1
|
Akagbosu CO, McCauley KE, Namasivayam S, Romero-Soto HN, O’Brien W, Bacorn M, Bohrnsen E, Schwarz B, Mistry S, Burns AS, Perez-Chaparro PJ, Chen Q, LaPoint P, Patel A, Krausfeldt LE, Subramanian P, Sellers BA, Cheung F, Apps R, Douagi I, Levy S, Nadler EP, Hourigan SK. Gut microbiome shifts in adolescents after sleeve gastrectomy with increased oral-associated taxa and pro-inflammatory potential. Gut Microbes 2025; 17:2467833. [PMID: 39971742 PMCID: PMC11845021 DOI: 10.1080/19490976.2025.2467833] [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: 09/26/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Bariatric surgery is highly effective in achieving weight loss in children and adolescents with severe obesity, however the underlying mechanisms are incompletely understood, and gut microbiome changes are unknown. Here, we show that adolescents exhibit significant gut microbiome and metabolome shifts several months after laparoscopic vertical sleeve gastrectomy (VSG), with increased alpha diversity and notably with enrichment of oral-associated taxa. To assess causality of the microbiome/metabolome changes in phenotype, pre-VSG and post-VSG stool was transplanted into germ-free mice. Post-VSG stool was not associated with any beneficial outcomes such as adiposity reduction compared pre-VSG stool. However, post-VSG stool exhibited a potentially inflammatory phenotype with increased intestinal Th17 and decreased regulatory T cells. Concomitantly, we found elevated fecal calprotectin and an enrichment of proinflammatory pathways in a subset of adolescents post-VSG. We show that in some adolescents, microbiome changes post-VSG may have inflammatory potential, which may be of importance considering the increased incidence of inflammatory bowel disease post-VSG.
Collapse
Affiliation(s)
- Cynthia O. Akagbosu
- Department of Gastroenterology, Weill Cornell Medicine, New York, New York, USA
| | - Kathryn E. McCauley
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sivaranjani Namasivayam
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Hector N. Romero-Soto
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Wade O’Brien
- Dartmouth Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mickayla Bacorn
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Shreni Mistry
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew S. Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - P. Juliana Perez-Chaparro
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Qing Chen
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Phoebe LaPoint
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anal Patel
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lauren E. Krausfeldt
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian A. Sellers
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Foo Cheung
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Richard Apps
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Iyadh Douagi
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Shira Levy
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Suchitra K. Hourigan
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Bashar S, Tun HM, Ting JY, Hicks M, Mandhane PJ, Moraes TJ, Simons E, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. Impact of postpartum hospital length of stay on infant gut microbiota: a comprehensive analysis of vaginal and caesarean birth. J Hosp Infect 2025; 156:50-60. [PMID: 39510138 DOI: 10.1016/j.jhin.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/15/2024] [Accepted: 10/05/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND The primary concern with prolonged hospitalization following birth is the risk of acquiring hospital-acquired infections (HAIs) caused by opportunistic bacteria, which can alter the early establishment of gut microbiota. OBJECTIVE To assess the association between postpartum hospital length of stay (LOS) and the composition of gut microbiota at 3 and 12 months of age according to birth mode. METHODS In total, 1313 Canadian infants from the CHILD Cohort Study were involved in this study. Prolonged LOS was defined as ≥2 days following vaginal delivery (VD) and ≥3 days following caesarean section (CS). The gut microbiota of infants was characterized by Illumina 16S rRNA sequencing of faecal samples at 3-4 months and 12 months of age. FINDINGS Following prolonged LOS, VD infants with no exposure to intrapartum antibiotics had a higher abundance of bacteria known to cause HAIs in their gut, including Enterococcus spp. at 3 and 12 months, Citrobacter spp. at 3 months, and Clostridioides difficile at 12 months. Abundance of Enterococcus spp. or Citrobacter spp. at 3 months significantly mediated the association between LOS and low abundance of Bacteroidaceae, or higher Enterococcaeae/Bacteriodaceae or Enterobacterales/Bacteroidaceae abundance ratios at 12 months of age in VD infants without intrapartum antibiotic exposure. HAI-causing Enterobacterales were also more abundant in later infancy in infants with prolonged LOS following CS. In the absence of exclusive breastfeeding at 3 months or any breastfeeding at 12 months, Porphyromonadaceae (of Bacteroidota) were depleted in CS infants with prolonged LOS. CONCLUSIONS Prolonged hospital stay after birth is associated with infant gut dysbiosis.
Collapse
Affiliation(s)
- S Bashar
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - H M Tun
- The Jockey Club School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Microbiota I-Center, Hong Kong, China
| | - J Y Ting
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - M Hicks
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - P J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - T J Moraes
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Canada
| | - E Simons
- Section of Allergy and Immunology, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - S E Turvey
- Department of Pediatrics, BC Children's Hospital, Vancouver, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - P Subbarao
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Canada; Department of Medicine, McMaster University, Hamilton, Canada
| | - J A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - A L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, Canada.
| |
Collapse
|
3
|
Zha Z, Jia C, Zhou R, Yin Q, Hu Y, Huang Z, Peng L, Zhang Y, Qiu X, Chen Y, Zhong Y, Wang Y, Pang M, Lu S, Sheng C, Huang L. Clostridium difficile-derived membrane vesicles promote fetal growth restriction via inhibiting trophoblast motility through PPARγ/RXRα/ANGPTL4 axis. NPJ Biofilms Microbiomes 2024; 10:158. [PMID: 39741137 DOI: 10.1038/s41522-024-00630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/07/2024] [Indexed: 01/02/2025] Open
Abstract
Fetal growth restriction (FGR) is a common complication of pregnancy, which seriously endangers fetal health and still lacks effective therapeutic targets. Clostridium difficile (C. difficile) is associated with fetal birth weight, and its membrane vesicles (MVs) are pathogenic vectors. However, the role of C. difficile and its MVs in FGR remains unclear. Here we found that supplementation with C. difficile altered the characteristics of gut microbiota and reduced the birth weight in mice. Interestingly, C. difficile MVs entered placenta, inhibited trophoblast motility, and induced fetal weight loss in mice. Mechanistically, C. difficile MVs activated the PPAR pathway via enhancing the transcriptional activity of PPARγ promoter, consequently inhibiting trophoblast motility. Moreover, PPARγ expression was significantly elevated in FGR placenta, and negatively correlated with fetal birth weight. Together, our findings reveal the significance of C. difficile and its MVs in FGR, providing new insights into the mechanisms of FGR development.
Collapse
Affiliation(s)
- Zhiqiang Zha
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunhong Jia
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ruisi Zhou
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinlan Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhipeng Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linyu Peng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yichi Zhang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaowei Qiu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yawen Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Menglan Pang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shijing Lu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chao Sheng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Wang X, Cui H, Li N, Liu B, Zhang X, Yang J, Zheng JS, Qiao C, Liu HX, Hu J, Wen D. Impact of vaginal seeding on the gut microbiome of infants born via cesarean section: A systematic review. J Infect 2024; 89:106348. [PMID: 39537035 DOI: 10.1016/j.jinf.2024.106348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/05/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE This systematic review summarizes eight studies involving 558 cesarean section (CS)-born infants (274 exposed to vaginal seeding (VS), 284 not exposed) and 261 infants born vaginally to investigate the effect of VS on gut microbiome colonization and development in CS-born infants. METHODS This study adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Relevant articles published before March 6, 2024, were identified through systematic searches of PubMed, Scopus, and Web of Science. We included experimental studies that investigated changes in the gut microbiota of CS-born infants following VS and reported changes in the gut microbiota. The relationship between VS and the gut microbiota composition of CS-born infants was assessed. RESULTS VS may selectively influence the abundance of bacterial genera from various phyla, such as an increased relative abundance of Bacteroides and Lactobacillus, in the gut microbiome of CS-seeded infants compared to CS-non-seeded infants. Conflicting results mainly concern microbial diversity. CONCLUSIONS Current evidence indicates modest changes in the gut microbiome of CS-born infants following VS. However, further clinical studies are necessary to fully understand its impact on early-life health outcomes, particularly regarding potential microbiome alterations and associated health risks.
Collapse
Affiliation(s)
- Xiaochuan Wang
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Health Sciences Institute, China Medical University, Shenyang 110122, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, China
| | - Hong Cui
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China Medical University, Shenyang 110004, China
| | - Na Li
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China Medical University, Shenyang 110004, China
| | - Borui Liu
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Health Sciences Institute, China Medical University, Shenyang 110122, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, China
| | - Xiaoyan Zhang
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Health Sciences Institute, China Medical University, Shenyang 110122, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, China
| | - Jing Yang
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Health Sciences Institute, China Medical University, Shenyang 110122, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, China
| | - Ju-Sheng Zheng
- Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China; School of Life Sciences, Westlake University, Hangzhou, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Chong Qiao
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China Medical University, Shenyang 110004, China
| | - Hui-Xin Liu
- Health Sciences Institute, China Medical University, Shenyang 110122, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, China.
| | - Jiajin Hu
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Health Sciences Institute, China Medical University, Shenyang 110122, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, China.
| | - Deliang Wen
- Research Center of China Medical University Birth Cohort, China Medical University, Shenyang 110122, China; Health Sciences Institute, China Medical University, Shenyang 110122, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang 110122, China.
| |
Collapse
|
5
|
Yang R, Shi Z, Li Y, Huang X, Li Y, Li X, Chen Q, Hu Y, Li X. Research focus and emerging trends of the gut microbiome and infant: a bibliometric analysis from 2004 to 2024. Front Microbiol 2024; 15:1459867. [PMID: 39633813 PMCID: PMC11615055 DOI: 10.3389/fmicb.2024.1459867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Background Over the past two decades, gut microbiota has demonstrated unprecedented potential in human diseases and health. The gut microbiota in early life is crucial for later health outcomes. This study aims to reveal the knowledge collaboration network, research hotspots, and explore the emerging trends in the fields of infant and gut microbiome using bibliometric analysis. Method We searched the literature on infant and gut microbiome in the Web of Science Core Collection (WOSCC) database from 2004 to 2024. CiteSpace V (version: 6.3.R1) and VOSview (version: 1.6.20) were used to display the top authors, journals, institutions, countries, authors, keywords, co-cited articles, and potential trends. Results A total of 9,899 documents were retrieved from the Web of Science Core Collection. The United States, China, and Italy were the three most productive countries with 3,163, 1,510, and 660 publications. The University of California System was the most prolific institution (524 publications). Van Sinderen, Douwe from University College Cork of Ireland was the most impactful author. Many studies have focused on atopic dermatitis (AD), necrotizing enterocolitis (NEC), as well as the immune mechanisms and microbial treatments for these diseases, such as probiotic strains mixtures and human milk oligosaccharides (HMOs). The mother-to-infant microbiome transmission, chain fatty acids, and butyrate maybe the emerging trends. Conclusion This study provided an overview of the knowledge structure of infant and gut microbiome, as well as a reference for future research.
Collapse
Affiliation(s)
- Ru Yang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Zeyao Shi
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yuan Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xi Huang
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yingxin Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xia Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Qiong Chen
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yanling Hu
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaowen Li
- Department of Neonatology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
6
|
Hsu CY, Khachatryan LG, Younis NK, Mustafa MA, Ahmad N, Athab ZH, Polyanskaya AV, Kasanave EV, Mirzaei R, Karampoor S. Microbiota-derived short chain fatty acids in pediatric health and diseases: from gut development to neuroprotection. Front Microbiol 2024; 15:1456793. [PMID: 39439941 PMCID: PMC11493746 DOI: 10.3389/fmicb.2024.1456793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
The infant gut microbiota undergoes significant changes during early life, which are essential for immune system maturation, nutrient absorption, and metabolic programming. Among the various microbial metabolites, short-chain fatty acids (SCFAs), primarily acetate, propionate, and butyrate, produced through the fermentation of dietary fibers by gut bacteria, have emerged as critical modulators of host-microbiota interactions. SCFAs serve as energy sources for colonic cells and play pivotal roles in regulating immune responses, maintaining gut barrier integrity, and influencing systemic metabolic pathways. Recent research highlights the potential neuroprotective effects of SCFAs in pediatric populations. Disruptions in gut microbiota composition and SCFA production are increasingly associated with a range of pediatric health issues, including obesity, allergic disorders, inflammatory bowel disease (IBD), and neurodevelopmental disorders. This review synthesizes current knowledge on the role of microbiota-derived SCFAs in pediatric health, emphasizing their contributions from gut development to neuroprotection. It also underscores the need for further research to unravel the precise mechanisms by which SCFAs influence pediatric health and to develop targeted interventions that leverage SCFAs for therapeutic benefits.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, United States
| | - Lusine G. Khachatryan
- Department of Pediatric Diseases, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Techniques, University of Imam Jafar Al-Sadiq, College of Technology, Baghdad, Iraq
| | - Nabeel Ahmad
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
- Department of Biotechnology, School of Allied Sciences, Dev Bhoomi Uttarakhand University Dehradun, Uttarakhand, India
| | - Zainab H. Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Angelina V. Polyanskaya
- Department of Pediatric Diseases, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Elena Victorovna Kasanave
- Department of Pediatric Diseases, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Sha C, Jin Z, Ku SY, Kogosov AS, Yu S, Bergese SD, Hsieh H. Necrotizing Enterocolitis and Neurodevelopmental Impairments: Microbiome, Gut, and Brain Entanglements. Biomolecules 2024; 14:1254. [PMID: 39456187 PMCID: PMC11505939 DOI: 10.3390/biom14101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
There is significant communication and interdependence among the gut, the microbiome, and the brain during development. Diseases, such as necrotizing enterocolitis (NEC), highlight how injury to the immature gastrointestinal tract leads to long-term neurological consequences, due to vulnerabilities of the brain in the early stages of life. A better understanding of the developing gut-microbiota-brain axis is needed to both prevent and treat the devastating consequences of these disease processes. The gut-microbiota-brain axis is a bidirectional communication pathway that includes metabolic, nervous, endocrine, and immune components. In this review, we discuss gut development, microbiome colonization and maturation, and the interactions that influence neurodevelopment in the context of NEC. We describe the components of the gut-brain axis and how the microbiome is an integral member of this relationship. Finally, we explore how derangements within the microbiome and gut-microbiota-brain axis affect the normal development and function of the other systems and long-term neurodevelopmental consequences for patients.
Collapse
Affiliation(s)
- Cuilee Sha
- Department of Pharmacological Sciences, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA;
- Center for Nervous System Disorders, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Zhaosheng Jin
- Department of Anesthesiology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA;
| | - Stella Y. Ku
- Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Ann S. Kogosov
- Renaissance School of Medicine, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Sun Yu
- Department of Surgery, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA
| | - Sergio D. Bergese
- Department of Anesthesiology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA;
| | - Helen Hsieh
- Center for Nervous System Disorders, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
- Department of Surgery, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA
| |
Collapse
|
8
|
Akagbosu CO, McCauley KE, Namasivayam S, Romero-Soto HN, O’Brien W, Bacorn M, Bohrnsen E, Schwarz B, Mistry S, Burns AS, Perez-Chaparro PJ, Chen Q, LaPoint P, Patel A, Krausfeldt LE, Subramanian P, Sellers BA, Cheung F, Apps R, Douagi I, Levy S, Nadler EP, Hourigan SK. Gut microbiome shifts in adolescents after sleeve gastrectomy with increased oral-associated taxa and pro-inflammatory potential. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.16.24313738. [PMID: 39371172 PMCID: PMC11451705 DOI: 10.1101/2024.09.16.24313738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background Bariatric surgery is highly effective in achieving weight loss in children and adolescents with severe obesity, however the underlying mechanisms are incompletely understood, and gut microbiome changes are unknown. Objectives 1) To comprehensively examine gut microbiome and metabolome changes after laparoscopic vertical sleeve gastrectomy (VSG) in adolescents and 2) to assess whether the microbiome/metabolome changes observed with VSG influence phenotype using germ-free murine models. Design 1) A longitudinal observational study in adolescents undergoing VSG with serial stool samples undergoing shotgun metagenomic microbiome sequencing and metabolomics (polar metabolites, bile acids and short chain fatty acids) and 2) a human-to-mouse fecal transplant study. Results We show adolescents exhibit significant gut microbiome and metabolome shifts several months after VSG, with increased alpha diversity and notably with enrichment of oral-associated taxa. To assess causality of the microbiome/metabolome changes in phenotype, pre-VSG and post-VSG stool was transplanted into germ-free mice. Post-VSG stool was not associated with any beneficial outcomes such as adiposity reduction compared pre-VSG stool. However, post-VSG stool exhibited an inflammatory phenotype with increased intestinal Th17 and decreased regulatory T cells. Concomitantly, we found elevated fecal calprotectin and an enrichment of proinflammatory pathways in a subset of adolescents post-VSG. Conclusion We show that in some adolescents, microbiome changes post-VSG may have inflammatory potential, which may be of importance considering the increased incidence of inflammatory bowel disease post-VSG.
Collapse
Affiliation(s)
- Cynthia O Akagbosu
- Department of Gastroenterology. Weill Cornell Medicine. New York, New York, United States
| | - Kathryn E McCauley
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Sivaranjani Namasivayam
- Clinical Microbiome Unit. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Hector N Romero-Soto
- Clinical Microbiome Unit. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Wade O’Brien
- Dartmouth Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States
| | - Mickayla Bacorn
- Clinical Microbiome Unit. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Eric Bohrnsen
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, United States
| | - Benjamin Schwarz
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, United States
| | - Shreni Mistry
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Andrew S Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - P. Juliana Perez-Chaparro
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Qing Chen
- Clinical Microbiome Unit. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Phoebe LaPoint
- Clinical Microbiome Unit. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Anal Patel
- Clinical Microbiome Unit. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Lauren E Krausfeldt
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Brian A Sellers
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, United States
| | - Foo Cheung
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, United States
| | - Richard Apps
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, United States
| | - Iyadh Douagi
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, United States
| | - Shira Levy
- Clinical Microbiome Unit. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Evan P Nadler
- Evan P Nadler. ProCare Consultants, Washington DC, Washington DC, United States
| | - Suchitra K Hourigan
- Clinical Microbiome Unit. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
9
|
Zhang M, Qiao H, Yang S, Kwok LY, Zhang H, Zhang W. Human Breast Milk: The Role of Its Microbiota and Metabolites in Infant Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10665-10678. [PMID: 38691667 DOI: 10.1021/acs.jafc.3c07690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
This review explores the role of microorganisms and metabolites in human breast milk and their impact on neonatal health. Breast milk serves as both a primary source of nutrition for newborns and contributes to the development and maturation of the digestive, immunological, and neurological systems. It has the potential to reduce the risks of infections, allergies, and asthma. As our understanding of the properties of human milk advances, there is growing interest in incorporating its benefits into personalized infant nutrition strategies, particularly in situations in which breastfeeding is not an option. Future infant formula products are expected to emulate the composition and advantages of human milk, aligning with an evolving understanding of infant nutrition. The long-term health implications of human milk are still under investigation.
Collapse
Affiliation(s)
- Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Hui Qiao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Shuwei Yang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
10
|
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
|
11
|
Aagaard KM, Barkin SL, Burant CF, Carnell S, Demerath E, Donovan SM, Eneli I, Francis LA, Gilbert-Diamond D, Hivert MF, LeBourgeois MK, Loos RJF, Lumeng JC, Miller AL, Okely AD, Osganian SK, Ramirez AG, Trasande L, Van Horn LV, Wake M, Wright RJ, Yanovski SZ. Understanding risk and causal mechanisms for developing obesity in infants and young children: A National Institutes of Health workshop. Obes Rev 2024; 25:e13690. [PMID: 38204366 DOI: 10.1111/obr.13690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 10/02/2023] [Accepted: 11/21/2023] [Indexed: 01/12/2024]
Abstract
Obesity in children remains a major public health problem, with the current prevalence in youth ages 2-19 years estimated to be 19.7%. Despite progress in identifying risk factors, current models do not accurately predict development of obesity in early childhood. There is also substantial individual variability in response to a given intervention that is not well understood. On April 29-30, 2021, the National Institutes of Health convened a virtual workshop on "Understanding Risk and Causal Mechanisms for Developing Obesity in Infants and Young Children." The workshop brought together scientists from diverse disciplines to discuss (1) what is known regarding epidemiology and underlying biological and behavioral mechanisms for rapid weight gain and development of obesity and (2) what new approaches can improve risk prediction and gain novel insights into causes of obesity in early life. Participants identified gaps and opportunities for future research to advance understanding of risk and underlying mechanisms for development of obesity in early life. It was emphasized that future studies will require multi-disciplinary efforts across basic, behavioral, and clinical sciences. An exposome framework is needed to elucidate how behavioral, biological, and environmental risk factors interact. Use of novel statistical methods may provide greater insights into causal mechanisms.
Collapse
Affiliation(s)
- Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Shari L Barkin
- Department of Pediatrics, Children's Hospital of Richmond, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan Carnell
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ellen Demerath
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sharon M Donovan
- Division of Nutritional Sciences, University of Illinois, Urbana-Champaign, Illinois, USA
- Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Illinois, USA
| | - Ihuoma Eneli
- Center for Healthy Weight and Nutrition, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
- Center of Nutrition, Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Lori A Francis
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Diane Gilbert-Diamond
- Department of Epidemiology, Medicine and Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Monique K LeBourgeois
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Ruth J F Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Julie C Lumeng
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Alison L Miller
- Department of Health Behavior and Health Education, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Anthony D Okely
- School of Health and Society, Faculty of Arts, Social Sciences and Humanities, University of Wollongong, Wollongong, New South Wales, Australia
- llawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia
- Department of Sport, Food, and Natural Sciences, Western Norway University of Applied Sciences, Sogndal, Norway
| | - Stavroula K Osganian
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Amelie G Ramirez
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Leonardo Trasande
- Department of Pediatrics, New York University (NYU) School of Medicine, New York, New York, USA
- Department of Environmental Medicine, New York University (NYU) School of Medicine, New York, New York, USA
- Department of Population Health, New York University (NYU) School of Medicine, New York, New York, USA
| | - Linda V Van Horn
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois, USA
| | - Melissa Wake
- Population Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, New York, New York, USA
| | - Susan Z Yanovski
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
12
|
Cho H, Kim J, Kim S, Jeong HI, Kwon M, Kim HM, Shim JS, Kim K, Baek J, Kyung Y, Choi SJ, Oh SY, Bae J, Won HH, Kim J, Ahn K. Postpartum Maternal Anxiety Affects the Development of Food Allergy Through Dietary and Gut Microbial Diversity During Early Infancy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:154-167. [PMID: 38528383 DOI: 10.4168/aair.2024.16.2.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/29/2023] [Accepted: 12/25/2023] [Indexed: 03/27/2024]
Abstract
PURPOSE We aimed to investigate the mediating factors between maternal anxiety and the development of food allergy (FA) in children until 2 years from birth. METHODS In this longitudinal cohort of 122 mother-child dyads from pregnancy to 24 months of age, we regularly surveyed maternal psychological states, infant feeding data, and allergic symptoms and collected stool samples at 6 months of age for microbiome analysis. Considering the temporal order of data collection, we investigated serial mediating effects and indirect effects among maternal anxiety, dietary diversity (DD), gut microbial diversity, and FA using structural equation modeling. RESULTS Among the 122 infants, 15 (12.3%) were diagnosed with FA. Increased maternal anxiety between 3 and 6 months after delivery was associated with a lower DD score. Infants with low DD at 4 months showed low gut microbial richness, which was associated with FA development. When the infants were grouped into 4 subtypes, using consensus clustering of 13 gut bacteria significantly associated with maternal anxiety and DD, Prevotella, Eubacterium, Clostridiales and Lachnospiraceae were more abundant in the group with lower FA occurrence. CONCLUSIONS Postpartum maternal anxiety, mediated by reduced DD and gut microbial diversity, may be a risk factor for the development of FA in infants during the first 2 years of life.
Collapse
Affiliation(s)
- Hyunbin Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Jiwon Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sukyung Kim
- Department of Pediatrics, Hallym University Dongtan Sacred Heart Hospital, Hallym University School of Medicine, Hwaseong, Korea
| | - Hye-In Jeong
- Department of Pediatrics, Eulji University Hospital, Eulji University School of Medicine, Seoul, Korea
| | - Mijeong Kwon
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Mi Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Sun Shim
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Kyunga Kim
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jihyun Baek
- Department of Psychology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yechan Kyung
- Department of Pediatrics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jaewoong Bae
- R&D Institute, BioEleven Co., Ltd., Seoul, Korea
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.
| | - Jihyun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea.
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea
| |
Collapse
|
13
|
Peng Y, Tun HM, Ng SC, Wai HKF, Zhang X, Parks J, Field CJ, Mandhane P, Moraes TJ, Simons E, Turvey SE, Subbarao P, Brook JR, Takaro TK, Scott JA, Chan FKL, Kozyrskyj AL. Maternal smoking during pregnancy increases the risk of gut microbiome-associated childhood overweight and obesity. Gut Microbes 2024; 16:2323234. [PMID: 38436093 PMCID: PMC10913716 DOI: 10.1080/19490976.2024.2323234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
Childhood obesity is linked to maternal smoking during pregnancy. Gut microbiota may partially mediate this association and could be potential targets for intervention; however, its role is understudied. We included 1,592 infants from the Canadian Healthy Infants Longitudinal Development Cohort. Data on environmental exposure and lifestyle factors were collected prenatally and throughout the first three years. Weight outcomes were measured at one and three years of age. Stool samples collected at 3 and 12 months were analyzed by sequencing the V4 region of 16S rRNA to profile microbial compositions and magnetic resonance spectroscopy to quantify the metabolites. We showed that quitting smoking during pregnancy did not lower the risk of offspring being overweight. However, exclusive breastfeeding until the third month of age may alleviate these risks. We also reported that maternal smoking during pregnancy significantly increased Firmicutes abundance and diversity. We further revealed that Firmicutes diversity mediates the elevated risk of childhood overweight and obesity linked to maternal prenatal smoking. This effect possibly occurs through excessive microbial butyrate production. These findings add to the evidence that women should quit smoking before their pregnancies to prevent microbiome-mediated childhood overweight and obesity risk, and indicate the potential obesogenic role of excessive butyrate production in early life.
Collapse
Affiliation(s)
- Ye Peng
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hein M Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Department of Medicine and Therapeutics, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hogan Kok-Fung Wai
- HKU-Pasteur Research Pole, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Xi Zhang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Jaclyn Parks
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Catherine J Field
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Piush Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Theo J Moraes
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Stuart E Turvey
- Department of Pediatrics, Child and Family Research Institute, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jeffrey R Brook
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Tim K Takaro
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Francis KL Chan
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Namasivayam S, Tilves C, Ding H, Wu S, Domingue JC, Ruiz-Bedoya C, Shah A, Bohrnsen E, Schwarz B, Bacorn M, Chen Q, Levy S, Dominguez Bello MG, Jain SK, Sears CL, Mueller NT, Hourigan SK. Fecal transplant from vaginally seeded infants decreases intraabdominal adiposity in mice. Gut Microbes 2024; 16:2353394. [PMID: 38743047 PMCID: PMC11095576 DOI: 10.1080/19490976.2024.2353394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
Exposing C-section infants to the maternal vaginal microbiome, coined "vaginal seeding", partially restores microbial colonization. However, whether vaginal seeding decreases metabolic disease risk is unknown. Therefore, we assessed the effect of vaginal seeding of human infants on adiposity in a murine model. Germ-free mice were colonized with transitional stool from human infants who received vaginal seeding or control (placebo) seeding in a double-blind randomized trial. There was a reduction in intraabdominal adipose tissue (IAAT) volume in male mice that received stool from vaginally seeded infants compared to control infants. Higher levels of isoleucine and lower levels of nucleic acid metabolites were observed in controls and correlated with increased IAAT. This suggests that early changes in the gut microbiome and metabolome caused by vaginal seeding have a positive impact on metabolic health.
Collapse
Affiliation(s)
- Sivaranjani Namasivayam
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Curtis Tilves
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Hua Ding
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shaoguang Wu
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jada C Domingue
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camilo Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ankit Shah
- Inova Health System, Inova Women’s Hospital, Falls Church, VA, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases; National Institutes of Health, Hamilton, MT, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases; National Institutes of Health, Hamilton, MT, USA
| | - Mickayla Bacorn
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Qing Chen
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shira Levy
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maria Gloria Dominguez Bello
- Departments of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA
- Humans and the microbiome program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, Canada
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cynthia L Sears
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Suchitra K Hourigan
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Division of Pediatric Gastroenterology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
DuPont HL, Salge MMH. The Importance of a Healthy Microbiome in Pregnancy and Infancy and Microbiota Treatment to Reverse Dysbiosis for Improved Health. Antibiotics (Basel) 2023; 12:1617. [PMID: 37998819 PMCID: PMC10668833 DOI: 10.3390/antibiotics12111617] [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: 10/21/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The microbiome of newborn infants during the first 1000 days, influenced early on by their mothers' microbiome health, mode of delivery and breast feeding, orchestrates the education and programming of the infant's immune system and determines in large part the general health of the infant for years. METHODS PubMed was reviewed for maternal infant microbiome health and microbiota therapy in this setting with prebiotics, probiotics, vaginal seeding and fecal microbiota transplantation (FMT). RESULTS A healthy nonobese mother, vaginal delivery and strict breast feeding contribute to microbiome health in a newborn and young infant. With reduced microbiome diversity (dysbiosis) during pregnancy, cesarean delivery, prematurity, and formula feeding contribute to dysbiosis in the newborn. Microbiota therapy is an important approach to repair dysbiosis in pregnant women and their infants. Currently available probiotics can have favorable metabolic effects on mothers and infants, but these effects are variable. In research settings, reversal of infant dysbiosis can be achieved via vaginal seeding or FMT. Next generation probiotics in development should replace current probiotics and FMT. CONCLUSIONS The most critical phase of human microbiome development is in the first 2-3 years of life. Preventing and treating dysbiosis during pregnancy and early life can have a profound effect on an infant's later health.
Collapse
Affiliation(s)
- Herbert L. DuPont
- Division of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas, Houston, TX 77030, USA
- Department of Internal Medicine, University of Texas McGovern Medical School, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Kelsey Research Foundation, Houston, TX 77005, USA
| | | |
Collapse
|
16
|
Al KF, Allen L, Bedell S, Burton JP, de Vrijer B. Assessing the impact of pregnancy and birth factors on the maternal and infant microbiota. MICROBIOME RESEARCH REPORTS 2023; 2:29. [PMID: 38045923 PMCID: PMC10688794 DOI: 10.20517/mrr.2023.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/28/2023] [Accepted: 07/11/2023] [Indexed: 12/05/2023]
Abstract
Background: The microbiota acquired at birth is known to play an intimate role in later life health and disease and has been shown to be affected by the mode of birth. There has been recent interest in microbiota correction by maternal vaginal seeding in Cesarean section-born infants; however, the safety of this practice has been debated. The aim of this study was to assess how other factors, such as timing of sampling, maternal obesity, vaginal Group B Streptococcus colonization (GBS), and antibiotic exposure, affect the maternal and infant microbiota. Methods: Maternal vaginal and saliva samples were collected at three time periods: 35-37 weeks gestation (prenatal), within 24-36 hours after birth (birth), and at ~6 weeks postpartum. Infant saliva and stool samples were collected at ~6 weeks postpartum. 16S rRNA amplicon sequencing was utilized to assess the taxonomic and inferred functional compositions of the bacterial communities from both mothers and infants. Results: Samples from 36 mothers and 32 infants were obtained. Gestational age, breastfeeding, mode of birth, and gravidity were associated with taxonomic alterations in the infant samples, while obesity, antibiotic use, and GBS status were not. Maternal samples were predominantly affected by time, whereby significant alterations including increased microbial diversity were seen at birth and persisted to 6 weeks postpartum. Conclusion: This study provides information on the relationship between health and delivery factors and changes in vaginal and infant microbiota. These results may better direct clinicians and mothers in optimizing the infant microbiota towards health during infancy and later life.
Collapse
Affiliation(s)
- Kait F Al
- Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, Ontario N6A4V2, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario N6A3K7, Canada
| | - Laura Allen
- London Health Sciences Centre, London, Ontario N6A5W9, Canada
- Department of Obstetrics and Gynaecology, Division of Maternal Fetal Medicine, Western University, London, Ontario N6H5W9, Canada
| | - Samantha Bedell
- London Health Sciences Centre, London, Ontario N6A5W9, Canada
- Department of Obstetrics and Gynaecology, Division of Maternal Fetal Medicine, Western University, London, Ontario N6H5W9, Canada
| | - Jeremy P Burton
- Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, Ontario N6A4V2, Canada
- Department of Microbiology and Immunology, Western University, London, Ontario N6A3K7, Canada
- Division of Urology, Department of Surgery, Western University, London, Ontario N6A4V2, Canada
| | - Barbra de Vrijer
- London Health Sciences Centre, London, Ontario N6A5W9, Canada
- Department of Obstetrics and Gynaecology, Division of Maternal Fetal Medicine, Western University, London, Ontario N6H5W9, Canada
- Children’s Health Research Institute, London, Ontario N6C 4V3, Canada
| |
Collapse
|
17
|
Mueller NT, Differding MK, Sun H, Wang J, Levy S, Deopujari V, Appel LJ, Blaser MJ, Kundu T, Shah AA, Dominguez Bello MG, Hourigan SK. Maternal Bacterial Engraftment in Multiple Body Sites of Cesarean Section Born Neonates after Vaginal Seeding-a Randomized Controlled Trial. mBio 2023; 14:e0049123. [PMID: 37074174 PMCID: PMC10294643 DOI: 10.1128/mbio.00491-23] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/20/2023] Open
Abstract
Children delivered by elective, prelabor Cesarean section (C-section) are not exposed to the birth canal microbiota and, in relation to vaginally delivered children, show altered microbiota development. Perturbed microbial colonization during critical early-life windows of development alters metabolic and immune programming and is associated with an increased risk of immune and metabolic diseases. In nonrandomized studies, vaginal seeding of C-section-born neonates partially restores their microbiota colonization to that of their vaginally delivered counterparts, but without randomization, confounding factors cannot be excluded. In a double-blind, randomized, placebo-controlled trial, we determined the effect of vaginal seeding versus placebo seeding (control arm) on the skin and stool microbiota of elective, prelabor C-section-born neonates (n = 20) at 1 day and 1 month after birth. We also examined whether there were between-arm differences in engraftment of maternal microbes in the neonatal microbiota. In relation to the control arm, vaginal seeding increased mother-to-neonate microbiota transmission and caused compositional changes and a reduction in alpha diversity (Shannon Index) of the skin and stool microbiota. The neonatal skin and stool microbiota alpha diversity when maternal vaginal microbiota is provided is intriguing and highlights the need of larger randomized studies to determine the ecological mechanisms and effects of vaginal seeding on clinical outcomes. IMPORTANCE Children delivered by elective C-section are not exposed to the birth canal and show altered microbiota development. Impairing microbial colonization during early life alters metabolic and immune programming and is associated with an increased risk of immune and metabolic diseases. In a double-blind, randomized, placebo-controlled trial, we determined the effect of vaginal seeding on the skin and stool microbiota of elective C-section born neonates and found that vaginal seeding increased mother-to-neonate microbiota transmission and caused compositional changes and a reduction in the skin and stool microbiota diversity. The reduction of neonatal skin and stool microbiota diversity when maternal vaginal microbiota is provided is intriguing and highlights the need of larger randomized studies to determine the ecological mechanisms and effects of vaginal seeding on clinical outcomes.
Collapse
Affiliation(s)
- Noel T. Mueller
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, USA
| | - Moira K. Differding
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, USA
| | - Haipeng Sun
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey, USA
| | - Jincheng Wang
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey, USA
| | - Shira Levy
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Inova Children’s Hospital, Inova Health System, Falls Church, Virginia, USA
| | - Varsha Deopujari
- Inova Children’s Hospital, Inova Health System, Falls Church, Virginia, USA
| | - Lawrence J. Appel
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, USA
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, New Jersey, USA
| | - Tanima Kundu
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, New Jersey, USA
| | - Ankit A. Shah
- Inova Women’s Hospital, Inova Health System, Falls Church, Virginia, USA
| | - Maria Gloria Dominguez Bello
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey, USA
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Anthropology, Rutgers University, New Brunswick, New Jersey, USA
- Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey, USA
- Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada
| | - Suchitra K. Hourigan
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Inova Children’s Hospital, Inova Health System, Falls Church, Virginia, USA
| |
Collapse
|
18
|
Abstract
Early-life microbial colonization plays a key role in future health. In this issue of Cell Host & Microbe, Bogaert et al. unravel the complexities of mother-infant microbial seeding by examining multiple maternal and infant niches. Importantly, they describe "auxiliary" seeding pathways that may partially compensate when seeding patterns are perturbed.
Collapse
Affiliation(s)
- Suchitra K Hourigan
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Maria Gloria Dominguez-Bello
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901, USA; Department of Anthropology, Rutgers University, New Brunswick, NJ 08901, USA; Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ 08901, USA; Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada
| |
Collapse
|
19
|
Wu S, Ren L, Li J, Shen X, Zhou Q, Miao Z, Jia W, He F, Cheng R. Breastfeeding might partially contribute to gut microbiota construction and stabilization of propionate metabolism in cesarean-section infants. Eur J Nutr 2023; 62:615-631. [PMID: 36173468 DOI: 10.1007/s00394-022-03020-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE This study was aimed to determine how delivery mode and feeding pattern influence the infant's gut microbiota construction and the variation of fecal microbial metabolites from a birth cohort. METHODS Fecal samples collected from 61 full-term born Chinese infants at four time points: day 0, day 7, month 1, and month 3. Based on delivery mode (vaginal delivery [V] or cesarean section [C]) and feeding pattern (breastfeeding [B] or mixed feeding [M]), infants were divided into four groups, namely VB, CB, VM, and CM groups. The gut microbiota composition and bacterial diversity were assessed using 16S rRNA sequencing. Short-chain fatty acid (SCFA) concentrations were determined via gas chromatography-mass spectrometry (GC-MS). RESULTS The CM group had a significantly higher relative abundance of Firmicutes (day 0 and month 1), Enterococcaceae (month 3), and Enterococcus (month 3) than the VB group and a significantly higher abundance of Firmicutes (month 1) and Blautia (month 3) than the CB group. The VB and CB groups exhibited a stable SCFA variation and a significantly lower level of propionate compared with the VM and CM groups. All groups showed an intense transition of enterotypes within 1 month and became stable at 3 months. The correlation between SCFA and enterotypes showed a significant positive correlation between Bifidobacteriaceae and acetate in the CB group (day 7 and month 3) and a significant positive correlation between Clostridiaceae and butyrate in the CB and VB groups (day 7 and month 3), respectively. CONCLUSION These results indicated that C-section was associated with higher abundance of the phylum Firmicutes and family Enterococcaceae, and intense fluctuation of SCFA, at least propionate. And breastfeeding might partially contribute to gut microbiota construction and stabilization propionate metabolism in cesarean-section infants.
Collapse
Affiliation(s)
- Simou Wu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lei Ren
- Hebei Inatural Bio-Tech Co.,Ltd., Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Jinxing Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xi Shen
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qingqing Zhou
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhonghua Miao
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Wen Jia
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
20
|
Gutierrez MW, Mercer EM, Moossavi S, Laforest-Lapointe I, Reyna ME, Becker AB, Simons E, Mandhane PJ, Turvey SE, Moraes TJ, Sears MR, Subbarao P, Azad MB, Arrieta MC. Maturational patterns of the infant gut mycobiome are associated with early-life body mass index. Cell Rep Med 2023; 4:100928. [PMID: 36736319 PMCID: PMC9975311 DOI: 10.1016/j.xcrm.2023.100928] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 10/24/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023]
Abstract
Unlike the bacterial microbiome, the role of early-life gut fungi in host metabolism and childhood obesity development remains poorly characterized. To address this, we investigate the relationship between the gut mycobiome of 100 infants from the Canadian Healthy Infant Longitudinal Development (CHILD) Cohort Study and body mass index Z scores (BMIz) in the first 5 years of life. An increase in fungal richness during the first year of life is linked to parental and infant BMI. The relationship between richness pattern and early-life BMIz is modified by maternal BMI, maternal diet, infant antibiotic exposure, and bacterial beta diversity. Further, the abundances of Saccharomyces, Rhodotorula, and Malassezia are differentially associated with early-life BMIz. Using structural equation modeling, we determine that the mycobiome's contribution to BMIz is likely mediated by the bacterial microbiome. This demonstrates that mycobiome maturation and infant growth trajectories are distinctly linked, advocating for inclusion of fungi in larger pediatric microbiome studies.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Pediatrics, University of Calgary, Calgary, AB T2N 1N4, Canada; International Microbiome Center, University of Calgary, Calgary, AB T2N 1N4, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Emily M Mercer
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Pediatrics, University of Calgary, Calgary, AB T2N 1N4, Canada; International Microbiome Center, University of Calgary, Calgary, AB T2N 1N4, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Shirin Moossavi
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Pediatrics, University of Calgary, Calgary, AB T2N 1N4, Canada; International Microbiome Center, University of Calgary, Calgary, AB T2N 1N4, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Myrtha E Reyna
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Allan B Becker
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Elinor Simons
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Theo J Moraes
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Meghan B Azad
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Winnipeg, MB, Canada; Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Pediatrics, University of Calgary, Calgary, AB T2N 1N4, Canada; International Microbiome Center, University of Calgary, Calgary, AB T2N 1N4, Canada; Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
21
|
Natural Green Spaces, Sensitization to Allergens, and the Role of Gut Microbiota during Infancy. mSystems 2023; 8:e0119022. [PMID: 36790181 PMCID: PMC10134798 DOI: 10.1128/msystems.01190-22] [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: 02/16/2023] Open
Abstract
The environment plays an instrumental role in the developmental origins of health and disease. Protective features of the environment in the development of asthma and atopy have been insufficiently studied. We used data from the CHILD (Canadian Healthy Infant Longitudinal Development) Cohort Study to examine relationships between living near natural green spaces in early infancy in Edmonton, AB, Canada and the development of atopic sensitization at 1 year and 3 years of age in a cohort of 699 infants, and whether these associations were mediated by infant gut microbiota (measured using 16s V4 amplicon sequencing) at 4 months. The Urban Planning Land Vegetation Index (uPLVI) map of the City of Edmonton was used to assess infants' exposure to natural spaces based on their home postal codes, and atopic sensitization was assessed using skin prink testing (SPTs) for common food and inhalant allergens. Our findings suggest there is a protective effect of natural green space proximity on the development of multiple inhalant atopic sensitizations at 3 years (odds ratio = 0.28 [95% CI 0.09, 0.90]). This relationship was mediated by changes to Actinobacteria diversity in infant fecal samples taken at 4 months. We also found a positive association between nature proximity and sensitization to at least one food or inhaled allergen; this association was not mediated by gut microbiota. Together, these findings underscore the importance of promoting natural urban greenspace preservation to improve child health by reducing atopic disease susceptibility. IMPORTANCE Our findings highlight the importance of preserving natural green space in urban settings to prevent sensitization to environmental allergens and promote early-life gut microbiota pathways to this health benefit. These findings support a mediating role of gut microbiome compositions in health and disease susceptibility. This study used unique, accurate, and comprehensive methodology to classify natural space exposure via a high-resolution topographical map of foliage subtypes within the City of Edmonton limits. These methods are improvements from other methods previously used to classify natural space exposure, such as the normalized density vegetation index from satellite imagery, which is not able to distinguish anthropogenic from green space. The use of these methods and the associations found between natural green space exposure and atopic sensitization outcomes support their use in future studies. Our findings also provide many avenues for future research including longer term follow up of this cohort and investigation of a causal role of reduced Actinobacteria diversity on atopic sensitization development.
Collapse
|
22
|
Piva VM, De Grandis MC, Zuin IS, Angerilli V, Nappo F, Alfieri R, Ahcene Djaballah S, Murgioni S, Bergamo F, Fassan M, Valmasoni M, Lonardi S. ctDNA as promising tool for the assessment of minimal residual disease (MRD) and the need of an adjuvant treatment in gastroesophageal adenocarcinoma. Updates Surg 2023; 75:305-312. [PMID: 36272058 DOI: 10.1007/s13304-022-01379-2] [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: 03/29/2022] [Accepted: 09/12/2022] [Indexed: 01/24/2023]
Abstract
Gastroesophageal adenocarcinoma is a challenging disease due to its poor prognosis and the presence of few therapeutic options. For these reasons, it is mandatory to identify the subgroup of patients who are at high risk for relapse after curative-intention surgery. In the last years, liquid biopsy has aroused great interest in cancer treatment for its feasibility and the possibility to capture tumor heterogeneity in a real-time way. In postoperative setting, the interest is directed to the identification of Minimal Residual Disease (MRD), defined as isolated or small cluster of cancer cells that residues after curative-intention surgery, and are undetectable by conventional radiological and clinical exams. This review wants to summarize current evidence on the use of liquid biopsy in gastroesophageal cancer, focusing on the detection of ctDNA in the postoperative setting and its potential role as a guide for treatment decision.
Collapse
Affiliation(s)
- Vittoria Matilde Piva
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Maria Caterina De Grandis
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Irene Sole Zuin
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Surgery Clinic 1, Padova University Hospital, Padua, Italy
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Floriana Nappo
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Rita Alfieri
- Unit of Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Selma Ahcene Djaballah
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128, Padua, Italy
| | - Sabina Murgioni
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Francesca Bergamo
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Michele Valmasoni
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Surgery Clinic 1, Padova University Hospital, Padua, Italy
| | - Sara Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128, Padua, Italy.
| |
Collapse
|
23
|
Chen YY, Tun HM, Field CJ, Mandhane PJ, Moraes TJ, Simons E, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. Impact of Cesarean Delivery and Breastfeeding on Secretory Immunoglobulin A in the Infant Gut Is Mediated by Gut Microbiota and Metabolites. Metabolites 2023; 13:metabo13020148. [PMID: 36837767 PMCID: PMC9959734 DOI: 10.3390/metabo13020148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/19/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
How gut immunity in early life is shaped by birth in relation to delivery mode, intrapartum antibiotic prophylaxis (IAP) and labor remains undetermined. We aimed to address this gap with a study of secretory Immunoglobulin A (SIgA) in the infant gut that also tested SIgA-stimulating pathways mediated by gut microbiota and metabolites. Among 1017 Canadian full-term infants, gut microbiota of fecal samples collected at 3 and 12 months were profiled using 16S rRNA sequencing; C. difficile was quantified by qPCR; fecal metabolites and SIgA levels were measured by NMR and SIgA enzyme-linked immunosorbent assay, respectively. We assessed the putative causal relationships from birth events to gut microbiota and metabolites, and ultimately to SIgA, in statistical sequential mediation models, adjusted for maternal gravida status in 551 infants. As birth mode influences the ability to breastfeed, the statistical mediating role of breastfeeding status and milk metabolites was also evaluated. Relative to vaginal birth without maternal IAP, cesarean section (CS) after labor was associated with reduced infant gut SIgA levels at 3 months (6.27 vs. 4.85 mg/g feces, p < 0.05); this association was sequentially mediated through gut microbiota and metabolites of microbial or milk origin. Mediating gut microbiota included Enterobacteriaceae, C. difficile, and Streptococcus. The milk or microbial metabolites in CS-SIgA mediating pathways were galactose, fucose, GABA, choline, lactate, pyruvate and 1,2-propanediol. This cohort study documented the impact of birth on infant gut mucosal SIgA. It is the first to characterize gut microbe-metabolite mediated pathways for early-life SIgA maturation, pathways that require experimental verification.
Collapse
Affiliation(s)
- Yuan Yao Chen
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Hein M. Tun
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Piushkumar J. Mandhane
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Theo J. Moraes
- Department of Pediatrics and Physiology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3A 1S1, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, BC V6H 0B3, Canada
| | - Padmaja Subbarao
- Department of Pediatrics and Physiology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - James A. Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Anita L. Kozyrskyj
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
- Correspondence: ; Tel.: +1-780-248-5508
| |
Collapse
|
24
|
Liu Y, Li HT, Zhou SJ, Zhou HH, Xiong Y, Yang J, Zhou YB, Chen DJ, Liu JM. Effects of vaginal seeding on gut microbiota, body mass index, and allergy risks in infants born through cesarean delivery: a randomized clinical trial. Am J Obstet Gynecol MFM 2023; 5:100793. [PMID: 36334724 DOI: 10.1016/j.ajogmf.2022.100793] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Vaginal seeding-exposure of neonates to maternal vaginal fluids-has been proposed to improve the microbiota of infants born through cesarean delivery, but its impacts on the infants' subsequent health outcomes remain unclear. OBJECTIVE This study aimed to examine the impacts of vaginal seeding on gut microbiota, growth, and allergy risks in infants born through cesarean delivery. STUDY DESIGN This randomized controlled trial was conducted at Liuyang Maternal and Child Health Care Hospital in Hunan, China. We estimated that a minimum sample size of 106 was needed by assuming a standardized effect size of 0.6 for the primary outcomes, with a statistical power of 80%, a 2-sided type I error of 0.05, and an expected loss to follow-up rate of 15%. Finally, 120 singleton term pregnant women scheduled for cesarean delivery were enrolled from November 2018 to September 2019. Infant follow-up was completed in September 2021. The participants were randomized in a 1:1 ratio to the vaginal seeding group (n=60; infants were swabbed immediately after birth using gauze preincubated in maternal vagina) or the control group (n=60; routine standard care). The first set of primary outcomes was infant body mass index and body mass index z-scores at 6, 12, 18, and 24 months of age. The other primary outcome was the total allergy risk score at 18 months for 20 common allergens (each scored from 0-6 points). Characteristics of gut microbiota, overweight/obesity, and allergic diseases and symptoms were included as secondary outcomes. The main analyses were performed according to the modified intention-to-treat principle. RESULTS Of 120 infants, 117 were included in the analyses. Infant body mass index and body mass index z-scores did not significantly differ between the 2 groups at any of the 4 time points, with the largest difference in point estimates occurring at 6 months: the mean (standard deviation) body mass index was 17.5 (1.4) kg/m2 and 17.8 (1.8) kg/m2 in the vaginal seeding and control groups, respectively (mean difference, -0.31 kg/m2 [95% confidence interval, -0.91 to 0.28]; P=.30), and body mass index z-score was 0.2 (1.0) and 0.4 (1.1), respectively (mean difference, -0.20 [95% confidence interval, -0.58 to 0.18]; P=.31). The median total allergy risk score was 1.5 (interquartile range, 0.0-4.0) in the vaginal seeding group and 2.0 (interquartile range, 1.0-3.0) in the control group (median difference, 0.00 [95% confidence interval, -1.00 to 1.00]; P=.48). For infants from the vaginal seeding group, the relative abundance of genera Lactobacillus and Bacteroides in the gut microbiota was slightly yet nonsignificantly elevated at birth and 6 months, and the risk of overweight/obesity was lower at 6 months (0/57 vs 6/59; relative risk, 0.03 [95% confidence interval, 0.00-0.57]; P=.03) though not at subsequent time points. Other secondary outcomes did not differ between groups. No adverse events related to the intervention were reported. CONCLUSION For infants born through cesarean delivery, vaginal seeding has no significant impacts on the gut microbiota, growth, or allergy risks during the first 2 years of life.
Collapse
Affiliation(s)
- Yang Liu
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University Health Science Center; Haidian District, Beijing, China (Drs Y Liu, H Li, Y Zhou, and J Liu); Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center; Haidian District, Beijing, China (Drs Y Liu, H Li, Y Zhou, and J Liu)
| | - Hong-Tian Li
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University Health Science Center; Haidian District, Beijing, China (Drs Y Liu, H Li, Y Zhou, and J Liu); Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center; Haidian District, Beijing, China (Drs Y Liu, H Li, Y Zhou, and J Liu); Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Haidian District, Beijing, China (Drs H Li and J Liu).
| | - Shu-Jin Zhou
- Liuyang Maternal and Child Health Care Hospital, Changsha, Hunan, China (Drs S Zhou, H Zhou, and Y Xiong)
| | - Hui-Huang Zhou
- Liuyang Maternal and Child Health Care Hospital, Changsha, Hunan, China (Drs S Zhou, H Zhou, and Y Xiong)
| | - Ying Xiong
- Liuyang Maternal and Child Health Care Hospital, Changsha, Hunan, China (Drs S Zhou, H Zhou, and Y Xiong)
| | - Jing Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, China (Dr J Yang)
| | - Yu-Bo Zhou
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University Health Science Center; Haidian District, Beijing, China (Drs Y Liu, H Li, Y Zhou, and J Liu); Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center; Haidian District, Beijing, China (Drs Y Liu, H Li, Y Zhou, and J Liu)
| | - Dun-Jin Chen
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (Dr D Chen)
| | - Jian-Meng Liu
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University Health Science Center; Haidian District, Beijing, China (Drs Y Liu, H Li, Y Zhou, and J Liu); Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center; Haidian District, Beijing, China (Drs Y Liu, H Li, Y Zhou, and J Liu); Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Haidian District, Beijing, China (Drs H Li and J Liu).
| |
Collapse
|
25
|
Mani J, Levy S, Angelova A, Hazrati S, Fassnacht R, Subramanian P, Richards T, Niederhuber JE, Maxwell GL, Hourigan SK. Epidemiological and microbiome associations of Clostridioides difficile carriage in infancy and early childhood. Gut Microbes 2023; 15:2203969. [PMID: 37096914 PMCID: PMC10132246 DOI: 10.1080/19490976.2023.2203969] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
There has been an increase in the prevalence of Clostridioides difficile (C. diff) causing significant economic impact on the health care system. Although toxigenic C. diff carriage is recognized in infancy, there is limited data regarding its longitudinal trends, associated epidemiolocal risk factors and intestinal microbiome characteristics. The objectives of our longitudinal cohort study were to investigate temporal changes in the prevalence of toxigenic C.diff colonization in children up to 2 years, associated epidemiological and intestinal microbiome characteristics. Pregnant mothers were enrolled prenatally, and serial stool samples were collected from their children for 2 years. 2608 serial stool samples were collected from 817 children. 411/817 (50%) were males, and 738/817 (90%) were born full term. Toxigenic C.diff was detected in 7/569 (1%) of meconium samples, 116/624 (19%) of 2 m (month), 221/606 (37%) of 6 m, 227/574 (40%) of 12 m and 18/235 (8%) of 24 m samples. Infants receiving any breast milk at 6 m were less likely to be carriers at 2 m, 6 m and 12 m than those not receiving it. (p = 0.002 at 2 m, p < 0.0001 at 6 m, p = 0.022 at 12 m). There were no robust differences in the underlying alpha or beta diversity between those with and without toxigenic C. diff carriage at any timepoint, although small differences in the relative abundance of certain taxa were found. In this largest longitudinal cohort study to date, a high prevalence of toxigenic C. diff carrier state was noted. Toxigenic C. diff carrier state in children is most likely a transient component of the dynamic infant microbiome.
Collapse
Affiliation(s)
- Jyoti Mani
- Department of Pediatrics, Children’s National Medical Center, Washington, DC, USA
| | - Shira Levy
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Angelina Angelova
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Sahel Hazrati
- Women’s Service Line, Inova Health System, Falls Church, VA, USA
| | - Ryan Fassnacht
- Inova Children’s Hospital, Inova Health System, Falls Church, VA, USA
| | - Poorani Subramanian
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
- Inova Children’s Hospital, Inova Health System, Falls Church, VA, USA
| | - Tiana Richards
- Inova Children’s Hospital, Inova Health System, Falls Church, VA, USA
| | - John E. Niederhuber
- Inova Children’s Hospital, Inova Health System, Falls Church, VA, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Suchitra K. Hourigan
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
- Inova Children’s Hospital, Inova Health System, Falls Church, VA, USA
| |
Collapse
|
26
|
Bacorn M, Romero-Soto HN, Levy S, Chen Q, Hourigan SK. The Gut Microbiome of Children during the COVID-19 Pandemic. Microorganisms 2022; 10:microorganisms10122460. [PMID: 36557713 PMCID: PMC9783902 DOI: 10.3390/microorganisms10122460] [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: 11/19/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
The gut microbiome has been shown to play a critical role in maintaining a healthy state. Dysbiosis of the gut microbiome is involved in modulating disease severity and potentially contributes to long-term outcomes in adults with COVID-19. Due to children having a significantly lower risk of severe illness and limited sample availability, much less is known about the role of the gut microbiome in children with COVID-19. It is well recognized that the developing gut microbiome of children differs from that of adults, but it is unclear if this difference contributes to the different clinical presentations and complications. In this review, we discuss the current knowledge of the gut microbiome in children with COVID-19, with gut microbiome dysbiosis being found in pediatric COVID-19 but specific taxa change often differing from those described in adults. Additionally, we discuss possible mechanisms of how the gut microbiome may mediate the presentation and complications of COVID-19 in children and the potential role for microbial therapeutics.
Collapse
|
27
|
Uchida K, Iida K, Fujioka I, Hachimura S, Kaminuma O. Suppressive Effect of Lactococcus lactis subsp. cremoris YRC3780 on a Murine Model of Japanese Cedar Pollinosis. Pathogens 2022; 11:1347. [PMID: 36422599 PMCID: PMC9698324 DOI: 10.3390/pathogens11111347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/01/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2024] Open
Abstract
Accumulating evidence suggests that Lactococcus lactis subsp. cremoris YRC3780 isolated from kefir has the potential to alleviate allergic responses. Herein, we investigated the effect of YRC3780 on a murine model of Japanese cedar pollinosis (JCP). BALB/c mice immunized with cedar pollen extract (CPE) exhibited an increase in serum immunoglobulin E and developed nasal inflammatory responses including sneezing, nasal hyperresponsiveness, and nasal eosinophil accumulation upon intranasal allergen challenge. These responses were suppressed by the oral administration of YRC3780, although the effects on CPE-induced sneezing response and eosinophil infiltration were not statistically significant. Total fecal microbiota diversity was not affected by allergen immunization and challenge or by YRC3780 administration. However, the abundances of Bifidobacteriales, Veillonellaceae, Lactococcus, and Lactococcus lactis were larger and that of Bacteroides was smaller in YRC3780-treated mice compared with those in CPE-challenged and YRC3780-untreated mice. Our findings suggest the usefulness of YRC3780 for alleviating JCP.
Collapse
Affiliation(s)
- Kenji Uchida
- R&D Center, Yotsuba Milk Products Co., Ltd., Sapporo 061-1264, Japan
| | - Kenichi Iida
- R&D Center, Yotsuba Milk Products Co., Ltd., Sapporo 061-1264, Japan
| | - Ikumi Fujioka
- R&D Center, Yotsuba Milk Products Co., Ltd., Sapporo 061-1264, Japan
| | - Satoshi Hachimura
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Osamu Kaminuma
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
28
|
Matharu D, Ponsero AJ, Dikareva E, Korpela K, Kolho KL, de Vos WM, Salonen A. Bacteroides abundance drives birth mode dependent infant gut microbiota developmental trajectories. Front Microbiol 2022; 13:953475. [PMID: 36274732 PMCID: PMC9583133 DOI: 10.3389/fmicb.2022.953475] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background and aims Birth mode and other early life factors affect a newborn's microbial colonization with potential long-term health effects. Individual variations in early life gut microbiota development, especially their effects on the functional repertoire of microbiota, are still poorly characterized. This study aims to provide new insights into the gut microbiome developmental trajectories during the first year of life. Methods Our study comprised 78 term infants sampled at 3 weeks, 3 months, 6 months, and 12 months (n = 280 total samples), and their mothers were sampled in late pregnancy (n = 50). Fecal DNA was subjected to shotgun metagenomic sequencing. Infant samples were studied for taxonomic and functional maturation, and maternal microbiota was used as a reference. Hierarchical clustering on taxonomic profiles was used to identify the main microbiota developmental trajectories in the infants, and their associations with perinatal and postnatal factors were assessed. Results In line with previous studies, infant microbiota composition showed increased alpha diversity and decreased beta diversity by age, converging toward an adult-like profile. However, we did not observe an increase in functional alpha diversity, which was stable and comparable with the mother samples throughout all the sampling points. Using a de novo clustering approach, two main infant microbiota clusters driven by Bacteroidaceae and Clostridiaceae emerged at each time point. The clusters were associated with birth mode and their functions differed mainly in terms of biosynthetic and carbohydrate degradation pathways, some of which consistently differed between the clusters for all the time points. The longitudinal analysis indicated three main microbiota developmental trajectories, with the majority of the infants retaining their characteristic cluster until 1 year. As many as 40% of vaginally delivered infants were grouped with infants delivered by C-section due to their clear and persistent depletion in Bacteroides. Intrapartum antibiotics, any perinatal or postnatal factors, maternal microbiota composition, or other maternal factors did not explain the depletion in Bacteroides in the subset of vaginally born infants. Conclusion Our study provides an enhanced understanding of the compositional and functional early life gut microbiota trajectories, opening avenues for investigating elusive causes that influence non-typical microbiota development.
Collapse
Affiliation(s)
- Dollwin Matharu
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Alise J. Ponsero
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Biosystems Engineering and BIO5 Institute, University of Arizona, Tucson, AZ, United States
| | - Evgenia Dikareva
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kaija-Leena Kolho
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children's Hospital, Pediatric Research Center, University of Helsinki and HUS, Helsinki, Finland
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Willem M. de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
29
|
Mei H, Yang S, Peng A, Li R, Xiang F, Zheng H, Tan Y, Zhang Y, Zhou A, Zhang J, Xiao H. Development of the gut microbiota in healthy twins during the first 2 years of life and associations with body mass index z-score: Results from the Wuhan twin birth cohort study. Front Microbiol 2022; 13:891679. [PMID: 36060734 PMCID: PMC9433903 DOI: 10.3389/fmicb.2022.891679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
The gut microbiota undergoes rapid and vital changes to microbial community structure and the microbial-immune crosstalk during the first 3 years of life, which is thought to be involved in the pathobiology of later-life disease. Compared to single-born children, little is known about the gut microbiota of twins in early childhood. Based on the Wuhan Twin Birth Cohort study, 344 stool samples from 204 twin families were analyzed to investigate the difference in gut microbiota composition at 6, 12, and 24 months of age. Furthermore, this study evaluated the association between gut microbiota development curves and body mass index z-score (BMI_Z) curves at 6, 12, and 24 months of age. The predominant microbiota phyla identified in twins were Proteobacteria, Actinobacteriota, Firmicutes, Bacteroidota, and Verrucomicrobiota. The richness and diversity of gut microbiota increased from 6 to 24 months old (alpha diversity with p < 0.05). Beta diversity revealed 61 gut microbiota genera that were significantly different in relative abundance among the three age groups. Among the 61 gut microbiota genera, 30 distinct trajectory curves (DTCs) were generated by group-based trajectory models after log2 transformation of their relative abundance. Subsequently, Spearman correlation analysis revealed that only five gut microbiota DTC were correlated with the BMI_Z DTC. Therefore, we further examined the association between the five gut microbiota genera DTC and BMI_Z DTC using generalized estimation equation models. The results revealed a significant association between the DTC groups of Parabacteroides and that of BMI_Z (coefficient = 0.75, p = 0.04). The results of this study validated the hypothesis that the richness and diversity of gut microbiota developed with age in twins. Moreover, participants with a higher DTC of log2-transformed Parabacteroides had a higher BMI_Z DTC during the first 2 years of life. Further studies are needed to confirm the association between Parabacteroides and BMI_Z in other populations.
Collapse
Affiliation(s)
- Hong Mei
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Maternal and Child Health Care, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoping Yang
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - An’na Peng
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruizhen Li
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feiyan Xiang
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zheng
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Tan
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya Zhang
- Department of Maternal and Child Health Care, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ai’fen Zhou
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianduan Zhang
- Department of Maternal and Child Health Care, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Xiao
- Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Zheng J, Zhang L, Gao Y, Wu H, Zhang J. The dynamic effects of maternal high-calorie diet on glycolipid metabolism and gut microbiota from weaning to adulthood in offspring mice. Front Nutr 2022; 9:941969. [PMID: 35928844 PMCID: PMC9343994 DOI: 10.3389/fnut.2022.941969] [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: 05/12/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Dysbiosis of gut microbiota can contribute to the progression of diabetes and obesity. Previous studies have shown that maternal high-fat (HF) diet during the perinatal period can alter the microbiota and induce metabolic disorders at weaning. However, whether dysbiosis of gut microbiota and metabolism could be recovered by a normal diet after weaning and the dynamic changes of gut microbiota have not been fully studied. In this study, C57BL/6J female mice were fed with a normal chow (NC) or HF diet for 4 weeks preconception, during gestation, and until pup weaning. After weaning, male offspring were fed with an NC diet until 9 weeks of age. The microbiota of offspring at weaning and 9 weeks of age was collected for 16S rRNA gene amplicon sequencing. We found that dams fed with an HF diet showed glucose intolerance after lactation. Compared with the offspring from NC dams, the offspring from HF dams exhibited a higher body weight, hyperglycemia, glucose intolerance, hyperinsulinemia, hypercholesterolemia, and leptin resistance and lower adiponectin at weaning. Fecal analysis indicated altered microbiota composition between the offspring of the two groups. The decrease in favorable bacteria (such as norank f Bacteroidales S24-7 group) and increase in unfavorable bacteria (such as Lachnoclostridium and Desulfovibrio) were strongly associated with a disturbance of glucose and lipid metabolism. After 6 weeks of normal diet, no difference in body weight, glucose, and lipid profiles was observed between the offspring of the two groups. However, the microbiota composition of offspring in the HF group was still different from that in the NC group, and microbiota diversity was lower in offspring of the HF group. The abundance of Lactobacillus was lower in the offspring of the HF group. In conclusion, a maternal HF diet can induce metabolic homeostasis and gut microbiota disturbance in offspring at weaning. Gut microbiota dysbiosis can persist into adulthood in the offspring, which might have a role in the promotion of susceptibility to obesity and diabetes in the later life of the offspring.
Collapse
Affiliation(s)
- Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Honghua Wu
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| |
Collapse
|
31
|
Volker E, Tessier C, Rodriguez N, Yager J, Kozyrskyj A. Pathways of atopic disease and neurodevelopmental impairment: assessing the evidence for infant antibiotics. Expert Rev Clin Immunol 2022; 18:901-922. [PMID: 35822921 DOI: 10.1080/1744666x.2022.2101450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Epidemiologic studies are starting to report associations between antibiotic use in early life and neurodevelopmental disorders. Through mechanisms within the gut microbiota-brain axis, indeed, it is plausible that infant antibiotic treatment plays a role in the development of atopic disease and neurodevelopmental disorders. AREAS COVERED This narrative review summarizes and interprets published evidence on infant antibiotic use in future outcomes of atopic disease, and neurodevelopmental delay and disorders, including attention deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). To this end, we critically assess study bias from 2 main confounding factors, maternal/infant infection and infant feeding status. We also discuss common mechanisms that link atopy and neurodevelopment, and propose hypotheses related to immune activation and the gut microbiome. EXPERT OPINION Atopic disease and neurodevelopmental disorders share many risk factors and biological pathways. Infant antibiotic use has been linked to both disorders and is likely a marker for prenatal or infant infection. The mediating role of breastfeeding can also not be discounted. The exploration of causal pathways along the gut-brain axis leading towards neurodevelopmental impairment is evolving and of future interest.
Collapse
|
32
|
Horne RG, Freedman SB, Johnson-Henry KC, Pang XL, Lee BE, Farion KJ, Gouin S, Schuh S, Poonai N, Hurley KF, Finkelstein Y, Xie J, Williamson-Urquhart S, Chui L, Rossi L, Surette MG, Sherman PM. Intestinal Microbial Composition of Children in a Randomized Controlled Trial of Probiotics to Treat Acute Gastroenteritis. Front Cell Infect Microbiol 2022; 12:883163. [PMID: 35774405 PMCID: PMC9238408 DOI: 10.3389/fcimb.2022.883163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
Compositional analysis of the intestinal microbiome in pre-schoolers is understudied. Effects of probiotics on the gut microbiota were evaluated in children under 4-years-old presenting to an emergency department with acute gastroenteritis. Included were 70 study participants (n=32 placebo, n=38 probiotics) with stool specimens at baseline (day 0), day 5, and after a washout period (day 28). Microbiota composition and deduced functions were profiled using 16S ribosomal RNA sequencing and predictive metagenomics, respectively. Probiotics were detected at day 5 of administration but otherwise had no discernable effects, whereas detection of bacterial infection (P<0.001) and participant age (P<0.001) had the largest effects on microbiota composition, microbial diversity, and deduced bacterial functions. Participants under 1 year had lower bacterial diversity than older aged pre-schoolers; compositional changes of individual bacterial taxa were associated with maturation of the gut microbiota. Advances in age were associated with differences in gut microbiota composition and deduced microbial functions, which have the potential to impact health later in life.
Collapse
Affiliation(s)
- Rachael G. Horne
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Stephen B. Freedman
- Sections of Pediatric Emergency Medicine and Gastroenterology, Department of Pediatrics, Alberta Children’s Hospital, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Xiao-Li Pang
- Alberta Precision Laboratories – Public Health Laboratory (ProvLab), Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Bonita E. Lee
- Women and Children’s Research Institute, Stollery Children’s Hospital, University of Alberta, Edmonton, AB, Canada
| | - Ken J. Farion
- Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
| | - Serge Gouin
- Departments of Emergency Medicine and Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Suzanne Schuh
- Division of Emergency Medicine, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Naveen Poonai
- Division of Pediatric Emergency Medicine, London Children’s Hospital Health Science Centre, Department of Pediatrics, Western University, London, ON, Canada
| | - Katrina F. Hurley
- Pediatric Emergency Medicine, Izaak Walton Killam (IWK) Children’s Hospital, Dalhousie University, Halifax, NS, Canada
| | - Yaron Finkelstein
- Division of Emergency Medicine, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jianling Xie
- Pediatric Emergency Medicine, Izaak Walton Killam (IWK) Children’s Hospital, Dalhousie University, Halifax, NS, Canada
| | - Sarah Williamson-Urquhart
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Alberta Children’s Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Linda Chui
- Alberta Precision Laboratories – Public Health Laboratory (ProvLab), Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Laura Rossi
- Department of Biochemistry and Biomedical Sciences, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Michael G. Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Philip M. Sherman
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- *Correspondence: Philip M. Sherman,
| |
Collapse
|
33
|
Tao W, Cao W, Yu B, Chen H, Gong R, Luorong Q, Luo J, Yao L, Zhang D. Hawk tea prevents high-fat diet-induced obesity in mice by activating the AMPK/ACC/SREBP1c signaling pathways and regulating the gut microbiota. Food Funct 2022; 13:6056-6071. [PMID: 35437540 DOI: 10.1039/d1fo04260b] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Scope: Hawk tea, a non-Camellia tea, is an ancient tea drink from southwest China and has been proven to exhibit significant hypoglycaemic and lipid-lowering effects. The aim of this study was to evaluate whether Hawk tea extract (HTE) can improve obesity induced by a high-fat diet (HFD) in a mouse model and to determine whether its anti-obesity effects are related to improvements in lipid metabolism and the gut microbiota. Methods and results: We tested the ability of HTE to prevent obesity and regulate gut microbiota in C57BL/6J mice fed with a HFD. We found that HTE significantly reduced body weight, fat deposition, serum triglyceride (TG), total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels, and significantly increased serum levels of high-density lipoprotein cholesterol (HDL-C) induced by HFD. HTE also increased the levels of AMPK and ACC phosphorylation, up-regulated the expression of CPT-1, and downregulated the expression of SREBP1c and FAS. In addition, the administration of HTE significantly altered the composition of the gut microbiota, reduced the ratio of Firmicutes to Bacteroidetes, increased the relative abundance of Akkermansia muciniphila, Bacteroides-vulgatus, and Faecalibaculum_rodentium, and decreased the relative abundance of Desulfovibrionaceae and Lachnospiraceae. Conclusions: Collectively, our data demonstrate that HTE can prevent HFD-induced obesity by regulating the AMPK/ACC/SREBP1c signaling pathways and the gut microbiota.
Collapse
Affiliation(s)
- Wei Tao
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Weiguo Cao
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.,The Lab of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Bao Yu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Huan Chen
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ruixue Gong
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Quji Luorong
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Juan Luo
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ling Yao
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Dan Zhang
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
34
|
Hourigan SK, Dominguez-Bello MG, Mueller NT. Can maternal-child microbial seeding interventions improve the health of infants delivered by Cesarean section? Cell Host Microbe 2022; 30:607-611. [PMID: 35550663 DOI: 10.1016/j.chom.2022.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Maternal-child microbial seeding interventions expose Cesarean-section (C-section)-delivered infants to the maternal microbiome they bypass during Cesarean delivery. It is theorized such interventions restore the microbiome and normalize immune development to reduce the occurrence of C-section-associated inflammatory conditions. Here we discuss the rationale, evidence, and controversies surrounding such interventions.
Collapse
Affiliation(s)
- Suchitra K Hourigan
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Maria Gloria Dominguez-Bello
- Department of Biochemistry and Microbiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21218, USA
| |
Collapse
|
35
|
Saturio S, Nogacka AM, Alvarado-Jasso GM, Salazar N, de los Reyes-Gavilán CG, Gueimonde M, Arboleya S. Role of Bifidobacteria on Infant Health. Microorganisms 2021; 9:2415. [PMID: 34946017 PMCID: PMC8708449 DOI: 10.3390/microorganisms9122415] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/19/2022] Open
Abstract
Bifidobacteria are among the predominant microorganisms during infancy, being a dominant microbial group in the healthy breastfed infant and playing a crucial role in newborns and infant development. Not only the levels of the Bifidobacterium genus but also the profile and quantity of the different bifidobacterial species have been demonstrated to be of relevance to infant health. Although no definitive proof is available on the causal association, reduced levels of bifidobacteria are perhaps the most frequently observed alteration of the intestinal microbiota in infant diseases. Moreover, Bifidobacterium strains have been extensively studied by their probiotic attributes. This review compiles the available information about bifidobacterial composition and function since the beginning of life, describing different perinatal factors affecting them, and their implications on different health alterations in infancy. In addition, this review gathers exhaustive information about pre-clinical and clinical studies with Bifidobacterium strains as probiotics in neonates.
Collapse
Affiliation(s)
- Silvia Saturio
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Guadalupe M. Alvarado-Jasso
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
36
|
Schlender J, Behrens F, McParland V, Müller D, Wilck N, Bartolomaeus H, Holle J. Bacterial metabolites and cardiovascular risk in children with chronic kidney disease. Mol Cell Pediatr 2021; 8:17. [PMID: 34677718 PMCID: PMC8536815 DOI: 10.1186/s40348-021-00126-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular complications are the major cause of the marked morbidity and mortality associated with chronic kidney disease (CKD). The classical cardiovascular risk factors such as diabetes and hypertension undoubtedly play a role in the development of cardiovascular disease (CVD) in adult CKD patients; however, CVD is just as prominent in children with CKD who do not have these risk factors. Hence, the CKD-specific pathophysiology of CVD remains incompletely understood. In light of this, studying children with CKD presents a unique opportunity to analyze CKD-associated mechanisms of CVD more specifically and could help to unveil novel therapeutic targets. Here, we comprehensively review the interaction of the human gut microbiome and the microbial metabolism of nutrients with host immunity and cardiovascular end-organ damage. The human gut microbiome is evolutionary conditioned and modified throughout life by endogenous factors as well as environmental factors. Chronic diseases, such as CKD, cause significant disruption to the composition and function of the gut microbiome and lead to disease-associated dysbiosis. This dysbiosis and the accompanying loss of biochemical homeostasis in the epithelial cells of the colon can be the result of poor diet (e.g., low-fiber intake), medications, and underlying disease. As a result of dysbiosis, bacteria promoting proteolytic fermentation increase and those for saccharolytic fermentation decrease and the integrity of the gut barrier is perturbed (leaky gut). These changes disrupt local metabolite homeostasis in the gut and decrease productions of the beneficial short-chain fatty acids (SCFAs). Moreover, the enhanced proteolytic fermentation generates unhealthy levels of microbially derived toxic metabolites, which further accumulate in the systemic circulation as a consequence of impaired kidney function. We describe possible mechanisms involved in the increased systemic inflammation in CKD that is associated with the combined effect of SCFA deficiency and accumulation of uremic toxins. In the future, a more comprehensive and mechanistic understanding of the gut–kidney–heart interaction, mediated largely by immune dysregulation and inflammation, might allow us to target the gut microbiome more specifically in order to attenuate CKD-associated comorbidities.
Collapse
Affiliation(s)
- Julia Schlender
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, 13353, Berlin, Germany.,Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), 13125, Berlin, Germany
| | - Felix Behrens
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, 13353, Berlin, Germany.,Charité - Universitätsmedizin Berlin and Berlin Institute of Health, 10117, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, 13316, Berlin, Germany.,Institute of Physiology, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Victoria McParland
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), 13125, Berlin, Germany
| | - Dominik Müller
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, 13353, Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), 13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, 13316, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Internal Intensive Care Medicine, 10117, Berlin, Germany
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), 13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, 13316, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Internal Intensive Care Medicine, 10117, Berlin, Germany
| | - Johannes Holle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, 13353, Berlin, Germany. .,Experimental and Clinical Research Center (ECRC), a cooperation of Charité - Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), 13125, Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Berlin, 13316, Berlin, Germany.
| |
Collapse
|
37
|
Ríos-Covian D, Langella P, Martín R. From Short- to Long-Term Effects of C-Section Delivery on Microbiome Establishment and Host Health. Microorganisms 2021; 9:microorganisms9102122. [PMID: 34683443 PMCID: PMC8537978 DOI: 10.3390/microorganisms9102122] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 12/02/2022] Open
Abstract
The establishment of gut microbiota has been proven to be impacted by several factors during pregnancy, delivery, and neonate periods. The body of evidence describing C-section delivery (CSD) as one of the most disruptive events during early life has expanded in recent years, concluding that CSD results in a drastic change in microbiota establishment patterns. When comparing the gut microbiota composition of CSD babies with vaginally delivered (VD) babies, the former show a microbiome that closely resembles that found in the environment and the mother’s skin, while VD babies show a microbiome more similar to the vaginal microbiome. Although these alterations of normal gut microbiota establishment tend to disappear during the first months of life, they still affect host health in the mid–long term since CSD has been correlated with a higher risk of early life infections and non-transmissible diseases, such as inflammatory diseases, allergies, and metabolic diseases. In recent years, this phenomenon has also been studied in other mammals, shedding light on the mechanisms involved in the effects of a CSD on host health. In addition, strategies to revert the disruptions in gut microbiomes caused by a CSD are currently in the process of development and evaluation. In this review, we discuss the recent advances in CSD research, from the alteration of gut microbiota establishment to the possible effects on host health during early life and development.
Collapse
|
38
|
Pagano F, Conti MG, Boscarino G, Pannucci C, Dito L, Regoli D, Di Chiara M, Battaglia G, Prota R, Cinicola B, Zicari AM, Aloi M, Oliva S, Terrin G. Atopic Manifestations in Children Born Preterm: A Long-Term Observational Study. CHILDREN-BASEL 2021; 8:children8100843. [PMID: 34682108 PMCID: PMC8534898 DOI: 10.3390/children8100843] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 08/24/2021] [Accepted: 09/22/2021] [Indexed: 01/01/2023]
Abstract
(1) Background: Preterm birth exposes the infant to the known risk factors for atopic diseases. We aimed to study the neonatal risk factors and to describe the clinical manifestations of atopy, including the march of symptoms, in a cohort of preschool children born preterm. (2) Methods: We enrolled neonates with gestational age < 32 weeks or birth weight < 1500 g. We classified patients in cases and controls according to the presence of at least one atopic manifestation. (3) Results: We observed 72 cases and 93 controls. Multivariate models showed that the administration of more than one cycle of antibiotics (B 0.902, p = 0.026) and gestational diabetes (B 1.207, p = 0.035) influence the risk of atopy in babies born preterm. In addition, risk of atopic dermatitis was influenced by gestational age < 29 weeks (B -1.710, p = 0.025) and gestational diabetes (B 1.275, p = 0.027). The risk of wheeze was associated with familiarity for asthma (B 1.392, p = 0.022) and the administration of more than one cycle of antibiotics (B 0.969, p = 0.025). We observed a significant reduction in the rate of atopic manifestation after 2 years of life (33.9% vs. 23.8%, p < 0.05). (4) Conclusions: Modifiable (gestational diabetes, antibiotics use) and unmodifiable (familiarity for asthma) conditions influence the risk of atopy in babies born preterm. Extreme prematurity reduces the risk of atopic dermatitis. Preterm babies showed a peculiar atopic march.
Collapse
Affiliation(s)
- Federica Pagano
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Maria Giulia Conti
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Giovanni Boscarino
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Chiara Pannucci
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Lucia Dito
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Daniela Regoli
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Maria Di Chiara
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Giuseppe Battaglia
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Rita Prota
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Bianca Cinicola
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Anna Maria Zicari
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Marina Aloi
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Salvatore Oliva
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
| | - Gianluca Terrin
- Department of Maternal and Child Health, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy; (F.P.); (M.G.C.); (G.B.); (C.P.); (L.D.); (D.R.); (M.D.C.); (G.B.); (R.P.); (B.C.); (A.M.Z.); (M.A.); (S.O.)
- Correspondence: ; Tel.: +39-064-997-2536
| |
Collapse
|
39
|
Chen YY, Zhao X, Moeder W, Tun HM, Simons E, Mandhane PJ, Moraes TJ, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. Impact of Maternal Intrapartum Antibiotics, and Caesarean Section with and without Labour on Bifidobacterium and Other Infant Gut Microbiota. Microorganisms 2021; 9:microorganisms9091847. [PMID: 34576741 PMCID: PMC8467529 DOI: 10.3390/microorganisms9091847] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022] Open
Abstract
Background and Aims: Few studies consider the joint effect of multiple factors related to birth, delivery mode, intrapartum antibiotic prophylaxis and the onset of labour, on the abundance of Bifidobacterium and the quantity of this genus and its species Bifidobacterium longum subsp. infantis in the infant gut microbiota. We implemented such a study. Methods: Among 1654 Canadian full-term infants, the gut microbiota of faecal samples collected at 3 months were profiled by 16S rRNA sequencing; the genus Bifidobacterium and Bifidobacterium longum subsp. infantis were quantified by qPCR. Associations between Bifidobacterium and other gut microbiota were examined by Spearman’s rank correlation. Results: Following vaginal birth, maternal IAP exposure was associated with reduced absolute quantities of bifidobacteria among vaginally delivered infants (6.80 vs. 7.14 log10 (gene-copies/g faeces), p < 0.05), as well as their lowered abundance relative to other gut microbiota. IAP differences in infant gut bifidobacterial quantity were independent of maternal pre-pregnancy body-mass-index (BMI), and remarkably, they were limited to breastfed infants. Pre-pregnancy BMI adjustment revealed negative associations between absolute quantities of bifidobacteria and CS with or without labour in non-breastfed infants, and CS with labour in exclusively breastfed infants. Significant correlations between Bifidobacterium abundance and other microbial taxa were observed. Conclusions: This study documented the impact of the birth mode and feeding status on the abundance of gut Bifidobacterium, and pointed to the important ecological role of the genus Bifidobacterium in gut microbiota due to its strong interaction with other gut microbiota in early infancy.
Collapse
Affiliation(s)
- Yuan Yao Chen
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada; (Y.Y.C.); (X.Z.); (H.M.T.); (P.J.M.)
| | - Xin Zhao
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada; (Y.Y.C.); (X.Z.); (H.M.T.); (P.J.M.)
| | - Wolfgang Moeder
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 1R4, Canada; (W.M.); (J.A.S.)
| | - Hein M. Tun
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada; (Y.Y.C.); (X.Z.); (H.M.T.); (P.J.M.)
- HKU-Pasteur Research Pole, School of Public Health, University of Hong Kong, Hong Kong SAR 999077, China
| | - Elinor Simons
- Department of Pediatrics and Child Health, Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Piushkumar J. Mandhane
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada; (Y.Y.C.); (X.Z.); (H.M.T.); (P.J.M.)
| | - Theo J. Moraes
- Department of Pediatrics and Physiology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; (T.J.M.); (P.S.)
| | - Stuart E. Turvey
- Department of Pediatrics, Child and Family Research Institute, BC Children’s Hospital, University of British Columbia, Vancouver, BC V5Z 4H4, Canada;
| | - Padmaja Subbarao
- Department of Pediatrics and Physiology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; (T.J.M.); (P.S.)
| | - James A. Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 1R4, Canada; (W.M.); (J.A.S.)
| | - Anita L. Kozyrskyj
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 1C9, Canada; (Y.Y.C.); (X.Z.); (H.M.T.); (P.J.M.)
- Correspondence: Anita Kozyrskyj
| |
Collapse
|
40
|
Lewis CR, Bonham KS, McCann SH, Volpe AR, D’Sa V, Naymik M, De Both MD, Huentelman MJ, Lemery-Chalfant K, Highlander SK, Deoni SCL, Klepac-Ceraj V. Family SES Is Associated with the Gut Microbiome in Infants and Children. Microorganisms 2021; 9:1608. [PMID: 34442687 PMCID: PMC8398307 DOI: 10.3390/microorganisms9081608] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/12/2021] [Accepted: 07/21/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND While early life exposures such as mode of birth, breastfeeding, and antibiotic use are established regulators of microbiome composition in early childhood, recent research suggests that the social environment may also exert influence. Two recent studies in adults demonstrated associations between socioeconomic factors and microbiome composition. This study expands on this prior work by examining the association between family socioeconomic status (SES) and host genetics with microbiome composition in infants and children. METHODS Family SES was used to predict a latent variable representing six genera abundances generated from whole-genome shotgun sequencing. A polygenic score derived from a microbiome genome-wide association study was included to control for potential genetic associations. Associations between family SES and microbiome diversity were assessed. RESULTS Anaerostipes, Bacteroides, Eubacterium, Faecalibacterium, and Lachnospiraceae spp. significantly loaded onto a latent factor, which was significantly predicted by SES (p < 0.05) but not the polygenic score (p > 0.05). Our results indicate that SES did not predict alpha diversity but did predict beta diversity (p < 0.001). CONCLUSIONS Our results demonstrate that modifiable environmental factors influence gut microbiome composition at an early age. These results are important as our understanding of gut microbiome influences on health continue to expand.
Collapse
Affiliation(s)
- Candace R. Lewis
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (M.N.); (M.D.D.B.); (M.J.H.)
| | - Kevin S. Bonham
- Department of Biological Sciences, Wellesley College, Wellesley, MA 02481, USA; (K.S.B.); (S.H.M.)
| | - Shelley Hoeft McCann
- Department of Biological Sciences, Wellesley College, Wellesley, MA 02481, USA; (K.S.B.); (S.H.M.)
| | - Alexandra R. Volpe
- Advanced Baby Imaging Lab, Hasbro Children’s Hospital, Rhode Island Hospital, Providence, RI 02903, USA; (A.R.V.); (V.D.); (S.C.L.D.)
| | - Viren D’Sa
- Advanced Baby Imaging Lab, Hasbro Children’s Hospital, Rhode Island Hospital, Providence, RI 02903, USA; (A.R.V.); (V.D.); (S.C.L.D.)
- Department of Pediatrics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Marcus Naymik
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (M.N.); (M.D.D.B.); (M.J.H.)
| | - Matt D. De Both
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (M.N.); (M.D.D.B.); (M.J.H.)
| | - Matthew J. Huentelman
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (M.N.); (M.D.D.B.); (M.J.H.)
| | | | - Sarah K. Highlander
- Pathogen and Microbiome Division, Translational Genomics Research Institute North (TGen), Flagstaff, AZ 86005, USA;
| | - Sean C. L. Deoni
- Advanced Baby Imaging Lab, Hasbro Children’s Hospital, Rhode Island Hospital, Providence, RI 02903, USA; (A.R.V.); (V.D.); (S.C.L.D.)
- Department of Pediatrics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- MNCH D&T, Bill and Melinda Gates Foundation, Seattle, WA 98109, USA
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA 02481, USA; (K.S.B.); (S.H.M.)
| |
Collapse
|