1
|
Naspolini NF, Schüroff PA, Vanzele PAR, Pereira-Santos D, Valim TA, Bonham KS, Fujita A, Passos-Bueno MR, Beltrão-Braga PCB, Carvalho ACPLF, Klepac-Ceraj V, Polanczyk GV, Campos AC, Taddei CR. Exclusive breastfeeding is associated with the gut microbiome maturation in infants according to delivery mode. Gut Microbes 2025; 17:2493900. [PMID: 40237336 PMCID: PMC12005435 DOI: 10.1080/19490976.2025.2493900] [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: 10/23/2024] [Revised: 01/29/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
Exclusive breastfeeding (EBF) plays a crucial role in infant gut microbiome assembly and development. However, few studies have investigated the effects of EBF in restoring a perturbed microbiome. In this study, we applied whole metagenomic sequencing to assess the gut microbiome assembly in 525 Brazilian infants from 3 to 9 months of age of the Germina Cohort, demonstrating the early determinants of microbial taxonomy and function modulation. Our analysis shows that EBF alters the relative abundance of genes related to the microbiome taxonomy and function, with effects varying by delivery mode. EBF alters the pattern of carbohydrates, lipid metabolism, and cell structure pathways depending on the delivery mode. The microbiome age is closer to chronological infant age in EBF than in non-EBF infants, meaning a lower microbiome maturation index (MMI). Using a complementary machine learning approach, we show that Escherichia coli, Ruminococcus gnavus, and Clostridium neonatale, as well as vitamin K and o-antigen pathways contribute strongly to EBF prediction. Moreover, EBF influences the microbiome maturation in early life, toward a microbiome age more similar to the chronological infant's age.
Collapse
Affiliation(s)
| | - Paulo A. Schüroff
- School of Arts, Sciences and Humanity, University of Sao Paulo, Sao Paulo, Brazil
| | - Pedro A. R. Vanzele
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Davi Pereira-Santos
- Department of Applied Mathematics and Statistics, Institute of Mathematics and Computer Sciences, University of Sao Paulo, Sao Carlos, Brazil
- Departamento Acadêmico de Computação, Universidade Tecnológica Federal do Paraná (UTFPR), Câmpus Medianeira, Medianeira, Brazil
| | - Tamires Amabili Valim
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Kevin S. Bonham
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - André Fujita
- Division of Network AI Statistics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Computer Science, Institute of Mathematics and Statistics, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Rita Passos-Bueno
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Patricia C. B. Beltrão-Braga
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Laboratory of Disease Modeling, Institut Pasteur de Sao Paulo, Sao Paulo, Brazil
| | - André C. P. L. F. Carvalho
- Department of Applied Mathematics and Statistics, Institute of Mathematics and Computer Sciences, University of Sao Paulo, Sao Carlos, Brazil
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Guilherme V. Polanczyk
- Department of Psychiatry, Faculdade de Medicina FMUSP, University of Sao Paulo, Sao Paulo, Brazil
| | - Alline C. Campos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Carla R. Taddei
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Division of Clinical Laboratory, University Hospital - University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Samarra A, Renwick S, Arzamasov AA, Rodionov DA, Spann K, Cabrera-Rubio R, Acuna-Gonzalez A, Martínez-Costa C, Hall L, Segata N, Osterman AL, Bode L, Collado MC. Human milk oligosaccharide metabolism and antibiotic resistance in early gut colonizers: insights from bifidobacteria and lactobacilli in the maternal-infant microbiome. Gut Microbes 2025; 17:2501192. [PMID: 40340669 PMCID: PMC12068340 DOI: 10.1080/19490976.2025.2501192] [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: 11/26/2024] [Revised: 03/22/2025] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
Breast milk, rich in human milk oligosaccharides (HMOs), supports the early-life colonization of beneficial bacteria such as bifidobacteria and lactobacilli, potentially reducing early-life antibiotic resistance. However, antibiotic treatment may interfere with the beneficial functions of HMO-degrading bacteria. This study investigated the metabolism of HMOs by bifidobacteria and lactobacilli isolated from human milk and mother-infant paired fecal samples, along with their antibiotic resistance profiles. Understanding these species- and sample-type-specific interactions will provide valuable insights into how bioactive components in human milk may shape the infant resistome during early life. A total of 39 Bifidobacterium and 14 Lactobacillaceae strains were isolated from paired mother-infant fecal and breast milk samples. Whole genome sequencing (WGS) allowed functional predictions on the HMO metabolism abilities and the resistance genotype of each strain. In vitro HMO utilization was assessed using growth kinetics assays combined with HMO glycoprofiling in culture supernatant. The minimum inhibitory concentration (MIC) was also determined for each strain. HMO metabolism by the bifidobacteria was species-specific. Bifidobacterium bifidum (B. bifidum) and Bifidobacterium longum subsp. infantis (B. infantis) exhibited the highest capacity for HMO degradation, consistent with genomic predictions. In contrast, lactobacilli were unable to degrade HMOs in vitro but were predicted to metabolize the by-products of HMO degradation. Phenotypic analysis revealed that B. bifidum strains had the lowest levels of antibiotic resistance, while Bifidobacterium animalis subsp. lactis (B. lactis) strains were resistant to most tested antibiotics. Overall, B. bifidum demonstrated the strongest HMO-degrading ability while remaining the most antibiotic-susceptible species. Early-life colonizing bifidobacterial species possess the essential machinery required to degrade HMOs and are highly susceptible to antibiotics. A better understanding of these dynamics could inform clinical strategies to protect and restore the infant gut microbiome, particularly in neonates exposed to antibiotics.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| | - Simone Renwick
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, USA
| | - Aleksandr A. Arzamasov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Dmitry A. Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Kennedy Spann
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| | - Antia Acuna-Gonzalez
- Food, Microbiome and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, Valencia, Spain
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, Valencia, Spain
| | - Lindsay Hall
- Food, Microbiome and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
| | - Andrei L. Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Lars Bode
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, USA
- Human Milk Institute, University of California San Diego, La Jolla, CA, USA
| | - MCarmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
5
|
Deleu S, Sabino J. Personalized Dietary Approaches to Optimizing Intestinal Microbial Health and Homeostasis. Gastroenterol Clin North Am 2025; 54:317-331. [PMID: 40348490 DOI: 10.1016/j.gtc.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Diet has a profound impact in human health, which is partly driven by changes in the intestinal microbiota. Several associations between dietary intake and the intestinal microbiota composition and function have been described. Namely, the Mediterranean diet is associated with beneficial bacteria, while the intake of ultraprocessed foods is linked to dysbiosis. It is, therefore, very tempting to tailor dietary approaches to the individual needs of the microbiota; however, high-quality prospective data are lacking. Provisionally, a diet rich in fruits and vegetables and low in ultraprocessed foods is recommended to improve the intestinal microbiota composition and function.
Collapse
Affiliation(s)
- Sara Deleu
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, Rome 00168, Italy
| | - João Sabino
- Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
6
|
Wang S, Zheng C, Bu C, Guo D, Zhang C, Xie Q, Pan J, Sun J, Chen W, Jiang S, Zhai Q. Role of sn-2 palmitate on the development of the infant gut microbiome: A metagenomic insight. Food Res Int 2025; 211:116488. [PMID: 40356145 DOI: 10.1016/j.foodres.2025.116488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/26/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
The infant gut microbiome, which develops from birth, has profound and lasting effects on human health. Its establishment in early life is influenced by events such as delivery mode and feeding type. This study examined the effects of formula milk enriched with sn-2 palmitate on the gut microbiota of healthy term infants. We conducted a 16-week comparative analysis of three feeding groups: infants receiving high sn-2 palmitate formula (n = 30), regular vegetable oil formula (n = 32), and breast milk (n = 30). Using shotgun metagenomic sequencing of fecal samples, we performed a comprehensive assessment of the gut microbiota. While overall microbial composition and diversity were comparable across groups, the functional profile of the microbiome in infants receiving sn-2 palmitate-enriched formula more closely resembled that of breastfed infants compared to the control formula group. This similarity extended to microbial species interactions, virulence gene abundance, and metabolic pathway expression patterns. In addition, sn-2 palmitate promoted the proliferation of Bifidobacterium breve and enhanced the robustness of the gut microbial ecology. Notably, the phylogenetic analysis of B. breve strains in the sn-2 palmitate group showed closer alignment with the breastfed group compared to the control group. These findings suggest that sn-2 palmitate-enriched formula may confer gut microbiota functional benefits that more closely resemble those of breast milk compared to control formula milk. This study provides scientific evidence for the development of future functional infant formulas.
Collapse
Affiliation(s)
- Shumin Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chengdong Zheng
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China
| | - Chaozhi Bu
- Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, Jiangsu 214002, China
| | - Danying Guo
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qinggang Xie
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China
| | - Jiancun Pan
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China
| | - Jianguo Sun
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Shilong Jiang
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China; PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Xueyuan Road 38, Haidian, Beijing 100083, China.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
7
|
Dash HR, Patel A. Genealogically bewildered individuals and forensic identification: a review of current and emerging solutions. Int J Legal Med 2025:10.1007/s00414-025-03513-2. [PMID: 40411594 DOI: 10.1007/s00414-025-03513-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 05/10/2025] [Indexed: 05/26/2025]
Abstract
The increasing use of assisted reproductive technologies (ART) with donor gametes is driven by rising infertility rates, delayed parenthood, and the need to prevent hereditary diseases. Greater social acceptance of diverse family structures, advancements in reproductive medicine, and improving success rates also contribute. Accessibility, affordability, and cross-border reproductive care further expand ART's reach, making donor gametes a preferred option for many individuals and couples worldwide. The widespread application of ART has led to an increasing number of donor-conceived individuals, many of whom are now reaching reproductive maturity. This demographic shift introduces significant challenges for traditional forensic genetic identification methods, which rely on biological reference samples from genetically related individuals. The absence of such samples complicates the identification process, particularly for individuals conceived via gamete donation or adoption, where biological and legal parentage are incongruent. Conventional forensic genetic analyses, including short tandem repeat (STR) and single nucleotide polymorphism (SNP) profiling of autosomal, Y-chromosome, X-chromosome, and mitochondrial DNA, exhibit limited efficacy in these scenarios. While these methods can sometimes identify individuals conceived using a single donor gamete, they are insufficient for cases involving dual donor gametes or mitochondrial replacement therapy. Emerging methodologies such as forensic genetic genealogy, DNA methylation profiling, and human microbiome analysis offer innovative approaches but necessitate further clinical validation and standardization.
Collapse
Affiliation(s)
- Hirak Ranjan Dash
- Department of Forensic Science, National Forensic Sciences University, Delhi Campus, New Delhi, 110085, India.
- School of Forensic Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, 752050, India.
| | - Anubhuti Patel
- Department of Reproductive Medicine and the Center for Human Reproduction, IMS and SUM Hospital, Bhubaneswar, Odisha, 751003, India
| |
Collapse
|
8
|
Lewis N, Lagopoulos J, Villani A. Gut-Brain Inflammatory Pathways in Attention-Deficit/Hyperactivity Disorder: The Role and Therapeutic Potential of Diet. Metabolites 2025; 15:335. [PMID: 40422911 DOI: 10.3390/metabo15050335] [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: 04/01/2025] [Revised: 05/05/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a common childhood-onset neurodevelopmental disorder that often persists into adulthood, leading to various adverse outcomes. Its underlying pathology is multifactorial, involving neurotransmitter imbalances, gut microbiota alterations, and oxidative and inflammatory dysregulation. Diet, a key environmental modifier of gut ecology, is consistently poorer in individuals with ADHD, with multiple nutrients implicated in its pathophysiology. This review examines the role of specific nutrients such as omega-3 fatty acids, key micronutrients, and potentially harmful dietary components, as well as broader dietary patterns, particularly the Western diet and Mediterranean diet (MedDiet), in relation to ADHD symptoms. It also evaluates both whole-diet and supplement-based clinical interventions, supporting the growing recognition of nutrition as a safe and relatively affordable modifiable factor in ADHD management. Additionally, the biological mechanisms linking diet to ADHD are reviewed, highlighting strong evidence for the involvement of gut dysbiosis and inflammatory processes. Despite the well-documented antioxidant, anti-inflammatory, and microbiome benefits of the MedDiet, direct research investigating its role in ADHD remains limited. Most whole-diet approaches to date have focused on elimination diets, leaving a significant gap in understanding the potential role of the MedDiet in ADHD management. Therefore, this review outlines preliminary evidence supporting the MedDiet and its key components as modulators of ADHD-related biological pathways, indicating its potential as a therapeutic approach. However, further research is required to rigorously evaluate its clinical efficacy. Finally, the limitations of observational and interventional nutritional research in ADHD are discussed, along with recommendations for future research directions.
Collapse
Affiliation(s)
- Naomi Lewis
- School of Health, University of the Sunshine Coast, 90 Sippy Downs Dr., Sippy Downs, QLD 4556, Australia
- Thompson Institute, University of the Sunshine Coast, 12 Innovation Pkwy., Birtinya, QLD 4575, Australia
| | - Jim Lagopoulos
- Thompson Brain and Mind Healthcare, Eccles Blvd., Birtinya, QLD 4575, Australia
| | - Anthony Villani
- School of Health, University of the Sunshine Coast, 90 Sippy Downs Dr., Sippy Downs, QLD 4556, Australia
| |
Collapse
|
9
|
Anantharaman K, Martin C. Genetic keys to microbial gut colonization. Cell Host Microbe 2025; 33:605-607. [PMID: 40373743 DOI: 10.1016/j.chom.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/17/2025]
Abstract
What enables certain microbes to successfully colonize the mammalian gut? In a recent issue of Cell, Liu et al. present a microbial cross-species genomic framework that identifies conserved genetic determinants of gut residency. This study provides a roadmap for developing more effective microbiome-based therapeutics, advancing our understanding of host-microbe interactions.
Collapse
Affiliation(s)
- Karthik Anantharaman
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA; Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA; Department of Data Science and AI, Indian Institute of Technology Madras, Chennai, TN, India.
| | - Cody Martin
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA; Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Lei Q, Cheng Z, Jiang M, Ma Q, Gong X, Huo Y, Lin M. Effects of Saccharomyces cerevisiae fermentation products on growth performance, fecal short chain fatty acids, and microbiota of pre-weaning calves. Anim Biosci 2025; 38:955-967. [PMID: 39483010 PMCID: PMC12062800 DOI: 10.5713/ab.24.0340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/01/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE This research aims to explore the effects of incorporating Saccharomyces cerevisiae fermentation products (SCFP) on growth performance, nutrient digestibility, antioxidant capacity, fecal short-chain fatty acids, and microbial composition of pre-weaning calves. METHODS Twenty Holstein calves, 10 days old and weighing an average of 48.63±0.91 kg, were randomly assigned to either the control group (CON) or the SCFP group, with 10 calves in each group. The CON group received only a basal diet, while the SCFP group received the starter diet supplemented with 5 g/head/d of SCFP products. The pre-trial period lasted for 5 days, followed by a main experimental period of 45 days. RESULTS The SCFP group had significantly higher final weight, average daily gain, and feed efficiency compared to the CON group (p<0.05). Moreover, the SCFP group exhibited increased apparent digestibility of dry matter, crude protein, ether extract, acid detergent fiber, Ca, and P (p<0.05). Additionally, supplementation with SCFP led to elevated content of growth hormone, insulin-like growth factor-1, and glucagon-like peptide-1 in serum. The inclusion of SCFP also raised serum catalase content and reduced serum malondialdehyde content in pre-weaning calves. Furthermore, SCFP supplementation influenced the composition of intestinal microflora by decreasing Actinobacteriota abundance and increasing the abundance of Ruminococcus, Lachnospiraceae_AC2044_group, Parabacteroides, and Butyricimonas. CONCLUSION The addition of SCFP has a positive impact on the growth performance, nutrient digestibility, antioxidant capacity, and intestinal microflora composition of pre-weaning calves.
Collapse
Affiliation(s)
- Qian Lei
- College of Animal Science and Technology, Yangzhou University, Yangzhou,
China
| | - Zhiqiang Cheng
- College of Animal Science and Technology, Yangzhou University, Yangzhou,
China
| | - Maocheng Jiang
- College of Animal Science and Technology, Yangzhou University, Yangzhou,
China
| | - Qianbo Ma
- College of Animal Science and Technology, Yangzhou University, Yangzhou,
China
| | - Xiaoxiao Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou,
China
| | - Yongjiu Huo
- College of Animal Science and Technology, Yangzhou University, Yangzhou,
China
| | - Miao Lin
- College of Animal Science and Technology, Yangzhou University, Yangzhou,
China
| |
Collapse
|
13
|
Lemons JMS, Narrowe AB, Firrman J, Mahalak KK, Liu L, Higgins S, Moustafa AM, Baudot A, Deyaert S, Van den Abbeele P. The food additive butylated hydroxyanisole minimally affects the human gut microbiome ex vivo. Food Chem 2025; 473:143037. [PMID: 39919360 DOI: 10.1016/j.foodchem.2025.143037] [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: 09/26/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
Butylated hydroxyanisole (BHA) continues to raise consumer concerns. All previous evaluations of this additive have failed to consider its effect on the gut microbiome, even though it enters the colon. An ex vivo model was used to assess the effect of BHA on microbial communities from 24 donors, aged infants to older adults. A dose of 0.35 g/L BHA elicited no statistically significant changes in the functional outputs or community structure for any age group. Although not large enough to affect community diversity, there were some significant decreases at the phylum level. Among the genes most significantly affected by treatment with BHA across age groups are those involved in lipopolysaccharide synthesis and bacterial electron transport encoded by Bacteroidota, Proteobacteria, and Verrucomicrobiota. Given what is known about the intracellular activity of BHA, these genes may hint at a mechanism behind BHA's evident, but minimally detrimental effect on the gut microbiota.
Collapse
Affiliation(s)
- Johanna M S Lemons
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA.
| | - Adrienne B Narrowe
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Jenni Firrman
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Karley K Mahalak
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - LinShu Liu
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Stephanie Higgins
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ahmed M Moustafa
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Microbial Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aurélien Baudot
- Cryptobiotix, Technologiepark-Zwijnaarde 82, Ghent 9052, Belgium
| | - Stef Deyaert
- Cryptobiotix, Technologiepark-Zwijnaarde 82, Ghent 9052, Belgium
| | | |
Collapse
|
14
|
Behl A, Sharma KK. Xenobiotics mediated modulation of gut microbiota and its role in lifestyle diseases: a critical appraisal on exposomics. Lett Appl Microbiol 2025; 78:ovaf067. [PMID: 40312786 DOI: 10.1093/lambio/ovaf067] [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: 01/31/2025] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/03/2025]
Abstract
Gastrointestinal tract of humans provides a niche to thousands of microbes, referred as gut microbiota (GM). GM establishes an intricate relationship with other organs via gut-organ axis, and modulates host health. The structure and functioning of these gut microbes can be influenced by the type of external exposome an individual experiences. Depending upon GM perturbations and host genotype, this can result in variable health implications. On the other hand, the huge arsenal of enzymes possessed by GM can chemically alter the xenobiotic structure. Its consequences can be numerous, including formation of harmful metabolites that cause organ damage, reversal of host detoxification pathways, or favourable health effects. Additionally, GM-mediated bio-transformation of pharmaceuticals can alter their pharmacokinetics and pharmacodynamics, potentially yielding variable drug responses, resulting into prolonged or ineffective treatments. To address this bi-facial relationship and the pivotal role of GM, this review incorporates recent in vitro, in vivo, and multiomics studies. It also suggests the need of machine learning approaches to decode the complex host-microbiota-xenobiotics interactions. These knowledge will aid in comprehending recent rise in chronic lifestyle-diseases which poses a huge burden on the health sector, and can also be a learning curve in making formulations and therapies for personalized treatment.
Collapse
Affiliation(s)
- Arush Behl
- Laboratory of Enzymology and Gut Microbiology, Maharshi Dayanand University, Rohtak 124001, India
| | - Krishna Kant Sharma
- Laboratory of Enzymology and Gut Microbiology, Maharshi Dayanand University, Rohtak 124001, India
| |
Collapse
|
15
|
Bao M, Wu R, Li J, Tang R, Song C. Research summary, possible mechanisms and perspectives of gut microbiota changes causing precocious puberty. Front Nutr 2025; 12:1596654. [PMID: 40352262 PMCID: PMC12061974 DOI: 10.3389/fnut.2025.1596654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
The increasing global incidence of precocious puberty, linked to environmental, metabolic, and genetic factors, necessitates innovative therapies beyond gonadotropin-releasing hormone (GnRH) analogs. Accumulating evidence implicates gut microbiota dysbiosis as a pivotal regulator of pubertal timing via interactions with hormone metabolism (e.g., estrogen reactivation via β-glucuronidase), neuroendocrine pathways (nitric oxide signaling), and immune-inflammatory responses. This review delineates taxonomic alterations in central precocious puberty (CPP) and obesity-related subtypes, including Streptococcus enrichment and Alistipes depletion, alongside functional shifts in microbial metabolite production. Mechanistic insights highlight microbiota-driven modulation of the hypothalamic-pituitary-gonadal (HPG) axis, leptin/insulin dynamics, and epigenetic regulation. Emerging interventions-probiotics, fecal microbiota transplantation (FMT), and dietary modifications-demonstrate efficacy in preclinical models and early clinical studies for delaying puberty onset and restoring hormonal balance. Translational efforts to validate these strategies are critical for addressing the clinical and psychosocial challenges posed by precocious puberty, positioning gut microbiota modulation as a novel therapeutic frontier in pediatric endocrinology.
Collapse
Affiliation(s)
- Maorong Bao
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Ba’nan Hospital Affiliated to Chongqing Medical University, Chongqing, China
| | - Jingwei Li
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Runan Tang
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Cui Song
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Koebnick C, Mukhopadhyay S, Getahun D, Tartof SY, Xiang AH, Puopolo KM, Sidell MA. The Contributions of delivery mode and intrapartum antibiotic exposure to body mass index among children through 5 years of age. Eur J Obstet Gynecol Reprod Biol 2025; 311:113984. [PMID: 40315685 DOI: 10.1016/j.ejogrb.2025.113984] [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: 08/05/2024] [Revised: 04/12/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025]
Abstract
OBJECTIVE To investigate independent effects of group B Streptococcus (GBS) intrapartum antibiotic prophylaxis (IAP) and cesarean delivery (CD) on body mass index (BMI) during early childhood. STUDY DESIGN Retrospective cohort study of infants (n = 157,820) born 2007-2015 in an integrated healthcare system. Exposures were delivery mode (CD or vaginal delivery [VD]) and GBS IAP exposure. CD was further divided into elective CD (without labor or rupture of membrane [ROM]) or unscheduled CD (following labor and/or ROM). BMI over 5 years was compared using non-linear multivariate models with B-splines, adjusted for demographics, maternal medical and obstetrical factors, and childhood antibiotic exposure. RESULTS At age 5 years, unscheduled CD without GBS IAP (Δ BMI 0.11 kg/m2, 95 % CI 0.06 to 0.16, p < 0.0001) and unscheduled CD with GBS IAP (Δ BMI 0.24 kg/m2, 95 % CI 0.11 to 0.36 kg/m2, p = 0.0002) were positively associated with higher BMI compared to their VD counterparts, respectively. No BMI difference was observed between children born by elective versus unscheduled CD. GBS IAP exposure was positively associated with increased BMI compared to non-exposed births in both VD (Δ BMI 0.07 kg/m2, 95 % CI 0.02 to 0.13 kg/m2, p = 0.0125) and CD (Δ BMI 0.22 kg/m2, 95 % CI 0.09 to 0.35 kg/m2, p = 0.0009). CONCLUSIONS Based on our findings, the widespread administration of GBS IAP and birth by cesarean delivery independently contribute to a significant upshift in body weight early in life that compares to or is higher than the annual increase in BMI in U.S. children on a population level.
Collapse
Affiliation(s)
- Corinna Koebnick
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States; Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena CA, United States.
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, United States; Clinical Futures, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
| | - Darios Getahun
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States; Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena CA, United States
| | - Sara Y Tartof
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States; Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena CA, United States
| | - Anny H Xiang
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States; Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena CA, United States
| | - Karen M Puopolo
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, United States; Clinical Futures, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, United States
| | - Margo A Sidell
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States
| |
Collapse
|
17
|
Hartert T, Kvysgaard JN, Thaver L, Suara-Istanbouli A, Allinson JP, Zar HJ. Understanding the childhood origins of asthma and chronic obstructive pulmonary disease: Insights from birth cohorts and studies across the life-span. J Allergy Clin Immunol 2025:S0091-6749(25)00419-1. [PMID: 40252849 DOI: 10.1016/j.jaci.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Birth cohorts have identified modifiable risk factors for asthma and respiratory health in children and adults, demonstrating the important role and pathways through which early-life events influence not only child outcomes but also adult health, disease, and mortality. This focused literature update from 2021 to 2024 summarizes birth cohort studies across the life-span that contribute to our understanding of risk factors for and the childhood origins of asthma and chronic obstructive pulmonary disease that may inform prevention efforts. We conclude that there are critical periods of developmental plasticity and susceptibility during which early-life events and exposures likely have the greatest impact on the development of asthma and chronic obstructive lung disease phenotypes, and that there are important prenatal and early childhood exposures, which, if modified, might be candidates for improving respiratory health across the life-span. Birth cohorts have been and will continue to be critical to advancing our understanding of lung health and disease across the life-span, including asthma and chronic obstructive pulmonary disease. As child mortality declines and the human population ages, data from birth cohort studies are needed to inform strategies for optimizing healthy longevity, including the investment in understanding the lifelong consequences of adverse prenatal and early childhood exposures.
Collapse
Affiliation(s)
- Tina Hartert
- Department of Medicine and Pediatrics, Vanderbilt University Medical Center, Nashville, Tenn.
| | - Julie Nyholm Kvysgaard
- Department of Pediatrics, Copenhagen Prospective Studies on Asthma in Childhood, Herlev, and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Linesri Thaver
- Department of Pediatrics & Child Health and the SA-MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Aisha Suara-Istanbouli
- Department of Medicine and Pediatrics, Vanderbilt University Medical Center, Nashville, Tenn
| | | | - Heather J Zar
- Department of Pediatrics & Child Health and the SA-MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
18
|
Upadhyay R, Mani S, Sevanan M. Microbiome-based dietary supplements for better development and healthy brain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:329-368. [PMID: 40414637 DOI: 10.1016/bs.irn.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Microbiome-based dietary supplements have gained attention for their role in enhancing brain development and cognitive health. The gut microbiome influences neurological functions through the gut-brain axis, impacting neurotransmitter production, immune regulation, and metabolic pathways. Dysbiosis is linked to neurological disorders such as Alzheimer's, Parkinson's, and autism spectrum disorders. This chapter explores dietary interventions targeting the microbiome, emphasising probiotics, prebiotics, and postbiotics. Additionally, AI and machine learning are transforming microbiome research by enabling personalised supplementation strategies tailored to individual gut profiles. Ethical challenges, including data privacy and algorithmic bias, are also discussed. Advances in big data analytics and predictive modelling are paving the way for precision-targeted interventions to optimise brain health. While microbiome-based therapies hold great promise, further clinical validation and regulatory frameworks are needed to ensure their efficacy and accessibility. This chapter highlights the future potential of microbiome-targeted strategies in neuroprotection and cognitive well-being.
Collapse
Affiliation(s)
- Riddhi Upadhyay
- Division of Biotechnology, Karunya Institute of Technology and Sciences (Deemed University), Coimbatore, Tamil Nadu, India
| | - Sugumar Mani
- Palamur Biosciences Private Limited, Mahabubnagar, Telangana, India
| | - Murugan Sevanan
- Division of Biotechnology, Karunya Institute of Technology and Sciences (Deemed University), Coimbatore, Tamil Nadu, India.
| |
Collapse
|
19
|
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
|
20
|
Ito K, Islam J, Sakurai K, Koyama S, Matsuo A, Okano K, Hirakawa R, Furukawa M, Nochi T. Breast milk stabilizes bacterial communities in the large intestine even after weaning. Biochem Biophys Res Commun 2025; 756:151585. [PMID: 40068432 DOI: 10.1016/j.bbrc.2025.151585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
The development and maintenance of a balanced microbiota is crucial for human health. Milk contains immune factors that not only protect offspring from infectious diseases but also play an important role in promoting the development and maintenance of the microbiota. However, the persisting effects of milk-derived immune factors on the maintenance of the microbiota after weaning have not been carefully examined. In this study, a cross-fostering model was employed using immunocompetent (IC) and immunodeficient (ID) mice in which one-half of the pups born from two dams were replaced. As a result, breast milk from the IC dam (IC milk) affected the development of the microbiota during lactation and maintained it even after weaning in the large intestine of the ID pups. The large intestinal microbiota of ID pups raised on IC milk remained similar to that of normal IC pups. Under normal conditions, the genus Mucispirillum was closely associated with other bacteria, forming a diverse bacterial community in the large intestine. In the small intestine, there were no differences in the microbiota before weaning, regardless of whether IC or ID milk was consumed. By contrast, significant differences were observed in the small intestinal microbiota between IC and ID mice after weaning; however, this was dependent on the immune-related characteristics of offspring (rather than milk-derived immune factors). These results indicate that breast milk plays an important role in the large (not small) intestine of offspring to create and maintain a diverse microbiota with a balanced bacterial network even after weaning.
Collapse
Affiliation(s)
- Kaori Ito
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Jahidul Islam
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Kota Sakurai
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Saeka Koyama
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Graduate Program in Food Science, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | | | - Kunihiro Okano
- GENODAS Co., Ltd. Miyagi, 980-8572, Japan; Department of Biological Environment, Faculty of Bioresource Sciences, Akita Prefectural University, Akita, 010-0195, Japan
| | - Ryota Hirakawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Graduate Program in Food Science, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Department of Animal Bioscience, University of Guelph, Ontario, N1G 2W1, Canada; Center for Professional Development, Institute for Excellence in Higher Education, Tohoku University, Miyagi, 980-8576, Japan.
| |
Collapse
|
21
|
Shiver AL, Sun J, Culver R, Violette A, Wynter C, Nieckarz M, Mattiello SP, Sekhon PK, Bottacini F, Friess L, Carlson HK, Wong DPGH, Higginbottom S, Weglarz M, Wang W, Knapp BD, Guiberson E, Sanchez J, Huang PH, Garcia PA, Buie CR, Good BH, DeFelice B, Cava F, Scaria J, Sonnenburg JL, Van Sinderen D, Deutschbauer AM, Huang KC. Genome-scale resources in the infant gut symbiont Bifidobacterium breve reveal genetic determinants of colonization and host-microbe interactions. Cell 2025; 188:2003-2021.e19. [PMID: 40068681 DOI: 10.1016/j.cell.2025.02.010] [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: 08/29/2023] [Revised: 08/08/2024] [Accepted: 02/13/2025] [Indexed: 03/27/2025]
Abstract
Bifidobacteria represent a dominant constituent of human gut microbiomes during infancy, influencing nutrition, immune development, and resistance to infection. Despite interest in bifidobacteria as a live biotic therapy, our understanding of colonization, host-microbe interactions, and the health-promoting effects of bifidobacteria is limited. To address these major knowledge gaps, we used a large-scale genetic approach to create a mutant fitness compendium in Bifidobacterium breve. First, we generated a high-density randomly barcoded transposon insertion pool and used it to determine fitness requirements during colonization of germ-free mice and chickens with multiple diets and in response to hundreds of in vitro perturbations. Second, to enable mechanistic investigation, we constructed an ordered collection of insertion strains covering 1,462 genes. We leveraged these tools to reveal community- and diet-specific requirements for colonization and to connect the production of immunomodulatory molecules to growth benefits. These resources will catalyze future investigations of this important beneficial microbe.
Collapse
Affiliation(s)
- Anthony L Shiver
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Jiawei Sun
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Rebecca Culver
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Arvie Violette
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Char Wynter
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Marta Nieckarz
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Science for Life Laboratory (SciLifeLab), Umeå University, Umeå 90187, Sweden
| | - Samara Paula Mattiello
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA; College of Mathematics and Science, The University of Tennessee Southern, Pulaski, TN 38478, USA
| | - Prabhjot Kaur Sekhon
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA; Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74074, USA; Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Francesca Bottacini
- School of Microbiology, University College Cork, Cork, Ireland; Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Lisa Friess
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hans K Carlson
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Daniel P G H Wong
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Steven Higginbottom
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Meredith Weglarz
- Stanford Shared FACS Facility, Center for Molecular and Genetic Medicine, Stanford University, Stanford, CA 94305, USA
| | - Weigao Wang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Benjamin D Knapp
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Emma Guiberson
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Chemistry and Biochemistry, Middlebury College, Middlebury, VT 05753, USA
| | - Juan Sanchez
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Po-Hsun Huang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Paulo A Garcia
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Cullen R Buie
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Benjamin H Good
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | | | - Felipe Cava
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Science for Life Laboratory (SciLifeLab), Umeå University, Umeå 90187, Sweden
| | - Joy Scaria
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA; Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74074, USA
| | - Justin L Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Douwe Van Sinderen
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Adam M Deutschbauer
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Zhang ZJ, Gao R, Lu YT, Zuo ZL, Li YH, Liu S, Song SY, Wang Y, Lai H. Factors affecting dysbiosis of the gut microbiota in the elderly and the progress of interventions in traditional Chinese and Western medicine. Front Cell Infect Microbiol 2025; 15:1529347. [PMID: 40196043 PMCID: PMC11973376 DOI: 10.3389/fcimb.2025.1529347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
As the population ages, intestinal health in the elderly has become a key area of concern, with gut microbiota dysbiosis emerging as a significant issue. This review summarizes the factors influencing dysbiosis and interventions from both traditional Chinese medicine (TCM) and Western medicine, offering a reference for future research. A comprehensive search of global databases up to March 2024 identified 617 original studies on gut microbiota dysbiosis in individuals aged 65 and older. After applying strict PRISMA guidelines, 20 articles met the inclusion criteria. Key findings are summarized in four areas: 1) the definition and mechanisms of dysbiosis, 2) evaluation tools for gut microbiota imbalance, 3) factors contributing to dysbiosis in the elderly, and 4) pharmacological treatments. Both TCM and Western medicine offer unique advantages in managing gut microbiota dysbiosis, and the choice of intervention should be tailored to the individual's condition. Future research should focus on optimizing integrated TCM and Western medicine approaches to improve outcomes for elderly patients with gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Zhi-Jun Zhang
- Hepatological surgery department, The People’s Hospital of Wenjiang Chengdu, Chengdu, China
| | - Ru Gao
- Nursing Department, The People’s Hospital of Wenjiang Chengdu, Chengdu, China
| | - Yu-Tong Lu
- Nursing Department, The People’s Hospital of Wenjiang Chengdu, Chengdu, China
| | - Zhi-Liang Zuo
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yu-Huan Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Shan Liu
- Nursing Department, The People’s Hospital of Wenjiang Chengdu, Chengdu, China
| | - Si-Yuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan, Chengdu, China
| | - Hongyan Lai
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing, China
| |
Collapse
|
25
|
DeVeaux AL, Hall-Moore C, Shaikh N, Wallace M, Burnham CAD, Schnadower D, Kuppermann N, Mahajan P, Ramilo O, Tarr PI, Dantas G, Schwartz DJ. Metagenomic signatures of extraintestinal bacterial infection in the febrile term infant gut microbiome. MICROBIOME 2025; 13:82. [PMID: 40128855 PMCID: PMC11931804 DOI: 10.1186/s40168-025-02079-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 03/04/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Extraintestinal bacterial infections (EBIs), e.g., urinary tract infection, bacteremia, and meningitis, occur in approximately 10% of febrile infants younger than 60 days. Although many EBI-causing species commonly reside in the infant gut, proof that the digestive system is a pre-infection habitat remains unestablished. RESULTS We studied a cohort of febrile term infants < 60 days old who presented to one of thirteen US emergency departments in the Pediatric Emergency Care Applied Research Network from 2016 to 2019. Forty EBI cases and 74 febrile controls matched for age, sex, and race without documented EBIs were selected for analysis. Shotgun sequencing was performed of the gut microbiome and of strains cultured from the gut and extraintestinal site(s) of EBI cases, including blood, urine, and/or cerebrospinal fluid. Using a combination of EBI isolate genomics and fecal metagenomics, we detected an intestinal strain presumptively isogenic to the EBI pathogen (> 99.999% average nucleotide identity) in 63% of infants with EBIs. Although there was no difference in gut microbiome diversity between cases and controls, we observed significantly increased Escherichia coli relative abundance in the gut microbiome of infants with EBIs caused by E. coli. Infants with E. coli infections who were colonized by the putatively isogenic pathogen strain had significantly higher E. coli phylogroup B2 abundance in their gut, and their microbiome was more likely to contain virulence factor loci associated with adherence, exotoxin production, and nutritional/metabolic function. CONCLUSIONS The intestine plausibly serves as a reservoir for EBI pathogens in a subset of febrile term infants, prompting consideration of new opportunities for surveillance and EBI prevention among colonized, pre-symptomatic infants. Video Abstract.
Collapse
Affiliation(s)
- Anna L DeVeaux
- The Edison Family Center for Genome Sciences and 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
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Meghan Wallace
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Carey-Ann D Burnham
- Department of Pediatrics, 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 Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - David Schnadower
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Kuppermann
- Departments of Pediatrics and Emergency Medicine, The George Washington School of Medicine and Health Sciences, and Children'S National Hospital, Washington, DC, USA
| | - Prashant Mahajan
- Departments of Emergency Medicine and Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Octavio Ramilo
- Department of Infectious Diseases, St. Jude Children'S Research Hospital, Memphis, TN, 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
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, 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 Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Drew J Schwartz
- 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 Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
26
|
Wang Y, Yang J, Lai X, Leung ASY, Xing Y, Wong GWK. Early Origins of Asthma and Allergies: Clues From Studies in China. Clin Exp Allergy 2025. [PMID: 40087850 DOI: 10.1111/cea.70033] [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: 10/14/2024] [Revised: 02/28/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
Asthma and allergies have emerged as some of the most common chronic diseases, particularly in developed countries. Epidemiological studies have consistently demonstrated that children growing up in farming/rural environments are less likely to develop these conditions. Over the past three decades, China has experienced unprecedented economic development and urbanisation, accompanied by a rapid rise in the prevalence of allergic disorders. Despite the substantial number of affected individuals, allergy management in China remains inconsistent and often inadequate, compounded by variations in diagnostic criteria and limited healthcare access in less developed regions. Furthermore, the vast population, regional disparities, and methodological inconsistencies in data collection have hindered the acquisition of comprehensive, large-scale epidemiological data. This review examines the factors contributing to asthma and allergies from their early origins, focusing on modifiable factors from a specific perspective of China. Factors related to traditional lifestyle, such as early-life exposure to agricultural farming and poultry, diverse dietary patterns, and early introduction of allergenic foods, appear to offer protection against allergies. Conversely, exposure to open-fire cooking, incense burning, tobacco smoke, as well as early-life antibiotic use and perinatal factors like Caesarean section delivery and prematurity may represent potential risks. A clear understanding of the role of these factors would pave the way for developing effective interventions to mitigate the substantial health and socioeconomic burdens associated with asthma and allergies.
Collapse
Affiliation(s)
- Yike Wang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Jing Yang
- Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Xueling Lai
- Shenzhen Guangming Maternal & Child Healthcare Hospital, Shenzhen, China
| | - Agnes Sze-Yin Leung
- Department of Paediatrics, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Yuhan Xing
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
- Department of Paediatrics, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Gary Wing-Kin Wong
- Department of Paediatrics, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
27
|
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
|
28
|
Shitrit IB, Sheiner E, Pariente G, Sergienko R, Wainstock T. Mode of delivery among preterm twins and offspring health, a retrospective cohort study. Eur J Pediatr 2025; 184:234. [PMID: 40063132 PMCID: PMC11893663 DOI: 10.1007/s00431-025-06060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025]
Abstract
Although cesarean delivery (CD) has been linked to long-term health risks in singleton infants, the impact of delivery mode on long-term health outcomes in preterm twins remains underexplored. A retrospective cohort study was conducted at a tertiary medical center in Israel from 1991 to 2021, comparing preterm twins vaginally delivered (VD) versus cesarean section, excluding cases with congenital malformations or perinatal deaths. Kaplan-Meier survival curves were used to compare the cumulative incidence, and Cox proportional hazards models were applied to adjust for potential confounders. Four thousand twenty-eight preterm twin offspring were included, with 1703 (42%) VD and 2325 (58%) by CD. Preterm twins delivered by CD had a higher incidence of respiratory morbidities (42% vs. 35% in the VD group, p < 0.001), with an adjusted Hazard Ratio (aHR) of 1.15 (95%CI 1.02-1.30). CD was associated with an increased incidence of neurologic morbidities (22% vs. 17% in the VD group, p < 0.001), with an aHR of 1.16 (95%CI 1.02-1.36). CD was associated with a higher incidence of infectious morbidities (69% vs. 62%, p < 0.001), with an aHR of 1.10 (95%CI 1.01-1.21). Gastrointestinal morbidities were more pronounced in the CD group (29% vs. 25%, p < 0.001), but the multivariable analysis did not reach significance (aHR = 1.10, 95%CI 0.95-1.27). Sub-analyses of elective-uncomplicated deliveries showed consistent results for most morbidities. CONCLUSIONS Cesarean delivery in preterm twins is associated with long-term respiratory, neurologic, infectious and gastrointestinal morbidities of the offspring. The findings suggest the potential benefits of vaginal over cesarean deliveries regarding offspring long-term health complications. WHAT IS KNOWN • Studies on singleton births show that cesarean delivery may increase respiratory, infectious, neurological and gastrointestinal outcomes remains inconsistent across term and preterm deliveries. • Cesarean delivery rates remain high despite recommendations to reduce their frequency, yet data on the association between cesarean delivery and morbidity among twins, particularly in small for gestational age twins, is limited. WHAT IS NEW • This is the first large-scale study demonstrating that cesarean delivery in preterm twins increases the odds of respiratory, neurologic, infectious and gastrointestinal long-term morbidities up to age 18. • The higher rates of respiratory, neurologic, infectious, and gastrointestinal complications persist even in uncomplicated cesarean deliveries.
Collapse
Affiliation(s)
- Itamar Ben Shitrit
- Department of Epidemiology, Biostatistics and Community Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Clinical Research Center, Soroka University Medical Center, PO Box 151, 84101, Be'er-Sheva, Israel.
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Emergency Pediatrics Department, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Eyal Sheiner
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Gali Pariente
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ruslan Sergienko
- Department of Epidemiology, Biostatistics and Community Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Health Policy and Management, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tamar Wainstock
- Department of Epidemiology, Biostatistics and Community Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
29
|
Li S, Liu J, Zhang X, Gu Q, Wu Y, Tao X, Tian T, Pan G, Chu M. The Potential Impact of Antibiotic Exposure on the Microbiome and Human Health. Microorganisms 2025; 13:602. [PMID: 40142495 PMCID: PMC11944296 DOI: 10.3390/microorganisms13030602] [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: 02/06/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Antibiotics are a cornerstone of modern medicine, saving countless lives. However, their widespread use presents two major challenges. First, antibiotic-induced changes in the microbiome can disrupt immune function, increasing the susceptibility to diseases associated with these alterations. Second, prolonged antibiotic use fosters the proliferation of antibiotic resistance genes, leading to the emergence of resistant strains and threatening our ability to control infections. These challenges highlight an urgent global health crisis, necessitating in-depth investigation into the multifaceted effects of antibiotic exposure on microbiome dynamics and human health. In this review, we explore the potential effects of antibiotic exposure on the microbiome and its implications for overall health. Additionally, we examine the role of emerging technologies in addressing these challenges and in shaping future antibiotic development. Our goal is to provide insights that will inform more effective public health strategies and interventions aimed at mitigating the adverse consequences of antibiotic use, restoring microbial balance, and improving overall health outcomes.
Collapse
Affiliation(s)
- Siqi Li
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Jiahao Liu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Xinyang Zhang
- School of Medical, Nantong University, Nantong 226019, China;
| | - Qihong Gu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Yutong Wu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Xiaobo Tao
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Tian Tian
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Gongbu Pan
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS 7005, Australia
| | - Minjie Chu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| |
Collapse
|
30
|
Zimmermann P, Kurth S, Giannoukos S, Stocker M, Bokulich NA. NapBiome trial: Targeting gut microbiota to improve sleep rhythm and developmental and behavioural outcomes in early childhood in a birth cohort in Switzerland - a study protocol. BMJ Open 2025; 15:e092938. [PMID: 40032396 PMCID: PMC11877202 DOI: 10.1136/bmjopen-2024-092938] [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: 08/27/2024] [Accepted: 02/08/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION The gut-brain axis plays a crucial role in the regulation and development of psychological and physical processes. The first year of life is a critical period for the development of the gut microbiome, which parallels important milestones in establishing sleep rhythm and brain development. Growing evidence suggests that the gut microbiome influences sleep, cognition and early neurodevelopment. For term-born and preterm-born infants, difficulties in sleep regulation may have consequences on health. Identifying effective interventions on the gut-brain axis in early life is likely to have long-term implications for the health and development of at-risk infants. METHODS AND ANALYSES In this multicentre, four-group, double-blinded, placebo (PLC)-controlled randomised trial with a factorial design, 120 preterm-born and 260 term-born infants will be included. The study will investigate whether the administration of daily synbiotics or PLC for a duration of 3 months improves sleep patterns and neurodevelopmental outcomes up to 2 years of age. The trial will also: (1) determine the association between gut microbiota, sleep patterns and health outcomes in children up to 2 years of age; and (2) leverage the interactions between gut microbiota, brain and sleep to develop new intervention strategies for at-risk infants. ETHICS AND DISSEMINATION The NapBiome trial has received ethical approval by the Committee of Northwestern and Central Switzerland and Canton Vaud, Switzerland (#2024-01681). Outcomes will be disseminated through publication and will be presented at scientific conferences. Metagenomic data will be shared through the European Nucleotide Archive. TRIAL REGISTRATION NUMBER The US National Institutes of Health NCT06396689.
Collapse
Affiliation(s)
- Petra Zimmermann
- Department of Community Health and Department of Paediatrics, Fribourg Hospital, University of Fribourg, Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Salome Kurth
- Department of Psychology, University of Fribourg, Fribourg, Switzerland
| | - Stamatios Giannoukos
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Martin Stocker
- Neonatology, Children's Hospital Lucerne, Lucerne, Switzerland
| | - Nicholas A Bokulich
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Johnson MJ, Lazarus SK, Bennett AE, Tovar-Salazar A, Robertson CE, Kofonow JM, Li S, McCollister B, Nunes MC, Madhi SA, Frank DN, Weinberg A. Gut microbiota and other factors associated with increased T cell regulation in HIV-exposed uninfected infants. Front Immunol 2025; 16:1533003. [PMID: 40098966 PMCID: PMC11911520 DOI: 10.3389/fimmu.2025.1533003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Infants exposed to HIV and uninfected (HEUs) are at higher risk of infectious morbidity than HIV-unexposed uninfected infants (HUUs). Multiple immune defects of unknown origin were observed in HEUs. We hypothesized that HEUs have more regulatory and inhibitory checkpoint-expressing T cells (Treg, Tici) than HUUs, which may dampen their immune defenses against pathogens. Method We used flow cytometry to measure 25 Treg/Tici subsets in HEUs and HUUs at birth, 6, 28, and 62 weeks of life. We used maternal and infant gut microbiome data reported in a previous study to establish correlations with the Treg/Tici. Results At birth, 3 Treg subsets, including the prototypic CD4+FOXP3+ and CD4+FOXP3+CD25+, had higher frequencies in 123 HEUs than in 117 HUUs, and 3 subsets had higher frequencies in HUUs. At 28 and 62 weeks of age, 5 Treg/Tici subsets had higher proportions in HEUs than HUUs. The frequencies of the Treg/Tici subsets that diverged between HEUs and HUUs at birth correlated with differential relative abundances of bacterial taxa in the maternal gut microbiome. The Treg/Tici subsets with significantly different frequencies at subsequent visits correlated with the concurrent composition of the infant gut microbiome. In vitro, treatment of HUU peripheral blood mononuclear cells (PBMC) with bacterial taxa most abundant in HEUs expanded Treg/Tici subsets with higher frequencies in HEUs than HUUs, recapitulating the in vivo correlations. Conversely, in vitro treatment of HEU PBMC did not increase Treg/Tici frequencies. Other factors that correlated with increased Treg/Tici frequencies were low maternal CD4+ T cells in HEUs at birth and male sex in the HUUs at 28 weeks of life. Discussion This study shows that maternal and infant gut dysbiosis are central to the increase in Treg/Tici in HEUs and may be targeted by mitigating interventions.
Collapse
Affiliation(s)
- Michael J. Johnson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sarah K. Lazarus
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Ashlynn E. Bennett
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Adriana Tovar-Salazar
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Charles E. Robertson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer M. Kofonow
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Shaobing Li
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Bruce McCollister
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Marta C. Nunes
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit and Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A. Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit and Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Daniel N. Frank
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Adriana Weinberg
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
32
|
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
|
33
|
Di Napoli A, Pasquini L, Visconti E, Vaccaro M, Rossi-Espagnet MC, Napolitano A. Gut-brain axis and neuroplasticity in health and disease: a systematic review. LA RADIOLOGIA MEDICA 2025; 130:327-358. [PMID: 39718685 DOI: 10.1007/s11547-024-01938-0] [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: 04/19/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
The gut microbiota emerged as a potential modulator of brain connectivity in health and disease. This systematic review details current evidence on the gut-brain axis and its influence on brain connectivity. The initial set of studies included 532 papers, updated to January 2024. Studies were selected based on employed techniques. We excluded reviews, studies without connectivity focus, studies on non-human subjects. Forty-nine papers were selected. Employed techniques in healthy subjects included 15 functional magnetic resonance imaging studies (fMRI), 5 diffusion tensor imaging, (DTI) 1 electroencephalography (EEG), 6 structural magnetic resonance imaging, 2 magnetoencephalography, 1 spectroscopy, 2 arterial spin labeling (ASL); in patients 17 fMRI, 6 DTI, 2 EEG, 9 structural MRI, 1 transcranial magnetic stimulation, 1 spectroscopy, 2 R2*MRI. In healthy subjects, the gut microbiota was associated with connectivity of areas implied in cognition, memory, attention and emotions. Among the tested areas, amygdala and temporal cortex showed functional and structural differences based on bacteria abundance, as well as frontal and somatosensory cortices, especially in patients with inflammatory bowel syndrome. Several studies confirmed the connection between microbiota and brain functions in healthy subjects and patients affected by gastrointestinal to renal and psychiatric diseases.
Collapse
Affiliation(s)
- Alberto Di Napoli
- Neuroradiology Unit, NESMOS Department, Sant'Andrea Hospital, La Sapienza University, 00189, Rome, Italy
| | - Luca Pasquini
- Radiology Department, Memorial Sloan Kettering Cancer Center, New York City, 10065, USA.
- Radiology Department, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, 06510, USA.
| | | | - Maria Vaccaro
- Medical Physics Department, Bambino Gesù Children's Hospital, 00165, Rome, Italy
| | | | - Antonio Napolitano
- Medical Physics Department, Bambino Gesù Children's Hospital, 00165, Rome, Italy
| |
Collapse
|
34
|
Zhang X, Tao T, Li P, Zhao Y, Cao H, Tao Y, Ma L, Zhou X, Jia H, Wu Y, Zhuo R, Yue G, Yang Y, Chen G, Yi X, Zhou H. Comparison of Robot-assisted Laparoscopic Extravesical Ureteral Reimplantation for Primary Vesicoureteral Reflux in Infants Under One Year of Age and Older Children. J Pediatr Surg 2025; 60:162114. [PMID: 39740279 DOI: 10.1016/j.jpedsurg.2024.162114] [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: 10/06/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 01/02/2025]
Abstract
PURPOSE To assess the surgical outcomes of Robot-Assisted Laparoscopic Extravesical Ureteral Reimplantation (RALUR-EV) in infants under one year of age with primary vesicoureteral reflux (VUR) as compared to older children. MATERIALS AND METHODS A retrospective analysis was conducted on 48 children with VUR who underwent unilateral or bilateral RALUR-EV between June 2018 and December 2022. Patients were divided into two groups: Group A (25 infants under one year) and Group B (23 children over one year). Preoperative evaluations included voiding cystourethrogram (VCUG), diuretic renography, ultrasonography, magnetic resonance urography, and urodynamic studies. Standard follow-up was conducted postoperatively, and both the resolution of reflux and any complications were meticulously documented. RESULTS All procedures were completed using the Da Vinci Surgical System without conversion to open surgery or major intraoperative complications. Group A had significantly smaller bladder capacities compared to Group B (70 [60, 90] ml vs. 150 [90, 200] ml, P <0.001) and a higher proportion of refractory febrile UTIs preoperatively (88.00 % vs. 60.87 %, P = 0.030). No significant differences in Operation time, estimated blood loss, or postoperative complications were observed between the groups. The success rates of reflux resolution were 96.00 % in Group A and 95.65 % in Group B. The length of hospital stay was significantly shorter in Group A (5 [4, 6] days vs. 7 [6, 10] days, P = 0.001). CONCLUSION RALUR-EV for the treatment of VUR in infants under one year of age is safe and effective. Compared to older children, the procedure does not increase intraoperative risks, and postoperative outcomes are comparable.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China; Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Tian Tao
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Pin Li
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yang Zhao
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Hualin Cao
- Department of Urology, Nan Xi Shan Hospital of Guangxi Zhuangzu Autonomous Region, Guilin, China
| | - Yuandong Tao
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lifei Ma
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaoguang Zhou
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Hongshuai Jia
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China; Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Yangyang Wu
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China; Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Ran Zhuo
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China; Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Guang Yue
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China; Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Yanpei Yang
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China; Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Guilong Chen
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China; Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Xiaoyu Yi
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Huixia Zhou
- Department of Pediatric Urology, Department of Senior Pediatrics, The Seventh Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China; Medical School of Chinese People's Liberation Army (PLA), Beijing, China.
| |
Collapse
|
35
|
Kadam O, Dalai S, Chauhan B, Guru RR, Mitra S, Raytekar N, Kumar R. Nanobiotechnology Unveils the Power of Probiotics: A Comprehensive Review on the Synergistic Role of Probiotics and Advanced Nanotechnology in Enhancing Geriatric Health. Cureus 2025; 17:e80478. [PMID: 40225478 PMCID: PMC11990693 DOI: 10.7759/cureus.80478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
The geriatric population, comprising ages 65 and above, encounters distinct health obstacles because of physiological changes and heightened vulnerability to diseases. New technologies are being investigated to tackle the intricate health requirements of this population. Recent advancements in probiotics and nanotechnology offer promising strategies to enhance geriatric health by improving nutrient absorption, modulating gut microbiota, and delivering targeted therapeutic agents. Probiotics play a crucial role in maintaining gut homeostasis, reducing inflammation, and supporting metabolic functions. However, challenges such as limited viability and efficacy in harsh gastrointestinal conditions hinder their therapeutic potential. Advanced nanotechnology can overcome these constraints by enhancing the efficacy of probiotics through nano-encapsulation, controlled delivery, and improvement of bioavailability. This review explores the synergistic potential of probiotics and advanced nanotechnology in addressing age-related health concerns. It highlights key developments in probiotic formulations, nano-based delivery systems, and their combined impact on gut health, immunity, and neuroprotection. The convergence of probiotics and nanotechnology represents a novel and transformative approach to promoting healthy aging, paving the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Onkar Kadam
- Biotechnology, Symbiosis Centre for Nanoscience and Nanotechnology, Symbiosis International (Deemed) University, Pune, IND
| | - Swayamprava Dalai
- Biotechnology, Symbiosis Centre for Nanoscience and Nanotechnology, Symbiosis International (Deemed) University, Pune, IND
| | - Bhawna Chauhan
- School of Biotech Engineering and Food Technology, Chandigarh University, Chandigarh, IND
| | - Rashmi Ranjan Guru
- Hospital Administration, All India Institute of Medical Sciences, Jodhpur, Jodhpur, IND
- Hospital Administration, Postgraduate Institute of Medical Education and Research, Chandigarh, Chandigarh, IND
| | - Subhodip Mitra
- Hospital Administration, All India Institute of Medical Sciences, Kalyani, Kolkata, IND
| | - Namita Raytekar
- Medical Technology, Symbiosis Institute of Health Sciences, Pune, IND
| | - Rahul Kumar
- Hospital Administration, Symbiosis University Hospital & Research Centre, Pune, IND
| |
Collapse
|
36
|
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
|
37
|
Ioannou A, Berkhout MD, Geerlings SY, Belzer C. Akkermansia muciniphila: biology, microbial ecology, host interactions and therapeutic potential. Nat Rev Microbiol 2025; 23:162-177. [PMID: 39406893 DOI: 10.1038/s41579-024-01106-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 01/03/2025]
Abstract
Akkermansia muciniphila is a gut bacterium that colonizes the gut mucosa, has a role in maintaining gut health and shows promise for potential therapeutic applications. The discovery of A. muciniphila as an important member of our gut microbiome, occupying an extraordinary niche in the human gut, has led to new hypotheses on gut health, beneficial microorganisms and host-microbiota interactions. This microorganism has established a unique position in human microbiome research, similar to its role in the gut ecosystem. Its unique traits in using mucin sugars and mechanisms of action that can modify host health have made A. muciniphila a subject of enormous attention from multiple research fields. A. muciniphila is becoming a model organism studied for its ability to modulate human health and gut microbiome structure, leading to commercial products, a genetic model and possible probiotic formulations. This Review provides an overview of A. muciniphila and Akkermansia genus phylogeny, ecophysiology and diversity. Furthermore, the Review discusses perspectives on ecology, strategies for harnessing beneficial effects of A. muciniphila for human mucosal metabolic and gut health, and its potential as a biomarker for diagnostics and prognostics.
Collapse
Affiliation(s)
- Athanasia Ioannou
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Maryse D Berkhout
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Sharon Y Geerlings
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands.
| |
Collapse
|
38
|
Wang Z, Yang Y, Kümmel S, Richnow HH, Nijenhuis I, Vogt C. Heterotrophic nitrate reduction potential of an aquifer microbial community from psychrophilic to thermophilic conditions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 967:178716. [PMID: 39946872 DOI: 10.1016/j.scitotenv.2025.178716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 03/05/2025]
Abstract
High temperature-aquifer thermal energy storage (HT-ATES) aims at the seasonal storage and extraction of large quantities of heat in the subsurface. However, the impacts of temperature fluctuations caused by HT-ATES toward biodiversity and ecosystem services in the subsurface environment with respect to the nitrogen cycle remain unclear. Hence, understanding possible temperature adaptation mechanisms of aquifer microbial communities is crucial to assess potential environmental risks associated with HT-ATES. In this study, we investigated the effects of temperatures between 12 °C and 80 °C on a pristine aquifer microbial community and its capacity to reduce nitrate, a common global groundwater contaminant. 13C-labeled acetate was used as easily consumable carbon and energy source for nitrate reduction, allowing precise activity measurement by analysis of released 13CO2. We observed nitrate reduction coupled to acetate mineralization at 12 °C, 25 °C, 38 °C, 45 °C and 60 °C but not at 80 °C. The rates of acetate mineralization at 12 °C to 38 °C were significantly higher than rates at 45 °C and 60 °C. Temperature significantly affected the composition of the acetate-mineralizing, nitrate-reducing microbial communities. Members of the families Pseudomonadaceae and Comamonadaceae mainly developed in enrichments incubated at 12 °C and 25 °C, whereas phylotypes affiliated to Rhizobiaceae dominated at 38 °C. At 45 °C and 60 °C, phylotypes belonging to Symbiobacteriaceae, Paenibacillaceae and Planococcaceae mainly developed. These findings indicate that the indigenous aquifer microbiome can maintain the ability to reduce nitrate over a wide temperature range, providing support that HT-ATES may allow thermal energy storage while simultaneously attenuating nitrate pollution.
Collapse
Affiliation(s)
- Zhenyu Wang
- Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research - UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Yonggang Yang
- School of Agricultural and Biological Engineering, Foshan University, Foshan 528000, China
| | - Steffen Kümmel
- Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research - UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Hans-Hermann Richnow
- Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research - UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Ivonne Nijenhuis
- Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research - UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Carsten Vogt
- Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research - UFZ, Permoserstraße 15, 04318 Leipzig, Germany.
| |
Collapse
|
39
|
Pattaroni C, Marsland BJ, Harris NL. Early-Life Host-Microbial Interactions and Asthma Development: A Lifelong Impact? Immunol Rev 2025; 330:e70019. [PMID: 40099971 PMCID: PMC11917194 DOI: 10.1111/imr.70019] [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: 10/01/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Childhood is a multifactorial disease, and recent research highlights the influence of early-life microbial communities in shaping disease risk. This review explores the roles of the gut and respiratory microbiota in asthma development, emphasizing the importance of early microbial exposure. The gut microbiota has been particularly well studied, with certain taxa like Faecalibacterium and Bifidobacterium linked to asthma protection, whereas short-chain fatty acids produced by gut microbes support immune tolerance through the gut-lung axis. In contrast, the respiratory microbiota, though low in biomass, shows consistent associations between early bacterial colonization by Streptococcus, Moraxella, and Haemophilus and increased asthma risk. The review also addresses the emerging roles of the skin microbiota and environmental fungi in asthma, though findings remain inconsistent. Timing is a critical factor, with early-life disruptions, such as antibiotic use, potentially leading to increased asthma risk. Despite significant advances, there are still unresolved questions about the long-term consequences of early microbial perturbations, particularly regarding whether microbial dysbiosis is a cause or consequence of asthma. This review integrates current findings, highlighting the need for deeper investigation into cross-organ interactions and early microbial exposures to understand childhood asthma pathophysiology.
Collapse
Affiliation(s)
- Céline Pattaroni
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Benjamin J. Marsland
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Nicola L. Harris
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
40
|
Ke H, Yao H, Wei P. Advances in research on gut microbiota and allergic diseases in children. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100362. [PMID: 40123594 PMCID: PMC11930230 DOI: 10.1016/j.crmicr.2025.100362] [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] [Indexed: 03/25/2025] Open
Abstract
Epidemiological studies indicate a rising prevalence of allergic diseases, now recognized as a major global public health concern. In children, the progression of these diseases often follows the "atopic march," beginning with eczema, followed by food allergies, allergic rhinitis, and asthma. Recent research has linked gut microbiota dysbiosis to the development of allergic diseases in children. The gut microbiota, a crucial component of human health, plays a vital role in maintaining overall well-being, highlighting its potential in preventing and modifying the course of allergic diseases. This review examines the relationship between childhood allergic diseases and gut microbiota, drawing on the latest evidence. We first elaborated the concepts of allergic diseases and gut microbiota, followed by a discussion of the developmental trajectory of the gut microbiota in healthy children. This review further explored the richness, diversity, and composition of the gut microbiota, as well as specific microbial taxa associated with allergic disease. Lastly, we discussed the current status and future potential of probiotic interventions in managing pediatric allergic diseases.
Collapse
Affiliation(s)
- Heng Ke
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Hongbing Yao
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ping Wei
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
41
|
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
|
42
|
Eisen A, Kiernan MC. The Neonatal Microbiome: Implications for Amyotrophic Lateral Sclerosis and Other Neurodegenerations. Brain Sci 2025; 15:195. [PMID: 40002527 PMCID: PMC11852589 DOI: 10.3390/brainsci15020195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Most brain development occurs in the "first 1000 days", a critical period from conception to a child's second birthday. Critical brain processes that occur during this time include synaptogenesis, myelination, neural pruning, and the formation of functioning neuronal circuits. Perturbations during the first 1000 days likely contribute to later-life neurodegenerative disease, including sporadic amyotrophic lateral sclerosis (ALS). Neurodevelopment is determined by many events, including the maturation and colonization of the infant microbiome and its metabolites, specifically neurotransmitters, immune modulators, vitamins, and short-chain fatty acids. Successful microbiome maturation and gut-brain axis function depend on maternal factors (stress and exposure to toxins during pregnancy), mode of delivery, quality of the postnatal environment, diet after weaning from breast milk, and nutritional deficiencies. While the neonatal microbiome is highly plastic, it remains prone to dysbiosis which, once established, may persist into adulthood, thereby inducing the development of chronic inflammation and abnormal excitatory/inhibitory balance, resulting in neural excitation. Both are recognized as key pathophysiological processes in the development of ALS.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Matthew C. Kiernan
- Neuroscience Research Australia, University of New South Wales, Randwick, Sydney, NSW 2031, Australia;
| |
Collapse
|
43
|
Nai S, Song J, Su W, Liu X. Bidirectional Interplay Among Non-Coding RNAs, the Microbiome, and the Host During Development and Diseases. Genes (Basel) 2025; 16:208. [PMID: 40004537 PMCID: PMC11855195 DOI: 10.3390/genes16020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
It is widely known that the dysregulation of non-coding RNAs (ncRNAs) and dysbiosis of the gut microbiome play significant roles in host development and the progression of various diseases. Emerging evidence has highlighted the bidirectional interplay between ncRNAs and the gut microbiome. This article aims to review the current understanding of the molecular mechanisms underlying the crosstalk between ncRNAs, especially microRNA (miRNA), and the gut microbiome in the context of development and diseases, such as colorectal cancer, inflammatory bowel diseases, neurological disorders, obesity, and cardiovascular disease. Ultimately, this review seeks to provide a foundation for exploring the potential roles of ncRNAs and gut microbiome interactions as biomarkers and therapeutic targets for clinical diagnosis and treatment, such as ncRNA mimics, antisense oligonucleotides, and small-molecule compounds, as well as probiotics, prebiotics, and diets.
Collapse
Affiliation(s)
| | | | | | - Xiaoqian Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (S.N.); (J.S.); (W.S.)
| |
Collapse
|
44
|
Wu Z, Chen X, Han F, Leeansyah E. MAIT cell homing in intestinal homeostasis and inflammation. SCIENCE ADVANCES 2025; 11:eadu4172. [PMID: 39919191 PMCID: PMC11804934 DOI: 10.1126/sciadv.adu4172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025]
Abstract
Mucosa-associated invariant T (MAIT) cells are a large population of unconventional T cells widely distributed in the human gastrointestinal tract. Their homing to the gut is central to maintaining mucosal homeostasis and immunity. This review discusses the potential mechanisms that guide MAIT cells to the intestinal mucosa during homeostasis and inflammation, emphasizing the roles of chemokines, chemokine receptors, and tissue adhesion molecules. The potential influence of the gut microbiota on MAIT cell homing to different regions of the human gut is also discussed. Last, we introduce how organoid technology offers a potentially valuable approach to advance our understanding of MAIT cell tissue homing by providing a more physiologically relevant model that mimics the human gut tissue. These models may enable a detailed investigation of the gut-specific homing mechanisms of MAIT cells. By understanding the regulation of MAIT cell homing to the human gut, potential avenues for therapeutic interventions targeting gut inflammatory conditions such as inflammatory bowel diseases (IBD) may emerge.
Collapse
Affiliation(s)
- Zhengyu Wu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Xingchi Chen
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Fei Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Edwin Leeansyah
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
45
|
Kennedy EA, Weagley JS, Kim AH, Antia A, DeVeaux AL, Baldridge MT. Bacterial community assembly of specific pathogen-free neonatal mice. MICROBIOME 2025; 13:46. [PMID: 39920864 PMCID: PMC11804086 DOI: 10.1186/s40168-025-02043-8] [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: 01/08/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Neonatal mice are frequently used to model diseases that affect human infants. Microbial community composition has been shown to impact disease progression in these models. Despite this, the maturation of the early-life murine microbiome has not been well-characterized. We address this gap by characterizing the assembly of the bacterial microbiota of C57BL/6 and BALB/c litters from birth to adulthood across multiple independent litters. RESULTS The fecal microbiome of young pups is dominated by only a few pioneering bacterial taxa. These taxa are present at low levels in the microbiota of multiple maternal body sites, precluding a clear identification of maternal source. The pup microbiota begins diversifying after 14 days, coinciding with the beginning of coprophagy and the consumption of solid foods. Pup stool bacterial community composition and diversity are not significantly different from dams from day 21 onwards. Short-read shotgun sequencing-based metagenomic profiling of young pups enabled the assembly of metagenome-assembled genomes for strain-level analysis of these pioneer Ligilactobacillus, Streptococcus, and Proteus species. CONCLUSIONS Assembly of the murine microbiome occurs over the first weeks of postnatal life and is largely complete by day 21. This detailed view of bacterial community development across multiple commonly employed mouse strains informs experimental design, allowing researchers to better target interventions before, during, or after the maturation of the bacterial microbiota. The source of pioneer bacterial strains appears heterogeneous, as the most abundant taxa identified in young pup stool were found at low levels across multiple maternal body sites, suggesting diverse routes for seeding of the murine microbiome. Video Abstract.
Collapse
Affiliation(s)
- Elizabeth A Kennedy
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - James S Weagley
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew H Kim
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna L DeVeaux
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan T Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
46
|
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
|
47
|
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
|
48
|
Khatib MA, Aljaaly EA, Albajri E, Khalifa NA, Khateeb S, Ajabnoor SM, Radhwan D, Aljohani K, Hussein AY. Prevalence of Functional Gastrointestinal Disorders and Associated Risk Factors Among Preschool Children in the City of Jeddah and Surrounding Areas: A Cross-Sectional Study. Diagnostics (Basel) 2025; 15:242. [PMID: 39941171 PMCID: PMC11817443 DOI: 10.3390/diagnostics15030242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Functional gastrointestinal disorders (FGIDs) affect children's daily activities and overall performance due to gastrointestinal symptoms. This study assesses the prevalence and types of FGIDs in children living in Jeddah City and its countryside. It also examines factors that contribute to the incidence of these disorders and their impact on children's lifestyles. Methods: This cross-sectional study was conducted among 285 mothers of preschool children enrolled in kindergartens during the academic year 2020-2021. The Rome IV Diagnostic Questionnaire was sent out online through kindergartens to be filled out by the children's mothers. The questionnaire assessed the prevalence of FGIDs subjectively through symptoms and their frequency. Results: Among the 285 participants, 9% (n = 27) fit the diagnostic criteria for FGIDs. Common FGIDs included functional constipation, 3.5% (n = 10); postprandial distress syndrome, 2.4% (n = 7); functional abdominal pain-not otherwise specified, 1% (n = 3); and functional epigastric pain, 0.7% (n = 2). Significant risk factors for developing FGIDs among the children in the sample included being a preterm baby (p < 0.01), being previously diagnosed with a gastrointestinal condition (p < 0.010), having a family history of diarrhea or nausea and vomiting (p < 0.001 and p < 0.01, respectively), skipping lunch at kindergarten (p < 0.01), and having pre-existing food allergies (p < 0.01). Conclusions: FGIDs were prevalent among 9% of children in Jeddah City and its countryside. Functional constipation was the most common disorder. Factors associated with FGIDs in children included preterm birth, being previously diagnosed with a GI condition, a family history of gastrointestinal conditions, irregular eating habits, and food allergies.
Collapse
Affiliation(s)
- Mai A. Khatib
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.A.A.); (E.A.); (S.M.A.); (D.R.); (K.A.); (A.Y.H.)
- Food, Nutrition, and Lifestyle Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Obesity and Lifestyle Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Saudi Diabetes Research Group, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Elham A. Aljaaly
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.A.A.); (E.A.); (S.M.A.); (D.R.); (K.A.); (A.Y.H.)
- Medical Nutrition Therapy Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Eram Albajri
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.A.A.); (E.A.); (S.M.A.); (D.R.); (K.A.); (A.Y.H.)
- Obesity and Lifestyle Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nahlaa A. Khalifa
- Department of Respiratory Therapy, Faculty of Medical Rehabilitation Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Saleh Khateeb
- Faculty of Clinical Sciences, Fakeeh College for Medical Sciences, Jeddah 23323, Saudi Arabia;
| | - Sarah M. Ajabnoor
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.A.A.); (E.A.); (S.M.A.); (D.R.); (K.A.); (A.Y.H.)
- Food, Nutrition, and Lifestyle Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Daniah Radhwan
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.A.A.); (E.A.); (S.M.A.); (D.R.); (K.A.); (A.Y.H.)
| | - Khawlah Aljohani
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.A.A.); (E.A.); (S.M.A.); (D.R.); (K.A.); (A.Y.H.)
| | - Aisha Y. Hussein
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.A.A.); (E.A.); (S.M.A.); (D.R.); (K.A.); (A.Y.H.)
| |
Collapse
|
49
|
Zhu J, He M, Li S, Lei Y, Xiang X, Guo Z, Wang Q. Shaping oral and intestinal microbiota and the immune system during the first 1,000 days of life. Front Pediatr 2025; 13:1471743. [PMID: 39906673 PMCID: PMC11790674 DOI: 10.3389/fped.2025.1471743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
The first 1, 000 days of life, from the fetal stage of a woman's pregnancy to 2 years of age after the baby is born, is a critical period for microbial colonization of the body and development of the immune system. The immune system and microbiota exhibit great plasticity at this stage and play a crucial role in subsequent development and future health. Two-way communication and interaction between immune system and microbiota is helpful to maintain human microecological balance and immune homeostasis. Currently, there is a growing interest in the important role of the microbiota in the newborn, and it is believed that the absence or dysbiosis of human commensal microbiota early in life can have lasting health consequences. Thus, this paper summarizes research advances in the establishment of the oral and intestinal microbiome and immune system in early life, emphasizing the substantial impact of microbiota diversity in the prenatal and early postnatal periods, and summarizes that maternal microbes, mode of delivery, feeding practices, antibiotics, probiotics, and the environment shape the oral and intestinal microbiota of infants in the first 1, 000 days of life and their association with the immune system.
Collapse
Affiliation(s)
- Jie Zhu
- Institute of Infection, Immunology and Tumor Microenvironment, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Mingxin He
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Simin Li
- Institute of Infection, Immunology and Tumor Microenvironment, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Yumeng Lei
- Institute of Infection, Immunology and Tumor Microenvironment, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaochen Xiang
- Institute of Infection, Immunology and Tumor Microenvironment, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Zhi Guo
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Medical College, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Renk H, Schoppmeier U, Müller J, Kuger V, Neunhoeffer F, Gille C, Peter S. Oxygenation and intestinal perfusion and its association with perturbations of the early life gut microbiota composition of children with congenital heart disease. Front Microbiol 2025; 15:1468842. [PMID: 39881980 PMCID: PMC11775010 DOI: 10.3389/fmicb.2024.1468842] [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: 07/22/2024] [Accepted: 12/10/2024] [Indexed: 01/31/2025] Open
Abstract
Background Early life gut microbiota is known to shape the immune system and has a crucial role in immune homeostasis. Only little is known about composition and dynamics of the intestinal microbiota in infants with congenital heart disease (CHD) and potential influencing factors. Methods We evaluated the intestinal microbial composition of neonates with CHD (n = 13) compared to healthy controls (HC, n = 30). Fecal samples were analyzed by shotgun metagenomics. Different approaches of statistical modeling were applied to assess the impact of influencing factors on variation in species composition. Unsupervised hierarchical clustering of the microbial composition of neonates with CHD was used to detect associations of distinct clusters with intestinal tissue oxygenation and perfusion parameters, obtained by the "oxygen to see" (O2C) method. Results Overall, neonates with CHD showed an intestinal core microbiota dominated by the genera Enterococcus (27%) and Staphylococcus (20%). Furthermore, a lower abundance of the genera Bacteroides (8% vs. 14%), Parabacteroides (1% vs. 3%), Bifidobacterium (4% vs. 12%), and Escherichia (8% vs. 23%) was observed in CHD compared to HCs. CHD patients that were born by vaginal delivery showed a lower fraction of the genera Bacteroides (15% vs. 21%) and Bifidobacterium (7% vs. 22%) compared to HCs and in those born by cesarean section, these genera were not found at all. In infants with CHD, we found a significant impact of oxygen saturation (SpO2) on relative abundances of the intestinal core microbiota by multivariate analysis of variance (F[8,2] = 24.9, p = 0.04). Statistical modeling suggested a large proportional shift from a microbiota dominated by the genus Streptococcus (50%) in conditions with low SpO2 towards the genus Enterococcus (61%) in conditions with high SpO2. We identified three distinct compositional microbial clusters, corresponding neonates differed significantly in intestinal blood flow and global gut perfusion. Conclusion Early life differences in gut microbiota of CHD neonates versus HCs are possibly linked to oxygen levels. Delivery method may affect microbiota stability. However, further studies are needed to assess the effect of potential interventions including probiotics or fecal transplants on early life microbiota perturbations in neonates with CHD.
Collapse
Affiliation(s)
- Hanna Renk
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University Children’s Hospital Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
- Department of Pediatric Cardiology, Pulmonology and Pediatric Intensive Care Medicine, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Ulrich Schoppmeier
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| | - Jennifer Müller
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - Vanessa Kuger
- Department of Pediatric Cardiology, Pulmonology and Pediatric Intensive Care Medicine, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Felix Neunhoeffer
- Department of Pediatric Cardiology, Pulmonology and Pediatric Intensive Care Medicine, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, Heidelberg, Germany
| | - Silke Peter
- German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| |
Collapse
|