1
|
Qi C, Li Z, Tu H, Sun F, Guo W, Di C, He R, Ze X, Zhang L, Gao R, Hu P, Yang W, Li K, Liu J, Pan X, Jin Z, Sun J. 2'-FL and cross-feeding bifidobacteria reshaped the gut microbiota of infants with atopic dermatitis ex vivo and prevented dermatitis in mice post-microbiota transplantation through retinol metabolism activation. Gut Microbes 2025; 17:2474148. [PMID: 40025650 PMCID: PMC11881859 DOI: 10.1080/19490976.2025.2474148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
2'-Fucosyllactose (2'-FL), a predominant human milk oligosaccharide, plays a crucial role in the development of the infant gut microbiota and immune system. However, the microbiota of infants with atopic dermatitis (AD) often has difficulty utilizing 2'-FL. Here, we found that strains from human milk, Bifidobacterium bifidum FN120 and Bifidobacterium longum subsp. longum FN103, utilized 2'-FL for growth by cross-feeding. Through an ex vivo continuous fermentation system, we found that 2'-FL and cross-feeding bifidobacteria synergistically enhanced the production of short-chain fatty acids (SCFAs), particularly acetate and propionate, while reshaping the gut microbiota in infants with AD. The reshaped microbiota was then transplanted into oxazolone-induced mice. We observed that AD symptoms in mice were effectively prevented, with significant changes in the ileum microbiota and increased intestinal SCFA levels. RNA sequencing analysis of Peyer's patches in the small intestine revealed activation of the retinol metabolic pathway. Nontargeted metabolomics analysis revealed a significant increase in plasma retinoate levels, which correlated markedly with AD-related markers. Collectively, our study demonstrated that supplementation with cross-feeding bifidobacteria and 2'-FL reshaped the gut microbiota, activated retinol metabolic pathways, promoted immune tolerance, and thereby prevented AD. Our findings provide novel insights into the therapeutic potential of combining prebiotics and probiotics to modulate the gut - skin axis and support immune tolerance in early life, offering a promising strategy for infantile AD management and prevention.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Zhongxia Li
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Huayu Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Fang Sun
- Pediatrics, Jiaozhou Maternal and Child Health and Family Planning Service Center, Qingdao, China
| | - Wenbo Guo
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Can Di
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Ruikun He
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Xiaolei Ze
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Lintao Zhang
- Pediatrics, Jiaozhou Maternal and Child Health and Family Planning Service Center, Qingdao, China
| | - Ruijuan Gao
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Pengyue Hu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Wenjing Yang
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Kexin Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jiayi Liu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Xiaonan Pan
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Zilu Jin
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Tüsüz Önata E, Özdemir Ö. Fecal microbiota transplantation in allergic diseases. World J Methodol 2025; 15:101430. [DOI: 10.5662/wjm.v15.i2.101430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/27/2024] Open
Abstract
Microorganisms such as bacteria, fungi, viruses, parasites living in the human intestine constitute the human intestinal microbiota. Dysbiosis refers to compositional and quantitative changes that negatively affect healthy gut microbiota. In recent years, with the demonstration that many diseases are associated with dysbiosis, treatment strategies targeting the correction of dysbiosis in the treatment of these diseases have begun to be investigated. Faecal microbiota transplantation (FMT) is the process of transferring faeces from a healthy donor to another recipient in order to restore the gut microbiota and provide a therapeutic benefit. FMT studies have gained popularity after probiotic, prebiotic, symbiotic studies in the treatment of dysbiosis and related diseases. FMT has emerged as a potential new therapy in the treatment of allergic diseases as it is associated with the maintenance of intestinal microbiota and immunological balance (T helper 1/T helper 2 cells) and thus suppression of allergic responses. In this article, the definition, application, safety and use of FMT in allergic diseases will be discussed with current data.
Collapse
Affiliation(s)
- Ece Tüsüz Önata
- Division of Pediatric Allergy and Immunology, Medical Faculty, Sakarya University, Adapazarı 54100, Sakarya, Türkiye
| | - Öner Özdemir
- Division of Pediatric Allergy and Immunology, Medical Faculty, Sakarya University, Adapazarı 54100, Sakarya, Türkiye
| |
Collapse
|
3
|
Schoultz I, Claesson MJ, Dominguez‐Bello MG, Fåk Hållenius F, Konturek P, Korpela K, Laursen MF, Penders J, Roager H, Vatanen T, Öhman L, Jenmalm MC. Gut microbiota development across the lifespan: Disease links and health-promoting interventions. J Intern Med 2025; 297:560-583. [PMID: 40270478 PMCID: PMC12087861 DOI: 10.1111/joim.20089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
The gut microbiota plays a pivotal role in human life and undergoes dynamic changes throughout the human lifespan, from infancy to old age. During our life, the gut microbiota influences health and disease across life stages. This review summarizes the discussions and presentations from the symposium "Gut microbiota development from infancy to old age" held in collaboration with the Journal of Internal Medicine. In early infancy, microbial colonization is shaped by factors such as mode of delivery, antibiotic exposure, and milk-feeding practices, laying the foundation for subsequent increased microbial diversity and maturation. Throughout childhood and adolescence, microbial maturation continues, influencing immune development and metabolic health. In adulthood, the gut microbiota reaches a relatively stable state, influenced by genetics, diet, and lifestyle. Notably, disruptions in gut microbiota composition have been implicated in various inflammatory diseases-including inflammatory bowel disease, Type 1 diabetes, and allergies. Furthermore, emerging evidence suggests a connection between gut dysbiosis and neurodegenerative disorders such as Alzheimer's disease. Understanding the role of the gut microbiota in disease pathogenesis across life stages provides insights into potential therapeutic interventions. Probiotics, prebiotics, and dietary modifications, as well as fecal microbiota transplantation, are being explored as promising strategies to promote a healthy gut microbiota and mitigate disease risks. This review focuses on the gut microbiota's role in infancy, adulthood, and aging, addressing its development, stability, and alterations linked to health and disease across these critical life stages. It outlines future research directions aimed at optimizing the gut microbiota composition to improve health.
Collapse
Affiliation(s)
- Ida Schoultz
- School of Medical SciencesFaculty of Medicine and Health Örebro UniversityOrebroSweden
| | | | - Maria Gloria Dominguez‐Bello
- Department of Biochemistry & Microbiology and of AnthropologyRutgers University–New BrunswickNew BrunswickNew JerseyUSA
| | - Frida Fåk Hållenius
- Department of Food Technology, Engineering and NutritionLund UniversityLundSweden
| | - Peter Konturek
- Department of Medicine, Thuringia Clinic SaalfeldTeaching Hospital of the University JenaJenaGermany
| | - Katri Korpela
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | | | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtthe Netherlands
| | - H. Roager
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenFrederiksbergDenmark
| | - Tommi Vatanen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFinland
- Department of Microbiology, Faculty of Agriculture and ForestryUniversity of HelsinkiHelsinkiFinland
- Research Program for Clinical and Molecular Metabolism, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Liggins InstituteUniversity of AucklandAucklandNew Zealand
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maria C. Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden
| |
Collapse
|
4
|
Imoto N, Kano C, Morita H, Hirota T, Amanuma F, Maruyama H, Nojiri S, Watanabe S. Impact of antimicrobial exposure at delivery and siblings on early Bifidobacterium succession and allergy development up to 24 months of age. BMC Microbiol 2025; 25:332. [PMID: 40426074 DOI: 10.1186/s12866-025-04056-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Allergic diseases such as asthma, eczema, and food allergies are rising globally. The infant gut microbiota, particularly the dominance of Bifidobacterium, shapes immune development and allergy risk. In Japan-where Bifidobacterium prevalence is notably high-longitudinal investigations focusing on the pre-weaning period, when external influences are relatively limited, remain scarce. Therefore, based on consistent hypotheses and findings from previous studies, we investigated how two important early factors-antibiotic exposure at birth and the presence of older siblings-influence the gut environment in early infancy and subsequent allergy development. RESULTS In a prospective cohort of 121 Japanese infants, stool samples were collected at seven time points from birth through 24 months. We quantified the relative abundances of Bifidobacterium, Bacteroides, Clostridium, and Faecalibacterium and recorded allergic outcomes at 2 years. Both antimicrobial exposure at delivery and sibling presence significantly altered gut microbiota composition and overall diversity in early infancy. Although the full cohort showed no consistent diversity or Bifidobacterium differences by allergic status, in several subgroups where these two factors were excluded, infants who had an allergy by 24 months exhibited marked shifts in early gut microbiota community structure-particularly in beta diversity-and reduced Bifidobacterium occupancy during the pre-weaning period (1-6 months) versus non-allergic peers. Moreover, infants whose gut microbiota was initially affected by these factors showed a recovery in diversity after weaning, a rebound more pronounced in non-allergic individuals. CONCLUSIONS These findings indicate that both the initial community configuration and its capacity to rebound after perturbation are critical determinants of allergy risk. By focusing on dynamic changes through weaning and adjusting for decisive confounders, this study refines insight beyond prior cross-sectional work. Early interventions that preserve or restore microbial diversity and Bifidobacterium dominance may therefore offer a promising strategy to mitigate allergic disease development.
Collapse
Affiliation(s)
- Naruaki Imoto
- Advanced Research Institute for Health Science, Juntendo University, Bunkyo Ward, Tokyo, 113-8421, Japan.
| | - Chie Kano
- Core Technology Laboratories, Asahi Quality & Innovations Ltd, Midori, Moriya, 302- 0106, Ibaraki, Japan
| | - Hiroto Morita
- Core Technology Laboratories, Asahi Quality & Innovations Ltd, Midori, Moriya, 302- 0106, Ibaraki, Japan
| | - Tatsuhiko Hirota
- Core Technology Laboratories, Asahi Quality & Innovations Ltd, Midori, Moriya, 302- 0106, Ibaraki, Japan
| | - Fumitaka Amanuma
- Department of Paediatrics, Department of Neonatology, Iwate Prefectural Iwai Hospital, Ichinoseki, 029-0192, Iwate, Japan
| | - Hidekazu Maruyama
- Department of Paediatrics, Department of Neonatology, Iwate Prefectural Iwai Hospital, Ichinoseki, 029-0192, Iwate, Japan
| | - Shuko Nojiri
- Clinical Research Support Centre, Juntendo University, Bunkyo Ward, Tokyo, 113-8421, Japan
| | - Shin Watanabe
- Advanced Research Institute for Health Science, Juntendo University, Bunkyo Ward, Tokyo, 113-8421, Japan
| |
Collapse
|
5
|
Gallant R, Reza S, Wiemels JL, Greaves M. Microbiome and pediatric leukemia, diabetes, and allergies: Systematic review and meta-analysis. PLoS One 2025; 20:e0324167. [PMID: 40392825 PMCID: PMC12091780 DOI: 10.1371/journal.pone.0324167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 04/21/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Despite the different pathologies and genetic susceptibilities of childhood ALL, T1DM and allergies, these conditions share epidemiological risk factors related to timing of infectious exposures and acquisition of the gut microbiome in infancy. We have assessed whether lower microbiome diversity (Shannon Index) and shared genus/species profiles are associated with pediatric ALL, allergies, and T1DM. METHODS AND FINDINGS Literature search was performed using PubMed, Embase, Cochrane, and Web of Science databases. Case-control, meta-analyses, and cohort studies were considered for inclusion. Inclusion criteria: (i) subjects age 1-18 years at diagnosis, (ii) reports effect of microbiome measured prior to/at time of diagnosis/first intervention (iii) outcome of ALL, allergies, asthma, or T1DM, (iv) English text. Exclusion criteria: (i) age < 1 or >18 years at diagnosis, (ii) Down Syndrome-associated ALL, (iii) non-English text, (iv) reviews, pre-print, or abstracts, (v) heavily biased studies. Abstract and full text screening were performed by two independent reviewers. Data extraction was performed by one reviewer following PRISMA guidelines. Data were pooled using a random-effects model. Eighty-eight studies were included in the analysis, with seventy-seven in the qualitative analysis and 54 in the meta-analysis. Cases were found to have lower alpha-diversity than controls in ALL (SMD:-0.78, 95%CI:-1.21, -0.34), T1DM (SMD:-1.26, 95%CI:-3.49, 0.96), eczema (SMD:-0.34, 95%CI:-0.56, -0.12), atopy (SMD:-0.06, 95%CI:-0.34, 0.22), asthma (SMD:-0.37, 95%CI:-1.16, 0.42), and food allergy (SMD:-0.11, 95%CI:-0.63, 0.41). CONCLUSIONS These results highlight similarities in the microbiome diversity and composition of children with ALL, T1DM, and allergies. This is compatible with a common risk factor related to immune priming in infancy and highlights the gut microbiome as a potentially modifiable risk factor and preventative strategy for these childhood diseases.
Collapse
Affiliation(s)
- Rachel Gallant
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
- Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Samiha Reza
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Joseph L. Wiemels
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, University of Southern California Keck School of Medicine, Los Angeles, California, United States of America
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
6
|
Liu X, Luo Y, Chen X, Wu M, Xu X, Tian J, Gao Y, Zhu J, Wang Z, Zhou Y, Zhang Y, Wang X, Li W, Lu Q, Yao X. Fecal microbiota transplantation against moderate-to-severe atopic dermatitis: A randomized, double-blind controlled explorer trial. Allergy 2025; 80:1377-1388. [PMID: 39470619 DOI: 10.1111/all.16372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) is a novel treatment for inflammatory diseases. Herein, we assess its safety, efficacy, and immunological impact in patients with moderate-to-severe atopic dermatitis (AD). METHODS In this randomized, double-blind, placebo-controlled clinical trial, we performed the efficacy and safety assessment of FMT for moderate-to-severe adult patients with AD. All patients received FMT or placebo once a week for 3 weeks, in addition to their standard background treatments. Patients underwent disease severity assessments at weeks 0, 1, 2, 4, 8, 12, and 16, and blood and fecal samples were collected for immunologic analysis and metagenomic shotgun sequencing, respectively. Safety was monitored throughout the trial. RESULTS Improvements in eczema area and severity index (EASI) scores and percentage of patients achieving EASI 50 (50% reduction in EASI score) were greater in patients treated with FMT than in placebo-treated patients. No serious adverse reactions occurred during the trial. FMT treatment decreased the Th2 and Th17 cell proportions among the peripheral blood mononuclear cells, and the levels of TNF-α, and total IgE in serum. By contrast, the expression levels of IL-12p70 and perforin on NK cells were increased. Moreover, FMT altered the abundance of species and functional pathways of the gut microbiota in the patients, especially the abundance of Megamonas funiformis and the pathway for 1,4-dihydroxy-6-naphthoate biosynthesis II. CONCLUSION FMT was a safe and effective therapy in moderate-to-severe adult patients with AD; the treatment changed the gut microbiota compositions and functions.
Collapse
Affiliation(s)
- Xiaochun Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yang Luo
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xingyu Chen
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Mingyang Wu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiaoqiang Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Jingru Tian
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yingxia Gao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jun Zhu
- 01life Institute, Shenzhen, China
| | | | - Yuan Zhou
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yu Zhang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiaokai Wang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Wei Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Qianjin Lu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
7
|
Manninen J, Saarenpää M, Roslund M, Galitskaya P, Sinkkonen A. Microbial communities on dry natural rocks are richer and less stressed than those on man-made playgrounds. Microbiol Spectr 2025; 13:e0193024. [PMID: 40202313 PMCID: PMC12054085 DOI: 10.1128/spectrum.01930-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
In modern urbanized societies, the incidence of major immune-mediated diseases is several times higher than before World War II. A potential explanation is that these diseases are triggered by limited possibilities to be exposed to rich environmental microbiota. This requires that the urban environment hosts less and poorer microbiota than the natural environment. The current study was designed to test the assumption that urban man-made environments host less and poorer environmental microbiota, compared to natural habitats. We selected two types of dry environments, natural rocks and playground rubber mats, both of which were used daily and extensively by children. In quantitative PCR and next-generation sequencing, bacterial abundance and richness were higher on the natural rocks than the rubber mats. Altogether, 67 amplicon sequence variants (ASVs) belonging mostly to Actinobacteria and Proteobacteria were indicative of rock microbiota, while three ASVs were indicative of rubber mats. Interestingly, bacteria formed more complex networks on rubber mats than natural rocks. Based on the literature, this indicates that the studied artificial dry environment is more challenging and stressful for bacterial communities than dry natural rocks. The results support the hypothesis that urban man-made environments host poor microbial communities, which is in accordance with the biodiversity hypothesis of immune-mediated diseases.IMPORTANCEThe current study provides new evidence that artificial urban play environments host poor microbial communities and provide a stressful environment for microbes, as compared to dry natural rocks. Through this, the current study underlines the need to enhance microbial diversity in urban areas, especially in outdoor play environments, which have a crucial role in providing essential microbial exposure for the development of children's immune system. This research can potentially offer guidance for urban planning and public health strategies that support planetary health.
Collapse
Affiliation(s)
- J. Manninen
- Faculty of Biological and Environmental Sciences, Ecosystems and Environment Research programme, University of Helsinki, Helsinki, Finland
| | - M. Saarenpää
- Faculty of Biological and Environmental Sciences, Ecosystems and Environment Research programme, University of Helsinki, Helsinki, Finland
| | - M. Roslund
- Natural Resources Institute Finland Luke, Helsinki, Finland
| | - P. Galitskaya
- Research Institute for Environmental Studies, Parede, Portugal
| | - A. Sinkkonen
- Natural Resources Institute Finland Luke, Helsinki, Finland
| |
Collapse
|
8
|
Chen CC, Yeh YM, Chen KJ, Chang HJ, Cheng ML, Lo CJ, Lai HC. Gut Microbiota and Related Metabolites in Children With Egg White Sensitization. Pediatr Infect Dis J 2025; 44:299-309. [PMID: 39637305 DOI: 10.1097/inf.0000000000004628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
BACKGROUND We hypothesized that food sensitization in children could be linked to specific gut microbiota. The objective of this study is to assess a group of children with egg white sensitization (ES) from the microbiological and biochemical-metabolic standpoint, applying the microbiota and metabolomics approach to studying the intestinal contents of the feces. METHODS Twenty-eight toddlers with ES (mean age 13.08 months) and 24 healthy controls (mean age 12.85 months) were recruited for feces collection, serum IgE measurement, gut microbiota and metabolomics analysis. Individual microbial diversity and composition were analyzed via targeting the 16S rRNA gene hypervariable V3-V5 regions. The metabolite profiles of human feces were explored by 1 H nuclear magnetic resonance. RESULTS Children with ES exhibited relatively high levels of Firmicutes at the phylum level and relatively low levels of Bacteroidetes. Moreover, children with ES exhibited significantly reduced overall gut microbiota diversity and richness compared with healthy children. At the family level, we observed significant increases in the numbers of Clostridiaceae, Lachnospiraceae, Pasteurellaceae and Ruminococcaceae in children with ES. Egg white sensitivity increases orotic acid, nicotinate, methyl succinate, urocanic acid, xanthine, amino acids (tyrosine, lysine, tryptophan, phenylalanine) and short-chain fatty acids ( n -butyrate, valerate) levels according to the results of metabolomics analysis. CONCLUSIONS In summary, some specific families and genera (dysbiosis) are enriched in the gut microbiota, and increases in the mean concentrations of organic compounds in the fecal metabolite profile are associated with ES in children. These findings may provide evidence of potential strategies to control the development of ES or other atopies by modifying the gut microbiota.
Collapse
Affiliation(s)
- Chien-Chang Chen
- From the Division of Gastroenterology, Department of Pediatrics, Chang Gung Children's Hospital, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yuan-Ming Yeh
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kun-Jei Chen
- From the Division of Gastroenterology, Department of Pediatrics, Chang Gung Children's Hospital, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Hung-Ju Chang
- From the Division of Gastroenterology, Department of Pediatrics, Chang Gung Children's Hospital, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Mei-Ling Cheng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine
- Metabolomics Core Laboratory, Healthy Aging Research Center
| | - Chi-Jen Lo
- Department of Medical Biotechnology and Laboratory Science, College of Medicine
- Metabolomics Core Laboratory, Healthy Aging Research Center
| | - Hsin-Chih Lai
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
9
|
Díez-Madueño K, Montero I, Fernández-Gosende M, Martínez-Álvarez N, Hidalgo-Cantabrana C, de la Cueva Dobao P, Coto-Segura P. Compositional and Functional Profile of Gut Microbiota in a Cohort of Adult Spanish Patients with Atopic Dermatitis Using Metagenomics: A Cross-Sectional Study. Dermatitis 2025. [PMID: 40111891 DOI: 10.1089/derm.2024.0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Background: The role of gut dysbiosis in the pathophysiology of atopic dermatitis (AD) through immune system (IS) imbalance is a novel line of investigation currently under discussion. This study aimed to characterize compare the composition and functional profile of the gut microbiota (GM) between adults with AD and healthy individuals. Methods: Observational cross-sectional study, where fecal samples from 70 adults (38 patients and 32 controls) were analyzed using metagenomics and bioinformatics. Results: Differences between the GM of patients with AD and healthy individuals were demonstrated. Reduced microbial diversity was found in subjects with AD. Bacterial species with lower abundance primarily belonged to the families Ruminococcaceae, Akkermansiaceae, and Methanobacteriaceae. Several microbial metabolic pathways were found to be decreased in patients with AD, including amino acid biosynthesis, vitamin biosynthesis, fatty acids and lipids biosynthesis, and energy metabolism. Conclusion: Adults with AD exhibited a distinct GM compared to healthy individuals. Changes were demonstrated both compositionally and functionally. Further investigation is mandatory to elucidate the potential link and causal relationship between gut dysbiosis and AD, which may be crucial for a deeper understanding of the disease's pathophysiology and the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Kevin Díez-Madueño
- From the Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | | | | | | | | | - Pablo de la Cueva Dobao
- From the Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | - Pablo Coto-Segura
- Dermatology Department, Hospital Vital Álvarez Buylla, Mieres, Spain
| |
Collapse
|
10
|
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
|
11
|
Anania C, Matys V, Marra S, De Canditiis D, Olivero F, Carraro C, Giugliano A, Zicari AM, Piccioni MG. Effect of Supplementation with a Specific Probiotic ( Bifidobacterium bifidum PRL2010) in Pregnancy for the Prevention of Atopic Dermatitis in Children: Preliminary Results of a Randomized Trial. Nutrients 2025; 17:673. [PMID: 40005000 PMCID: PMC11858544 DOI: 10.3390/nu17040673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by the appearance of recurrent eczematous lesions and intense itching. The World Allergy Organization (WAO) suggested the administration of probiotics in pregnant women at high risk of allergies in their children. OBJECTIVES Our study aims to evaluate the role of administering the Bifidobacterium bifidum strain PRL2010 during pregnancy and breastfeeding in preventing and/or reducing the severity of AD manifestations in children. METHODS It is a monocentric, randomized, double-blind, placebo-controlled trial with probiotic/placebo administration since the 36th week of gestation to mothers with atopy or a family history of atopy; the effects were evaluated over the first 12 months of the children's lives. RESULTS No severe adverse effects due to probiotic intake were reported in our cohort. Although proportionally fewer children with AD were in the probiotic group, the statistical analysis showed no significant differences between the probiotic and placebo groups. However, infants who developed the most severe forms of AD in the probiotic group showed a better clinical course during follow-up compared to those in the placebo group. CONCLUSIONS In conclusion, administering the probiotic Bifidobacterium bifidum strain PRL2010 during pregnancy and breastfeeding is safe and potentially beneficial; further large-scale studies may confirm its usefulness in improving the clinical manifestation of AD in children with a family history of atopy.
Collapse
Affiliation(s)
- Caterina Anania
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.M.); (S.M.); (C.C.); (A.G.); (A.M.Z.); (M.G.P.)
| | - Viviana Matys
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.M.); (S.M.); (C.C.); (A.G.); (A.M.Z.); (M.G.P.)
| | - Simona Marra
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.M.); (S.M.); (C.C.); (A.G.); (A.M.Z.); (M.G.P.)
| | | | | | - Carlo Carraro
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.M.); (S.M.); (C.C.); (A.G.); (A.M.Z.); (M.G.P.)
| | - Anna Giugliano
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.M.); (S.M.); (C.C.); (A.G.); (A.M.Z.); (M.G.P.)
| | - Anna Maria Zicari
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.M.); (S.M.); (C.C.); (A.G.); (A.M.Z.); (M.G.P.)
| | - Maria Grazia Piccioni
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.M.); (S.M.); (C.C.); (A.G.); (A.M.Z.); (M.G.P.)
| |
Collapse
|
12
|
Chakravarty K, Gaur S, Kumar R, Jha NK, Gupta PK. Exploring the Multifaceted Therapeutic Potential of Probiotics: A Review of Current Insights and Applications. Probiotics Antimicrob Proteins 2025; 17:341-363. [PMID: 39069588 DOI: 10.1007/s12602-024-10328-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
The interplay between human health and the microbiome has gained extensive attention, with probiotics emerging as pivotal therapeutic agents due to their vast potential in treating various health issues. As significant modulators of the gut microbiota, probiotics are crucial in maintaining intestinal homeostasis and enhancing the synthesis of short-chain fatty acids. Despite extensive research over the past decades, there remains an urgent need for a comprehensive and detailed review that encapsulates probiotics' latest insights and applications. This review focusses on the multifaceted roles of probiotics in promoting health and preventing disease, highlighting the complex mechanisms through which these beneficial bacteria influence both gut flora and the human body at large. This paper also explores probiotics' neurological and gastrointestinal applications, focussing on their significant impact on the gut-brain axis and their therapeutic potential in a broad spectrum of pathological conditions. Current innovations in probiotic formulations, mainly focusing on integrating genomics and biotechnological advancements, have also been comprehensively discussed herein. This paper also critically examines the regulatory landscape that governs probiotic use, ensuring safety and efficacy in clinical and dietary settings. By presenting a comprehensive overview of recent studies and emerging trends, this review aims to illuminate probiotics' extensive therapeutic capabilities, leading to future research and clinical applications. However, besides extensive research, further advanced explorations into probiotic interactions and mechanisms will be essential for developing more targeted and effective therapeutic strategies, potentially revolutionizing health care practices for consumers.
Collapse
Affiliation(s)
- Kashyapi Chakravarty
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, 201309, India
| | - Smriti Gaur
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, 201309, India.
| | - Rohit Kumar
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Niraj Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 602105, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, 140401, India
| | - Piyush Kumar Gupta
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India.
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, 248002, India.
| |
Collapse
|
13
|
Crabtree D, Seidler K, Barrow M. Pathophysiological mechanisms of gut dysbiosis and food allergy and an investigation of probiotics as an intervention for atopic disease. Clin Nutr ESPEN 2025; 65:189-204. [PMID: 39571752 DOI: 10.1016/j.clnesp.2024.11.019] [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: 10/26/2024] [Accepted: 11/15/2024] [Indexed: 12/08/2024]
Abstract
BACKGROUND AND AIMS Epidemiological studies have associated reduced bacterial diversity and abundance and food allergy. This mechanistic review investigated the link between gut dysbiosis and food allergy with a focus on the role of short-chain fatty acids (SCFAs) in modulating T-cells. T-cell differentiation poses an opportunity to direct the immune cells towards an anergic regulatory T cell (Treg) or allergic T helper 2 (Th2) response. Probiotic intervention to prevent and/or treat atopic disease symptoms through this mechanistic pathway was explored. METHODOLOGY A narrative review was conducted following a three-stage systematic literature search of EMBASE and Medline databases. Ninety-six of 571 papers were accepted and critically appraised using ARRIVE and SIGN50 forms. Thematic analysis identified key pathophysiological mechanisms within the narrative of included papers. RESULTS Preclinical studies provided compelling evidence for SCFAs' modulation of T-cell differentiation, which may act through G-protein coupled receptors 41, 43 and 109a and histone deacetylase inhibition. Foxp3 transcription factor was implicated in the upregulation of Tregs. Human probiotic intervention studies aimed at increasing SCFAs and Tregs and preventing atopic disease showed inconclusive results. However, evidence for probiotic intervention in children with cow's milk protein allergy (CMPA) was more promising and warrants further investigation. CONCLUSION Preclinical evidence suggests that the mechanism of gut dysbiosis and reduced SCFAs may skew T-cell differentiation towards a Th2 response, thus inducing allergy symptoms. Probiotic trials were inconclusive: probiotics were predominantly unsuccessful in the prevention of allergic disease, however, may be able to modulate food allergy symptoms in infants with CMPA.
Collapse
Affiliation(s)
- Danielle Crabtree
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| | - Karin Seidler
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| | - Michelle Barrow
- Centre for Nutrition Education and Lifestyle Management, PO Box 3739, Wokingham, RG40 9UA, UK.
| |
Collapse
|
14
|
Dera N, Kosińska-Kaczyńska K, Żeber-Lubecka N, Brawura-Biskupski-Samaha R, Massalska D, Szymusik I, Dera K, Ciebiera M. Impact of Early-Life Microbiota on Immune System Development and Allergic Disorders. Biomedicines 2025; 13:121. [PMID: 39857705 PMCID: PMC11762082 DOI: 10.3390/biomedicines13010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Introduction: The shaping of the human intestinal microbiota starts during the intrauterine period and continues through the subsequent stages of extrauterine life. The microbiota plays a significant role in the predisposition and development of immune diseases, as well as various inflammatory processes. Importantly, the proper colonization of the fetal digestive system is influenced by maternal microbiota, the method of pregnancy completion and the further formation of the microbiota. In the subsequent stages of a child's life, breastfeeding, diet and the use of antibiotics influence the state of eubiosis, which determines proper growth and development from the neonatal period to adulthood. The literature data suggest that there is evidence to confirm that the intestinal microbiota of the infant plays an important role in regulating the immune response associated with the development of allergic diseases. However, the identification of specific bacterial species in relation to specific types of reactions in allergic diseases is the basic problem. Background: The main aim of the review was to demonstrate the influence of the microbiota of the mother, fetus and newborn on the functioning of the immune system in the context of allergies and asthma. Methods: We reviewed and thoroughly analyzed the content of over 1000 articles and abstracts between the beginning of June and the end of August 2024. Over 150 articles were selected for the detailed study. Results: The selection was based on the PubMed National Library of Medicine search engine, using selected keywords: "the impact of intestinal microbiota on the development of immune diseases and asthma", "intestinal microbiota and allergic diseases", "the impact of intrauterine microbiota on the development of asthma", "intrauterine microbiota and immune diseases", "intrauterine microbiota and atopic dermatitis", "intrauterine microbiota and food allergies", "maternal microbiota", "fetal microbiota" and "neonatal microbiota". The above relationships constituted the main criteria for including articles in the analysis. Conclusions: In the present review, we showed a relationship between the proper maternal microbiota and the normal functioning of the fetal and neonatal immune system. The state of eubiosis with an adequate amount and diversity of microbiota is essential in preventing the development of immune and allergic diseases. The way the microbiota is shaped, resulting from the health-promoting behavior of pregnant women, the rational conduct of the medical staff and the proper performance of the diagnostic and therapeutic process, is necessary to maintain the health of the mother and the child. Therefore, an appropriate lifestyle, rational antibiotic therapy as well as the way of completing the pregnancy are indispensable in the prevention of the above conditions. At the same time, considering the intestinal microbiota of the newborn in relation to the genera and phyla of bacteria that have a potentially protective effect, it is worth noting that the use of suitable probiotics and prebiotics seems to contribute to the protective effect.
Collapse
Affiliation(s)
- Norbert Dera
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
| | - Katarzyna Kosińska-Kaczyńska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Center of Postgraduate Medical Education, 02-781 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Robert Brawura-Biskupski-Samaha
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Diana Massalska
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| | - Iwona Szymusik
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Kacper Dera
- Pediatric Ward, Department of Pediatrics, Center of Postgraduate Medical Education, Bielański Hospital, 01-809 Warsaw, Poland
| | - Michał Ciebiera
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| |
Collapse
|
15
|
Du B, Shama A, Zhang Y, Chen B, Bu Y, Chen PA, Lin C, Liu J, Zheng J, Li Z, Chen Q, Sun Y, Fu X. Gut microbiota and plasma metabolites in pregnant mothers and infant atopic dermatitis: A multi-omics study. World Allergy Organ J 2025; 18:101017. [PMID: 39850616 PMCID: PMC11754505 DOI: 10.1016/j.waojou.2024.101017] [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: 06/10/2024] [Revised: 11/08/2024] [Accepted: 12/02/2024] [Indexed: 01/25/2025] Open
Abstract
Background Many studies reported the influence of infants' gut microbiota on atopic dermatitis (AD) postnatally, yet the role of maternal gut microbiota and plasma metabolites in infants' AD remains largely unexplored. Methods Sixty-three pregnant mother-infants were enrolled and followed after childbirth in Guangzhou, China. Demographic information, maternal stool and plasma samples, and records for infants' AD were collected. Maternal gut microbiota/metabolome and plasma metabolome were profiled using shotgun metagenomics and non-targeted metabolomics. Logistic regression and multi-omics analysis were used to explore characteristic maternal gut microbiota in the AD and health groups. Results The α-diversity of maternal gut microbiota in health group was significantly higher than AD group (Shannon diversity P = 0.02, Simpson diversity P = 0.04). Short-chain fatty acids (SCFAs) producing microorganisms, including Faecalibacterium, Roseburia, Butyricicoccus, and Ruminococcus, as well as the abundance of phenylalanine, tyrosine, and tryptophan biosynthesis pathway, were enriched in health group (LDA>2 and P < 0.05). Virulent factors (VFs) involved in immune modulation were enriched in the health group, while VFs involving in adhesin were enriched in the AD group (P < 0.05). Metabolomic analysis showed that a polyunsaturated fatty acid/linoleic acid, 13S-hydroxyoctadecadienoic, were negatively associated with AD in both the gut and plasma samples (FDR<0.05). Several other linoleic acids and flavonoids were negatively associated with AD (FDR<0.05). Neural network analysis revealed that microorganisms enriched in health group may produce these protective fatty acids. Conclusions Our findings show that maternal gut microorganisms/metabolites and plasma metabolites during pregnancy impact subsequent pathogenesis of infants AD. This illuminates new strategies against early AD in offspring.
Collapse
Affiliation(s)
- Bingqian Du
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102200, PR China
| | - Aga Shama
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
| | - Yi Zhang
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
| | - Baolan Chen
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
| | - Yongqi Bu
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
| | - Pei-an Chen
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
| | - Chuzhi Lin
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
| | - Jie Liu
- Maternity and Child Health Hospital of Baiyun District, Guangzhou, 510400, Guangdong, PR China
| | - Juan Zheng
- Maternity and Child Health Hospital of Baiyun District, Guangzhou, 510400, Guangdong, PR China
| | - Zhenjun Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102200, PR China
| | - Qingsong Chen
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
| | - Yu Sun
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China
| | - Xi Fu
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, PR China
| |
Collapse
|
16
|
Palmer DJ, Cuthbert AR, Sullivan TR, Pretorius RA, Garssen J, Rueter K, Jenmalm MC, Keelan JA, Silva D, Prescott SL. Effects of pregnancy and lactation prebiotics supplementation on infant allergic disease: A randomized controlled trial. J Allergy Clin Immunol 2025; 155:144-152. [PMID: 39173718 DOI: 10.1016/j.jaci.2024.08.009] [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: 04/21/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Ingestion of prebiotics during pregnancy and lactation may have immunomodulatory benefits for the developing fetal and infant immune system and provide a potential dietary strategy to reduce the risk of allergic diseases. OBJECTIVE We sought to determine whether maternal supplementation with dietary prebiotics reduces the risk of allergic outcomes in infants with hereditary risk. METHODS We undertook a double-blind randomized controlled trial in which pregnant women were allocated to consume prebiotics (14.2 g daily of galacto-oligosaccharides and fructo-oligosaccharides in the ratio 9:1) or placebo (8.7 g daily of maltodextrin) powder from less than 21 weeks' gestation until 6 months postnatal during lactation. Eligible women had infants with a first-degree relative with a history of medically diagnosed allergic disease. The primary outcome was medically diagnosed infant eczema by age 1 year, and secondary outcomes included allergen sensitization, food allergy, and recurrent wheeze by age 1 year. RESULTS A total of 652 women were randomized between June 2016 and November 2021 (329 in the prebiotics group and 323 in the placebo group). There was no significant difference between groups in the percentage of infants with medically diagnosed eczema by age 1 year (prebiotics 31.5% [103 of 327 infants] vs placebo 32.6% [105 of 322 infants]; adjusted relative risk, 0.98; 95% CI, 0.77-1.23; P = .84). Secondary outcomes and safety measures also did not significantly differ between groups. CONCLUSIONS We found little evidence that maternal prebiotics supplementation during pregnancy and lactation reduces the risk of medically diagnosed infant eczema by age 1 year in infants who are at hereditary risk of allergic disease.
Collapse
Affiliation(s)
- Debra J Palmer
- Telethon Kids Institute, The University of Western Australia, Nedlands, Australia; School of Medicine, The University of Western Australia, Crawley, Australia.
| | - Alana R Cuthbert
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Thomas R Sullivan
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, Australia; School of Public Health, The University of Adelaide, Adelaide, Australia
| | - Rachelle A Pretorius
- Telethon Kids Institute, The University of Western Australia, Nedlands, Australia; School of Medicine, The University of Western Australia, Crawley, Australia
| | - Johan Garssen
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Nutricia Research, Utrecht, The Netherlands
| | - Kristina Rueter
- School of Medicine, The University of Western Australia, Crawley, Australia; Department of Immunology and Dermatology, Perth Children's Hospital, Nedlands, Australia
| | - Maria C Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Jeffrey A Keelan
- School of Medicine, The University of Western Australia, Crawley, Australia; School of Biomedical Sciences, The University of Western Australia, Crawley, Australia
| | - Desiree Silva
- Telethon Kids Institute, The University of Western Australia, Nedlands, Australia; School of Medicine, The University of Western Australia, Crawley, Australia; Joondalup Health Campus, Joondalup, Australia; Edith Cowan University, Perth, Australia
| | - Susan L Prescott
- Telethon Kids Institute, The University of Western Australia, Nedlands, Australia; School of Medicine, The University of Western Australia, Crawley, Australia; Department of Immunology and Dermatology, Perth Children's Hospital, Nedlands, Australia; Nova Institute for Health, Baltimore, Md
| |
Collapse
|
17
|
Díez-Madueño K, de la Cueva Dobao P, Torres-Rojas I, Fernández-Gosende M, Hidalgo-Cantabrana C, Coto-Segura P. Gut Dysbiosis and Adult Atopic Dermatitis: A Systematic Review. J Clin Med 2024; 14:19. [PMID: 39797102 PMCID: PMC11721037 DOI: 10.3390/jcm14010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Research on the relationship between gut microbiota (GM) and atopic dermatitis (AD) has seen a growing interest in recent years. The aim of this systematic review was to determine whether differences exist between the GM of adults with AD and that of healthy adults (gut dysbiosis). Methods: We conducted a systematic review based on the PRISMA guidelines (Preferred Reporting Items for Systematic Reviews and Meta-Analyses). The search was performed using PubMed, EMBASE, and Web of Science. Observational and interventional studies were analyzed. Results: Although the studies showed heterogeneous results, some distinguishing characteristics were found in the intestinal microbial composition of adults with dermatitis. Even though no significant differences in diversity were found between healthy and affected adults, certain microorganisms, such as Bacteroidales, Enterobacteriaceae, and Clostridium (perfringens), were more characteristic of the fecal microbiota in adults with AD. Healthy individuals exhibited lower abundances of aerobic bacteria and higher abundances of short-chain fatty acid-producing species and polyamines. Clinical trials showed that the consumption of probiotics (Bifidobacterium and/or Lactobacillus), fecal microbiota transplants, and balneotherapy modified the fecal microbiota composition of participants and were associated with significant improvements in disease management. Conclusions: In anticipation of forthcoming clinical trials, it is essential to conduct meta-analyses that comprehensively evaluate the effectiveness and safety of interventions designed to modify intestinal flora in the context of AD. Preliminary evidence suggests that certain interventions may enhance adult AD management.
Collapse
Affiliation(s)
- Kevin Díez-Madueño
- Dermatology Department, Hospital Universitario Infanta Leonor, Complutense University of Madrid, 28040 Madrid, Spain;
- School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Pablo de la Cueva Dobao
- Dermatology Department, Hospital Universitario Infanta Leonor, Complutense University of Madrid, 28040 Madrid, Spain;
- School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Isabel Torres-Rojas
- Allergy Department, Hospital Universitario Infanta Sofía, 28702 Alcobendas, Spain;
| | | | | | - Pablo Coto-Segura
- Dermatology Department, Hospital Vital Álvarez Buylla, 33611 Mieres, Spain;
| |
Collapse
|
18
|
Liu F, Zhang H, Fan L, Yu Q, Wang S. Hotspots and development trends of gut microbiota in atopic dermatitis: A bibliometric analysis from 1988 to 2024. Medicine (Baltimore) 2024; 103:e40931. [PMID: 39686442 PMCID: PMC11651439 DOI: 10.1097/md.0000000000040931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a prevalent inflammatory skin condition that commonly occurs in children. More and more scientific evidence suggests that gut microbiota plays an important role in the pathogenesis of AD, whereas there is no article providing a comprehensive summary and analysis. We aimed to analyze documents on AD and gut microbiota and identify hotspots and development trends in this field. METHODS Articles and reviews in the field of AD and gut microbiota from January 1, 1988 to October 20, 2024 were obtained from the Web of Science Core Collection database. Biblioshiny was utilized for evaluating and visualizing the core authors, journals, countries, documents, trend topics, and hotspots in this field. RESULTS Among 1672 documents, it indicated that the number of annual publications generally increased. The United States had the highest production, impact, and international collaboration. Journal of Allergy and Clinical Immunology was the journal of the maximum publications. Based on keyword co-occurrence and clustering analysis, "stratum-corneum lipids," "probiotics," "prebiotics," "fecal microbiota transplantation," "phage therapy," "short chain fatty-acids," "biologic therapy," and "skin inflammation" represented current trend topics. The pathological and molecular mechanisms and associated therapeutic methods for AD and gut microbiota were the research hotspots. The incorporation of microbiota-based therapies alongside conventional treatments can contribute to better clinical outcomes. CONCLUSION We highlighted that gut microbiota may exacerbate symptoms of AD through various aspects, including immunity, metabolites, and neuroendocrine pathways. More efforts are required to investigate the safety and efficacy of gut microbial management methods for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Fang Liu
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Haipeng Zhang
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Lina Fan
- Hangzhou Linping District Maternal and Child Health Care Hospital, Hangzhou, China
| | - Qi Yu
- Chengdong College, Northeast Agricultural University, Harbin, China
| | - Siqiao Wang
- Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Wang XZ, Huang JL, Zhang J, Li QH, Zhang PP, Wu C, Jia YY, Su H, Sun X. Fecal microbiota transplantation as a new way for OVA-induced atopic dermatitis of juvenile mice. Int Immunopharmacol 2024; 142:113183. [PMID: 39298815 DOI: 10.1016/j.intimp.2024.113183] [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: 10/24/2023] [Revised: 07/31/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Children all over the world suffer from atopic dermatitis (AD), a prevalent condition that impairs their health. Corticosteroids, which have long-term negative effects, are frequently used to treat AD. There has been a growing body of research on the gut microbiota's function in AD. Nevertheless, the function and underlying mechanisms of fecal microbiota transplantation (FMT) in AD children remain to be established. Therefore, in order to assess the preventive effects of FMT treatment on AD and investigate the mechanisms, we constructed an ovalbumin (OVA)-induced juvenile mouse AD model in this investigation. This study explored the role and mechanism of FMT treatment in AD through 16S RNA sequencing, pathological histological staining, molecular biology, and Flow cytometry. Results demonstrated that the FMT treatment improved the gut microbiota's diversity and composition, bringing it back to a level similar to that of a close donor. Following FMT treatment, OVA-specific antibodies were inhibited, immunoglobulin (Ig) E production was decreased, the quantity of mast cells and eosinophils was decreased, and specific inflammatory markers in the skin and serum were decreased. Further mechanistic studies revealed that FMT treatment induced CD103+ DCs and programmed cell death ligand 1 (PD-L1)/programmed cell death 1 (PD-1) expression in skin-draining lymph nodes and promoted Treg production to induce immune tolerance and suppress skin inflammation. Meanwhile, changes in the gut microbiota were substantially correlated with Th2 cytokines, OVA-specific antibodies, and PD-L1/PD-1. In conclusion, FMT regulates the Th1/Th2 immunological balance and the gut microbiota. It may also inhibit AD-induced allergy responses through the PD-L1/PD-1 pathway, and providing a unique idea and possibly a fresh approach to the treatment of AD.
Collapse
Affiliation(s)
- Xing-Zhi Wang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Jin-Li Huang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Juan Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Qiu-Hong Li
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Pan-Pan Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Cheng Wu
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Yuan-Yuan Jia
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China.
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
20
|
Li X, Khan I, Han R, Huang G, Xia W, Yin L, Leong WK, Su L, Law BYK, Wong VKW, Wu Q, Guo X, Hsiao WLW. Gynostemma pentaphyllum saponins shield mice from peanut allergy by modulation of gut microbiota: A novel approach for peanut allergy management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156101. [PMID: 39522254 DOI: 10.1016/j.phymed.2024.156101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Food allergies, particularly peanut (PN) allergies, are a growing concern, with fatal anaphylaxis incidents often reported. While palforzia is the sole FDA-approved drug for managing PN allergies, it is not universally effective. PURPOSE This study aimed to investigate the potential of Gynostemma pentaphyllum saponins (GpS) as a novel therapeutic agent for PN allergy through modulation of gut microbiota, addressing the limitations of current treatments. METHODS To elucidate the role of GpS on peanut allergy, we first built a PN-sensitized C57BL/6J model mice. Through comprehensive sequencing analysis, we identified Parabacteroides distasonis as a key bacterium triggering PN sensitization. Employing the same mouse model, GpS was evaluated for its effects on anaphylactic symptoms, serum immunoglobulin levels, and allergy-related biomarkers. 16S rRNA sequencing and transcriptomic analysis were applied to investigate the impact of GpS on the host's gut epithelium and microbiome. RESULTS GpS treatment effectively reduced anaphylactic symptoms in PN-sensitized mice, as shown by decreased IgG1, total IgE, and PN-specific IgE levels. It also modulated the immune response by suppressing proinflammatory cytokines (IL-1β, IFN-γ, IL-21) and chemokines (CCL5, CCL12, CCL17, CCL22), while enhancing anti-inflammatory cytokines (IL-4, IL-10, IL-12, IL-13). Fecal microbial transplant from GpS-treated Model mice to PN-sensitized mice displayed anti-peanut allergy effects. Additionally, the administration of GpS-enhanced bacteria (Clostridium aldenese or Lactobacillus murinus), alleviated anaphylactic symptoms and reduced serum allergy markers in PN-sensitized mice. CONCLUSION To conclude, we revealed the intestinal environment, signaling molecules, mucosal cytokines, and commensal microbial profiles in the peanut-sensitized mouse model. We further presented evidence for the protective effect of GpS against PN allergen sensitization by downregulating a series of food-allergy-associated biomarkers and cytokines via the modulation of gut bacteria. More importantly, supported by both in vitro and in vivo experiments, we demonstrated that the protective effect of GpS against PN-allergy is through the enhancement of two commensal bacteria, Clostridium aldenese, and Lactobacillus murinus.
Collapse
Affiliation(s)
- Xiaoang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Ruixuan Han
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Guoxin Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Wenrui Xia
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lin Yin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Wai Kit Leong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lu Su
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Qiang Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xiaoling Guo
- Foshan Maternal and Child Health Research Institute, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, China
| | - W L Wendy Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Foshan Maternal and Child Health Research Institute, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, China.
| |
Collapse
|
21
|
Pahirah N, Narkwichean A, Taweechotipatr M, Wannaiampikul S, Duang-Udom C, Laosooksathit W. Comparison of Gut Microbiomes Between Neonates Born by Cesarean Section and Vaginal Delivery: Prospective Observational Study. BIOMED RESEARCH INTERNATIONAL 2024; 2024:8302361. [PMID: 39640900 PMCID: PMC11620805 DOI: 10.1155/bmri/8302361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Background: Balanced diversity and abundance of gut microbiome play important roles in human health, including neonatal health. Though not established, there is evidence that the delivery route could alter the diversity of neonatal gut microbiomes. Objective: The objective of the study was to investigate the differences in the gut microbiomes of neonates delivered via cesarean section compared to those born by vaginal delivery and to identify the predominant microbial taxa present in each group. Study Design: A prospective observational study of 281 healthy neonates born between February 2021 and April 2023 at Her Royal Highness Maha Chakri Sirindhorn Medical Center, Srinakharinwirot University, Thailand, was performed. The study population was divided into two groups: 139 neonates born via vaginal delivery and 141 neonates born via cesarean section. The microbiota composition of each neonate's fecal sample was identified by using 16S ribosomal ribonucleic acid metagenomic sequencing. Results: Neonates delivered vaginally exhibited a gut microbiome with higher abundance and diversity than those delivered by cesarean delivery. Bifidobacterium was the dominant genus in both groups. Bifidobacterium breve was the dominant species and was significantly higher in cesarean-delivered neonates compared to those delivered vaginally (24.0% and 9.2%, respectively) (p < 0.001). However, the taxonomy of only 89 (64.0%) and 44 (31.43%) fecal samples could be identified from the vaginal and cesarean delivery groups, respectively. Conclusion: Route of delivery is associated with neonatal gut microbiome abundance and diversity. Neonates delivered via vaginal delivery exhibited higher diversity but lower abundance of the dominant species in the gut microbiome. Trial Registration: Thai Clinical Trials Registry identifier: TCTR20221024003.
Collapse
Affiliation(s)
- Nichapat Pahirah
- Department of Obstetrics and Gynecology, Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | - Amarin Narkwichean
- Department of Obstetrics and Gynecology, Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | - Malai Taweechotipatr
- Department of Microbiology, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | - Sivaporn Wannaiampikul
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | | | - Wipada Laosooksathit
- Department of Obstetrics and Gynecology, Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| |
Collapse
|
22
|
Ljung A, Gio-Batta M, Hesselmar B, Imberg H, Rabe H, Nowrouzian FL, Johansen S, Törnhage CJ, Lindhagen G, Ceder M, Lundell AC, Rudin A, Wold AE, Adlerberth I. Gut microbiota markers in early childhood are linked to farm living, pets in household and allergy. PLoS One 2024; 19:e0313078. [PMID: 39602375 PMCID: PMC11602077 DOI: 10.1371/journal.pone.0313078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Children growing up on farms or with pets have a lower risk of developing allergy, which may be linked to their gut microbiota development during infancy. METHODS Children from the FARMFLORA birth cohort (N = 65), of whom 28 (43%) lived on a dairy farm and 40 (62%) had pets, provided fecal samples at intervals from 3 days to 18 months of age. Gut microbiota composition was characterized using quantitative microbial culture of various typical anaerobic and facultatively anaerobic bacteria, with colonization rate and population counts of bacterial groups determined at the genus or species level. Allergy was diagnosed at three and eight years of age by experienced pediatricians. Generalized estimating equations were used to identify associations between farm residence or pet ownership, gut microbiota development and allergy. Adjustments were made for important potential confounders. RESULTS Growing up on a farm was associated with a higher ratio of anaerobic to facultative bacteria in the first week, smaller Escherichia coli populations in colonized children in the first months of life and less frequent colonization by Clostridioides difficile at 12 months of age. Having pets in the household was associated with more frequent colonization by Bifidobacterium, Lactobacillus and Bacteroides in the first months. A higher ratio of anaerobic to facultative bacteria at one week of age, early colonization by Bifidobacterium, Lactobacillus and Bacteroides, and reduced carriage of C. difficile at 4-12 months of age all correlated negatively with subsequent allergy diagnosis. CONCLUSIONS Our findings indicate that lower rates of allergy in children growing up on farms or with pets may be related to early establishment of typical anaerobic commensals in their gut microbiota. However, further studies are needed to validate our observations in this small birth cohort study.
Collapse
Affiliation(s)
- Annika Ljung
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Monica Gio-Batta
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bill Hesselmar
- Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Imberg
- Statistiska Konsultgruppen, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hardis Rabe
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Forough L. Nowrouzian
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Carl-Johan Törnhage
- Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Pediatric Clinic, Skaraborg Hospital, Skövde, Sweden
| | | | | | - Anna-Carin Lundell
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Rudin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Agnes E. Wold
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingegerd Adlerberth
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
23
|
Soto Ocaña J, Friedman ES, Keenan O, Bayard NU, Ford E, Tanes C, Munneke MJ, Beavers WN, Skaar EP, Bittinger K, Zemel BS, Wu GD, Zackular JP. Metal availability shapes early life microbial ecology and community succession. mBio 2024; 15:e0153424. [PMID: 39440978 PMCID: PMC11558993 DOI: 10.1128/mbio.01534-24] [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: 05/18/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
The gut microbiota plays a critical role in human health and disease. Microbial community assembly and succession early in life are influenced by numerous factors. In turn, assembly of this microbial community is known to influence the host, including immune system development, and has been linked to outcomes later in life. To date, the role of host-mediated nutritional immunity and metal availability in shaping microbial community assembly and succession early in life has not been explored in depth. Using a human infant cohort, we show that the metal-chelating protein calprotectin is highly abundant in infants. Taxa previously shown to be successful early colonizers of the infant gut, such as Enterococcus, Enterobacteriaceae, and Bacteroides, are highly resistant to experimental metal starvation in culture. Lactobacillus, meanwhile, is highly susceptible to metal restriction, pointing to a possible mechanism by which host-mediated metal limitation shapes the fitness of early colonizing taxa in the infant gut. We further demonstrate that formula-fed infants harbor markedly higher levels of metals in their gastrointestinal tract compared to breastfed infants. Formula-fed infants with high levels of metals harbor distinct microbial communities compared to breastfed infants, with higher levels of Enterococcus, Enterobacter, and Klebsiella, taxa which show increased resistance to the toxic effects of high metal concentrations. These data highlight a new paradigm in microbial community assembly and suggest an unappreciated role for nutritional immunity and dietary metals in shaping the earliest colonization events of the microbiota.IMPORTANCEEarly life represents a critical window for microbial colonization of the human gastrointestinal tract. Surprisingly, we still know little about the rules that govern the successful colonization of infants and the factors that shape the success of early life microbial colonizers. In this study, we report that metal availability is an important factor in the assembly and succession of the early life microbiota. We show that the host-derived metal-chelating protein, calprotectin, is highly abundant in infants and successful early life colonizers can overcome metal restriction. We further demonstrate that feeding modality (breastmilk vs formula) markedly impacts metal levels in the gut, potentially influencing microbial community succession. Our work suggests that metals, a previously unexplored aspect of early life ecology, may play a critical role in shaping the early events of microbiota assembly in infants.
Collapse
Affiliation(s)
- Joshua Soto Ocaña
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elliot S. Friedman
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Orlaith Keenan
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nile U. Bayard
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eileen Ford
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Matthew J. Munneke
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - William N. Beavers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- The Center for Microbial Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Babette S. Zemel
- Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Gary D. Wu
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph P. Zackular
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Center for Microbial Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Luo X, Wang H, Liu H, Chen Y, Tian L, Ji Q, Xie D. Effects of probiotics on the prevention and treatment of children with allergic rhinitis: a meta-analysis of randomized controlled trials. Front Pediatr 2024; 12:1352879. [PMID: 39421038 PMCID: PMC11484092 DOI: 10.3389/fped.2024.1352879] [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: 12/14/2023] [Accepted: 07/30/2024] [Indexed: 10/19/2024] Open
Abstract
Background and aim Recent studies have demonstrated the anti-allergic effects of probiotics in humans. However, their role in preventing and treating pediatric allergic rhinitis has not been thoroughly investigated. This study aimed to systematically review the efficacy and preventive effects of probiotics on pediatric allergic rhinitis. Methods We systematically searched PubMed, Embase, the Cochrane Central Register of Controlled Trials, and Web of Science databases for all relevant studies on probiotics and pediatric allergic rhinitis. Studies meeting the inclusion criteria were included, data were extracted, and meta-analyses were performed. Results A total of 28 studies with 4,765 participants were included in this study. The pooled results showed that the use of probiotics was associated with a significant improvement in total nose symptom scores (SMD, -2.27; 95% CI, -3.26 to -1.29; P < 0.00001), itchy nose scores (SMD, -0.44; 95% CI, -0.80 to -0.07; P = 0.02), sneezing scores (SMD, -0.47; 95% CI, -0.84 to -0.10; P = 0.01), eye symptoms (SMD, -3.77; 95% CI, -5.47 to -2.07; P < 0.00001), and Pediatric Rhinoconjunctivitis Quality of Life Questionnaire (SMD, -2.52; 95% CI, -4.12 to -0.92; P < 00001). However, the use of probiotics was not associated with the incidence of allergic rhinitis (RR, 0.9; 95% CI, 0.74-1.08; P = 0.26). Conclusions The present study demonstrated that probiotics were effective and safe for improving pediatric allergic rhinitis symptoms and quality of life. However, probiotics could not prevent pediatric allergic rhinitis.
Collapse
Affiliation(s)
- Xinyi Luo
- The First Affiliated Hospital of Chengdu Medical College Clinical Medical College, Chengdu, Sichuan, China
| | - Huan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Huixia Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yue Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Li Tian
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qing Ji
- Department of Otolaryngology, Chengdu First People’s Hospital, Chengdu, Sichuan, China
| | - Dengpiao Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Pantazi AC, Nori W, Kassim MAK, Balasa AL, Mihai CM, Chisnoiu T, Mihai L, Ungureanu A, Frecus CE, Chirila SI, Cambrea SC. Gut microbiota profile and atopic dermatitis in the first year of life. J Med Life 2024; 17:948-952. [PMID: 39720170 PMCID: PMC11665751 DOI: 10.25122/jml-2024-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/27/2024] [Indexed: 12/26/2024] Open
Abstract
The connection between the immune response and the composition of gut microbiota has been associated with an increased prevalence of atopic dermatitis in the first year of life. The study aimed to investigate gut microbiota characteristics in infants with atopic dermatitis compared to healthy infants to better understand the link between early-life microbiota composition and the development of atopic dermatitis. The study analyzed the intestinal microbiota of 121 infants with clinical signs of atopic dermatitis, divided into Group I (infants with atopic dermatitis) and Group II (healthy controls). The study showed that infants with atopic dermatitis presented increased values of proteolytic bacteria mainly represented by Enterobacter species (P = 0.041), Klebsiella species (P = 0.038), and Escherichia coli (P = 0.013), with significantly decreased levels of acidifying bacteria represented by Enterococcus species, Lactobacillus and Bifidobacterium (P < 0.05) and normal levels of Clostridium species, Candida albicans, Mould fungi and Geotrichum species. This study highlights distinct differences in the gut microbiota of infants with atopic dermatitis, providing insights into the dynamic intestinal ecosystem during early life for future personalized therapeutic strategies.
Collapse
Affiliation(s)
- Alexandru Cosmin Pantazi
- Faculty of Medicine, Ovidius University, Constanta, Romania
- Department of Pediatrics, Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Wassan Nori
- College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | | | - Adriana Luminita Balasa
- Faculty of Medicine, Ovidius University, Constanta, Romania
- Department of Pediatrics, Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Cristina Maria Mihai
- Faculty of Medicine, Ovidius University, Constanta, Romania
- Department of Pediatrics, Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Tatiana Chisnoiu
- Faculty of Medicine, Ovidius University, Constanta, Romania
- Department of Pediatrics, Clinical Emergency Hospital of Constanta, Constanta, Romania
| | - Larisia Mihai
- Faculty of Medicine, Ovidius University, Constanta, Romania
- Department of Pediatrics, Clinical Emergency Hospital of Constanta, Constanta, Romania
| | | | - Corina Elena Frecus
- Faculty of Medicine, Ovidius University, Constanta, Romania
- Department of Pediatrics, Clinical Emergency Hospital of Constanta, Constanta, Romania
| | | | | |
Collapse
|
26
|
Sansonetti PJ, Doré J. [The human microbiome proofed by the Anthropocene: from correlation to causality and intervention]. Med Sci (Paris) 2024; 40:757-765. [PMID: 39450961 DOI: 10.1051/medsci/2024121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Abstract
The deleterious effects of human activities on biodiversity in the vegetal and animal world, and on climate changes are now well-established facts. However, little is yet known on the impact of human activities on microbial diversity on the planet and more specifically on the human microbiota Large implementation of metagenomics allows exaustive microbial cataloguing with broad spatio-temporal resolution of human microbiota. A reduction in bacterial richness and diversity in the human microbiota, particularly in the intestinal tract, is now established and particularly obvious in the most industrialized regions of the planet. Massive, uncontrolled use of antibiotics, drastic changes in traditional food habits and some elements of the "global exposome" that remain to identify are usually considered as stressors accounting for this situation of "missing microbes". As a consequence, a dysbiotic situation develops, a "dysbiosis" being characterized by the erosion of the central core of shared bacterial species across individuals and the development of opportunistic "pathobionts" in response to a weaker barrier capacity of these impoverished microbiota. The current challenge is to establish a causality link between the extension of these dysbiotic situations and the steady emergence of epidemic, non-communicable diseases such as asthma, allergy, obesity, diabetes, autoimmune diseases and some cancers. Experimental animal models combined with controlled, prospective clinical interventions are in demand to consolidate causality links, with the understanding that in the deciphering of the mechanisms of alteration of the human-microbiome symbiosis resides a novel exciting chapter of medicine: "microbial medicine".
Collapse
Affiliation(s)
| | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS Jouy-en-Josas France
| |
Collapse
|
27
|
Özdemіr E, Öksüz L. Effect of Staphylococcus aureus colonization and immune defects on the pathogenesis of atopic dermatitis. Arch Microbiol 2024; 206:410. [PMID: 39302484 DOI: 10.1007/s00203-024-04134-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/26/2024] [Accepted: 09/07/2024] [Indexed: 09/22/2024]
Abstract
Atopic dermatitis (AD) is a common and recurrent skin disease characterized by skin barrier dysfunction, inflammation and chronic pruritus, with wide heterogeneity in terms of age of onset, clinical course and persistence over the lifespan. Although the pathogenesis of the disease are unclear, epidermal barrier dysfunction, immune and microbial dysregulation, and environmental factors are known to be critical etiologies in AD pathology. The skin microbiota represents an ecosystem consisting of numerous microbial species that interact with each other as well as host epithelial cells and immune cells. Although the skin microbiota benefits the host by supporting the basic functions of the skin and preventing the colonization of pathogens, disruption of the microbial balance (dysbiosis) can cause skin diseases such as AD. Although AD is a dermatological disease, recent evidence has shown that changes in microbiota composition in the skin and intestine contribute to the pathogenesis of AD. Environmental factors that contribute to skin barrier dysfunction and microbial dysbiosis in AD include allergens, diet, irritants, air pollution, epigenetics and microbial exposure. Knowing the microbial combination of intestin, as well as the genetic and epigenetic determinants associated with the development of autoantibodies, may help elucidate the pathophysiology of the disease. The skin of patients with AD is characterized by microbial dysbiosis as a result of reduced microbial diversity and overgrowth of the pathogens such as Staphylococcus aureus. Recent studies have revealed the importance of building a strong immune response against microorganisms during childhood and new mechanisms of microbial community dynamics in modulating the skin microbiome. Numerous microorganisms are reported to modulate host response through communication with keratinocytes, specific immune cells and adipocytes to improve skin health and barrier function. This growing insight into bioactive substances in the skin microbiota has led to novel biotherapeutic approaches targeting the skin surface for the treatment of AD. This review will provide an updated overview of the skin microbiota in AD and its complex interaction with immune response mechanisms, as well as explore possible underlying mechanisms in the pathogenesis of AD and provide insights into new therapeutic developments for the treatment of AD. It also focuses on restoring skin microbial homeostasis, aiming to reduce inflammation by repairing the skin barrier.
Collapse
Affiliation(s)
- Evrim Özdemіr
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
- Institute of Graduate Studies in Health Sciences, Istanbul University, Istanbul, Turkey.
| | - Lütfiye Öksüz
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
28
|
Wang Y, Wang B, Sun S, Wang Z. Mapping the relationship between atopic dermatitis and gut microbiota: a bibliometric analysis, 2014-2023. Front Microbiol 2024; 15:1400657. [PMID: 39296293 PMCID: PMC11408322 DOI: 10.3389/fmicb.2024.1400657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Background Atopic dermatitis (AD) is a chronic inflammatory skin condition affecting a significant portion of the population, with prevalence rates of 25% in children and 7-10% in adults. AD not only poses physical challenges but also profoundly impacts patients' mental well-being and quality of life. The stability of gut microbiota is crucial for overall health and can influence AD progression by modulating immune function, skin barrier integrity, and neuroendocrine signaling, which may be an effective target for the prevention and treatment of AD. Thus, exploring the interactions between AD and gut microbiota, particularly in infants, can provide insights into potential preventive and therapeutic strategies. This study aimed to explore the correlation between AD and gut microbiota while providing an overview of current research trends and emerging areas of interest in this field. Methods A comprehensive search was conducted on the Web of Science Core Collection (WOSCC) for relevant publications from January 1, 2014, to December 31, 2023. English-language articles and reviews were included. Two investigators independently screened the publications, and visual analysis was performed using CiteSpace, VOSviewer, Scimago Graphica, and Microsoft Excel software. Results A total of 804 articles were included, showing a significant increase in publications over the past decade. The United States, Wageningen University, and University Ulsan (represented by Hong SJ) had the highest number of published papers. Nutrients was the journal with the most publications, while the Journal of Allergy and Clinical Immunology had the highest number of citations and centrality among co-cited journals. Keyword visualization analysis identified "atopic dermatitis" and "gut microbiota" as central themes. Notably, there has been a notable shift in research focus over the years, with early studies concentrating on "Fecal microbiota," "caesarean section," and "first 6 months," while recent studies have highlighted the roles of "cells," "dysbiosis," and "prebiotics." This shift indicates growing interest in the underlying mechanisms and potential therapeutic interventions related to the intestinal microecology in AD treatment. Conclusion The field of AD and gut microbiota research has evolved significantly, with an increasing focus on understanding the intricate interactions between gut microbiota and AD pathogenesis. Recent years have witnessed increased interest in understanding the relationship between AD and gut microbiota, with researchers conducting extensive studies exploring various aspects of this connection. This review analyzes research trends over the past decade, highlighting trends and hotspots in the study of AD, particularly in infants, and the role of microbiota. This review serves as a valuable reference for future investigations, aiming to provide deeper insights into this burgeoning field and suggests directions for future research.
Collapse
Affiliation(s)
- Yilin Wang
- Department of Dermatology, The 83rd Group Army Hospital of the PLA, Xinxiang, China
| | - Bingkun Wang
- Department of Dermatology, The 83rd Group Army Hospital of the PLA, Xinxiang, China
| | - Shiyou Sun
- Department of Dermatology, The 83rd Group Army Hospital of the PLA, Xinxiang, China
| | - Zhongzhi Wang
- Department of Dermatology, Shanghai Fourth People 's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Chang R, Chen HY, Hung YM, Huang JY, Wei JCC. Time-dependent risk of atopic dermatitis following nontyphoidal Salmonella infection. Postgrad Med J 2024; 100:649-656. [PMID: 38565127 DOI: 10.1093/postmj/qgae041] [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: 01/17/2024] [Revised: 02/18/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND The pathogenesis of atopic dermatitis (AD) remains unclear. Nontyphoidal Salmonella (NTS) infection might trigger immune-mediated reactions. We aimed to examine NTS and the risk of subsequent AD. METHODS From 2002 to 2015, eligible patients (aged 0-100 years) with NTS were identified. NTS and non-NTS groups were matched at a 1:10 ratio on age and sex. We utilized conditional multivariable Cox proportional hazard models to estimate the adjusted hazard ratio (aHR) and 95% confidence interval (CI) for AD development. Subgroup analyses were conducted based on age, sex, and severity of NTS infection. We utilized landmark analysis to explore the time-dependent hazard of AD following NTS. RESULTS In the NTS group (N = 6624), 403 developed AD. After full adjustment of demographics and comorbidities, the NTS group had a higher risk of AD than the reference group (aHR = 1.217, 95% CI = 1.096-1.352). Age-stratified analysis revealed that NTS group exhibited an elevated risk compared to the reference group, particularly among those aged 13-30 years (aHR = 1.25, 95% CI = 1.017-1.559), individuals aged 31-50 years (aHR = 1.388, 95% CI = 1.112-1.733), those aged 51-70 years (aHR = 1.301, 95% CI = 1.008-1.679), and individuals aged 71 years and over (aHR = 1.791, 95% CI = 1.260-2.545). Severe NTS was associated with a higher risk of AD than the reference group (aHR = 2.411, 95% CI = 1.577-3.685). Landmark analysis showed generally consistent findings. CONCLUSIONS Minimizing exposure to NTS infection may represent a prospective strategy for averting the onset and progression of atopic dermatitis.
Collapse
Affiliation(s)
- Renin Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 95050, Taiwan
| | - Hui-Yuan Chen
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Yao-Min Hung
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital Taitung Branch, 40201, Taiwan
| | - Jing-Yang Huang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Division of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung 40432, Taiwan
- Department of Nursing, Chung Shan Medical University, Taichung 40721, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
30
|
Javed U, Podury S, Kwon S, Liu M, Kim DH, Fallahzadeh A, Li Y, Khan AR, Francois F, Schwartz T, Zeig-Owens R, Grunig G, Veerappan A, Zhou J, Crowley G, Prezant DJ, Nolan A. Biomarkers of Airway Disease, Barrett's and Underdiagnosed Reflux Noninvasively (BAD-BURN) in World Trade Center exposed firefighters: a case-control observational study protocol. BMC Gastroenterol 2024; 24:255. [PMID: 39123126 PMCID: PMC11312152 DOI: 10.1186/s12876-024-03294-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/12/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Particulate matter exposure (PM) is a cause of aerodigestive disease globally. The destruction of the World Trade Center (WTC) exposed first responders and inhabitants of New York City to WTC-PM and caused obstructive airways disease (OAD), gastroesophageal reflux disease (GERD) and Barrett's Esophagus (BE). GERD not only diminishes health-related quality of life but also gives rise to complications that extend beyond the scope of BE. GERD can incite or exacerbate allergies, sinusitis, bronchitis, and asthma. Disease features of the aerodigestive axis can overlap, often necessitating more invasive diagnostic testing and treatment modalities. This presents a need to develop novel non-invasive biomarkers of GERD, BE, airway hyperreactivity (AHR), treatment efficacy, and severity of symptoms. METHODS Our observational case-cohort study will leverage the longitudinally phenotyped Fire Department of New York (FDNY)-WTC exposed cohort to identify Biomarkers of Airway Disease, Barrett's and Underdiagnosed Reflux Noninvasively (BAD-BURN). Our study population consists of n = 4,192 individuals from which we have randomly selected a sub-cohort control group (n = 837). We will then recruit subgroups of i. AHR only ii. GERD only iii. BE iv. GERD/BE and AHR overlap or v. No GERD or AHR, from the sub-cohort control group. We will then phenotype and examine non-invasive biomarkers of these subgroups to identify under-diagnosis and/or treatment efficacy. The findings may further contribute to the development of future biologically plausible therapies, ultimately enhance patient care and quality of life. DISCUSSION Although many studies have suggested interdependence between airway and digestive diseases, the causative factors and specific mechanisms remain unclear. The detection of the disease is further complicated by the invasiveness of conventional GERD diagnosis procedures and the limited availability of disease-specific biomarkers. The management of reflux is important, as it directly increases risk of cancer and negatively impacts quality of life. Therefore, it is vital to develop novel noninvasive disease markers that can effectively phenotype, facilitate early diagnosis of premalignant disease and identify potential therapeutic targets to improve patient care. TRIAL REGISTRATION Name of Primary Registry: "Biomarkers of Airway Disease, Barrett's and Underdiagnosed Reflux Noninvasively (BADBURN)". Trial Identifying Number: NCT05216133 . Date of Registration: January 31, 2022.
Collapse
Affiliation(s)
- Urooj Javed
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University Grossman School of Medicine (NYUGSoM), New Bellevue, 16 North Room 20 (Lab), 462 1st Avenue, New York, NY, 10016, USA
| | - Sanjiti Podury
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University Grossman School of Medicine (NYUGSoM), New Bellevue, 16 North Room 20 (Lab), 462 1st Avenue, New York, NY, 10016, USA
| | - Sophia Kwon
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University Grossman School of Medicine (NYUGSoM), New Bellevue, 16 North Room 20 (Lab), 462 1st Avenue, New York, NY, 10016, USA
| | - Mengling Liu
- Department of Population Health, Division of Biostatistics, NYUGSoM, New York, NY, USA
| | - Daniel H Kim
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University Grossman School of Medicine (NYUGSoM), New Bellevue, 16 North Room 20 (Lab), 462 1st Avenue, New York, NY, 10016, USA
| | - Aida Fallahzadeh
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University Grossman School of Medicine (NYUGSoM), New Bellevue, 16 North Room 20 (Lab), 462 1st Avenue, New York, NY, 10016, USA
| | - Yiwei Li
- Department of Population Health, Division of Biostatistics, NYUGSoM, New York, NY, USA
| | - Abraham R Khan
- Center for Esophageal Health, NYUGSoM, New York, NY, 10016, USA
- Department of Medicine, Division of Gastroenterology, NYUGSoM, New York, NY, 10016, USA
| | - Fritz Francois
- Department of Medicine, Division of Gastroenterology, NYUGSoM, New York, NY, 10016, USA
| | - Theresa Schwartz
- Fire Department of New York, Bureau of Health Services, Brooklyn, NY, 1120, USA
| | - Rachel Zeig-Owens
- Fire Department of New York, Bureau of Health Services, Brooklyn, NY, 1120, USA
| | - Gabriele Grunig
- Department of Medicine, Division of Environmental Medicine, NYUGSoM, New York, NY, 10010, USA
| | - Arul Veerappan
- Department of Medicine, Division of Environmental Medicine, NYUGSoM, New York, NY, 10010, USA
| | - Joanna Zhou
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University Grossman School of Medicine (NYUGSoM), New Bellevue, 16 North Room 20 (Lab), 462 1st Avenue, New York, NY, 10016, USA
| | - George Crowley
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University Grossman School of Medicine (NYUGSoM), New Bellevue, 16 North Room 20 (Lab), 462 1st Avenue, New York, NY, 10016, USA
| | - David J Prezant
- Fire Department of New York, Bureau of Health Services, Brooklyn, NY, 1120, USA
| | - Anna Nolan
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, New York University Grossman School of Medicine (NYUGSoM), New Bellevue, 16 North Room 20 (Lab), 462 1st Avenue, New York, NY, 10016, USA.
- Fire Department of New York, Bureau of Health Services, Brooklyn, NY, 1120, USA.
- Department of Medicine, Division of Environmental Medicine, NYUGSoM, New York, NY, 10010, USA.
| |
Collapse
|
31
|
Zou X, Zou X, Gao L, Zhao H. Gut microbiota and psoriasis: pathogenesis, targeted therapy, and future directions. Front Cell Infect Microbiol 2024; 14:1430586. [PMID: 39170985 PMCID: PMC11335719 DOI: 10.3389/fcimb.2024.1430586] [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: 05/13/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024] Open
Abstract
Background Psoriasis is one of the most common autoimmune skin diseases. Increasing evidence shows that alterations in the diversity and function of microbiota can participate in the pathogenesis of psoriasis through various pathways and mechanisms. Objective To review the connection between microbial changes and psoriasis, how microbial-targeted therapy can be used to treat psoriasis, as well as the potential of prebiotics, probiotics, synbiotics, fecal microbiota transplantation, diet, and Traditional Chinese Medicine as supplementary and adjunctive therapies. Methods Literature related to the relationship between psoriasis and gut microbiota was searched in PubMed and CNKI. Results Adjunct therapies such as dietary interventions, traditional Chinese medicine, and probiotics can enhance gut microbiota abundance and diversity in patients with psoriasis. These therapies stimulate immune mediators including IL-23, IL-17, IL-22, and modulate gamma interferon (IFN-γ) along with the NF-kB pathway, thereby suppressing the release of pro-inflammatory cytokines and ameliorating systemic inflammatory conditions. Conclusion This article discusses the direction of future research and clinical treatment of psoriasis from the perspective of intestinal microbiota and the mechanism of traditional Chinese medicine, so as to provide clinicians with more comprehensive diagnosis and treatment options and bring greater hope to patients with psoriasis.
Collapse
Affiliation(s)
- Xinyan Zou
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| | - Xinfu Zou
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Longxia Gao
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| | - Hanqing Zhao
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, China
| |
Collapse
|
32
|
Changhai L, Zaichun W, Bo Y, Dan L, Shaohua W. Micronutrients and Allergic Diseases: A Mendelian Randomization Study. Int Arch Allergy Immunol 2024; 186:41-51. [PMID: 39106841 DOI: 10.1159/000540376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/09/2024] [Indexed: 08/09/2024] Open
Abstract
INTRODUCTION Previous studies have indicated a controversy regarding the association between dietary micronutrient concentrations and the risk of allergic diseases. In this study, we employed Mendelian randomization (MR) analysis using data from two samples to investigate the causal relationship between circulating micronutrient concentrations and three allergic diseases. METHODS In this study, we considered 16 circulating micronutrients as exposure variables (beta carotene, calcium, copper, folate, iron, lycopene, magnesium, phosphorus, selenium, vitamin A1, vitamin B6, vitamin B12, vitamin C, vitamin D, vitamin E, and zinc); and three common allergic diseases (allergic asthma [AA], atopic dermatitis [AD], and allergic rhinitis [AR]) as outcomes. The inverse variance weighted (IVW) method was primarily applied for MR analysis, supplemented by MR-Egger and weighted-median methods to corroborate the IVW results; and sensitivity analysis was conducted to ensure the robustness of the MR assumptions. RESULTS Our results revealed that an increase in serum phosphorus and zinc concentrations may diminish the risk of AA, while for AD an increase in serum zinc concentration may reduce the risk, but an increase in serum vitamin C concentration may elevate the risk. As for AR, an increase in serum phosphorus and selenium concentrations appeared to be associated with a reduced risk. We did not find evidence for an association between other micronutrients and the risk of allergic diseases. CONCLUSION Our study indicates that an increase in serum phosphorus and zinc concentrations may reduce the risk of AA, while an increase in serum zinc concentration may reduce the risk of AD, but an increase in serum vitamin C concentration may elevate the risk of AD. An increase in serum phosphorus and selenium concentrations is associated with a reduced risk of AR. This provides additional support for research on the effects of micronutrients on allergic diseases.
Collapse
Affiliation(s)
- Long Changhai
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China,
| | - Wu Zaichun
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Bo
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Li Dan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wang Shaohua
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
33
|
Xiang ST, Zhou C, Zhao K, Ma Y, Huang R, Peng Y, Tang Y, Yang F, Qiu J. Association of metals with early postnatal gut microbiota among infants admitted to the neonatal intensive care unit. Int J Hyg Environ Health 2024; 261:114410. [PMID: 38925082 DOI: 10.1016/j.ijheh.2024.114410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/02/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
The gut microbiota is closely related to infant health. However, the impact of environmental factors on the gut microbiota has not been widely investigated, particularly in vulnerable populations such as infants admitted to the neonatal intensive care unit (NICU). This study investigated the association between exposure to 12 metals and the composition of the gut microbiota in infants admitted to the NICU. Metal concentrations were determined in serum samples obtained from 107 infants admitted to the NICU at Hunan Children's hospital, China. Gut microbiota data were derived from 16S rRNA sequencing using stool samples. Generalized linear regression (GLR) models and Bayesian kernel machine regression (BKMR) analyses were used to estimate the associations between metals and both alpha-diversity indices and bacterial taxa. The GLR models showed that tin correlated negatively with the Shannon index (β = -0.55, 95% conficence interval [CI]: -0.79, -0.30, PFDR< 0.001) and positively with the Simpson index (β = 0.26, 95% CI: 0.13, 0.39, PFDR< 0.001). The BKMR analysis yielded similar results, showing that tin had the largest posterior inclusion probability for both the Shannon (0.986) and the Simpson (0.796) indices. Tin, cadmium, mercury, lead, and thallium were associated with changes in one or more taxa at the genus level. The BKMR analysis also revealed a negative correlation between metal mixtures and Clostridium_sensu_stricto, and tin contibuted mostly to the negative correlation. Early postnatal exposure to metals were associated with differences in the microbiome among infants admitted to the NICU. However, as the study was cross-sectional, these relationships must be confirmed in further studies.
Collapse
Affiliation(s)
- Shi-Ting Xiang
- Pediatrics Research Institute of Hunan Province, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's hospital), Changsha, 410007, China
| | - Changci Zhou
- Academy of Pediatrics, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Kunyan Zhao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ye Ma
- Department of Neonatology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's hospital), Changsha, China
| | - Ruiwen Huang
- Department of Neonatology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's hospital), Changsha, China
| | - Yunlong Peng
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, 215123, China
| | - Yan Tang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Jun Qiu
- Pediatrics Research Institute of Hunan Province, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's hospital), Changsha, 410007, China.
| |
Collapse
|
34
|
Marques-Mejias A, Bartha I, Ciaccio CE, Chinthrajah RS, Chan S, Hershey GKK, Hui-Beckman JW, Kost L, Lack G, Layhadi JA, Leung DYM, Marshall HF, Nadeau KC, Radulovic S, Rajcoomar R, Shamji MH, Sindher S, Brough HA. Skin as the target for allergy prevention and treatment. Ann Allergy Asthma Immunol 2024; 133:133-143. [PMID: 38253125 DOI: 10.1016/j.anai.2023.12.030] [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: 08/21/2023] [Revised: 11/27/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024]
Abstract
The fact that genetic and environmental factors could trigger disruption of the epithelial barrier and subsequently initiate a TH2 inflammatory cascade conversely proposes that protecting the same barrier and promoting adequate interactions with other organs, such as the gut, may be crucial for lowering the risk and preventing atopic diseases, particularly, food allergies. In this review, we provide an overview of structural characteristics that support the epithelial barrier hypothesis in patients with atopic dermatitis, including the most relevant filaggrin gene mutations, the recent discovery of the role of the transient receptor potential vanilloid 1, and the role involvement of the microbiome in healthy and damaged skin. We present experimental and human studies that support the mechanisms of allergen penetration, particularly the dual allergen exposure and the outside-in, inside-out, and outside-inside-outside hypotheses. We discuss classic skin-targeted therapies for food allergy prevention, including moisturizers, steroids, and topical calcineurin inhibitors, along with pioneering trials proposed to change their current use (Prevention of Allergy via Cutaneous Intervention and Stopping Eczema and ALlergy). We provide an overview of the novel therapies that enhance the skin barrier, such as probiotics and prebiotics topical application, read-through drugs, direct and indirect FLG replacement, and interleukin and janus kinases inhibitors. Last, we discuss the newer strategies for preventing and treating food allergies in the form of epicutaneous immunotherapy and the experimental use of single-dose of adeno-associated virus vector gene immunotherapy.
Collapse
Affiliation(s)
- Andreina Marques-Mejias
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom
| | - Irene Bartha
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom
| | - Christina E Ciaccio
- Department of Pediatrics, The University of Chicago, Chicago, Illinois; Department of Medicine, The University of Chicago, Chicago, Illinois
| | - R Sharon Chinthrajah
- Department of Medicine, and Sean N Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California
| | - Susan Chan
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Laurie Kost
- Department of Medicine, and Sean N Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California
| | - Gideon Lack
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Janice A Layhadi
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - Hannah F Marshall
- Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom
| | - Kari C Nadeau
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Suzana Radulovic
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Reena Rajcoomar
- Department of Medicine, and Sean N Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California
| | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sayantani Sindher
- Department of Medicine, and Sean N Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California
| | - Helen A Brough
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
35
|
Masi AC, Stewart CJ. Role of breastfeeding in disease prevention. Microb Biotechnol 2024; 17:e14520. [PMID: 38946112 PMCID: PMC11214977 DOI: 10.1111/1751-7915.14520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024] Open
Abstract
Human milk provides the infant with many bioactive factors, including immunomodulating components, antimicrobials and prebiotics, which modulate the infant microbiome and immune system maturation. As a result, breastfeeding can impact infant health from infancy, through adolescence, and into adulthood. From protecting the infant from infections, to reducing the risk of obesity, type 1 diabetes and childhood leukaemia, many positive health outcomes are observed in infants receiving breastmilk. For the mother, breastfeeding protects against postpartum bleeding and depression, increases weight loss, and long-term lowers the risk of type 2 diabetes, breast and ovarian cancer, and cardiovascular diseases. Beyond infants and mothers, the wider society is also impacted because of avoidable costs relating to morbidity and mortality derived from a lack of human milk exposure. In this review, Medline was used to search for relevant articles to discuss the health benefits of breastfeeding and its societal impact before exploring future recommendations to enhance our understanding of the mechanisms behind breastfeeding's positive effects and promote breastfeeding on a global scale.
Collapse
Affiliation(s)
- Andrea C. Masi
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | | |
Collapse
|
36
|
Schoos AMM. Atopic diseases-Diagnostics, mechanisms, and exposures. Pediatr Allergy Immunol 2024; 35:e14198. [PMID: 39016386 DOI: 10.1111/pai.14198] [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: 10/12/2023] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024]
Abstract
Epidemiological data suggest that atopic diseases begin in early life and that most cases present clinically during early childhood. The diseases are highly prevalent and increase as communities adopt western lifestyles. Disentangling the pathophysiological mechanisms leading to disease debut is necessary to identify beneficial/harmful exposures so that successful prevention and treatment can be generated. The objective of this review is to explore the definition of atopy and mechanisms of atopic diseases, and to investigate the importance of environmental factors in early life, prior to disease development. First, the distribution of sIgE levels in children is investigated, as this is one of the main criteria for the definition of atopy. Thereafter, it is explored how studies of parental atopic status, sensitization patterns, and early debut and severity of atopic dermatitis have substantiated the theory of an early-life window of opportunity for intervention that precedes the development of atopic diseases in childhood. Then, it is examined whether early-life exposures such as breastfeeding, dogs, cats, and house dust mites in the home perinatally constitute important influencers in this crucial time of life. Finally, it is discussed how these findings could be validated in randomized controlled trials, which might prepare the ground for improved diagnostics and prevention strategies to mitigate the current atopic pandemic.
Collapse
Affiliation(s)
- Ann-Marie Malby Schoos
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Kim TH, Shin JS, Kim SY, Kim J. Association of Previous Antibiotics Use and Kawasaki Disease: A Cohort Study of 106,908 Patients. Pediatr Infect Dis J 2024; 43:643-650. [PMID: 38534913 DOI: 10.1097/inf.0000000000004335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
BACKGROUND Microbial imbalance in the gut from antibiotic use may be an etiologic factor of Kawasaki disease (KD). We aimed to identify the association between the use of antibiotics and the development of KD, considering various antibiotic profiles. METHODS A population-based, case-control study was performed using data from the Health Insurance Review and Assessment Service database. Children <5 years of age, who were diagnosed with KD between 2016 and 2019, were identified. Propensity score-matched controls were selected from the general population in a 1:5 ratio. Four separate study cohorts were created according to different periods of antibiotic use: (1) within 28 days and (2) 12 months after birth and (3) within 6 months and (4) 12 months from the index date. Profiles regarding antibiotic use were compared between patients with KD and matched controls. RESULTS We included 17,818 patients with KD and 89,090 matched controls. Use of antibiotics within 6 months [odds ratio (OR): 1.18; 95% confidence interval (CI): 1.12-1.26] and 12 months (OR: 1.23; 95% CI: 1.14-1.32) from the index date were associated with the development of KD. The association between antibiotic use and KD was most prominent in patients who had received 3 or more types of antibiotics within 12 months from the index date (OR: 1.26; 95% CI: 1.17-1.37). CONCLUSIONS Antibiotic use within the preceding 6 or 12 months was associated with KD. Alteration in gut microbiota due to antibiotic usage might play a role in the development of KD.
Collapse
Affiliation(s)
- Tae-Hwan Kim
- From the Spine Center, Department of Orthopedics, Hallym University Sacred Heart Hospital
| | - Ji Seong Shin
- Division of Infection, Department of Pediatrics, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Sin Young Kim
- Division of Infection, Department of Pediatrics, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jihye Kim
- Division of Infection, Department of Pediatrics, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
38
|
Hick E, Suárez M, Rey A, Mantecón L, Fernández N, Solís G, Gueimonde M, Arboleya S. Personalized Nutrition with Banked Human Milk for Early Gut Microbiota Development: In Pursuit of the Perfect Match. Nutrients 2024; 16:1976. [PMID: 38999725 PMCID: PMC11243202 DOI: 10.3390/nu16131976] [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: 05/23/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
The correct initial colonization and establishment of the gut microbiota during the early stages of life is a key step, with long-lasting consequences throughout the entire lifespan of the individual. This process is affected by several perinatal factors; among them, feeding mode is known to have a critical role. Breastfeeding is the optimal nutrition for neonates; however, it is not always possible, especially in cases of prematurity or early pathology. In such cases, most commonly babies are fed with infant formulas in spite of the official nutritional and health international organizations' recommendation on the use of donated human milk through milk banks for these cases. However, donated human milk still does not totally match maternal milk in terms of infant growth and gut microbiota development. The present review summarizes the practices of milk banks and hospitals regarding donated human milk, its safety and quality, and the health outcomes in infants fed with donated human milk. Additionally, we explore different alternatives to customize pasteurized donated human milk with the aim of finding the perfect match between each baby and banked milk for promoting the establishment of a beneficial gut microbiota from the early stages of life.
Collapse
Affiliation(s)
- Emilia Hick
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Marta Suárez
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Alejandra Rey
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
| | - Laura Mantecón
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Nuria Fernández
- Pediatrics Service, University Hospital of Cabueñes (CAB-SESPA), 33394 Gijón, Spain
| | - Gonzalo Solís
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
39
|
Zhang S, Fang X, Xu B, Zhou Y, Li F, Gao Y, Luo Y, Yao X, Liu X. Comprehensive analysis of phenotypes and transcriptome characteristics reveal the best atopic dermatitis mouse model induced by MC903. J Dermatol Sci 2024; 114:104-114. [PMID: 38806322 DOI: 10.1016/j.jdermsci.2024.05.003] [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: 01/05/2024] [Revised: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Although several mouse models of exogenous-agent-induced atopic dermatitis (AD) are currently available, the lack of certainty regarding their similarity with human AD has limited their scientific value. Thus, comprehensive evaluation of the characteristics of mouse models and their similarity with human AD is essential. OBJECTIVE To compare six different exogenous-agent-induced AD mouse models and find out the optimum models for study. METHODS Female BALB/c mice underwent induction of AD-like dermatitis by MC903 alone or in combination with ovalbumin (OVA), dinitrofluorobenzene (DNFB) alone or in combination with OVA, OVA alone, or Staphylococcus aureus. Gross phenotype, total immunoglobulin E (IgE) level, histopathological manifestations, and skin lesion transcriptome were analyzed, and metagenomic sequencing of the gut microbiome was performed. RESULTS The DNFB plus OVA model showed the highest disease severity, while the OVA model showed the lowest severity. The MC903 and MC903 plus OVA models showed high expression of T-helper (Th)2- and Th17-related genes; the DNFB and DNFB plus OVA models showed upregulation of Th1-, Th2-, and Th17-related genes; while the S. aureus inoculation model showed more enhanced Th1 and Th17 immune responses. In contrast to the other models, the OVA-induced model showed the lowest expression levels of inflammation-related genes, while the MC903 model shared the largest overlap with human AD profiles. The intestinal microbiota of all groups showed significant differences after modeling. CONCLUSION Each AD mouse model exhibited different characteristics. The MC903 model was the best to recapitulate most features of human AD among these exogenous-agent-induced AD models.
Collapse
Affiliation(s)
- Shan Zhang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Xiaokai Fang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Beilei Xu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yuan Zhou
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Fang Li
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yuwen Gao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yang Luo
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China.
| | - Xiaochun Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China.
| |
Collapse
|
40
|
Gamal NA, Shoaib MA, Farag AG, Stark R, Tso S. A non-inferiority clinical trial comparing probiotics and oral corticosteroids for the management of acute exacerbation of atopic dermatitis patients. SKIN HEALTH AND DISEASE 2024; 4:e373. [PMID: 38846682 PMCID: PMC11150755 DOI: 10.1002/ski2.373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
A prospective controlled pilot study on the feasibility of utilization of a probiotic mixture for management of acute exacerbation of atopic dermatitis (AD). Patients were allocated to either standard of care (SOC) therapy with tapering dose of steroids or a probiotic mixture over 3 weeks. After the 3-week intervention, patients on steroids achieved significantly higher clinical response rates and significantly deeper response as measured by the change in SCORAD score. No gut microbiome changes could be appreciated in either group after the treatment period. We could conclude that probiotics cannot replace SOC therapy for the management of acute exacerbation of AD.
Collapse
Affiliation(s)
- Nahla A. Gamal
- Department of DermatologyMenoufia UniversityShebin ElkomEgypt
- South Warwickshire NHS Foundation TrustWarwickUK
| | | | - Azza G. Farag
- Department of DermatologyMenoufia UniversityShebin ElkomEgypt
| | - Richard Stark
- Bioinformatics Research Technology PlatformUniversity of WarwickCoventryUK
| | - Simon Tso
- South Warwickshire NHS Foundation TrustWarwickUK
| |
Collapse
|
41
|
Javed U, Podury S, Kwon S, Liu M, Kim D, Fallah Zadeh A, Li Y, Khan A, Francois F, Schwartz T, Zeig-Owens R, Grunig G, Veerappan A, Zhou J, Crowley G, Prezant D, Nolan A. Biomarkers of Airway Disease, Barrett's and Underdiagnosed Reflux Noninvasively (BAD-BURN): a Case-Control Observational Study Protocol. RESEARCH SQUARE 2024:rs.3.rs-4355584. [PMID: 38798396 PMCID: PMC11118699 DOI: 10.21203/rs.3.rs-4355584/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND Particulate matter exposure (PM) is a cause of aerodigestive disease globally. The destruction of the World Trade Center (WTC) exposed fifirst responders and inhabitants of New York City to WTC-PM and caused obstructive airways disease (OAD), gastroesophageal Refux disease (GERD) and Barrett's Esophagus (BE). GERD not only diminishes health-related quality of life but also gives rise to complications that extend beyond the scope of BE. GERD can incite or exacerbate allergies, sinusitis, bronchitis, and asthma. Disease features of the aerodigestive axis can overlap, often necessitating more invasive diagnostic testing and treatment modalities. This presents a need to develop novel non-invasive biomarkers of GERD, BE, airway hyperreactivity (AHR), treatment efficacy, and severity of symptoms. METHODS Our observational case-cohort study will leverage the longitudinally phenotyped Fire Department of New York (FDNY)-WTC exposed cohort to identify Biomarkers of Airway Disease, Barrett's and Underdiagnosed Refux Noninvasively (BAD-BURN). Our study population consists of n = 4,192 individuals from which we have randomly selected a sub-cohort control group (n = 837). We will then recruit subgroups of i. AHR only ii. GERD only iii. BE iv. GERD/BE and AHR overlap or v. No GERD or AHR, from the sub-cohort control group. We will then phenotype and examine non-invasive biomarkers of these subgroups to identify under-diagnosis and/or treatment efficacy. The findings may further contribute to the development of future biologically plausible therapies, ultimately enhance patient care and quality of life. DISCUSSION Although many studies have suggested interdependence between airway and digestive diseases, the causative factors and specific mechanisms remain unclear. The detection of the disease is further complicated by the invasiveness of conventional GERD diagnosis procedures and the limited availability of disease-specific biomarkers. The management of Refux is important, as it directly increases risk of cancer and negatively impacts quality of life. Therefore, it is vital to develop novel noninvasive disease markers that can effectively phenotype, facilitate early diagnosis of premalignant disease and identify potential therapeutic targets to improve patient care. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05216133; January 18, 2022.
Collapse
Affiliation(s)
- Urooj Javed
- New York University Grossman School of Medicine (NYUGSoM)
| | - Sanjiti Podury
- New York University Grossman School of Medicine (NYUGSoM)
| | - Sophia Kwon
- New York University Grossman School of Medicine (NYUGSoM)
| | - Mengling Liu
- New York University Grossman School of Medicine (NYUGSoM)
| | - Daniel Kim
- New York University Grossman School of Medicine (NYUGSoM)
| | | | - Yiwei Li
- New York University Grossman School of Medicine (NYUGSoM)
| | - Abraham Khan
- New York University Grossman School of Medicine (NYUGSoM)
| | - Fritz Francois
- New York University Grossman School of Medicine (NYUGSoM)
| | | | | | | | - Arul Veerappan
- New York University Grossman School of Medicine (NYUGSoM)
| | - Joanna Zhou
- New York University Grossman School of Medicine (NYUGSoM)
| | - George Crowley
- New York University Grossman School of Medicine (NYUGSoM)
| | - David Prezant
- New York University Grossman School of Medicine (NYUGSoM)
| | - Anna Nolan
- New York University Grossman School of Medicine (NYUGSoM)
| |
Collapse
|
42
|
Vänni P, Turunen J, Äijälä VK, Tapiainen VV, Paalanne M, Pokka T, Paalanne N, Tejesvi MV, Ruuska TS. Gut Mycobiome in Atopic Dermatitis and in Overweight Young Children: A Prospective Cohort Study in Finland. J Fungi (Basel) 2024; 10:333. [PMID: 38786688 PMCID: PMC11122208 DOI: 10.3390/jof10050333] [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: 02/18/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Gut bacterial alterations have been previously linked to several non-communicable diseases in adults, while the association of mycobiome is not well understood in these diseases, especially in infants and children. Few studies have been conducted on the association between gut mycobiome and non-communicable diseases in children. We investigated gut mycobiome composition using 194 faecal samples collected at birth, 6 months after birth, and 18 months after birth in relation to atopic dermatitis (AD) and overweight diagnoses at the age of 18 or 36 months. The mycobiome exhibited distinct patterns, with Truncatella prevalent in the meconium samples of both overweight and non-overweight groups. Saccharomyces took precedence in overweight cases at 6 and 18 months, while Malassezia dominated non-overweight samples at 6 months. Saccharomyces emerged as a consistent high-abundance taxon across groups that had dermatitis and were overweight. We found a weak association between gut mycobiome and AD at birth and overweight at 18 months when using machine learning (ML) analyses. In ML, unidentified fungi, Alternaria, Rhodotorula, and Saccharomyces, were important for classifying AD, while Saccharomyces, Thelebolus, and Dothideomycetes were important for classifying overweight. Gut mycobiome might be associated with the development of AD and overweight in children.
Collapse
Affiliation(s)
- Petri Vänni
- Research Unit of Clinical Medicine, University of Oulu, 8000 Oulu, Finland; (J.T.); (M.P.); (T.P.); (N.P.); (M.V.T.); (T.S.R.)
| | - Jenni Turunen
- Research Unit of Clinical Medicine, University of Oulu, 8000 Oulu, Finland; (J.T.); (M.P.); (T.P.); (N.P.); (M.V.T.); (T.S.R.)
- Biocenter Oulu, University of Oulu, 8000 Oulu, Finland
| | - Ville K. Äijälä
- Department of Paediatrics and Adolescent Medicine, Oulu University Hospital, 8000 Oulu, Finland; (V.K.Ä.); (V.V.T.)
| | - Vilja V. Tapiainen
- Department of Paediatrics and Adolescent Medicine, Oulu University Hospital, 8000 Oulu, Finland; (V.K.Ä.); (V.V.T.)
| | - Marika Paalanne
- Research Unit of Clinical Medicine, University of Oulu, 8000 Oulu, Finland; (J.T.); (M.P.); (T.P.); (N.P.); (M.V.T.); (T.S.R.)
- Department of Paediatrics and Adolescent Medicine, Oulu University Hospital, 8000 Oulu, Finland; (V.K.Ä.); (V.V.T.)
| | - Tytti Pokka
- Research Unit of Clinical Medicine, University of Oulu, 8000 Oulu, Finland; (J.T.); (M.P.); (T.P.); (N.P.); (M.V.T.); (T.S.R.)
- Research Service Unit, Oulu University Hospital, 8000 Oulu, Finland
| | - Niko Paalanne
- Research Unit of Clinical Medicine, University of Oulu, 8000 Oulu, Finland; (J.T.); (M.P.); (T.P.); (N.P.); (M.V.T.); (T.S.R.)
- Department of Paediatrics and Adolescent Medicine, Oulu University Hospital, 8000 Oulu, Finland; (V.K.Ä.); (V.V.T.)
| | - Mysore V. Tejesvi
- Research Unit of Clinical Medicine, University of Oulu, 8000 Oulu, Finland; (J.T.); (M.P.); (T.P.); (N.P.); (M.V.T.); (T.S.R.)
- Ecology and Genetics, Faculty of Science, University of Oulu, 8000 Oulu, Finland
| | - Terhi S. Ruuska
- Research Unit of Clinical Medicine, University of Oulu, 8000 Oulu, Finland; (J.T.); (M.P.); (T.P.); (N.P.); (M.V.T.); (T.S.R.)
- Biocenter Oulu, University of Oulu, 8000 Oulu, Finland
- Department of Paediatrics and Adolescent Medicine, Oulu University Hospital, 8000 Oulu, Finland; (V.K.Ä.); (V.V.T.)
| |
Collapse
|
43
|
Paller AS, Scharschmidt TC, Kezic S, Irvine AD. Preclinical Atopic Dermatitis Skin in Infants: An Emerging Research Area. J Invest Dermatol 2024; 144:1001-1009. [PMID: 38573278 DOI: 10.1016/j.jid.2024.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 04/05/2024]
Abstract
Whereas clinically apparent atopic dermatitis (AD) can be confirmed by validated diagnostic criteria, the preclinical phenotype of infants who eventually develop AD is less well-characterized. Analogous to unaffected or nonlesional skin in established AD, clinically normal-appearing skin in infants who will develop clinical AD has distinct changes. Prospective studies have revealed insights into this preclinical AD phenotype. In this study, we review the structural, immunologic, and microbiome nature of the preclinical AD phenotype. Determination of markers that predict the development of AD will facilitate targeting of interventions to prevent the development or reduce the severity of AD in infants.
Collapse
Affiliation(s)
- Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Sanja Kezic
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Alan D Irvine
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
44
|
Hilliard MA, Sela DA. Transmission and Persistence of Infant Gut-Associated Bifidobacteria. Microorganisms 2024; 12:879. [PMID: 38792709 PMCID: PMC11124121 DOI: 10.3390/microorganisms12050879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Bifidobacterium infantis are the primary colonizers of the infant gut, yet scientific research addressing the transmission of the genus Bifidobacterium to infants remains incomplete. This review examines microbial reservoirs of infant-type Bifidobacterium that potentially contribute to infant gut colonization. Accordingly, strain inheritance from mother to infant via the fecal-oral route is likely contingent on the bifidobacterial strain and phenotype, whereas transmission via the vaginal microbiota may be restricted to Bifidobacterium breve. Additional reservoirs include breastmilk, horizontal transfer from the environment, and potentially in utero transfer. Given that diet is a strong predictor of Bifidobacterium colonization in early life and the absence of Bifidobacterium is observed regardless of breastfeeding, it is likely that additional factors are responsible for bifidobacterial colonization early in life.
Collapse
Affiliation(s)
- Margaret A. Hilliard
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - David A. Sela
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
- Department of Nutrition, University of Massachusetts, Amherst, MA 01003, USA
- Department of Microbiology, University of Massachusetts, Amherst, MA 01003, USA
- Department of Microbiology & Physiological Systems and Center for Microbiome Research, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
45
|
Mohammad S, Karim MR, Iqbal S, Lee JH, Mathiyalagan R, Kim YJ, Yang DU, Yang DC. Atopic dermatitis: Pathophysiology, microbiota, and metabolome - A comprehensive review. Microbiol Res 2024; 281:127595. [PMID: 38218095 DOI: 10.1016/j.micres.2023.127595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/11/2023] [Accepted: 12/24/2023] [Indexed: 01/15/2024]
Abstract
Atopic dermatitis (AD) is a prevalent inflammatory skin condition that commonly occurs in children. Genetics, environment, and defects in the skin barrier are only a few of the factors that influence how the disease develops. As human microbiota research has advanced, more scientific evidence has shown the critical involvement of the gut and skin bacteria in the pathogenesis of atopic dermatitis. Microbiome dysbiosis, defined by changed diversity and composition, as well as the development of pathobionts, has been identified as a potential cause for recurring episodes of atopic dermatitis. Gut dysbiosis causes "leaky gut syndrome" by disrupting the epithelial lining of the gut, which allows bacteria and other endotoxins to enter the bloodstream and cause inflammation. The same is true for the disruption of cutaneous homeostasis caused by skin dysbiosis, which enables bacteria and other pathogens to reach deeper skin layers or even systemic circulation, resulting in inflammation. Furthermore, it is now recognized that the gut and skin microbiota releases both beneficial and toxic metabolites. Here, this review covers a range of topics related to AD, including its pathophysiology, the microbiota-AD connection, commonly used treatments, and the significance of metabolomics in AD prevention, treatment, and management, recognizing its potential in providing valuable insights into the disease.
Collapse
Affiliation(s)
- Shahnawaz Mohammad
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Md Rezaul Karim
- Department of Biopharmaceutical Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Safia Iqbal
- Department of Biopharmaceutical Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Microbiology, Varendra Institute of Biosciences, Affiliated by Rajshahi University, Natore, Rajshahi 6400, Bangladesh
| | - Jung Hyeok Lee
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Yeon Ju Kim
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Dong Uk Yang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| | - Deok Chun Yang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| |
Collapse
|
46
|
Tai SK, Lin YH, Lin CH, Lin MC. Antibiotic exposure during pregnancy increases risk for childhood atopic diseases: a nationwide cohort study. Eur J Med Res 2024; 29:189. [PMID: 38504329 PMCID: PMC10953187 DOI: 10.1186/s40001-024-01793-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/13/2024] [Indexed: 03/21/2024] Open
Abstract
PURPOSE The prevalence of atopic diseases has increased in recent decades. A possible link between antibiotic use during pregnancy and childhood atopic disease has been proposed. The aim of this study is to explore the association of antibiotic exposure during pregnancy with childhood atopic diseases from a nationwide, population-based perspective. METHODS This was a nationwide population-based cohort study. Taiwan's National Health Insurance Research Database was the main source of data. The pairing of mothers and children was achieved by linking the NHIRD with the Taiwan Maternal and Child Health Database. This study enrolled the first-time pregnancies from 2004 to 2010. Infants of multiple delivery, preterm delivery, and death before 5 years old were excluded. All participants were followed up at least for 5 years. Antenatal antibiotics prescribed to mothers during the pregnancy period were reviewed. Children with more than two outpatient visits, or one admission, with a main diagnosis of asthma, allergic rhinitis, or atopic dermatitis were regarded as having an atopic disease. RESULTS A total of 900,584 children were enrolled in this study. The adjusted hazard ratios of antibiotic exposure during pregnancy to childhood atopic diseases were 1.12 for atopic dermatitis, 1.06 for asthma, and 1.08 for allergic rhinitis, all of which reached statistical significance. The trimester effect was not significant. There was a trend showing the higher the number of times a child was prenatally exposed to antibiotics, the higher the hazard ratio was for childhood atopic diseases. CONCLUSIONS Prenatal antibiotic exposure might increase the risk of childhood atopic diseases in a dose-dependent manner.
Collapse
Affiliation(s)
- Sheng-Kang Tai
- Department of Pediatrics, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Yi-Hsuan Lin
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ching-Heng Lin
- Department of Medical Research, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sec. 4, Taichung, 40705, Taiwan
| | - Ming-Chih Lin
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Food and Nutrition, Providence University, Taichung, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| |
Collapse
|
47
|
Barman M, Gio-Batta M, Andrieux L, Stråvik M, Saalman R, Fristedt R, Rabe H, Sandin A, Wold AE, Sandberg AS. Short-chain fatty acids (SCFA) in infants' plasma and corresponding mother's milk and plasma in relation to subsequent sensitisation and atopic disease. EBioMedicine 2024; 101:104999. [PMID: 38340558 PMCID: PMC10869761 DOI: 10.1016/j.ebiom.2024.104999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Short-chain fatty acids (SCFAs) in intestinal contents may influence immune function, while less is known about SCFAs in blood plasma. The aims were to investigate the relation between infants' and maternal plasma SCFAs, as well as SCFAs in mother's milk, and relate SCFA concentrations in infant plasma to subsequent sensitisation and atopic disease. METHODS Infant plasma (N = 148) and corresponding mother's milk and plasma were collected four months postpartum. Nine SCFA (formic, acetic, propionic, isobutyric, butyric, succinic, valeric, isovaleric, and caproic acid) were analysed by UPLC-MS. At 12 months of age, atopic disease was diagnosed by a pediatric allergologist, and sensitisation was measured by skin prick test. All families participated in the Swedish birth cohort NICE (Nutritional impact on Immunological maturation during Childhood in relation to the Environment). FINDINGS Infants with sensitisation, atopic eczema, or food allergy had significantly lower concentrations of five, three, and two SCFAs, respectively, in plasma at four months. Logistic regressions models showed significant negative associations between formic, succinic, and caproic acid and sensitisation [ORadj (95% CI) per SD: 0.41 (0.19-0.91); 0.19 (0.05-0.75); 0.25 (0.09-0.66)], and between acetic acid and atopic eczema [0.42 (0.18-0.95)], after adjusting for maternal allergy. Infants' and maternal plasma SCFA concentrations correlated strongly, while milk SCFA concentrations were unrelated to both. Butyric and caproic acid concentrations were enriched around 100-fold, and iso-butyric and valeric acid around 3-5-fold in mother's milk, while other SCFAs were less prevalent in milk than in plasma. INTERPRETATION Butyric and caproic acid might be actively transported into breast milk to meet the needs of the infant, although mechanistic studies are needed to confirm this. The negative associations between certain SCFAs on sensitisation and atopic disease adds to prior evidence regarding their immunoregulatory potential. FUNDING Swedish Research Council (Nr. 2013-3145, 2019-0137 and 2023-02217 to A-S.S.), Swedish Research Council for Health, Working Life and Welfare FORTE, Nr 2018-00485 to A.W.), The Swedish Asthma and Allergy Association's Research Fund (2020-0020 to A.S.).
Collapse
Affiliation(s)
- Malin Barman
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden.
| | - Monica Gio-Batta
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Léna Andrieux
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden; Département de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, 69342 Lyon Cedex 07, France
| | - Mia Stråvik
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Robert Saalman
- Institute of Clinical Sciences, Department of Pediatrics, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Rikard Fristedt
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Hardis Rabe
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Anna Sandin
- Department of Clinical Science, Pediatrics, Sunderby Research Unit, Umeå University, Umeå 901 87, Sweden
| | - Agnes E Wold
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Ann-Sofie Sandberg
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
48
|
Shen F, Gao C, Wang M, Ding X, Zhao H, Zhou M, Mao J, Kuai L, Li B, Wang D, Zhang H, Ma X. Therapeutic effects of the Qingre-Qushi recipe on atopic dermatitis through the regulation of gut microbiota and skin inflammation. Heliyon 2024; 10:e26063. [PMID: 38380039 PMCID: PMC10877368 DOI: 10.1016/j.heliyon.2024.e26063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Accumulating evidence has highlighted a strong association between gut microbiota and the occurrence, development, prevention, and treatment of atopic dermatitis (AD). The regulation of gut microbial dysbiosis by oral traditional Chinese medicine (TCM) has garnered significant attention. In the treatment of AD, the TCM formula Qingre-Qushi Recipe (QRQS) has demonstrated clinical efficacy. However, both the therapeutic mechanisms of QRQS and its impact on gut microbiota remain unclear. Thus, our study aimed to assess the efficacy of QRQS and evaluate its influence on the composition and diversity of gut microbiota in AD animal models. First, we investigated the therapeutic effect of QRQS on AD using two animal models: filaggrin-deficient mice (Flaky tail, ft/ft) and MC903-induced AD-like mice. Subsequently, we explored its influence on the composition and diversity of gut microbiota. Our results demonstrated that QRQS treatment ameliorated the symptoms in both ft/ft mice and MC903-induced AD-like mice. It also reduced the levels of serum IgE and pro-inflammatory cytokines, including IL-1β, IL-4, IL-5, IL-9, IL-13, IL-17A, and TNF-α. Furthermore, QRQS remarkably regulated gut microbiota diversity by increasing Lactobacillaceae and decreasing Bacteroidales. The inflammatory factors in peripheral serum of ft/ft mice showed a close correlation with gut microbiota, as determined using the Spearman correlation coefficient. Additionally, PICRUSt analysis revealed an enrichment in ascorbate and aldarate metabolism, fatty acid metabolism and biosynthesis, and propanoate metabolism in the QRQS group compared to the ft/ft group. Finally, we identified liquiritin as the primary active ingredient of QRQS using ultra-high-performance liquid chromatography-high-resolution mass spectrometry (UPLC-HRMS). Our findings revealed that QRQS improved AD-like symptoms and alleviated skin inflammation in ft/ft and MC903-induced mice. This suggests that modulating the gut microbiota may help elucidate its anti-inflammation activation mechanism, highlighting a new therapeutic strategy that targets the intestinal flora to prevent and treat AD.
Collapse
Affiliation(s)
- Fang Shen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Chunjie Gao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Mingxia Wang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Xiaojie Ding
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Hang Zhao
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Mi Zhou
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jingyi Mao
- Department of Dermatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Le Kuai
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Bin Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
- Institute of Dermatology, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Dongming Wang
- Department of Dermatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huimin Zhang
- Department of Dermatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
- Department of Dermatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| |
Collapse
|
49
|
Chan OM, Xu W, Cheng NS, Leung ASY, Ching JYL, Fong BLY, Cheong PK, Zhang L, Chan FKL, Ng SC, Leung TF. A novel infant microbiome formula (SIM03) improved eczema severity and quality of life in preschool children. Sci Rep 2024; 14:3168. [PMID: 38326388 PMCID: PMC10850179 DOI: 10.1038/s41598-024-53848-w] [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: 09/14/2023] [Accepted: 02/06/2024] [Indexed: 02/09/2024] Open
Abstract
Altered gut microbiome composition has been reported in children with eczema and interventions that restore beneficial bacteria in the gut may improve eczema. This open-label pilot study aimed to investigate the efficacy of a novel infant microbiome formula (SIM03) in young children with eczema. Pre-school Chinese children aged 1-5 years old with eczema received SIM03 twice daily for three months. The novelty of SIM03 consists of both the use of a patented microencapsulation technology to protect the viability of unique Bifidobacterium bifidum and Bifidobacterium breve strains identified through big data analysis of large metagenomic datasets of young Chinese children. Paired stool samples at baseline and following SIM03 were analyzed by metagenomics sequencing. Generalized estimating equation was used to analyze changes in eczema severity, skin biophysical parameters, quality of life and stool microbiome. Twenty children aged 3.0 ± 1.6 years (10 with severe eczema) were recruited. Treatment compliance was ≥ 98%. SCORing Atopic Dermatitis score decreased significantly at two months (P = 0.008) and three months (P < 0.001), while quality of life improved significantly at 1, 2, and 3 months. The relative abundance of B. breve and microbial pathways on acetate and acetyl-CoA synthesis were enriched in stool samples at one month (P = 0.0014). Children who demonstrated increased B. bifidum after SIM03 showed improvement in sleep loss (P = 0.045). Relative abundance of B. breve correlated inversely with eczema extent (P = 0.023) and intensity (P = 0.019) only among patients with increased B. breve at Month 3. No serious adverse event was observed. In conclusion, SIM03 is well tolerated. This patented microbiome formula improves disease severity and quality of life in young eczematous children by enhancing the delivery of B. bifidum and B. breve in the gut. SIM03 is a potential treatment option for childhood eczema.
Collapse
Affiliation(s)
- Oi Man Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Wenye Xu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Shatin, Hong Kong SAR, China
| | - Nam Sze Cheng
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Agnes Sze Yin Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jessica Yuet Ling Ching
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Shatin, Hong Kong SAR, China
| | - Brian Leong Yuen Fong
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Pui Kuan Cheong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Shatin, Hong Kong SAR, China
| | - Lin Zhang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Shatin, Hong Kong SAR, China
| | - Francis Ka Leung Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Shatin, Hong Kong SAR, China
| | - Siew Chien Ng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Shatin, Hong Kong SAR, China
| | - Ting Fan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China.
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
50
|
Ryguła I, Pikiewicz W, Grabarek BO, Wójcik M, Kaminiów K. The Role of the Gut Microbiome and Microbial Dysbiosis in Common Skin Diseases. Int J Mol Sci 2024; 25:1984. [PMID: 38396663 PMCID: PMC10889245 DOI: 10.3390/ijms25041984] [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: 01/05/2024] [Revised: 01/28/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Dermatoses are an increasingly common problem, particularly in developed countries. The causes of this phenomenon include genetic factors and environmental elements. More and more scientific reports suggest that the gut microbiome, more specifically its dysbiosis, also plays an important role in the induction and progression of diseases, including dermatological diseases. The gut microbiome is recognised as the largest endocrine organ, and has a key function in maintaining human homeostasis. In this review, the authors will take a close look at the link between the gut-skin axis and the pathogenesis of dermatoses such as atopic dermatitis, psoriasis, alopecia areata, and acne. The authors will also focus on the role of probiotics in remodelling the microbiome and the alleviation of dermatoses.
Collapse
Affiliation(s)
- Izabella Ryguła
- Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Wojciech Pikiewicz
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Beniamin Oskar Grabarek
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Michał Wójcik
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Konrad Kaminiów
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| |
Collapse
|