1
|
Xu R, Yu Y, Chen T. Exploring the dark side of probiotics to pursue light: Intrinsic and extrinsic risks to be opportunistic pathogens. Curr Res Food Sci 2025; 10:101044. [PMID: 40235735 PMCID: PMC11999689 DOI: 10.1016/j.crfs.2025.101044] [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: 01/26/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Probiotics, live microorganisms with multiple health benefits, have gained popularity for their roles in maintaining daily health and treating a variety of diseases. However, they have the potential to be opportunistic pathogens in some conditions. This review delves into the intrinsic and extrinsic risks associated with probiotics. Intrinsic risks involve the production of harmful substances, such as toxins and invasive factors, biofilm formation, bacteria emboli, antibiotic resistance with relevant genetic materials, genetic plasticity, and metabolic issues, while extrinsic risks include problems in regulatory oversight and public awareness, host health status and appropriately administration. It emphasizes the need for a balanced view of their therapeutic benefits and potential hazards, advocating for further research to understand the complex interactions between probiotics and the human microbiome, to optimize the safety and efficacy of probiotics.
Collapse
Affiliation(s)
- Ruiyan Xu
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yifeng Yu
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tingtao Chen
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
2
|
Yang L, Hui Y, Sangild PT, Kot WP, Aunsholt L, Zachariassen G, Jiang PP, Nielsen DS. Gut microbiota development in very preterm infants following fortification of human milk. mSystems 2025; 10:e0091624. [PMID: 39982063 PMCID: PMC11915873 DOI: 10.1128/msystems.00916-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
Very preterm infants (VPIs) are born with an immature gut and predisposed to gut microbiota dysbiosis-related diseases, for example, necrotizing enterocolitis. Although fortification of human milk is required for these infants, the optimal fortifier remains uncertain. Bovine colostrum (BC), rich in protein and bioactive components, could serve as an alternative to conventional fortifiers (CF). The gut microbiota (GM) of 225 VPIs fed human milk fortified with either BC or CF (FortiColos study, NCT03537365) was profiled by 16S rRNA gene amplicon sequencing of fecal samples collected before, and after 1 and 2 weeks of fortification. Birth mode exhibited transient effects on the microbial community structure shortly after birth, with cesarean section-born VPIs dominated by Firmicutes, whereas vaginally born VPIs were dominated by Proteobacteria. This birth mode-derived difference diminished with age and disappeared around 1 month after birth. Fortifier type affected the microbial community structure to a modest extent, but no specific taxa significantly differed between the BC and CF groups. Fecal pH, increased by BC, was positively correlated with Staphylococcus and Corynebacterium and negatively with Bifidobacterium abundance. Change in the relative abundance of Staphylococcus was negatively correlated with body weight gain. Collectively, fortification of human milk with BC or CF does influence the GM of VPIs but only to a modest extent during early life. Birth mode appears to have a significant, but temporary influence on the GM during this period.IMPORTANCEEarly life is a key period for gut microbiota (GM) establishment, where enteral feeding plays a significant role. This is also the case for infants born preterm, who, due to their immature gut, are at a high risk of developing GM dysbiosis-related diseases. Human milk is the optimal feed for preterm infants, but it requires fortification to reach adequate levels of especially protein. Only a few studies have investigated the impact of fortifiers on GM development in preterm infants. Here, we demonstrate that two different bovine milk-based fortifiers, bovine colostrum and a conventional fortifier based on mature bovine milk, exhibit limited effects on the microbial community structure of very preterm infants. These findings suggest that although great care in terms of optimally maturing the preterm infant GM should be taken, the choice of fortifier only has limited impact. In clinical practice, the choice of fortifier can thus be fully focussed on optimizing preterm infant nutrition.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT03537365.
Collapse
Affiliation(s)
- Lin Yang
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Yan Hui
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Per Torp Sangild
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Neonatology, Hans Christian Andersen Children’s Hospital, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Witold Piotr Kot
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lise Aunsholt
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gitte Zachariassen
- Department of Neonatology, Hans Christian Andersen Children’s Hospital, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Ping-Ping Jiang
- Section for Comparative Paediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | |
Collapse
|
3
|
Wang H, Chen D, Li H, Fu C, Fang L, Wang R, Xu J. Bifidobacterium regulates premature infant gut metabolites, reducing serum inflammatory factors: a randomised controlled trial. Pediatr Res 2025; 97:1171-1182. [PMID: 39271904 DOI: 10.1038/s41390-024-03552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/17/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Analyse the effects of Bifidobacterium BB-12 on intestinal metabolites and serum inflammatory factors in premature infants. METHODS 71 premature infants at gestational age of ≤32 weeks were randomly divided into the probiotic (n = 36) and control (n = 35) groups. Faecal and blood samples were collected from the two groups of premature infants at the 2nd and 4th week of life for intestinal metabolite detection and assessment of the level of the serum inflammatory markers TLR4, NF- κ B, IL-1β, and TNF- α. RESULTS Compared to the control group, the probiotic group contained more amino acids, these elements were enriched on multiple amino acid metabolic pathways, and the probiotic group showed significantly lower levels of the serum inflammatory markers TLR4, NF-κB, IL-1β, and TNF-α. Finally, the probiotic group showed a lower incidence of feeding intolerance. CONCLUSIONS The administration of Bifidobacterium BB-12 is associated with increasing the levels of glutamine, glutamic acid, and kynurenine in the gut of premature infants, and associated with reducing the levels of TLR4 and NF-κB in the serum, further decreasing the secretion of the pro-inflammatory factors IL-1β and TNF-α, and alleviating systemic inflammatory reactions, thereby reducing the incidence of feeding intolerance. IMPACT 1. The use of Bifidobacterium BB-12 in premature infants can increase the levels of amino acids in the intestine. 2. Increases in Bifidobacterium BB-12 may decrease the serum levels of TLR4, NF-κB, IL-1β, and TNF-α. 3. Kynurenine may improve the prognosis of preterm infants by reducing inflammation. 4. Bifidobacterium BB-12 may improve the feeding tolerance of premature infants, thus reducing the incidence of feeding intolerance.
Collapse
Affiliation(s)
- He Wang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Dongmei Chen
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Huamei Li
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Chunyan Fu
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Lingyu Fang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Ruiquan Wang
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China
| | - Jinglin Xu
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China.
- Department of Neonatology, Quanzhou Maternity and Children's Hospital, Quanzhou, Fujian Province, China.
| |
Collapse
|
4
|
Wasana WP, Waterland M, Everett DW, Thum C. Functional Significance of Probiotic Bacterial Interactions with Milk Fat Globules in a Human Host. Microorganisms 2025; 13:223. [PMID: 40005590 PMCID: PMC11857118 DOI: 10.3390/microorganisms13020223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
Dairy products often serve as matrices for delivering probiotic bacteria to humans through the diet; however, little is known about the impact of milk fat globules on the growth and survival of probiotic microorganisms. This review discusses current knowledge on the structure and functionality of the milk fat globule membrane (MFGM) and the structural components contributing to the mechanisms of interactions with probiotic bacteria. We analyzed studies published between 2001 and 2025 with reference to earlier foundational research on probiotics and MFGM structure to explore the functional significance of MFGM-probiotic interactions. Recent research indicates that the effects of MFGM interaction with bacteria are species-specific and may influence probiotic activity in the host, including enhancing probiotic viability during intestinal transit and modulating probiotic colonization. In general, research findings suggest that the MFGM holds potential for use as a probiotic carrier to the gut with beneficial health consequences.
Collapse
Affiliation(s)
- Withanage Prasadini Wasana
- Food Function and Physiology Team, AgResearch, Palmerston North 4410, New Zealand;
- Riddet Institute, Massey University, Palmerston North 4410, New Zealand;
| | - Mark Waterland
- School of Food Technology and Natural Sciences, Massey University, Palmerston North 4410, New Zealand;
| | - David W. Everett
- Riddet Institute, Massey University, Palmerston North 4410, New Zealand;
- School of Food Technology and Natural Sciences, Massey University, Palmerston North 4410, New Zealand;
| | - Caroline Thum
- Food Function and Physiology Team, AgResearch, Palmerston North 4410, New Zealand;
- Riddet Institute, Massey University, Palmerston North 4410, New Zealand;
| |
Collapse
|
5
|
Vievermanns K, Dierikx TH, Oldenburger NJ, Jamaludin FS, Niemarkt HJ, de Meij TGJ. Effect of probiotic supplementation on the gut microbiota in very preterm infants: a systematic review. Arch Dis Child Fetal Neonatal Ed 2024; 110:57-67. [PMID: 38925919 DOI: 10.1136/archdischild-2023-326691] [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: 11/27/2023] [Accepted: 05/13/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE There is increasing evidence that probiotic supplementation in very preterm infants decreases the risk of necrotising enterocolitis (NEC), sepsis and mortality. The underlying mechanisms, including effects on the gut microbiota, are largely unknown. We aimed to systematically review the available literature on the effects of probiotic supplementation in very preterm infants on gut microbiota development. DESIGN A systematic review in Medline, Embase, Cochrane Library, CINAHL and Web of Science. SETTING Neonatal intensive care unit. PATIENTS Premature infants. INTERVENTION Probiotic supplementation. MAIN OUTCOME MEASURES Gut microbiota. RESULTS A total of 1046 articles were screened, of which 29 were included. There was a large heterogeneity in study design, dose and type of probiotic strains, timepoints of sample collection and analysing techniques. Bifidobacteria and lactobacilli were the most used probiotic strains. The effects of probiotics on alpha diversity were conflicting; however, beta diversity was significantly different between probiotic-supplemented infants and controls in the vast majority of studies. In most studies, probiotic supplementation led to increased relative abundance of the supplemented strains and decreased abundance of genera such as Clostridium, Streptococcus, Klebsiella and Escherichia. CONCLUSIONS Probiotic supplementation to preterm infants seems to increase the relative abundance of the supplemented strains with a concurrent decrease of potentially pathogenic species. These probiotic-induced microbial alterations may contribute to the decreased risk of health complications such as NEC. Future trials, including omics technologies to analyse both microbiota composition and function linked to health outcomes, are warranted to identify the optimal mixture and dosing of probiotic strains. PROSPERO REGISTRATION NUMBER CRD42023385204.
Collapse
Affiliation(s)
- Kayleigh Vievermanns
- Pediatric Gastroenterology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Thomas H Dierikx
- Pediatric Gastroenterology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
- Microbiology, Maastricht UMC+, Maastricht, The Netherlands
| | | | - Faridi S Jamaludin
- Medical Library AMC, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Hendrik J Niemarkt
- Neonatology, Maxima Medisch Centrum locatie Veldhoven, Veldhoven, The Netherlands
- Electrical Engineering, TU Eindhoven, Eindhoven, The Netherlands
| | - Tim G J de Meij
- Pediatric Gastroenterology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
- Pediatric Gastroenterology, Emma children's hospital amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Yin L, Liu Y, Sunzi K, Huang D, Huang J, Tang L, Liu M. The posttraumatic growth of fathers of preterm infants: protocol for a qualitative study in China. Front Psychiatry 2024; 15:1444226. [PMID: 39279814 PMCID: PMC11393778 DOI: 10.3389/fpsyt.2024.1444226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Background Prematurity presents a significant life crisis for families, often exceeding their expectations. Fathers of premature infants face the burden of multiple caregiving roles and undergo psychological changes. When confronted with such crises, individuals often engage in self-evaluation and may experience positive transformations. This study aims to employ a qualitative research methodology to explore the experiences of fathers of preterm infants. Materials and methods A phenomenological approach design will be utilized, drawing upon semi-structured in-depth interviews informed by existing literature. Thematic analysis will be employed, adhering to the Consolidated Criteria for Reporting Qualitative Research (COREQ) guidelines. In-depth individual interviews, lasting 40-60 minutes, will be conducted with fathers of preterm infants to understand their experiences. The thematic analysis process will facilitate a comprehensive understanding of the factors contributing to post-traumatic growth among these fathers. This methodology provides a structured approach to investigating the experiences and influences on post-traumatic growth in fathers of preterm infants. Results This study will highlight changes in post-traumatic growth among fathers of preterm infants. Discussion Research on the post-traumatic growth (PTG) of fathers of preterm infants is crucial to understanding the unique challenges and psychological transformations they experience. This study aims to explore the factors contributing to PTG in these fathers and how cultural contexts in China influence this process. By elucidating these aspects, the findings can inform targeted interventions and support systems tailored to the needs of fathers of preterm infants. The results may also contribute to developing guidelines and policies to promote psychological well-being and resilience among this population in the healthcare system. Ethics and dissemination This study adheres to the International Ethical Guidelines for Biomedical Research and the Declaration of Helsinki. Approval has been obtained from the People's Hospital of Deyang Human Research Ethics Committee (No: 2019-04-150-K01). The research follows the principles of open science, and the findings will be published while ensuring participants' confidentiality.
Collapse
Affiliation(s)
- Lina Yin
- Department of Pediatric, People's Hospital of Deyang City, Sichuan, Deyang, China
| | - Yanli Liu
- Department of Pediatric, People's Hospital of Deyang City, Sichuan, Deyang, China
| | - Kejimu Sunzi
- Department of Pediatric, People's Hospital of Deyang City, Sichuan, Deyang, China
| | - Dandan Huang
- Department of Pediatric, People's Hospital of Deyang City, Sichuan, Deyang, China
| | - Jing Huang
- Department of Pediatric, People's Hospital of Deyang City, Sichuan, Deyang, China
| | - Liangmei Tang
- Department of Pediatric, People's Hospital of Deyang City, Sichuan, Deyang, China
| | - Minghui Liu
- Department of Pediatric, People's Hospital of Deyang City, Sichuan, Deyang, China
| |
Collapse
|
7
|
Liu L, Chen C, Li Y, Ao D, Wu J, Cai N, Li W, Xiang M. Dynamics alteration of the gut microbiota and faecal metabolomes in very low or extremely low birth weight infants: a Chinese single-center, prospective cohort study. Front Microbiol 2024; 15:1438213. [PMID: 39247697 PMCID: PMC11377216 DOI: 10.3389/fmicb.2024.1438213] [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: 05/28/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Objective The aim of this study is to comprehensively investigate the temporal dynamics of faecal gut microbiota and metabonomics in early postnatal with a focus on very low or extremely low birth weight (VLBW/ELBW) infants. Methods We collected faecal samples from 157 VLBW/ELBW infants at three time points: days 1, 14, and 28 in a prospective cohort study. The faecal microbial diversity, abundance, composition, and metabolomic analyses were determined using 16S rRNA sequencing and liquid chromatography tandem mass spectrometry (LC-MS/MS). Microbiome functional analyses were conducted utilizing PICRUSt2. The ecological association networks were employed to investigate the interactions between gut microbiota and identify the core genus within 28 days of birth, as well as to unveil correlations between taxa and metabolites. Result (1) The alpha diversity of gut microbiota significantly decreased from D1 to D28, accompanied by an interrupted trajectory lacking obligate anaerobes. At the phylum level, the 16S RNA sequencing results showed an increase in Proteobacteria and a decrease in Firmicutes and Bacteroidota from D1 to D28. At the genus level, there was a decrease in the relative abundance of Staphylococcus, Acinetobacter and Ureaplasma, with Klebsiella and Enterococcus emerging as the most abundant genera. (2) The analysis revealed a total of 561 metabolic markers that exhibited significant and distinct alterations between D1 and D14. (3) Ecological association networks revealed that the gut microbiota in D1 exhibited a significantly higher degree of microbial interactions compared to those in D14 and D28. Additionally, Enterococcus, Klebsiella, and Enterobacter were major contributors to the co-occurring network at these three time points. (4) Steroid hormone biosynthesis, including tetrahydrocortisone, androsterone glucuronide, androstenedione and etiocholanolone glucuronide, decreased within 28 days after birth. Conclusion We have successfully demonstrated a significant dysbiosis in the gut microbiota and a subsequent decrease in its diversity within 4 weeks postpartum in VLBW/ELBW infants. Monitoring the gut microbiota of VLBW/ELBW infants and promptly rectifying dysbiosis in the early stages may represent a potential therapeutic strategy.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chaohong Chen
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - YeShan Li
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Dang Ao
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiayuan Wu
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Nali Cai
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wen Li
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Min Xiang
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
8
|
Tsuchiya M, Tsuchiya S, Momma H, Ikeda R, Suzuki J, Nagatomi R, Yaegashi N, Arima T, Igarashi K. Impact of Habitual Yogurt Intake in Mother-Child Dyads on Incidence of Childhood Otitis Media: The Japan Environment and Children's Study (JECS). Probiotics Antimicrob Proteins 2024; 16:884-893. [PMID: 37195509 DOI: 10.1007/s12602-023-10086-2] [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] [Accepted: 05/04/2023] [Indexed: 05/18/2023]
Abstract
Otitis media, one of the most common childhood diseases, is characterized by inflammation or infection of the middle ear. Due to their ease of access, daily probiotics are recommended for the prevention of early childhood otitis media. This study aimed to assess the impact of probiotics on the incidence of otitis media using a dataset (n = 95,380) from the Japan Environment and Children's Study, a nationwide birth cohort study. After multiple imputations, the association between the incidence of otitis media in early childhood and the daily frequency of yogurt intake in children and mothers was examined using a generalized linear model after adjusting for several confounders. Repeated incidence of otitis media during the 2 years after birth was found in 14,874 participants (15.6%). Based on participants with the lowest frequency of yogurt intake ("almost never") as the reference group, risk ratios for otitis media incidence decreased with higher frequencies of yogurt intake in children at one year of age, but also independently in mothers during pregnancy. The lowest risk ratio (95% confidence interval) for otitis media incidence at six months of age was observed with the most frequent yogurt intake (once/day or more) (0.54 [0.46-0.63]). Additionally, although a similar association was observed in the subgroup of those with cleft lip and/or palate (CL/P), a high-risk group for severe recurrent otitis media, no statistical significance was observed. Thus, increased regular yogurt intake in both children and mothers was associated with a decrease of otitis media during early childhood.
Collapse
Affiliation(s)
| | - Shinobu Tsuchiya
- Department of Orthodontics and Speech Therapy for Craniofacial Anomalies, Tohoku University Hospital, Sendai, Japan
| | - Haruki Momma
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryoukichi Ikeda
- Department of Otolaryngology - Head and Neck Surgery, Iwate Medical School of University, Morioka, Japan
| | - Jun Suzuki
- Department of Otolaryngology - Head and Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryoichi Nagatomi
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Biomedical Engineering for Health & Welfare, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kaoru Igarashi
- Department of Orthodontics and Speech Therapy for Craniofacial Anomalies, Tohoku University Hospital, Sendai, Japan
- Division of Craniofacial Anomalies, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
9
|
Zhang D, Lan Y, Zhang J, Cao M, Yang X, Wang X. Effects of early-life gut microbiota on the neurodevelopmental outcomes of preterm infants: a multi-center, longitudinal observational study in China. Eur J Pediatr 2024; 183:1733-1740. [PMID: 38231236 DOI: 10.1007/s00431-024-05423-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/18/2024]
Abstract
To prospectively investigate associations between the features of gut microbiota at the fourth week after birth in preterm infants and neurodevelopment from 1 month of corrected age to 6 months of corrected age (MCA). Seventy-seven preterm infants were recruited from three NICUs of three tertiary hospitals between Apr 2021 to Sep 2022. Stool samples were collected during the fourth week after birth. Illumina MiSeq high-throughput sequencing technology was used to detect the composition and diversity of gut microbiota. Neurodevelopment assessments of preterm infants were conducted at 1, 3, and 6 MCA using the Ages and Stages Questionnaire, the third edition (ASQ-3). Spearman correlation, a generalized linear mixed model (GLMM), and permutational multivariate analysis of variance (PERMANOVA) analysis were used to horizontally and prospectively explore the associations between gut microbial and ASQ-3 dimension scores at each time point. The GLMM showed no significant associations between the alpha diversity and neurodevelopmental trajectory from 1 to 6 MCA. The beta diversity was significantly associated with gross motor scores at 1, 3, and 6 MCA (R2 = 0.067, p = 0.001; R2 = 0.039, p = 0.020; R2 = 0.031, p = 0.047); communication scores at 3 MCA (R2 = 0.030, p = 0.040); and fine motor scores at 6 MCA (R2 = 0.035, p = 0.022). After adjusting for covariates, the GLMM showed that the relative abundance of Klebsiella was negatively associated with gross motor score trajectory from 1 to 6 MCA (β = - 1.449; 95% CI, - 2.275 to - 0.572; p = 0.001), while the relative abundance of Lactobacillus displayed a positive association (β = 1.421; 95% CI, 0.139 to 2.702; p = 0.030). Moreover, the relative abundance of Streptococcus was negatively associated with fine motor trajectory from 1 to 6 MCA (β = - 1.669; 95% CI, - 3.305 to - 0.033; p = 0.046). CONCLUSION Our results suggest a possible association between the neonatal gut microbial diversity; the relative abundance of Klebsiella, Streptococcus, and Lactobacillus; and neurodevelopment from 1 to 6 MCA. In the future, clinical staff can focus on the window period of gut microbiota colonization, and implement probiotics targeted at the dominant genera to improve the neurodevelopment of preterm infants. WHAT IS KNOWN • In the fields of biology and medicine, current studies suggest that gut microbiota may play an important role in the critical window period of neurodevelopment through the gut-brain axis pathway. • Extensive preclinical research has implied the vital role of the initial gut colonization in the long-term neurodevelopment of children. WHAT IS NEW • The early-life gut microbiota was associated with neurodevelopment in preterm infants within 6 months of corrected age (MCA).
Collapse
Affiliation(s)
- Dan Zhang
- School of Nursing, Wuhan University, No. 115, Donghu Road, Wuchang District, Wuhan, 430071, People's Republic of China
| | - Yancong Lan
- School of Nursing, Wuhan University, No. 115, Donghu Road, Wuchang District, Wuhan, 430071, People's Republic of China
| | - Jun Zhang
- School of Nursing, Wuhan University, No. 115, Donghu Road, Wuchang District, Wuhan, 430071, People's Republic of China.
| | - Mi Cao
- School of Nursing, Wuhan University, No. 115, Donghu Road, Wuchang District, Wuhan, 430071, People's Republic of China
| | - Xinyi Yang
- School of Nursing, Wuhan University, No. 115, Donghu Road, Wuchang District, Wuhan, 430071, People's Republic of China
| | - Xia Wang
- Department of Pediatrics, Women and Children's Hospital, Zhongnan Hospital, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
10
|
Calvo LN, Greenberg RG, Gray KD. Safety and Effectiveness of Probiotics in Preterm Infants with Necrotizing Enterocolitis. Neoreviews 2024; 25:e193-e206. [PMID: 38556491 DOI: 10.1542/neo.25-3-e193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Although necrotizing enterocolitis is a leading cause of morbidity and mortality among preterm infants, its underlying pathophysiology is not fully understood. Gut dysbiosis, an imbalance between commensal and pathogenic microbes, in the preterm infant is likely a major contributor to the development of necrotizing enterocolitis. In this review, we will discuss the increasing use of probiotics in the NICU, an intervention aimed to mitigate alterations in the gut microbiome. We will review the existing evidence regarding the safety and effectiveness of probiotics, and their potential to reduce rates of necrotizing enterocolitis in preterm infants.
Collapse
Affiliation(s)
- Laura N Calvo
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Rachel G Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
- Duke Clinical Research Institute, Durham, NC
| | - Keyaria D Gray
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
| |
Collapse
|
11
|
Calvo LN, Greenberg RG, Gray KD. Safety and Effectiveness of Probiotics in Preterm Infants with Necrotizing Enterocolitis. Neoreviews 2024; 25:e193-e206. [PMID: 38556499 DOI: 10.1542/neo.25-4-e193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Although necrotizing enterocolitis is a leading cause of morbidity and mortality among preterm infants, its underlying pathophysiology is not fully understood. Gut dysbiosis, an imbalance between commensal and pathogenic microbes, in the preterm infant is likely a major contributor to the development of necrotizing enterocolitis. In this review, we will discuss the increasing use of probiotics in the NICU, an intervention aimed to mitigate alterations in the gut microbiome. We will review the existing evidence regarding the safety and effectiveness of probiotics, and their potential to reduce rates of necrotizing enterocolitis in preterm infants.
Collapse
Affiliation(s)
- Laura N Calvo
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Rachel G Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
- Duke Clinical Research Institute, Durham, NC
| | - Keyaria D Gray
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
| |
Collapse
|
12
|
Abu YF, Singh S, Tao J, Chupikova I, Singh P, Meng J, Roy S. Opioid-induced dysbiosis of maternal gut microbiota during gestation alters offspring gut microbiota and pain sensitivity. Gut Microbes 2024; 16:2292224. [PMID: 38108125 PMCID: PMC10730209 DOI: 10.1080/19490976.2023.2292224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
There has been a rapid increase in neonates born with a history of prenatal opioid exposure. How prenatal opioid exposure affects pain sensitivity in offspring is of interest, as this may perpetuate the opioid epidemic. While few studies have reported hypersensitivity to thermal pain, potential mechanisms have not been described. This study posits that alterations in the gut microbiome may underly hypersensitivity to pain in prenatally methadone-exposed 3-week-old male offspring, which were generated using a mouse model of prenatal methadone exposure. Fecal samples collected from dams and their offspring were subjected to 16s rRNA sequencing. Thermal and mechanical pain were assessed using the tail flick and Von Frey assays. Transcriptomic changes in whole brain samples of opioid or saline-exposed offspring were investigated using RNA-sequencing, and midbrain sections from these animals were subjected to qPCR profiling of genes related to neuropathic and inflammatory pain pathways. Prenatal methadone exposure increased sensitivity to thermal and mechanical pain and elevated serum levels of IL-17a. Taxonomical analysis revealed that prenatal methadone exposure resulted in significant alterations in fecal gut microbiota composition, including depletion of Lactobacillus, Bifidobacterium, and Lachnospiracea sp and increased relative abundance of Akkermansia, Clostridium sensu stricto 1, and Lachnoclostridium. Supplementation of the probiotic VSL#3 in dams rescued hypersensitivity to thermal and mechanical pain in prenatally methadone-exposed offspring. Similarly, cross-fostering prenatally methadone-exposed offspring to control dams also attenuated hypersensitivity to thermal pain in opioid-exposed offspring. Modulation of the maternal and neonatal gut microbiome with probiotics resulted in transcriptional changes in genes related to neuropathic and immune-related signaling in whole brain and midbrain samples of prenatally methadone-exposed offspring. Together, our work provides compelling evidence of the gut-brain-axis in mediating pain sensitivity in prenatally opioid-exposed offspring.
Collapse
Affiliation(s)
- Yaa F. Abu
- Department of Microbiology and Immunology, University of Miami, Miami, FL, USA
| | - Salma Singh
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Junyi Tao
- Department of Surgery, University of Miami, Miami, FL, USA
| | | | - Praveen Singh
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Jingjing Meng
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Sabita Roy
- Department of Surgery, University of Miami, Miami, FL, USA
| |
Collapse
|
13
|
Li L, Liu T, Shi Y. Treatment of preterm brain injury via gut-microbiota-metabolite-brain axis. CNS Neurosci Ther 2024; 30:e14556. [PMID: 38108213 PMCID: PMC10805406 DOI: 10.1111/cns.14556] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/06/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Brain injury in preterm infants potentially disrupts critical structural and functional connective networks in the brain. It is a major cause of neurological sequelae and developmental deficits in preterm infants. Interesting findings suggest that the gut microbiota (GM) and their metabolites contribute to the programming of the central nervous system (CNS) during developmental stages and may exert structural and functional effects throughout the lifespan. AIM To summarize the existing knowledge of the potential mechanisms related to immune, endocrine, neural, and blood-brain barrier (BBB) mediated by GM and its metabolites in neural development and function. METHODS We review the recent literature and included 150 articles to summarize the mechanisms through which GM and their metabolites work on the nervous system. Potential health benefits and challenges of relevant treatments are also discussed. RESULTS This review discusses the direct and indirect ways through which the GM may act on the nervous system. Treatment of preterm brain injury with GM or related derivatives, including probiotics, prebiotics, synbiotics, dietary interventions, and fecal transplants are also included. CONCLUSION This review summarizes mechanisms underlying microbiota-gut-brain axis and novel therapeutic opportunities for neurological sequelae in preterm infants. Optimizing the initial colonization and microbiota development in preterm infants may represent a novel therapy to promote brain development and reduce long-term sequelae.
Collapse
Affiliation(s)
- Ling Li
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Tianjing Liu
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Yongyan Shi
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
14
|
Pan LL, Sun Y, Dong X, Ren Z, Li B, Yang P, Zhang L, Sun J. Infant feces-derived Lactobacillus gasseri FWJL-4 mitigates experimental necrotizing enterocolitis via acetate production. Gut Microbes 2024; 16:2430541. [PMID: 39648298 PMCID: PMC11633162 DOI: 10.1080/19490976.2024.2430541] [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: 02/13/2024] [Revised: 09/02/2024] [Accepted: 11/11/2024] [Indexed: 12/10/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening disease in premature infants, characterized by high mortality. Recent studies increasingly highlight the role of gut dysbiosis in NEC pathogenesis. Although probiotics have shown some efficacy in preventing NEC, further research is needed to determine potential strains and approaches. In this study, we demonstrated that the novel probiotic strain Lactobacillus gasseri (L. gasseri) FWJL-4, isolated from the feces of healthy infants, significantly enhanced intestinal barrier function, providing substantial protection against NEC. This protective effect was attributed to elevated intestinal acetate levels. Notably, acetate supplementation alone was sufficient to mitigate NEC, mimicking the protective effects of L. gasseri FWJL-4. Mechanistically, we revealed that L. gasseri FWJL-4 inhibited necroptosis and preserved the number of the goblet cells and enterocytes through the production of the short-chain fatty acid acetate, via activation of the acetate receptors G protein-coupled receptor (GPR) 41 and GPR43. Our findings suggest that L. gasseri FWJL-4 enhances intestinal barrier function to protect against NEC, underscoring the potential of probiotic manipulation as a promising strategy for NEC prevention.
Collapse
Affiliation(s)
- Li-Long Pan
- Department of Neonatology, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yudong Sun
- Department of Neonatology, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaoliang Dong
- Department of Neonatology, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhengnan Ren
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Binbin Li
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ping Yang
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Le Zhang
- Department of Neonatology, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jia Sun
- Department of Neonatology, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
15
|
Yin P, Du T, Yi S, Zhang C, Yu L, Tian F, Chen W, Zhai Q. Response differences of gut microbiota in oligofructose and inulin are determined by the initial gut Bacteroides/Bifidobacterium ratios. Food Res Int 2023; 174:113598. [PMID: 37986462 DOI: 10.1016/j.foodres.2023.113598] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/07/2023] [Accepted: 10/13/2023] [Indexed: 11/22/2023]
Abstract
Prebiotics are known to modulate the gut microbiota, but there is host variability, mainly due to differences in carbohydrate-utilisation by gut microbiota. Bifidobacterium and Bacteroides are powerful carbohydrate-utilising bacteria, and the ratio of both is closely related to the utilisation of prebiotics. However, the differential impact of prebiotics on the composition and function of the gut microbiota and its metabolites in participants with different Bacteroides/Bifidobacterium (Ba/Bi) ratios have not been studied. Here, we conducted a 4-week randomised double-blind, parallel four-arm trial using two prebiotics (oligofructose and inulin) in two populations with high Ba/Bi (H) and low Ba/Bi (L). The response to prebiotics in both populations was influenced by the baseline microbiota background specificity. Notably, at an overall level, FOS was slightly better than inulin in modulating the gut microbiota. Difference in gut microbiota regulation by FOS across microbiota contexts were significant between the two groups. Butyric acid-producing bacteria were significantly more abundant in H and further elevated butyric acid and related metabolite levels, with H more likely to benefit from the FOS intervention. The two groups showed only metabolic differences in their response to inulin, with L showing a significant increase in propionic acid and being enriched in glycolysis functions, whereas H was enriched in amino acids and aminoglycolysis functions. Overall, these results provide a basis for selecting appropriate prebiotics for participants with different gut backgrounds.
Collapse
Affiliation(s)
- Pingping Yin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ting Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Shanrong Yi
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
16
|
Ma G, Shi Y, Meng L, Fan H, Tang X, Luo H, Wang D, Zhou J, Xiao X. Factors affecting the early establishment of neonatal intestinal flora and its intervention measures. Front Cell Infect Microbiol 2023; 13:1295111. [PMID: 38106467 PMCID: PMC10722192 DOI: 10.3389/fcimb.2023.1295111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
In recent years, it has become evident that early-life intestinal flora plays a pivotal role in determining human health. Consequently, it is imperative to explore the establishment of neonatal intestinal flora and its influencing factors. Early neonatal intestinal flora is influenced by a multitude of factors, including maternal and infant-related factors, as well as external environment. This review summarizes the colonization mechanism of intestinal flora in the early life of newborns and discussed their influence on the establishment of neonatal intestinal flora, taking into account factors such as delivery mode, gestational age and feeding mode. Additionally, this review delves into the natural or artificial reconstruction of intestinal flora colonization defects in infants born via cesarean section and premature infants, with the goal of establishing a theoretical foundation for preventing and treating issues related to neonatal intestinal flora colonization and associated diseases.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuguo Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lulu Meng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Haolong Fan
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments and Center for Composite Materials and Structures, Harbin Institute of Technology, Harbin, China
| | - Xiaomei Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Huijuan Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongju Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Juan Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
17
|
Reynolds HM, Bettini ML. Early-life microbiota-immune homeostasis. Front Immunol 2023; 14:1266876. [PMID: 37936686 PMCID: PMC10627000 DOI: 10.3389/fimmu.2023.1266876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
As the prevalence of allergy and autoimmune disease in industrialized societies continues to rise, improving our understanding of the mechanistic roles behind microbiota-immune homeostasis has become critical for informing therapeutic interventions in cases of dysbiosis. Of particular importance, are alterations to intestinal microbiota occurring within the critical neonatal window, during which the immune system is highly vulnerable to environmental exposures. This review will highlight recent literature concerning mechanisms of early-life microbiota-immune homeostasis as well as discuss the potential for therapeutics in restoring dysbiosis in early life.
Collapse
Affiliation(s)
| | - Matthew L. Bettini
- Department of Microbiology and Immunology, University of Utah, Salt Lake, UT, United States
| |
Collapse
|
18
|
Song H, Guo R, Sun X, Kou Y, Ma X, Chen Y, Song L, Yuan C, Wu Y. Xylooligosaccharides from corn cobs alleviate loperamide-induced constipation in mice via modulation of gut microbiota and SCFA metabolism. Food Funct 2023; 14:8734-8746. [PMID: 37694718 DOI: 10.1039/d3fo02688d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
This study aimed to optimize the structure and efficacy of xylooligosaccharides (XOSs) from corn cobs in constipated mice. Structural analysis revealed that XOSs from corn cobs were composed of β-Xyl-(1 →4)-[β-Xyl-(1→4)]n-α/β-Xyl (n = 0-5) without any other substituents. XOS administration significantly reduced the defecation time, increased the gastrointestinal transit rate, restored the gastrointestinal neurotransmitter imbalance, protected against oxidative stress, and reversed constipation-induced colonic inflammation. Fecal metabolite and microbiota analysis showed that XOS supplementation significantly increased short chain fatty acid (SCFA) levels and improved the gut microbial environment. These findings highlighted the potential of XOSs from corn cobs as an active ingredient for functional foods or as a therapeutic agent in constipation therapy.
Collapse
Affiliation(s)
- Hong Song
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Rui Guo
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xianbao Sun
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yuxing Kou
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xuan Ma
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yinan Chen
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Lihua Song
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Chunmei Yuan
- Yunnan Maoduoli Group Food Co., Ltd., Yuxi 653100, China
| | - Yan Wu
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
19
|
Park H, Park NY, Koh A. Scarring the early-life microbiome: its potential life-long effects on human health and diseases. BMB Rep 2023; 56:469-481. [PMID: 37605613 PMCID: PMC10547969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/30/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023] Open
Abstract
The gut microbiome is widely recognized as a dynamic organ with a profound influence on human physiology and pathology. Extensive epidemiological and longitudinal cohort studies have provided compelling evidence that disruptions in the early-life microbiome can have long-lasting health implications. Various factors before, during, and after birth contribute to shaping the composition and function of the neonatal and infant microbiome. While these alterations can be partially restored over time, metabolic phenotypes may persist, necessitating research to identify the critical period for early intervention to achieve phenotypic recovery beyond microbiome composition. In this review, we provide current understanding of changes in the gut microbiota throughout life and the various factors affecting these changes. Specifically, we highlight the profound impact of early-life gut microbiota disruption on the development of diseases later in life and discuss perspectives on efforts to recover from such disruptions. [BMB Reports 2023; 56(9): 469-481].
Collapse
Affiliation(s)
- Hyunji Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Na-Young Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Ara Koh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
20
|
Xiang Q, Yan X, Shi W, Li H, Zhou K. Early gut microbiota intervention in premature infants: Application perspectives. J Adv Res 2023; 51:59-72. [PMID: 36372205 PMCID: PMC10491976 DOI: 10.1016/j.jare.2022.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preterm birth is the leading cause of death in children under the age of five. One of the major factors contributing to the high risk of diseases and deaths in premature infants is the incomplete development of the intestinal immune system. The gut microbiota has been widely recognized as a critical factor in promoting the development and function of the intestinal immune system after birth. However, the gut microbiota of premature infants is at high risk of dysbiosis, which is highly associated with adverse effects on the development and education of the early life immune system. Early intervention can modulate the colonization and development of gut microbiota and has a long-term influence on the development of the intestinal immune system. AIM OF REVIEW This review aims to summarize the characterization, interconnection, and underlying mechanism of gut microbiota and intestinal innate immunity in premature infants, and to discuss the status, applicability, safety, and prospects of different intervention strategies in premature infants, thus providing an overview and outlook of the current applications and remaining gaps of early intervention strategies in premature infants. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, the gut microbiota of premature infants is at high risk of dysbiosis, resulting in dysfunctional intestinal immune system processes. Secondly, contributing roles of early intervention have been observed in improving the intestinal environment and promoting gut microbiota colonization, which is significant in the development and function of gut immunity in premature infants. Thirdly, different strategies of early intervention, such as probiotics, fecal microbiota transplantation, and nutrients, show different safety, applicability, and outcome in premature infants, and the underlying mechanism is complex and poorly understood.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, the first affiliated hospital of Southern University of Science and Technology of China, Shenzhen People's Hospital, Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
21
|
Luo Z, Chen A, Xie A, Liu X, Jiang S, Yu R. Limosilactobacillus reuteri in immunomodulation: molecular mechanisms and potential applications. Front Immunol 2023; 14:1228754. [PMID: 37638038 PMCID: PMC10450031 DOI: 10.3389/fimmu.2023.1228754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
Frequent use of hormones and drugs may be associated with side-effects. Recent studies have shown that probiotics have effects on the prevention and treatment of immune-related diseases. Limosilactobacillus reuteri (L. reuteri) had regulatory effects on intestinal microbiota, host epithelial cells, immune cells, cytokines, antibodies (Ab), toll-like receptors (TLRs), tryptophan (Try) metabolism, antioxidant enzymes, and expression of related genes, and exhibits antibacterial and anti-inflammatory effects, leading to alleviation of disease symptoms. Although the specific composition of the cell-free supernatant (CFS) of L. reuteri has not been clarified, its efficacy in animal models has drawn increased attention to its potential use. This review summarizes the effects of L. reuteri on intestinal flora and immune regulation, and discusses the feasibility of its application in atopic dermatitis (AD), asthma, necrotizing enterocolitis (NEC), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS), and provides insights for the prevention and treatment of immune-related diseases.
Collapse
Affiliation(s)
- Zichen Luo
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Ailing Chen
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Anni Xie
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Xueying Liu
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Renqiang Yu
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
22
|
Wala SJ, Ragan MV, Sajankila N, Volpe SG, Purayil N, Dumbauld Z, Besner GE. Probiotics and novel probiotic delivery systems. Semin Pediatr Surg 2023; 32:151307. [PMID: 37295299 DOI: 10.1016/j.sempedsurg.2023.151307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Necrotizing enterocolitis (NEC) is an infectious and inflammatory intestinal disease that is the most common surgical emergency in the premature patient population. Although the etiology of the disease is multifactorial, intestinal dysbiosis is a hallmark of this disease. Based on this, probiotics may play a therapeutic role in NEC by introducing beneficial bacteria with immunomodulating, antimicrobial, and anti-inflammatory functions into the gastrointestinal tract. Currently, there is no Food and Drug Administration (FDA)-approved probiotic for the prevention and treatment of NEC. All probiotic clinical studies to date have administered the bacteria in their planktonic (free-living) state. This review will discuss established probiotic delivery systems including planktonic probiotics, prebiotics, and synbiotics, as well as novel probiotic delivery systems such as biofilm-based and designer probiotics. We will also shed light on whether or not probiotic efficacy is influenced by administration with breast milk. Finally, we will consider the challenges associated with developing an FDA-approved probiotic for NEC.
Collapse
Affiliation(s)
- Samantha J Wala
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Mecklin V Ragan
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Nitin Sajankila
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Samuel G Volpe
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Nanditha Purayil
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Zachary Dumbauld
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Gail E Besner
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
23
|
Abstract
The last decade has witnessed a meteoric rise in research focused on characterizing the human microbiome and identifying associations with disease risk. The advent of sequencing technology has all but eradicated gel-based fingerprinting approaches for studying microbial ecology, while at the same time traditional microbiological culture is undergoing a renaissance. Although multiplexed high-throughput sequencing is relatively new, the discoveries leading to this are nearly 50 years old, coinciding with the inaugural Microbiology Society Fleming Prize lecture. It was an honour to give the 2022 Fleming Prize lecture and this review will cover the topics from that lecture. The focus will be on the bacterial community in early life, beginning with term infants before moving on to infants delivered prematurely. The review will discuss recent work showing how human milk oligosaccharides (HMOs), an abundant but non-nutritious component of breast milk, can modulate infant microbiome and promote the growth of Bifidobacterium spp. This has important connotations for preterm infants at risk of necrotizing enterocolitis, a devastating intestinal disease representing the leading cause of death and long-term morbidity in this population. With appropriate mechanistic studies, it may be possible to harness the power of breast milk bioactive factors and infant gut microbiome to improve short- and long-term health in infants.
Collapse
|
24
|
Therapeutic Potential of Gut Microbiota and Its Metabolite Short-Chain Fatty Acids in Neonatal Necrotizing Enterocolitis. Life (Basel) 2023; 13:life13020561. [PMID: 36836917 PMCID: PMC9959300 DOI: 10.3390/life13020561] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Short chain fatty acids (SCFAs), the principle end-products produced by the anaerobic gut microbial fermentation of complex carbohydrates (CHO) in the colon perform beneficial roles in metabolic health. Butyrate, acetate and propionate are the main SCFA metabolites, which maintain gut homeostasis and host immune responses, enhance gut barrier integrity and reduce gut inflammation via a range of epigenetic modifications in DNA/histone methylation underlying these effects. The infant gut microbiota composition is characterized by higher abundances of SCFA-producing bacteria. A large number of in vitro/vivo studies have demonstrated the therapeutic implications of SCFA-producing bacteria in infant inflammatory diseases, such as obesity and asthma, but the application of gut microbiota and its metabolite SCFAs to necrotizing enterocolitis (NEC), an acute inflammatory necrosis of the distal small intestine/colon affecting premature newborns, is scarce. Indeed, the beneficial health effects attributed to SCFAs and SCFA-producing bacteria in neonatal NEC are still to be understood. Thus, this literature review aims to summarize the available evidence on the therapeutic potential of gut microbiota and its metabolite SCFAs in neonatal NEC using the PubMed/MEDLINE database.
Collapse
|
25
|
Srinivasjois R, Gebremedhin A, Silva D, Rao S, Pereira G. Probiotic supplementation in neonates and long-term gut colonisation: A systematic review of randomised controlled trials. J Paediatr Child Health 2023; 59:212-217. [PMID: 36629072 DOI: 10.1111/jpc.16318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Probiotic supplementation in the neonatal period results in improved gut colonisation with probiotic bacteria in the short term. There is limited information on the long-term sustainability of this colonisation. AIMS To evaluate whether oral probiotic supplementation in the neonatal period results in sustained gut colonisation with probiotic bacteria at or beyond 6 months after its cessation. METHODS A systematic review of neonatal probiotic randomised controlled trials (RCTs) that reported on the stool microbiota during post-discharge follow-up was carried out using guidelines of the Cochrane neonatal group. RESULTS Four RCTs (n = 605 infants) were included in the review. The studies were heterogeneous in case selection, choice of probiotics, duration of supplementation, timing and the method of stool microbial analysis. Three RCTs (n = 471) showed the presence of intestinal probiotic bacteria at 6-12 months. The overall certainty of evidence was very low in view of small sample size, heterogeneity and identification only to the genus/species level. CONCLUSION Low certainty of evidence suggests that probiotic supplementation in the neonatal period may result in sustained gut colonisation 6-12 months post-cessation, but not at 24 months. Adequately powered, well-designed RCTs with strain-specific assays are needed in this area.
Collapse
Affiliation(s)
- Ravisha Srinivasjois
- Curtin School of Population Health, Curtin University, Bentley, Western Australia, Australia
- Department of Paediatrics and Neonatology, Joondalup Health Campus, Perth, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Division of Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Amanuel Gebremedhin
- Curtin School of Population Health, Curtin University, Bentley, Western Australia, Australia
- Enable Institute, Curtin University, Bentley, Western Australia, Australia
| | - Desiree Silva
- Department of Paediatrics and Neonatology, Joondalup Health Campus, Perth, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Shripada Rao
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Department of Neonatology, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Gavin Pereira
- Curtin School of Population Health, Curtin University, Bentley, Western Australia, Australia
- Enable Institute, Curtin University, Bentley, Western Australia, Australia
- Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
26
|
Mercer EM, Arrieta MC. Probiotics to improve the gut microbiome in premature infants: are we there yet? Gut Microbes 2023; 15:2201160. [PMID: 37122152 PMCID: PMC10153018 DOI: 10.1080/19490976.2023.2201160] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/06/2023] [Indexed: 05/02/2023] Open
Abstract
Gut microbiome maturation in infants born prematurely is uniquely influenced by the physiological, clinical, and environmental factors surrounding preterm birth and early life, leading to altered patterns of microbial succession relative to term infants during the first months of life. These differences in microbiome composition are implicated in acute clinical conditions that disproportionately affect preterm infants, including necrotizing enterocolitis (NEC) and late-onset sepsis (LOS). Probiotic supplementation initiated early in life is an effective prophylactic measure for preventing NEC, LOS, and other clinical concerns relevant to preterm infants. In parallel, reported benefits of probiotics on the preterm gut microbiome, metabolome, and immune function are beginning to emerge. This review summarizes the current literature on the influence of probiotics on the gut microbiome of preterm infants, outlines potential mechanisms by which these effects are exerted, and highlights important clinical considerations for determining the best practices for probiotic use in premature infants.
Collapse
Affiliation(s)
- Emily M. Mercer
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
27
|
Kaplina A, Zaikova E, Ivanov A, Volkova Y, Alkhova T, Nikiforov V, Latypov A, Khavkina M, Fedoseeva T, Pervunina T, Skorobogatova Y, Volkova S, Ulyantsev V, Kalinina O, Sitkin S, Petrova N. Intestinal microbiome changes in an infant with right atrial isomerism and recurrent necrotizing enterocolitis: A case report and review of literature. World J Clin Cases 2022; 10:10583-10599. [PMID: 36312470 PMCID: PMC9602219 DOI: 10.12998/wjcc.v10.i29.10583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/20/2022] [Accepted: 08/14/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a multifactorial disease that predominantly affects premature neonates. Intestinal dysbiosis plays a critical role in NEC pathogenesis in premature neonates. The main risk factor for NEC in term infants is mesenteric hypoperfusion associated with ductal-dependent congenital heart disease (CHD) that eventually leads to intestinal ischemia. The incidence of NEC in neonates with critical CHD is 6.8%-13%. However, the role of the intestinal microbiome in NEC pathogenesis in infants with ductal-dependent CHD remains unclear. CASE SUMMARY A male term neonate with right atrial isomerism underwent modified Blalock-Taussig shunt placement on the 14th day of life and had persistent mesenteric hypoperfusion after surgery. The patient had episodes of NEC stage IIA on the 1st and 28th days after cardiac surgery. Fecal microbial composition was analyzed before and after cardiac surgery by sequencing region V4 of the 16S rRNA gene. Before surgery, species belonging to genera Veillonella and Clostridia and class Gammaproteobacteria were detected, Bifidobacteriaceae showed a low abundance. The first NEC episode was associated with postoperative hemodynamic instability, intestinal ischemia-reperfusion injury during cardiopulmonary bypass, and a high abundance of Clostridium paraputrificum (Clostridium sensu stricto I) (56.1%). Antibacterial therapy after the first NEC episode resulted in increased abundance of Gammaproteobacteria, decreased abundance of Firmicutes, and low alpha diversity. These changes in the microbial composition promoted the growth of Clostridium sensu stricto I (72.0%) before the second NEC episode. CONCLUSION A high abundance of Clostridium sensu stricto I and mesenteric hypoperfusion may have contributed to NEC in the present case.
Collapse
Affiliation(s)
- Aleksandra Kaplina
- Research Laboratory of Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Ekaterina Zaikova
- Research Laboratory of Autoimmune and Autoinflammatory Diseases, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Artem Ivanov
- International Laboratory of Computer Technologies, ITMO University, St. Petersburg 197101, Russia
| | - Yulia Volkova
- Department of Cardiovascular Surgery for Children, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Alkhova
- Department of Neonatal Physiology with an ICU Ward, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Vladimir Nikiforov
- Pediatric Cardiac Intensive Care Unit, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Alexander Latypov
- Department of Cardiovascular Surgery for Children, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Marina Khavkina
- Neonatal and Preterm Special Care Unit (2nd Stage Care), Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Fedoseeva
- Research Laboratory of Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Pervunina
- Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Yulia Skorobogatova
- Express Laboratory of Perinatal Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Svetlana Volkova
- Clinical Diagnostic Laboratory, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Vladimir Ulyantsev
- International Laboratory of Computer Technologies, ITMO University, St. Petersburg 197101, Russia
| | - Olga Kalinina
- Research Laboratory of Autoimmune and Autoinflammatory Diseases, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Stanislav Sitkin
- Epigenetics and Metagenomics Group, Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
- Department of Internal Diseases, Gastroenterology and Dietetics, North-Western State Medical University named after I.I. Mechnikov, St. Petersburg 191015, Russia
| | - Natalia Petrova
- Research Laboratory of Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| |
Collapse
|
28
|
Jia Q, Yu X, Chang Y, You Y, Chen Z, Wang Y, Liu B, Chen L, Ma D, Xing Y, Tong X. Dynamic Changes of the Gut Microbiota in Preterm Infants With Different Gestational Age. Front Microbiol 2022; 13:923273. [PMID: 35847070 PMCID: PMC9279133 DOI: 10.3389/fmicb.2022.923273] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
The gut microbiota plays a key role in the pathogenesis of diseases affecting preterm infants and gestational age is one of the important factors which affect the gut microbiota of infants. To determine the characteristics of the gut microbiota in preterm infants of different gestational ages from birth to 1 year after birth, we collected 622 fecal samples from neonates of different gestational ages at different time points after birth. According to the gestational ages, the samples were divided into four groups, extremely preterm, very preterm, moderate to late preterm, and term group. Meconium and fecal samples at day 14, 28, 120, and 365 after birth were collected. 16S rRNA sequencing was performed and the composition and structure of the gut microbiota in preterm infants of different gestational age was compared with that of term infants. In our study, alpha diversity of meconium in extremely preterm group was higher than very preterm group, moderate to late preterm group and term group and alpha diversity of meconium in preterm group was decreased with increasing of gestational age. At day 14 to day 120 after birth, alpha diversity of term and moderate to late preterm group were significantly higher than other two preterm groups. However, moderate to late preterm group owned the highest alpha diversity which was higher than term group at day 365 after birth. Besides, the results shown the duration of opportunistic pathogen such as Klebsiella and Enterococcus which dominant colonization was different in different gestational age groups. As well as the probiotics, such as Bifidobacterium, which abundance enriched at different time point in different gestational age groups. We profiled the features of dynamic changes of gut microbiome from different gestational ages infants. The results of our research provide new insights for individualized interventions of specific microbes of preterm infants with different gestational ages at different time points after birth.
Collapse
Affiliation(s)
- Qiong Jia
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Xue Yu
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yanmei Chang
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yanxia You
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Zekun Chen
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Ying Wang
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Bin Liu
- National Engineering Center of Dairy for Maternal and Child Health, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Lijun Chen
- National Engineering Center of Dairy for Maternal and Child Health, Beijing Sanyuan Foods Co., Ltd., Beijing, China
| | - Defu Ma
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
29
|
Hitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J, Clavel T. Microbiome-based interventions to modulate gut ecology and the immune system. Mucosal Immunol 2022; 15:1095-1113. [PMID: 36180583 PMCID: PMC9705255 DOI: 10.1038/s41385-022-00564-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023]
Abstract
The gut microbiome lies at the intersection between the environment and the host, with the ability to modify host responses to disease-relevant exposures and stimuli. This is evident in how enteric microbes interact with the immune system, e.g., supporting immune maturation in early life, affecting drug efficacy via modulation of immune responses, or influencing development of immune cell populations and their mediators. Many factors modulate gut ecosystem dynamics during daily life and we are just beginning to realise the therapeutic and prophylactic potential of microbiome-based interventions. These approaches vary in application, goal, and mechanisms of action. Some modify the entire community, such as nutritional approaches or faecal microbiota transplantation, while others, such as phage therapy, probiotics, and prebiotics, target specific taxa or strains. In this review, we assessed the experimental evidence for microbiome-based interventions, with a particular focus on their clinical relevance, ecological effects, and modulation of the immune system.
Collapse
Affiliation(s)
- Thomas C A Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Lindsay J Hall
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, UK
- Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Sarah Kate Walsh
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | | | - Emma Slack
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | | | - Jens Walter
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany.
| |
Collapse
|
30
|
Yu X, Xing Y, Liu H, Chang Y, You Y, Dou Y, Liu B, Wang Q, Ma D, Chen L, Tong X. Effects of a Formula with scGOS/lcFOS (9:1) and Glycomacropeptide (GMP) Supplementation on the Gut Microbiota of Very Preterm Infants. Nutrients 2022; 14:nu14091901. [PMID: 35565868 PMCID: PMC9102166 DOI: 10.3390/nu14091901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 01/27/2023] Open
Abstract
Microbial colonization of very preterm (VPT) infants is detrimentally affected by the complex interplay of physiological, dietary, medical, and environmental factors. The aim of this study was to evaluate the effects of an infant formula containing the specific prebiotic mixture of scGOS/lcFOS (9:1) and glycomacropeptide (GMP) on the composition and function of VPT infants’ gut microbiota. Metagenomic analysis was performed on the gut microbiota of VPT infants sampled at four time points: 24 h before the trial and 7, 14, and 28 days after the trial. Functional profiling was aggregated into gut and brain modules (GBMs) and gut metabolic modules (GMMs) based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Enterococcus faecium, Escherichia coli, Klebsiella aerogenes, and Klebsiella pneumoniae were dominant species in both the test group and the control group. After the 4-week intervention, the abundance of Bifidobacterium in the test group was significantly increased. We found two GBMs (quinolinic acid synthesis and kynurenine degradation) and four GMMs (glutamine degradation, glyoxylate bypass, dissimilatory nitrate reduction, and preparatory phase of glycolysis) were significantly enriched in the test group, respectively. The results of this study suggested that formula enriched with scGOS/lcFOS (9:1) and GPM is beneficial to the intestinal microecology of VPT infants.
Collapse
Affiliation(s)
- Xue Yu
- School of Public Health, Peking University Health Science Center, Beijing 100191, China; (X.Y.); (Y.D.)
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
| | - Hui Liu
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
| | - Yanmei Chang
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
| | - Yanxia You
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
| | - Yuqi Dou
- School of Public Health, Peking University Health Science Center, Beijing 100191, China; (X.Y.); (Y.D.)
| | - Bin Liu
- National Engineering Center of Dairy for Maternal and Child Health, Beijing Sanyuan Foods Co., Ltd., Beijing 100163, China;
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Defu Ma
- School of Public Health, Peking University Health Science Center, Beijing 100191, China; (X.Y.); (Y.D.)
- Correspondence: (D.M.); (L.C.); (X.T.)
| | - Lijun Chen
- National Engineering Center of Dairy for Maternal and Child Health, Beijing Sanyuan Foods Co., Ltd., Beijing 100163, China;
- Correspondence: (D.M.); (L.C.); (X.T.)
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China; (Y.X.); (H.L.); (Y.C.); (Y.Y.)
- Correspondence: (D.M.); (L.C.); (X.T.)
| |
Collapse
|
31
|
Liu W, Zhou T, Tian J, Yu X, Ren C, Cao Z, Hou P, Zhang Q, Li A. Role of GDNF, GFRα1 and GFAP in a Bifidobacterium-Intervention Induced Mouse Model of Intestinal Neuronal Dysplasia. Front Pediatr 2022; 9:795678. [PMID: 35096711 PMCID: PMC8796853 DOI: 10.3389/fped.2021.795678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the effects of glial cell-derived neurotrophic factor (GDNF), GDNF family receptor alpha 1 (GFRα1), and glial fibrillary acidic protein (GFAP) on colonic motility in a mouse model of intestinal neuronal dysplasia by intervention with Bifidobacterium and to explore the influence of Bifidobacterium on enteric glial cells (EGCs). METHODS Western blotting and qRT-PCR were employed to detect the expression of GFRα1 and GFAP in colonic tissues of mice with or without Tlx2 mutations, and ELISA was used to detect the expression of GDNF in serum. IHC was used to detect the appearance of the ganglion cells. Subsequently, Tlx2 homozygous mutant (Tlx2-/-) mice were treated with Bifidobacterium. Colonic motility was measured before and after intervention by measuring the glass bead expelling time. The variations in abdominal circumference and GDNF, GFRα1, and GFAP expression were measured. In addition, 16SrRNA gene sequencing was performed to detect the abundance of the intestinal microbiota. RESULTS The mRNA and protein expression of GFRα1 and GFAP was decreased in the colonic tissues of Tlx2-/- mice and GDNF expression was decreased in serum compared with Tlx2+/- and WT mice. After confirming the colonization of Bifidobacterium by 16S rRNA gene sequencing, the expelling time and abdominal distension were ameliorated, and the expression of GFAP, GDNF, and GFRα1 was increased. CONCLUSIONS The expression of GDNF, GFRα1, and GFAP is associated with colonic motility. The altered expression of EGC-related factors suggested that Bifidobacterium may be involved in the EGC activation process. The amelioration of IND symptoms after intervention with Bifidobacterium prompted the elicitation of adjuvant therapy.
Collapse
Affiliation(s)
- Wei Liu
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tingting Zhou
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jinqiu Tian
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofang Yu
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuantao Ren
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Pediatric Surgery, Dezhou People's Hospital, Dezhou, China
| | - Zengcai Cao
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peimin Hou
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiangye Zhang
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Aiwu Li
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
32
|
Healy DB, Ryan CA, Ross RP, Stanton C, Dempsey EM. Clinical implications of preterm infant gut microbiome development. Nat Microbiol 2022; 7:22-33. [PMID: 34949830 DOI: 10.1038/s41564-021-01025-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Perturbations to the infant gut microbiome during the first weeks to months of life affect growth, development and health. In particular, assembly of an altered intestinal microbiota during infant development results in an increased risk of immune and metabolic diseases that can persist into childhood and potentially into adulthood. Most research into gut microbiome development has focused on full-term babies, but health-related outcomes are also important for preterm babies. The systemic physiological immaturity of very preterm gestation babies (born earlier than 32 weeks gestation) results in numerous other microbiome-organ interactions, the mechanisms of which have yet to be fully elucidated or in some cases even considered. In this Perspective, we compare assembly of the intestinal microbiome in preterm and term infants. We focus in particular on the clinical implications of preterm infant gut microbiome composition and discuss the prospects for microbiome diagnostics and interventions to improve the health of preterm babies.
Collapse
Affiliation(s)
- David B Healy
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| | - C Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,INFANT Research Centre, Cork University Hospital, Cork, Ireland
| |
Collapse
|