankston, Australia
| | - Anna Wrobel
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Emma Todd
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Wolfgang Marx
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Michael Berk
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Melbourne, Parkville, Australia
- Orygen Youth Health Research Centre and the Centre of Youth Mental Health, Melbourne, Australia
- The Florey Institute for Neuroscience and Mental Health, Parkville, Australia
- Department of Mental Health Drug and Alcohol Services, Barwon Health, Geelong, Australia
| | - Mojtaba Lotfaliany
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - David Castle
- School of Psychological Sciences, University of Tasmania, Hobart, Tasmania
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork and APC Microbiome, Cork, Ireland
| | - Eugene Athan
- Department of Mental Health Drug and Alcohol Services, Barwon Health, Geelong, Australia
| | - Christopher Hair
- Department of Mental Health Drug and Alcohol Services, Barwon Health, Geelong, Australia
| | - Andrew A Nierenberg
- Dauten Family Center for Bipolar Treatment Innovation, Department of Psychiatry, Massachusetts General Hospital, Boston, USA
- Department of Psychiatry, Harvard Medical School, Boston, USA
| | - Felice N Jacka
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Department of Immunology, Therapeutics, and Vaccines, James Cook University, Townsville, Australia
| | - Samantha Dawson
- IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| |
Collapse
|
32
|
Shibata R, Nakanishi Y, Suda W, Nakano T, Sato N, Inaba Y, Kawasaki Y, Hattori M, Shimojo N, Ohno H. Neonatal gut microbiota and risk of developing food sensitization and allergy. J Allergy Clin Immunol 2025; 155:932-946. [PMID: 39692676 DOI: 10.1016/j.jaci.2024.10.029] [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: 04/20/2024] [Revised: 09/20/2024] [Accepted: 10/24/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Food sensitization (FS) develops in early infancy and is a risk factor for subsequent food allergy (FA). Recent evidence suggests relationships of gut microbiota with FS and FA. However, little is known about the role of neonatal gut microbiota in the pathobiology of these manifestations. OBJECTIVES We sought to characterize gut microbiota in children using an enterotyping approach and determine the association of gut microbiota and the enterotypes with the development of FS and FA. METHODS We combined gut microbiome and fecal short-chain fatty acid data from 2 longitudinal birth-cohort studies in Japan, clustered the microbiome data from children who were 1 week to 7 years old and their mothers and identified enterotypes. We also determined the associations of gut microbiota and enterotypes with risks of developing FS and FA across the 2 studies using multivariable regression models. RESULTS Data from the 2563 microbiomes identified 6 enterotypes. More gut bacteria (eg, Bifidobacterium) in 1-month-old children showed significant relationships with the development of FS and FA than in 1-week-old children. Enterotypes at 1 month old consisted of Bacteroides-dominant, Klebsiella-dominant, and Bifidobacterium-dominant enterotypes. Bifidobacterium-dominant enterotypes with the highest fecal propionate concentration had the lowest risks of developing FS and FA, especially of hen egg white sensitization. Bifidobacterium-dominant enterotypes had lower risks at 2 years old in one study (vs Bacteroides-dominant enterotype, adjusted odds ratio [adjOR]: 0.10, 95% CI: 0.01-0.78; vs Klebsiella-dominant enterotype, adjOR: 0.10, 95% CI: 0.01-0.77) and at 9 months old in the other study (vs Bacteroides-dominant enterotype, adjOR: 0.33, 95% CI: 0.11-0.91). CONCLUSIONS In these birth-cohort studies, gut microbiome clustering identified distinct neonatal enterotypes with differential risks of developing FS and FA.
Collapse
Affiliation(s)
- Ryohei Shibata
- Laboratorie for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan; Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan.
| | - Yumiko Nakanishi
- Laboratorie for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Wataru Suda
- Laboratorie for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Taiji Nakano
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Noriko Sato
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Yosuke Inaba
- Clinical Research Center, Chiba University Hospital, Chiba City, Japan
| | - Yohei Kawasaki
- Faculty of Nursing, Japanese Red Cross College of Nursing, Tokyo, Japan
| | - Masahira Hattori
- Laboratorie for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Naoki Shimojo
- Center for Preventive Medical Sciences, Chiba University, Chiba City, Japan
| | - Hiroshi Ohno
- Laboratorie for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan; Laboratorie for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan.
| |
Collapse
|
33
|
Lamichhane S, Dickens AM, Buchacher T, Lou ET, Charron-Lamoureux V, Kattelus R, Karmacharya P, Pinto da Silva L, Kråkström M, Rasool O, Sen P, Walker C, Patan A, Gentry EC, Arzoomand A, Lakshmikanth T, Mikeš J, Mebrahtu A, Vatanen T, Raffatellu M, Zengler K, Hyötyläinen T, Xavier RJ, Brodin P, Lahesmaa R, Dorrestein PC, Knip M, Orešič M. Trajectories of microbiome-derived bile acids in early life - insights into the progression to islet autoimmunity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.18.25322275. [PMID: 40061321 PMCID: PMC11888530 DOI: 10.1101/2025.02.18.25322275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Recent studies reveal that gut microbes produce diverse bile acid conjugates, termed microbially conjugated bile acids (MCBAs). However, their regulation and health effects remain unclear. Here, we analyzed early-life MCBA patterns and their link to islet autoimmunity. We quantified 110 MCBAs in 303 stool samples collected longitudinally (3-36 months) from children who developed one or more islet autoantibodies and controls who remained autoantibody-negative. Stool MCBAs showed distinct age-dependent trajectories and correlated with gut microbiome composition. Altered levels of ursodeoxycholic and deoxycholic acid conjugates were linked to islet autoimmunity as well as modulated monocyte activation in response to immunostimulatory lipopolysaccharide and Th17/Treg cell balance. These findings suggest MCBAs influence immune development and type 1 diabetes risk.
Collapse
Affiliation(s)
- Santosh Lamichhane
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland
| | - Alex M Dickens
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- Department of Chemistry, University of Turku, Turku, 20500, Finland
| | - Tanja Buchacher
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Evita Tianai Lou
- Medical Research Council Laboratory of Medical Sciences (MRC LMS), Imperial College Hammersmith Campus, London, UK
- Department of Immunology and Inflammation, Imperial College London, W12 0NN, London, UK
| | - Vincent Charron-Lamoureux
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Roosa Kattelus
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Pragya Karmacharya
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
| | - Lucas Pinto da Silva
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
| | - Matilda Kråkström
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
| | - Omid Rasool
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
| | - Corinn Walker
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Abubaker Patan
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Emily C. Gentry
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Aron Arzoomand
- Department of women’s and children’s health, Karolinska Institutet, Stockholm, Sweden
| | - Tadepally Lakshmikanth
- Medical Research Council Laboratory of Medical Sciences (MRC LMS), Imperial College Hammersmith Campus, London, UK
- Department of Immunology and Inflammation, Imperial College London, W12 0NN, London, UK
- Department of women’s and children’s health, Karolinska Institutet, Stockholm, Sweden
| | - Jaromir Mikeš
- Department of women’s and children’s health, Karolinska Institutet, Stockholm, Sweden
| | - Aman Mebrahtu
- Medical Research Council Laboratory of Medical Sciences (MRC LMS), Imperial College Hammersmith Campus, London, UK
- Department of Immunology and Inflammation, Imperial College London, W12 0NN, London, UK
| | - Tommi Vatanen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
- Liggins Institute, University of Auckland, Auckland, New Zealand
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Manuela Raffatellu
- Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, 39 92093, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla, CA 92093, USA
| | - Karsten Zengler
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, 39 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | | | - Petter Brodin
- Medical Research Council Laboratory of Medical Sciences (MRC LMS), Imperial College Hammersmith Campus, London, UK
- Department of Immunology and Inflammation, Imperial College London, W12 0NN, London, UK
- Department of women’s and children’s health, Karolinska Institutet, Stockholm, Sweden
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Pieter C. Dorrestein
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, 39 92093, USA
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
- Department of Pediatrics, Center for Child Health Research, Tampere University Hospital, FI-33520 Tampere, Finland
| | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- Department of Life Technologies, University of Turku, Turku, FI-20014 Turku, Finland
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 702 81 Örebro, Sweden
| |
Collapse
|
34
|
Seco-Hidalgo V, Witney AA, Chico ME, Vaca M, Arevalo A, Schuyler AJ, Platts-Mills TAE, Ster IC, Cooper PJ. Rurality and relative poverty drive acquisition of a stable and diverse gut microbiome in early childhood in a non-industrialized setting. Sci Rep 2025; 15:5601. [PMID: 39955323 PMCID: PMC11830098 DOI: 10.1038/s41598-025-89224-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
There are limited longitudinal data from non-industrialized settings on patterns and determinants of gut bacterial microbiota development in early childhood. We analysed epidemiological data and stool samples collected from 60 children followed from early infancy to 5 years of age in a rural tropical district in coastal Ecuador. Data were collected longitudinally on a wide variety of individual, maternal, and household exposures. Extracted DNA from stool samples were analysed for bacterial microbiota using 16S rRNA gene sequencing. Both alpha and beta diversity indices suggested stable profiles towards 5 years of age. Greater alpha diversity and lower beta diversity were associated with factors typical of rural poverty including low household incomes, overcrowding, and greater agricultural and animal exposures. Consumption of unpasteurized milk was consistently associated with greater alpha diversity indices. Delivery method and antibiotic exposures during pregnancy and early childhood appeared to have limited effects on developmental trajectories of gut microbiota. Infants living in a non-industrialized setting in conditions of greater poverty and typically rural exposures appeared to acquire more rapidly a stable and diverse gut bacterial microbiome during childhood.
Collapse
Affiliation(s)
- Victor Seco-Hidalgo
- Institute of Infection and Immunity, St George's University of London, London, SW17 0RE, UK
| | - Adam A Witney
- Institute of Infection and Immunity, St George's University of London, London, SW17 0RE, UK
| | - Martha E Chico
- Fundación Ecuatoriana Para la Investigación en Salud, Quito, Ecuador
| | - Maritza Vaca
- Fundación Ecuatoriana Para la Investigación en Salud, Quito, Ecuador
| | - Andrea Arevalo
- Fundación Ecuatoriana Para la Investigación en Salud, Quito, Ecuador
| | - Alexander J Schuyler
- Division of Allergy & Clinical Immunology, University of Virginia, Charlottesville, VA, USA
| | | | - Irina Chis Ster
- Institute of Infection and Immunity, St George's University of London, London, SW17 0RE, UK
| | - Philip J Cooper
- Institute of Infection and Immunity, St George's University of London, London, SW17 0RE, UK.
- Escuela de Medicine, Universidad Internacional del Ecuador, Quito, Ecuador.
- Fundación Ecuatoriana Para la Investigación en Salud, Quito, Ecuador.
| |
Collapse
|
35
|
Yin X, Xiao M, Sun J, Feng J, Xia S, Li F, Liu X, Li J. Trajectory of gut microbiota before and after pediatric cardiopulmonary bypass surgery. Front Cell Infect Microbiol 2025; 14:1470925. [PMID: 40018264 PMCID: PMC11865025 DOI: 10.3389/fcimb.2024.1470925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/30/2024] [Indexed: 03/01/2025] Open
Abstract
Background Varied congenital heart disease (CHD) may induce gut microbiota dysbiosis due to intestinal hypoperfusion or/and hypoxemia. Microbiota dysbiosis has been found in preoperative infants and cardiopulmonary bypass (CPB) exacerbated it further. However, the trajectory of gut microbiota from pre- to early post-CPB and one-year later remains unexplored. We examined this trajectory in the two most common CHDs, i.e., left-to-right shunt (ventricular septal defect, VSD) vs. right-to-left shunt (tetralogy of Fallot, TOF). Methods We enrolled 13 infants with VSD and 11 with TOF, and collected fecal samples at pre- and early post-CPB. 10 and 12 age- and gender-matched healthy control infants were enrolled respectively. We also enrolled 13 and 9 gender- and CHD diagnosis- and operation-matched one-year post-CPB patients, and 8 age- and gender-matched healthy control children. 16S rRNA sequencing of fecal samples were performed. Results Compared to the control groups, both VSD and TOF pre-CPB groups had significantly increased Enterobacteriaceae and Shigella, and decreased Bifidobacterium (Ps ≤ 0.049). No significant change in microbial community diversity was observed between pre- and early post-CPB periods (Ps≥0.227). Compared with early post-CPB, one-year post-CPB groups had significantly increased short-chain fatty acids-producing microbes (Ps ≤ 0.025), and their microbial communities were close to that of the control group (Ps≥0.102). There was no significant difference in microbial communities between VSD and TOF groups in any of 3 periods (Ps≥0.055). Conclusion In children with VSD or TOF, gut microbiota dysbiosis existed preoperatively and were not significantly altered by CPB. One-year post-CPB, microbiota significantly improved towards normal. Similar microbial communities were found between children with VSD and TOF throughout the perioperative and long-term postoperative periods.
Collapse
Affiliation(s)
- Xi Yin
- Clinical Physiology Laboratory, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Minhua Xiao
- Department of Nutrition, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Jing Sun
- Department of Nutrition, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Jinqing Feng
- Clinical Physiology Laboratory, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Shuliang Xia
- Heart Center, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Fengxiang Li
- Heart Center, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Xihong Liu
- Department of Nutrition, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Jia Li
- Clinical Physiology Laboratory, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| |
Collapse
|
36
|
Liu T, Xu X, Wang T, Xu P. CRAmed: a conditional randomization test for high-dimensional mediation analysis in sparse microbiome data. Bioinformatics 2025; 41:btaf038. [PMID: 39880365 PMCID: PMC11821267 DOI: 10.1093/bioinformatics/btaf038] [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: 07/03/2024] [Revised: 12/20/2024] [Accepted: 01/25/2025] [Indexed: 01/31/2025] Open
Abstract
MOTIVATION Numerous microbiome studies have revealed significant associations between the microbiome and human health and disease. These findings have motivated researchers to explore the causal role of the microbiome in human complex traits and diseases. However, the complexities of microbiome data pose challenges for statistical analysis and interpretation of causal effects. RESULTS We introduced a novel statistical framework, CRAmed, for inferring the mediating role of the microbiome between treatment and outcome. CRAmed improved the interpretability of the mediation analysis by decomposing the natural indirect effect into two parts, corresponding to the presence-absence and abundance of a microbe, respectively. Comprehensive simulations demonstrated the superior performance of CRAmed in Recall, precision, and F1 score, with a notable level of robustness, compared to existing mediation analysis methods. Furthermore, two real data applications illustrated the effectiveness and interpretability of CRAmed. Our research revealed that CRAmed holds promise for uncovering the mediating role of the microbiome and understanding of the factors influencing host health. AVAILABILITY AND IMPLEMENTATION The R package CRAmed implementing the proposed methods is available online at https://github.com/liudoubletian/CRAmed.
Collapse
Affiliation(s)
- Tiantian Liu
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Jiangsu 211198, China
| | - Xiangnan Xu
- Chair of Statistics, Humboldt-Universität zu Berlin, Berlin 10099, Germany
| | - Tao Wang
- SJTU-Yale Joint Center of Biostatistics and Data Science, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Statistics, School of Mathematical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- MOE-LSC & CMA-Shanghai, Shanghai Jiao Tong University, Shanghai 200240, China
- MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Peirong Xu
- Department of Statistics, School of Mathematical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
37
|
Bashar S, Tun HM, Ting JY, Hicks M, Mandhane PJ, Moraes TJ, Simons E, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. Impact of postpartum hospital length of stay on infant gut microbiota: a comprehensive analysis of vaginal and caesarean birth. J Hosp Infect 2025; 156:50-60. [PMID: 39510138 DOI: 10.1016/j.jhin.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/15/2024] [Accepted: 10/05/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND The primary concern with prolonged hospitalization following birth is the risk of acquiring hospital-acquired infections (HAIs) caused by opportunistic bacteria, which can alter the early establishment of gut microbiota. OBJECTIVE To assess the association between postpartum hospital length of stay (LOS) and the composition of gut microbiota at 3 and 12 months of age according to birth mode. METHODS In total, 1313 Canadian infants from the CHILD Cohort Study were involved in this study. Prolonged LOS was defined as ≥2 days following vaginal delivery (VD) and ≥3 days following caesarean section (CS). The gut microbiota of infants was characterized by Illumina 16S rRNA sequencing of faecal samples at 3-4 months and 12 months of age. FINDINGS Following prolonged LOS, VD infants with no exposure to intrapartum antibiotics had a higher abundance of bacteria known to cause HAIs in their gut, including Enterococcus spp. at 3 and 12 months, Citrobacter spp. at 3 months, and Clostridioides difficile at 12 months. Abundance of Enterococcus spp. or Citrobacter spp. at 3 months significantly mediated the association between LOS and low abundance of Bacteroidaceae, or higher Enterococcaeae/Bacteriodaceae or Enterobacterales/Bacteroidaceae abundance ratios at 12 months of age in VD infants without intrapartum antibiotic exposure. HAI-causing Enterobacterales were also more abundant in later infancy in infants with prolonged LOS following CS. In the absence of exclusive breastfeeding at 3 months or any breastfeeding at 12 months, Porphyromonadaceae (of Bacteroidota) were depleted in CS infants with prolonged LOS. CONCLUSIONS Prolonged hospital stay after birth is associated with infant gut dysbiosis.
Collapse
Affiliation(s)
- S Bashar
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - H M Tun
- The Jockey Club School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Microbiota I-Center, Hong Kong, China
| | - J Y Ting
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - M Hicks
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - P J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - T J Moraes
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Canada
| | - E Simons
- Section of Allergy and Immunology, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - S E Turvey
- Department of Pediatrics, BC Children's Hospital, Vancouver, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - P Subbarao
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Canada; Department of Medicine, McMaster University, Hamilton, Canada
| | - J A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - A L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, Canada.
| |
Collapse
|
38
|
Li J, Xie Z, Yang L, Guo K, Zhou Z. The impact of gut microbiome on immune and metabolic homeostasis in type 1 diabetes: Clinical insights for prevention and treatment strategies. J Autoimmun 2025; 151:103371. [PMID: 39883994 DOI: 10.1016/j.jaut.2025.103371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
Type 1 diabetes (T1D) is a complex disease triggered by a combination of genetic and environmental factors, where abnormal autoimmune responses lead to progressive damage of the pancreatic β cells and severe glucose metabolism disorder. Recent studies have increasingly highlighted the close link between gut microbiota dysbiosis and the development of T1D. This review delves into existing population studies to explore the intricate interactions between the gut microbiota and the immune and metabolic homeostasis in T1D. It summarizes how changes in the structure and function of the gut microbiota are closely associated with the onset and progression of T1D across its natural course and clinical stages. More importantly, based on evidence accumulated from clinical observations and trials, we pioneer the discussion on gut microbiota-based T1D prevention and treatment strategies, this not only enriches our understanding of the complex pathological mechanisms of T1D but also provides potential directions for developing novel prevention and treatment strategies.
Collapse
Affiliation(s)
- Jiaqi Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lin Yang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Keyu Guo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
39
|
Benitez AJ, Tanes C, Friedman ES, Zackular JP, Ford E, Gerber JS, DeRusso PA, Kelly A, Li H, Elovitz MA, Wu GD, Zemel B, Bittinger K. Antibiotic exposure is associated with minimal gut microbiome perturbations in healthy term infants. MICROBIOME 2025; 13:21. [PMID: 39856742 PMCID: PMC11761179 DOI: 10.1186/s40168-024-01999-3] [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: 04/05/2024] [Accepted: 12/05/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND The evolving infant gut microbiome influences host immune development and later health outcomes. Early antibiotic exposure could impact microbiome development and contribute to poor outcomes. Here, we use a prospective longitudinal birth cohort of n = 323 healthy term African American children to determine the association between antibiotic exposure and the gut microbiome through shotgun metagenomics sequencing as well as bile acid profiles through liquid chromatography-mass spectrometry. RESULTS Stool samples were collected at ages 4, 12, and 24 months for antibiotic-exposed (n = 170) and unexposed (n = 153) participants. A short-term substudy (n = 39) collected stool samples at first exposure, and over 3 weeks following antibiotics initiation. Antibiotic exposure (predominantly amoxicillin) was associated with minimal microbiome differences, whereas all tested taxa were modified by breastfeeding. In the short-term substudy, we observed microbiome differences only in the first 2 weeks following antibiotics initiation, mainly a decrease in Bifidobacterium bifidum. The differences did not persist a month after antibiotic exposure. Four species were associated with infant age. Antibiotic exposure was not associated with an increase in antibiotic resistance gene abundance or with differences in microbiome-derived fecal bile acid composition. CONCLUSIONS Short-term and long-term gut microbiome perturbations by antibiotic exposure were detectable but substantially smaller than those associated with breastfeeding and infant age.
Collapse
Affiliation(s)
- Alain J Benitez
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
| | - Elliot S Friedman
- Division of Gastroenterology and Hepatology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Joseph P Zackular
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, and The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Eileen Ford
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey S Gerber
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Diseases, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Patricia A DeRusso
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrea Kelly
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hongzhe Li
- Department of Biostatistics, Informatics, and Epidemiology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michal A Elovitz
- Women's Biomedical Research Institute, Icahn School of Medicine, New York, NY, USA
| | - Gary D Wu
- Division of Gastroenterology and Hepatology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Babette Zemel
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA.
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Brown JA, Bashir H, Zeng MY. Lifelong partners: Gut microbiota-immune cell interactions from infancy to old age. Mucosal Immunol 2025:S1933-0219(25)00006-6. [PMID: 39862964 DOI: 10.1016/j.mucimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Our immune system and gut microbiota are intricately coupled from birth, both going through maturation during early life and senescence during aging almost in a synchronized fashion. The symbiotic relationship between the human host and microbiota is critically dependent on a healthy immune system to keep our microbiota in check, while the microbiota provides essential functions to promote the development and fitness of our immune system. The partnership between our immune system and microbiota is particularly important during early life, when microbial ligands and metabolites shape the development of the immune cells and immune tolerance; during aging, having sufficient beneficial gut bacteria is critical for the maintenance of intact mucosal barriers, immune metabolic fitness, and strong immunity against pathogens. The immune system during childhood is programmed, with the support of the microbiota, to develop robust immune tolerance, and limit autoimmunity and metabolic dysregulation, which are prevalent during aging. This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age, with the goal to gain a better understanding of potential strategies to leverage the gut microbiota to combat age-related immune decline.
Collapse
Affiliation(s)
- Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Hilal Bashir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, United States.
| |
Collapse
|
41
|
Wang M, Valizadegan N, Fields CJ, Donovan SM. Fecal Microbiome and Metabolomic Profiles of Mixed-Fed Infants Are More Similar to Formula-Fed than Breastfed Infants. Microorganisms 2025; 13:166. [PMID: 39858934 PMCID: PMC11767595 DOI: 10.3390/microorganisms13010166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Many infants consume both human milk and infant formula (mixed-fed); however, few studies have investigated how mixed feeding affects the gut microbiome composition and metabolic profiles compared to exclusive breastfeeding or formula feeding. Herein, how delivery mode and early nutrition affect the microbiome and metabolome of 6-week-old infants in the STRONG Kids2 cohort was investigated. Fecal samples were collected from exclusively breastfed (BF; n = 25), formula-fed (FF; n = 25) or mixed-fed (MF; n = 25) participants. Within each feeding group, infants were either delivered vaginally (VD; n = 13) or by Cesarean section (CS; n = 12). Feeding mode affects the fecal microbiome diversity, composition, and functional potential, as well as metabolomic profiles regardless of delivery mode. Alpha and beta diversity of MF differed from that of BF (p < 0.05) but were comparable to FF infants. Functional analyses have shown 117 potential metabolic pathways differed between BF and FF, 112 between BF and MF, and 8 between MF and FF infants (p < 0.05, q < 0.10). Fecal metabolomic profiles of MF and FF clustered together and separated from BF infants. In total, 543 metabolites differed between BF and FF, 517 between BF and MF, and 3 between MF and FF (p < 0.05, q < 0.10). Delivery mode affected overall microbial composition (p = 0.022) at the genus level and 24 potential functional pathways, with 16 pathways being higher in VD than CS infants (p < 0.05, q < 0.10). Metabolomic analysis identified 47 differential metabolites between CS and VD, with 39 being lower in CS than VD (p < 0.05, q < 0.10). In summary, fecal microbiota composition and function and metabolite profiles of 6-week-old MF infants are closer to FF than BF infants.
Collapse
Affiliation(s)
- Mei Wang
- Department of Food Science & Human Nutrition, University of Illinois, Urbana, IL 61801, USA;
| | - Negin Valizadegan
- High-Performance Biological Computing, University of Illinois, Urbana, IL 61801, USA; (N.V.); (C.J.F.)
| | - Christopher J. Fields
- High-Performance Biological Computing, University of Illinois, Urbana, IL 61801, USA; (N.V.); (C.J.F.)
| | - Sharon M. Donovan
- Department of Food Science & Human Nutrition, University of Illinois, Urbana, IL 61801, USA;
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
42
|
Fahur Bottino G, Bonham KS, Patel F, McCann S, Zieff M, Naspolini N, Ho D, Portlock T, Joos R, Midani FS, Schüroff P, Das A, Shennon I, Wilson BC, O'Sullivan JM, Britton RA, Murray DM, Kiely ME, Taddei CR, Beltrão-Braga PCB, Campos AC, Polanczyk GV, Huttenhower C, Donald KA, Klepac-Ceraj V. Early life microbial succession in the gut follows common patterns in humans across the globe. Nat Commun 2025; 16:660. [PMID: 39809768 PMCID: PMC11733223 DOI: 10.1038/s41467-025-56072-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: 07/25/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Characterizing the dynamics of microbial community succession in the infant gut microbiome is crucial for understanding child health and development, but no normative model currently exists. Here, we estimate child age using gut microbial taxonomic relative abundances from metagenomes, with high temporal resolution (±3 months) for the first 1.5 years of life. Using 3154 samples from 1827 infants across 12 countries, we trained a random forest model, achieving a root mean square error of 2.56 months. We identified key taxonomic predictors of age, including declines in Bifidobacterium spp. and increases in Faecalibacterium prausnitzii and Lachnospiraceae. Microbial succession patterns are conserved across infants from diverse human populations, suggesting universal developmental trajectories. Functional analysis confirmed trends in key microbial genes involved in feeding transitions and dietary exposures. This model provides a normative benchmark of "microbiome age" for assessing early gut maturation that may be used alongside other measures of child development.
Collapse
Affiliation(s)
| | - Kevin S Bonham
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Fadheela Patel
- University of Cape Town, Cape Town, Western Cape, South Africa
| | - Shelley McCann
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Michal Zieff
- University of Cape Town, Cape Town, Western Cape, South Africa
| | - Nathalia Naspolini
- School of Arts, Sciences and Humanity, University of São Paulo, São Paulo, SP, Brazil
| | - Daniel Ho
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Theo Portlock
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Raphaela Joos
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Firas S Midani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Paulo Schüroff
- School of Arts, Sciences and Humanity, University of São Paulo, São Paulo, SP, Brazil
| | - Anubhav Das
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Inoli Shennon
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Brooke C Wilson
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | | | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- INFANT Maternal and Child Health Centre, Dept of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Deirdre M Murray
- INFANT Maternal and Child Health Centre, Dept of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Mairead E Kiely
- INFANT Maternal and Child Health Centre, Dept of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Carla R Taddei
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, São Paulo, SP, Brazil
| | - Patrícia C B Beltrão-Braga
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, São Paulo, SP, Brazil
| | - Alline C Campos
- Pharmacology of Neuroplasticity Lab- Department of Pharmacology, Ribeirão Preto Medical School- University of São Paulo, São Paulo, SP, Brazil
| | - Guilherme V Polanczyk
- Division of Child & Adolescent Psychiatry, Department & Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA.
| |
Collapse
|
43
|
Zeng S, Zhou M, Mu D, Wang S. Clinical implications of maternal multikingdom transmissions and early-life microbiota. THE LANCET. MICROBE 2025:101042. [PMID: 39818230 DOI: 10.1016/j.lanmic.2024.101042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 01/18/2025]
Abstract
Mother-to-infant transmission of the bacteriome, virome, mycobiome, archaeome, and their mobile genetic elements has been recognised in nature as an important step for the infant to acquire and maintain a healthy early-life (from birth till age 3 years) microbiota. A comprehensive overview of other maternal multikingdom transmissions remains unavailable, except for that of the bacteriome. Associations between microorganisms and diseases throughout the human life span have been gradually discovered; however, whether these microorganisms are maternally derived and how they concomitantly interact with other microbial counterparts remain poorly understood. This Review first discusses the current understanding of maternal multikingdom transmissions, their contributions to the development of early-life microbiota, and the primary factors that influence the transmission processes. The clinical implications of the inherited microbiota on human health in early life have been emphasised upon next, along with highlighting of knowledge gaps that should be addressed in future research. Finally, interventions to restore typical vertical transmission or disturbed early-life microbiota have been discussed as potential therapeutic approaches.
Collapse
Affiliation(s)
- Shuqin Zeng
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Meicen Zhou
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Shaopu Wang
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
44
|
Chen-Liaw A, Aggarwala V, Mogno I, Haifer C, Li Z, Eggers J, Helmus D, Hart A, Wehkamp J, Lamousé-Smith ESN, Kerby RL, Rey FE, Colombel JF, Kamm MA, Olle B, Norman JM, Menon R, Watson AR, Crossette E, Terveer EM, Keller JJ, Borody TJ, Grinspan A, Paramsothy S, Kaakoush NO, Dubinsky MC, Faith JJ. Gut microbiota strain richness is species specific and affects engraftment. Nature 2025; 637:422-429. [PMID: 39604726 DOI: 10.1038/s41586-024-08242-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 10/17/2024] [Indexed: 11/29/2024]
Abstract
Despite the fundamental role of bacterial strain variation in gut microbiota function1-6, the number of unique strains of a species that can stably colonize the human intestine is still unknown for almost all species. Here we determine the strain richness (SR) of common gut species using thousands of sequenced bacterial isolates with paired metagenomes. We show that SR varies across species, is transferable by faecal microbiota transplantation, and is uniquely low in the gut compared with soil and lake environments. Active therapeutic administration of supraphysiologic numbers of strains per species increases recipient SR, which then converges back to the population average after dosing is ceased. Stratifying engraftment outcomes by high or low SR shows that SR predicts microbial addition or replacement in faecal transplants. Together, these results indicate that properties of the gut ecosystem govern the number of strains of each species colonizing the gut and thereby influence strain addition and replacement in faecal microbiota transplantation and defined live biotherapeutic products.
Collapse
Affiliation(s)
- Alice Chen-Liaw
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Varun Aggarwala
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Reliance Foundation Institution of Education and Research, Jio Institute, Navi Mumbai, India
| | - Ilaria Mogno
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Craig Haifer
- Concord Clinical School, University of Sydney, Sydney, New South Wales, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Zhihua Li
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph Eggers
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Drew Helmus
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amy Hart
- Janssen R&D, Spring House, PA, USA
| | | | | | - Robert L Kerby
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jean Frédéric Colombel
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael A Kamm
- Department of Gastroenterology and Medicine, St. Vincent's Hospital, Melbourne, Victoria, Australia
| | | | | | | | | | | | - Elisabeth M Terveer
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Josbert J Keller
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gastroenterology, Haaglanden Medical Center, The Hague, The Netherlands
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas J Borody
- Centre for Digestive Diseases, Sydney, New South Wales, Australia
| | - Ari Grinspan
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sudarshan Paramsothy
- Concord Clinical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Gastroenterology and Hepatology, Macquarie University Hospital, Sydney, New South Wales, Australia
| | - Nadeem O Kaakoush
- School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Marla C Dubinsky
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeremiah J Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
45
|
Han L, Lin C, Lan Y, Hua Y, Wu J, Fan Z, Li Y. Metagenomic Analysis of Gut Microbiome of Persistent Pulmonary Hypertension of the Newborn. Cardiovasc Toxicol 2025; 25:135-147. [PMID: 39680298 DOI: 10.1007/s12012-024-09949-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Persistent pulmonary hypertension of the newborn (PPHN) is one of the most common diseases in the neonatal intensive care unit which severely affects neonatal survival. Gut microbes play an increasingly important role in human health, but there are rarely reported how gut microbiota contribute to PPHN. In our study, the metagenomic sequencing of feces from 12 PPHN's neonates and 8 controls were performed to expose the relation between neonatal gut microbes and PPHN disease. Firstly, we found that the abundance of Actinobacteria, Proteobacteria, Bacteroidetes were significantly increased in PPHN compared with controls, but the Firmicutes components was reduced. And some pathogenic strains (like Vibrio metschnikovii) were significantly enriched in the PPHN compared with controls. Secondly, functional annotation of genes found that PPHN up-regulated transmembrane transport, but down-regulated ribosome and ATP binding. Lastly, microbial metabolic pathway enrichment analysis indicated that some metabolic pathway in PPHN were conflicting and contradictory, showed that an abnormally increased metabolism, disturbed protein synthesis and genomic instability in the PPHN neonate. Our results contribute to understanding the changes in the species and function of gut microbiota in PPHN, thus providing a theoretical basis for the explanation and treatment of PPHN.
Collapse
Affiliation(s)
- Linli Han
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuyang Lin
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yue Lan
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Yimin Hua
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinlin Wu
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, 20 3rd Section, Renmin S. Rd., Chengdu, 610041, Sichuan, China.
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China.
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, 24 1rd Section, Renmin S. Rd., Chengdu, 610065, Sichuan, China.
| | - Yifei Li
- Department of Pediatrics, Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, 20 3rd Section, Renmin S. Rd., Chengdu, 610041, Sichuan, China.
| |
Collapse
|
46
|
Dasari MR, Roche KE, Jansen D, Anderson J, Alberts SC, Tung J, Gilbert JA, Blekhman R, Mukherjee S, Archie EA. Social and environmental predictors of gut microbiome age in wild baboons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.605707. [PMID: 39131274 PMCID: PMC11312535 DOI: 10.1101/2024.08.02.605707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Mammalian gut microbiomes are highly dynamic communities that shape and are shaped by host aging, including age-related changes to host immunity, metabolism, and behavior. As such, gut microbial composition may provide valuable information on host biological age. Here we test this idea by creating a microbiome-based age predictor using 13,563 gut microbial profiles from 479 wild baboons collected over 14 years. The resulting "microbiome clock" predicts host chronological age. Deviations from the clock's predictions are linked to some demographic and socio-environmental factors that predict baboon health and survival: animals who appear old-for-age tend to be male, sampled in the dry season (for females), and have high social status (both sexes). However, an individual's "microbiome age" does not predict the attainment of developmental milestones or lifespan. Hence, in our host population, gut microbiome age largely reflects current, as opposed to past, social and environmental conditions, and does not predict the pace of host development or host mortality risk. We add to a growing understanding of how age is reflected in different host phenotypes and what forces modify biological age in primates.
Collapse
Affiliation(s)
- Mauna R. Dasari
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- California Academy of Sciences, San Francisco, CA, USA
| | - Kimberly E. Roche
- Program in Computational Biology and Bioinformatics, Duke University, Durham, NC, USA
| | - David Jansen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Jordan Anderson
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
| | - Susan C. Alberts
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
- Department of Biology, Duke University, Durham, NC, USA
- Duke University Population Research Institute, Duke University, Durham, NC, USA
| | - Jenny Tung
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
- Department of Biology, Duke University, Durham, NC, USA
- Duke University Population Research Institute, Duke University, Durham, NC, USA
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
- Canadian Institute for Advanced Research, Toronto, Ontario, Canada
- Faculty of Life Sciences, Institute of Biology, Leipzig University, Leipzig, Germany
| | - Jack A. Gilbert
- Department of Pediatrics and the Scripps Institution of Oceanography, University of California, San Diego, San Diego, CA, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Sayan Mukherjee
- Departments of Statistical Science, Mathematics, Computer Science, and Bioinformatics & Biostatistics, Duke University, Durham, NC, USA
- Center for Scalable Data Analytics and Artificial Intelligence, University of Leipzig, Leipzig Germany
- Max Planck Institute for Mathematics in the Natural Sciences, Leipzig, Germany
| | - Elizabeth A. Archie
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
47
|
Granato A, Renwick S, Yau C, Kong T, Daigneault MC, Knip M, Allen-Vercoe E, Danska JS. Analysis of early childhood intestinal microbial dynamics in a continuous-flow bioreactor. MICROBIOME 2024; 12:255. [PMID: 39639333 PMCID: PMC11619690 DOI: 10.1186/s40168-024-01976-w] [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: 08/15/2023] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND The human gut microbiota is inoculated at birth and undergoes a process of assembly and diversification during the first few years of life. Studies in mice and humans have revealed associations between the early-life gut microbiome and future susceptibility to immune and metabolic diseases. To resolve microbe and host contributing factors to early-life development and to disease states requires experimental platforms that support reproducible, longitudinal, and high-content analyses. RESULTS Here, we deployed a continuous single-stage chemostat culture model of the human distal gut to study gut microbiota from 18- to 24-month-old children integrating both culture-dependent and -independent methods. Chemostat cultures recapitulated multiple aspects of the fecal microbial ecosystem enabling investigation of relationships between bacterial strains and metabolic function, as well as a resource from which we isolated and curated a diverse library of early life bacterial strains. CONCLUSIONS We report the reproducible, longitudinal dynamics of early-life bacterial communities cultured in an advanced model of the human gut providing an experimental approach and a characterized bacterial resource to support future investigations of the human gut microbiota in early childhood.
Collapse
Affiliation(s)
- Alessandra Granato
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Simone Renwick
- Dept. of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
- Infant Center of Research Excellence, The Larsson-Rosenquist Foundation Mother-Milk, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Christopher Yau
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Dept. of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tiffany Kong
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Dept. of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Emma Allen-Vercoe
- Dept. of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Jayne S Danska
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.
- Dept. of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Dept. of Medicine Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
48
|
Seco-Hidalgo V, Witney A, Chico ME, Vaca M, Arevalo A, Schuyler AJ, Platts-Mills TA, Ster IC, Cooper PJ. Rurality and relative poverty drive acquisition of a stable and diverse gut microbiome in early childhood in a non-industrialized setting. RESEARCH SQUARE 2024:rs.3.rs-5361957. [PMID: 39678332 PMCID: PMC11643292 DOI: 10.21203/rs.3.rs-5361957/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
There are limited longitudinal data from non-industrialized settings on patterns and determinants of gut bacterial microbiota development in early childhood. We analysed epidemiological data and stool samples collected from 60 children followed from early infancy to 5 years of age in a rural tropical district in coastal Ecuador. Data were collected longitudinally on a wide variety of individual, maternal, and household exposures. Extracted DNA from stool samples were analyzed for bacterial microbiota using 16S rRNA gene sequencing. Both alpha and beta diversity indices suggested stable profiles towards 5 years of age. Greater alpha diversity and lower beta diversity were associated with factors typical of rural poverty including low household incomes, overcrowding, and greater agricultural and animal exposures, but not with birth mode or antibiotic exposures. Consumption of unpasteurized milk was consistently associated with greater alpha diversity indices. Infants living in a non-industrialized setting in conditions of greater poverty and typically rural exposures appeared to acquire more rapidly a stable and diverse gut bacterial microbiome during childhood.
Collapse
Affiliation(s)
| | | | | | - Maritza Vaca
- Fundación Ecuatoriana Para la Investigación en Salud
| | | | | | | | | | | |
Collapse
|
49
|
Frazier AN, Beck MR, Waldrip H, Koziel JA. Connecting the ruminant microbiome to climate change: insights from current ecological and evolutionary concepts. Front Microbiol 2024; 15:1503315. [PMID: 39687868 PMCID: PMC11646987 DOI: 10.3389/fmicb.2024.1503315] [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: 09/28/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
Ruminant livestock provide meat, milk, wool, and other products required for human subsistence. Within the digestive tract of ruminant animals, the rumen houses a complex and diverse microbial ecosystem. These microbes generate many of the nutrients that are needed by the host animal for maintenance and production. However, enteric methane (CH4) is also produced during the final stage of anaerobic digestion. Growing public concern for global climate change has driven the agriculture sector to enhance its investigation into CH4 mitigation. Many CH4 mitigation methods have been explored, with varying outcomes. With the advent of new sequencing technologies, the host-microbe interactions that mediate fermentation processes have been examined to enhance ruminant enteric CH4 mitigation strategies. In this review, we describe current knowledge of the factors driving ruminant microbial assembly, how this relates to functionality, and how CH4 mitigation approaches influence ecological and evolutionary gradients. Through the current literature, we elucidated that many ecological and evolutionary properties are working in tandem in the assembly of ruminant microbes and in the functionality of these microbes in methanogenesis. Additionally, we provide a conceptual framework for future research wherein ecological and evolutionary dynamics account for CH4 mitigation in ruminant microbial composition. Thus, preparation of future research should incorporate this framework to address the roles ecology and evolution have in anthropogenic climate change.
Collapse
Affiliation(s)
- A. Nathan Frazier
- Conservation and Production Research Laboratory, United States Department of Agriculture, Agricultural Research Service, Bushland, TX, United States
| | - Matthew R. Beck
- Conservation and Production Research Laboratory, United States Department of Agriculture, Agricultural Research Service, Bushland, TX, United States
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Heidi Waldrip
- Conservation and Production Research Laboratory, United States Department of Agriculture, Agricultural Research Service, Bushland, TX, United States
| | - Jacek A. Koziel
- Conservation and Production Research Laboratory, United States Department of Agriculture, Agricultural Research Service, Bushland, TX, United States
| |
Collapse
|
50
|
Yang KC, Tien WY, Cheng MF. Gut microbiota compositions in the carriers and noncarriers of third-generation cephalosporin-resistant Escherichia coli: A study among children in southern Taiwan. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:896-905. [PMID: 39261124 DOI: 10.1016/j.jmii.2024.08.010] [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: 04/16/2024] [Revised: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Antimicrobial resistance, particularly in third-generation cephalosporin-resistant (3GC-R) Escherichia coli (E. coli), poses major global health challenges and has various clinical implications. Researchers have explored the relationship between extended-spectrum β-lactamase-producing E. coli and gut microbiota composition, which influence host health and disease susceptibility, in adults. In this study, we analyzed gut microbiota composition in Taiwanese children by the colonization status of 3GC-R E. coli. METHODS This cross-sectional study included children (age, 0-6 years) from Kaohsiung, Taiwan. Fecal samples were subjected to microbiological and gut microbiome (full-length 16S rRNA sequencing) analyses. The antimicrobial susceptibility of E. coli colonies isolated from the samples was tested. Furthermore, gut microbiota compositions and diversity indices were compared between 3GC-R E. coli carriers and noncarriers. RESULTS Approximately 46% of all children aged <6 years carried 3GC-R E. coli. The abundances of Drancourtella, Romboutsia, and Desulfovibrio (genus level) were higher in carriers than in noncarriers. By contrast, the abundances of Odoribacteraceae (family level) and Sutterella (genus level) were higher in noncarriers than in carriers. No significant between-group difference was observed in alpha diversity. However, a significant between-group difference was noted in beta diversity (unweighted UniFrac analysis). CONCLUSION This is the first study that investigated differences in the gut microbiota between healthy 3GC-R E. coli carriers and noncarriers in children, suggesting potential mechanisms involving altered utilization of short-chain fatty acids and elevated succinate levels contributing to increased colonization of 3GC-R E. coli. The other taxa identified in this study may contribute to colonization resistance in the pediatric population.
Collapse
Affiliation(s)
- Keng-Chin Yang
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wan-Yu Tien
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ming-Fang Cheng
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Kaohsiung Veterans General Hospital Tainan Branch, Tainan, Taiwan.
| |
Collapse
|