1
|
Alswat AS. The Influence of the Gut Microbiota on Host Health: A Focus on the Gut-Lung Axis and Therapeutic Approaches. Life (Basel) 2024; 14:1279. [PMID: 39459579 PMCID: PMC11509314 DOI: 10.3390/life14101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
The human gut microbiota is a complex ecosystem harboring thousands of microbial strains that play a crucial role in maintaining the overall well-being of its host. The composition of the gut microbiota varies with age from infancy to adulthood and is influenced by dietary habits, environment, and genetic disposition. Recent advances in culture-independent techniques and nucleic acid sequencing have improved our understanding of the diversity of the gut microbiota. The microbial species present in the gut release short-chain fatty acids (SCFAs), which have anti-inflammatory properties. The gut microbiota also plays a substantial role in modulating the host's immune system, promoting immune tolerance, and maintaining homeostasis. The impact of the gut microbiota on the health of the host is quite evident, as gut dysbiosis has been linked to various diseases, including metabolic disorders, autoimmune diseases, allergies, and inflammatory bowel diseases. The gut microbiota has bidirectional communication with the respiratory system, creating the gut-lung axis, which has been associated with different respiratory diseases. Therapeutic approaches targeting the gut microbiota, such as probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation (FMT), aim to restore microbial balance and promote the growth of beneficial strains in the gut. Nonetheless, gaining knowledge of the complex interactions between the gut microbiota and the host is necessary to develop personalized medicine approaches and microbiota-based therapies for various conditions. This review summarizes studies related to the gut-lung axis with particular emphasis on the role of the microbiota. Future research directions are also discussed.
Collapse
Affiliation(s)
- Amal S Alswat
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
2
|
Tyrsin OY, Tyrsin DY, Nemenov DG, Ruzov AS, Odintsova VE, Koshechkin SI, D Amico L. Effect of Lactobacillus reuteri NCIMB 30351 drops on symptoms of infantile functional gastrointestinal disorders and gut microbiota in early infants: Results from a randomized, placebo-controlled clinical trial. Eur J Pediatr 2024; 183:2311-2324. [PMID: 38427038 DOI: 10.1007/s00431-024-05473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 03/02/2024]
Abstract
Infantile functional gastrointestinal disorders, such as colic, constipation, diarrhea, and gastroesophageal reflux (regurgitation), often occur in early infancy and, representing one of the causes of significant parental anxiety, lead to a significant strain on the healthcare resources. In this study, we aimed to evaluate the effects of Lactobacillus reuteri drops (L. reuteri NCIMB 30351) on the symptoms of infantile colic, constipation, diarrhea, and gastroesophageal reflux, as well as on the levels of intestinal microbiota in full-term newborns during the first months of life. A randomized, placebo-controlled, single-masked (blinded), post-marketing clinical study was conducted in two clinical units-Children's City Clinical Hospital of Moscow and Medical Center "St. Andrew's Hospitals-NEBOLIT" from March 2020 to May 2022 in 90 infants aged from 1 to 4 months (mean age (± SD) 12.3 ± 5.09 weeks; 53.3% females, 46.7% males). Patients with colic, regurgitation (single symptom or combination of several symptoms), and constipation or diarrhea were randomly allocated in two parallel arms to receive either 5 drops (2 × 108 colony forming unit) of L. reuteri NCIMB 30351 (n = 60) or masked placebo (n = 30) for 25 consecutive days. Two treatment arms had equal numbers of patients with constipation and diarrhea (n = 30 each). Daily crying times and their duration, evacuations, and regurgitations were recorded in a structured diary. The levels of gut microbiota were analyzed by deep sequencing of bacterial 16S rRNA gene. Infants with colic receiving supplementary L. reuteri NCIMB 30351 for 25 days had significant reduction in the numbers of colic (change from baseline - 6.3 (7.34) vs - 3.0 (7.29) in placebo, P < 0.05) and numbers of crying cases and mean duration of crying (decrease from baseline - 144 (70.7) minutes, lower in the diarrhea subgroup than in constipation infants, compared with - 80 (58.9) in placebo, P < 0.0001), as well as regurgitation numbers (decreased by - 4.8 (2.49) with L. reuteri vs - 3 (7.74) with placebo). We also observed increased numbers of evacuations in infants with constipation (L. reuteri 2.2 (2.4) vs 0.9 (1.06) in placebo, P < 0.05). There was a remarkable reduction of evacuations in infants with diarrhea, while not statistically significant. The analysis of bacterial 16S rRNA gene in the collected samples showed that L. reuteri positively influences the proportions of prevalent species, while it negatively affects both conditionally pathogenic and commensal microbes. Additional in vitro test for formation of Clostridium colonies in the presence of the probiotic demonstrated that L. reuteri effectively inhibits the growth of pathogenic Clostridium species. No adverse events were reported in this study. Conclusion: The uptake of L. reuteri NCIMB 30351 leads to a significant reduction in the number of regurgitations, feeding-induced constipations, and diarrhea as well as mean daily numbers of crying and crying duration in infants during the first months of life. Our results suggest that L. reuteri NCIMB 30351 represents a safe and effective treatment for colic in newborns. Trial registration: ClinicalTrials.gov : NCT04262648. What is Known: • Infantile functional gastrointestinal disorders, such as colic, constipation, diarrhea, and gastroesophageal reflux (regurgitation), often occur in early infancy and, represent one of the causes of significant parental anxiety. • A number of studies have shown that both the composition and diversity of the intestinal microbiota play important roles in the development and function of the gastrointestinal tract. What is New: • The uptake of L. reuteri NCIMB 30351 leads to a significant reduction in the number of regurgitations, feeding-induced constipations, and diarrhea as well as mean daily numbers of crying and crying duration in infants during the first months of life. • L. reuteri positively influences the proportions of prevalent species, while it negatively affects both conditionally pathogenic and commensal microbes in gut microbiota.
Collapse
|
3
|
Román L, Melis-Arcos F, Pröschle T, Saa PA, Garrido D. Genome-scale metabolic modeling of the human milk oligosaccharide utilization by Bifidobacterium longum subsp. infantis. mSystems 2024; 9:e0071523. [PMID: 38363147 PMCID: PMC10949479 DOI: 10.1128/msystems.00715-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/10/2024] [Indexed: 02/17/2024] Open
Abstract
Bifidobacterium longum subsp. infantis is a representative and dominant species in the infant gut and is considered a beneficial microbe. This organism displays multiple adaptations to thrive in the infant gut, regarded as a model for human milk oligosaccharides (HMOs) utilization. These carbohydrates are abundant in breast milk and include different molecules based on lactose. They contain fucose, sialic acid, and N-acetylglucosamine. Bifidobacterium metabolism is complex, and a systems view of relevant metabolic pathways and exchange metabolites during HMO consumption is missing. To address this limitation, a refined genome-scale network reconstruction of this bacterium is presented using a previous reconstruction of B. infantis ATCC 15967 as a template. The latter was expanded based on an extensive revision of genome annotations, current literature, and transcriptomic data integration. The metabolic reconstruction (iLR578) accounted for 578 genes, 1,047 reactions, and 924 metabolites. Starting from this reconstruction, we built context-specific genome-scale metabolic models using RNA-seq data from cultures growing in lactose and three HMOs. The models revealed notable differences in HMO metabolism depending on the functional characteristics of the substrates. Particularly, fucosyl-lactose showed a divergent metabolism due to a fucose moiety. High yields of lactate and acetate were predicted under growth rate maximization in all conditions, whereas formate, ethanol, and 1,2-propanediol were substantially lower. Similar results were also obtained under near-optimal growth on each substrate when varying the empirically observed acetate-to-lactate production ratio. Model predictions displayed reasonable agreement between central carbon metabolism fluxes and expression data across all conditions. Flux coupling analysis revealed additional connections between succinate exchange and arginine and sulfate metabolism and a strong coupling between central carbon reactions and adenine metabolism. More importantly, specific networks of coupled reactions under each carbon source were derived and analyzed. Overall, the presented network reconstruction constitutes a valuable platform for probing the metabolism of this prominent infant gut bifidobacteria.IMPORTANCEThis work presents a detailed reconstruction of the metabolism of Bifidobacterium longum subsp. infantis, a prominent member of the infant gut microbiome, providing a systems view of its metabolism of human milk oligosaccharides.
Collapse
Affiliation(s)
- Loreto Román
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melis-Arcos
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tomás Pröschle
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pedro A. Saa
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute for Mathematical and Computational Engineering, Pontificia Universidad Católica de Chile, Vicuña Mackenna, Santiago, Chile
| | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
4
|
González A, Fullaondo A, Odriozola A. Impact of evolution on lifestyle in microbiome. ADVANCES IN GENETICS 2024; 111:149-198. [PMID: 38908899 DOI: 10.1016/bs.adgen.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
This chapter analyses the interaction between microbiota and humans from an evolutionary point of view. Long-term interactions between gut microbiota and host have been generated as a result of dietary choices through coevolutionary processes, where mutuality of advantage is essential. Likewise, the characteristics of the intestinal environment have made it possible to describe different intrahost evolutionary mechanisms affecting microbiota. For its part, the intestinal microbiota has been of great importance in the evolution of mammals, allowing the diversification of dietary niches, phenotypic plasticity and the selection of host phenotypes. Although the origin of the human intestinal microbial community is still not known with certainty, mother-offspring transmission plays a key role, and it seems that transmissibility between individuals in adulthood also has important implications. Finally, it should be noted that certain aspects inherent to modern lifestyle, including refined diets, antibiotic intake, exposure to air pollutants, microplastics, and stress, could negatively affect the diversity and composition of our gut microbiota. This chapter aims to combine current knowledge to provide a comprehensive view of the interaction between microbiota and humans throughout evolution.
Collapse
Affiliation(s)
- Adriana González
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Asier Fullaondo
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Adrián Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
5
|
Moore ML, Ford JL, Schladweiler MC, Dye JA, Jackson TW, Miller CN. Gut metabolic changes during pregnancy reveal the importance of gastrointestinal region in sample collection. Metabolomics 2024; 20:40. [PMID: 38460019 PMCID: PMC11168590 DOI: 10.1007/s11306-024-02099-x] [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: 01/08/2024] [Accepted: 02/06/2024] [Indexed: 03/11/2024]
Abstract
INTRODUCTION Studies of gastrointestinal physiology and the gut microbiome often consider the influence of intestinal region on experimental endpoints. However, this same consideration is not often applied to the gut metabolome. Understanding the contribution of gut regionality may be critically important to the rapidly changing metabolic environments, such as during pregnancy. OBJECTIVES We sought to characterize the difference in the gut metabolome in pregnant mice stratified by region-comparing the small intestine, cecum, and feces. Pre-pregnancy feces were collected to understand the influence of pregnancy on the fecal metabolome. METHODS Feces were collected from CD-1 female mice before breeding. On gestation day (GD) 18, gut contents were collected from the small intestine, cecum, and descending colon. Metabolites were analyzed with LC-MS/MS using the Biocrates MetaboINDICATOR™ MxP® Quant 500 kit. RESULTS Of the 104 small molecule metabolites meeting analysis criteria, we found that 84 (81%) were differentially abundant based on gut region. The most significant regional comparison observed was between the cecum and small intestines, with 52 (50%) differentially abundant metabolites. Pregnancy itself altered 41 (39.4%) fecal small molecule metabolites. CONCLUSIONS The regional variation observed in the gut metabolome are likely due to the microbial and physiological differences between the different parts of the intestines. Additionally, pregnancy impacts the fecal metabolome, which may be due to evolving needs of both the dam and fetus.
Collapse
Affiliation(s)
- Makala L Moore
- Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Jermaine L Ford
- Chemical Characterization and Exposure Division, Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Mette C Schladweiler
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Janice A Dye
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Thomas W Jackson
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA.
| | - Colette N Miller
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA.
| |
Collapse
|
6
|
Meeusen E, Cao L, Delsing DJ, Groeneveld A, Heerikhuisen M, Schuren F, Boltje TJ. Gram-scale chemical synthesis of galactosyllactoses and their impact on infant gut microbiota in vitro. Org Biomol Chem 2024; 22:2091-2097. [PMID: 38363206 PMCID: PMC10917138 DOI: 10.1039/d3ob02069j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/14/2024] [Indexed: 02/17/2024]
Abstract
Galactooligosaccharides (GOS) are widely used as a supplement in infant nutrition to mimic the beneficial effects found in prebiotic human milk oligosaccharides (HMOs). However, the complexity of the GOS mixture makes it challenging to ascertain which of the GOS components contribute most to their health benefits. Galactosyllactoses (GLs) are lactose-based trisaccharides containing a β-galactopyranosyl residue at the 3'-position (3'galactosyllactose, 3'-GL), 4'-position (4'-galactosyllactose, 4'-GL), or the 6'-position (6'-galactosyllactose, 6'-GL). These GLs are of particular interest as they are present in both GOS mixtures and human milk at early stages of lactation. However, research on the potential health benefits of these individual GLs has been limited. Gram quantities are needed to assess their health benefits but these GLs are not readily available at this scale. In this study, we report the gram-scale chemical synthesis of 3'-GL, 4'-GL, and 6'-GL. All three galactosyllactoses were obtained on a gram scale in good purity from cheap and commercially available lactose. Furthermore, in vitro incubation of GLs with infant faecal microbiota demonstrates that the GLs were able to increase the abundance of Bifidobacterium and stimulate short chain fatty acid production.
Collapse
Affiliation(s)
- Evy Meeusen
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands.
| | - Linqiu Cao
- FrieslandCampina N.V., Amersfoort, The Netherlands
| | | | | | - Margreet Heerikhuisen
- Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Frank Schuren
- Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Thomas J Boltje
- Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands.
| |
Collapse
|
7
|
Erçelik HC, Kaya V. The effects of fermented food consumption in pregnancy on neonatal and infant health: An integrative review. J Pediatr Nurs 2024; 75:173-179. [PMID: 38160470 DOI: 10.1016/j.pedn.2023.12.019] [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: 07/05/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE This study was carried out to determine the effects of fermented food in maternal diet during pregnancy on neonatal and infant health. INTRODUCTION Fermented food consumption positively affects microbiota development. It is widely acknowledged that maternal microbiota is a crucial component in the microbiota formation of the newborn. However, the short-term and long-term effects of fermented food consumption during pregnancy on newborns/infants have not been fully investigated so far. INCLUSION CRITERIA The study included studies that were randomized controlled, quasi-experimental, pre-test and post-test controlled, cohort, descriptive and qualitative studies published in English with full-text access and with "moderate" or "strong" scores in quality assessment. METHODS The researchers conducted research on Pubmed, Google Scholar, Web of Science, Scopus, Clinical Keys, Cochrane and Ebsco-Host databases without any time limitation. RESULTS As a result, 1419 articles were reviewed and five studies were selected among which two studies demonstrated that fermented food consumption during pregnancy may reduce the risk of atopic dermatitis in the infant, and another study indicated that it may reduce the risk of food protein-induced allergic proctocolitis. One cohort study also reported that fermented food consumption during pregnancy improved sleep duration while another cohort study pointed out that it increased the birth weight of infants. CONCLUSION Evidence supports the positive effects of including fermented foods in pregnancy nutrition on neonatal and infant health. Fermented products can be added to the daily diet as an alternative to probiotic supplements. By adding these foods to the nutritional guidelines, awareness of pregnant women can be raised.
Collapse
Affiliation(s)
- Hamide Coşkun Erçelik
- Süleyman Demirel University Research and Application Hospital, Health Research and Application Center, Isparta, Turkey; Pamukkale University Health Science Faculty, Department of Pediatric Nursing, Denizli, Turkey.
| | - Vildan Kaya
- Süleyman Demirel University Research and Application Hospital, Health Research and Application Center, Isparta, Turkey.
| |
Collapse
|
8
|
Zhang W, Zhang Q, Cao Z, Zheng L, Hu W. Physiologically Based Pharmacokinetic Modeling in Neonates: Current Status and Future Perspectives. Pharmaceutics 2023; 15:2765. [PMID: 38140105 PMCID: PMC10747965 DOI: 10.3390/pharmaceutics15122765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Rational drug use in special populations is a clinical problem that doctors and pharma-cists must consider seriously. Neonates are the most physiologically immature and vulnerable to drug dosing. There is a pronounced difference in the anatomical and physiological profiles be-tween neonates and older people, affecting the absorption, distribution, metabolism, and excretion of drugs in vivo, ultimately leading to changes in drug concentration. Thus, dose adjustments in neonates are necessary to achieve adequate therapeutic concentrations and avoid drug toxicity. Over the past few decades, modeling and simulation techniques, especially physiologically based pharmacokinetic (PBPK) modeling, have been increasingly used in pediatric drug development and clinical therapy. This rigorously designed and verified model can effectively compensate for the deficiencies of clinical trials in neonates, provide a valuable reference for clinical research design, and even replace some clinical trials to predict drug plasma concentrations in newborns. This review introduces previous findings regarding age-dependent physiological changes and pathological factors affecting neonatal pharmacokinetics, along with their research means. The application of PBPK modeling in neonatal pharmacokinetic studies of various medications is also reviewed. Based on this, we propose future perspectives on neonatal PBPK modeling and hope for its broader application.
Collapse
Affiliation(s)
| | | | | | - Liang Zheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| |
Collapse
|
9
|
Chang YS, Li CW, Chen L, Wang XA, Lee MS, Chao YH. Early Gut Microbiota Profile in Healthy Neonates: Microbiome Analysis of the First-Pass Meconium Using Next-Generation Sequencing Technology. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1260. [PMID: 37508757 PMCID: PMC10377966 DOI: 10.3390/children10071260] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Gut microbiome development during early life has significant long-term effects on health later in life. The first-pass meconium is not sterile, and it is important to know the initial founder of the subsequent gut microbiome. However, there is limited data on the microbiota profile of the first-pass meconium in healthy neonates. To determine the early gut microbiota profile, we analyzed 39 samples of the first-pass meconium from healthy neonates using 16S rRNA sequencing. Our results showed a similar profile of the microbiota composition in the first-pass meconium samples. Pseudomonas was the most abundant genus in most samples. The evenness of the microbial communities in the first-pass meconium was extremely poor, and the average Shannon diversity index was 1.31. An analysis of the relationship between perinatal characteristics and the meconium microbiome revealed that primigravidae babies had a significantly higher Shannon diversity index (p = 0.041), and the Bacteroidales order was a biomarker for the first-pass meconium of these neonates. The Shannon diversity index was not affected by the mode of delivery, maternal intrapartum antibiotic treatment, prolonged rupture of membranes, or birth weight. Our study extends previous research with further characterization of the gut microbiome in very early life.
Collapse
Affiliation(s)
- Yi-Sheng Chang
- Department of Research and Development, AllBio Life Incorporation, Taichung 402, Taiwan
| | - Chang-Wei Li
- Department of Research and Development, AllBio Life Incorporation, Taichung 402, Taiwan
| | - Ling Chen
- Department of Research and Development, AllBio Life Incorporation, Taichung 402, Taiwan
| | - Xing-An Wang
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Maw-Sheng Lee
- Department of Obstetrics and Gynecology, Lee Women's Hospital, Taichung 406, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Yu-Hua Chao
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Clinical Pathology, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| |
Collapse
|
10
|
De Sales-Millán A, Aguirre-Garrido JF, González-Cervantes RM, Velázquez-Aragón JA. Microbiome-Gut-Mucosal-Immune-Brain Axis and Autism Spectrum Disorder (ASD): A Novel Proposal of the Role of the Gut Microbiome in ASD Aetiology. Behav Sci (Basel) 2023; 13:548. [PMID: 37503995 PMCID: PMC10376175 DOI: 10.3390/bs13070548] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder characterised by deficits in social interaction and communication, as well as restricted and stereotyped interests. Due of the high prevalence of gastrointestinal disorders in individuals with ASD, researchers have investigated the gut microbiota as a potential contributor to its aetiology. The relationship between the microbiome, gut, and brain (microbiome-gut-brain axis) has been acknowledged as a key factor in modulating brain function and social behaviour, but its connection to the aetiology of ASD is not well understood. Recently, there has been increasing attention on the relationship between the immune system, gastrointestinal disorders and neurological issues in ASD, particularly in relation to the loss of specific species or a decrease in microbial diversity. It focuses on how gut microbiota dysbiosis can affect gut permeability, immune function and microbiota metabolites in ASD. However, a very complete study suggests that dysbiosis is a consequence of the disease and that it has practically no effect on autistic manifestations. This is a review of the relationship between the immune system, microbial diversity and the microbiome-gut-brain axis in the development of autistic symptoms severity and a proposal of a novel role of gut microbiome in ASD, where dysbiosis is a consequence of ASD-related behaviour and where dysbiosis in turn accentuates the autistic manifestations of the patients via the microbiome-gut-brain axis in a feedback circuit.
Collapse
Affiliation(s)
- Amapola De Sales-Millán
- División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Lerma, Lerma 52006, Estado de Mexico, Mexico
| | - José Félix Aguirre-Garrido
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana-Lerma, Lerma 52006, Estado de Mexico, Mexico
| | - Rina María González-Cervantes
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana-Lerma, Lerma 52006, Estado de Mexico, Mexico
| | | |
Collapse
|
11
|
Khatoon S, Kalam N, Rashid S, Bano G. Effects of gut microbiota on neurodegenerative diseases. Front Aging Neurosci 2023; 15:1145241. [PMID: 37323141 PMCID: PMC10268008 DOI: 10.3389/fnagi.2023.1145241] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/12/2023] [Indexed: 06/17/2023] Open
Abstract
A progressive degradation of the brain's structure and function, which results in a reduction in cognitive and motor skills, characterizes neurodegenerative diseases (NDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). The morbidity linked to NDs is growing, which poses a severe threat to human being's mental and physical ability to live well. The gut-brain axis (GBA) is now known to have a crucial role in the emergence of NDs. The gut microbiota is a conduit for the GBA, a two-way communication system between the gut and the brain. The myriad microorganisms that make up the gut microbiota can affect brain physiology by transmitting numerous microbial chemicals from the gut to the brain via the GBA or neurological system. The synthesis of neurotransmitters, the immunological response, and the metabolism of lipids and glucose have all been demonstrated to be impacted by alterations in the gut microbiota, such as an imbalance of helpful and harmful bacteria. In order to develop innovative interventions and clinical therapies for NDs, it is crucial to comprehend the participation of the gut microbiota in these conditions. In addition to using antibiotics and other drugs to target particular bacterial species that may be a factor in NDs, this also includes using probiotics and other fecal microbiota transplantation to maintain a healthy gut microbiota. In conclusion, the examination of the GBA can aid in understanding the etiology and development of NDs, which may benefit the improvement of clinical treatments for these disorders and ND interventions. This review indicates existing knowledge about the involvement of microbiota present in the gut in NDs and potential treatment options.
Collapse
Affiliation(s)
- Saima Khatoon
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Nida Kalam
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Summya Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Gulnaz Bano
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
12
|
Martin AJ, Serebrinsky-Duek K, Riquelme E, Saa PA, Garrido D. Microbial interactions and the homeostasis of the gut microbiome: the role of Bifidobacterium. MICROBIOME RESEARCH REPORTS 2023; 2:17. [PMID: 38046822 PMCID: PMC10688804 DOI: 10.20517/mrr.2023.10] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 12/05/2023]
Abstract
The human gut is home to trillions of microorganisms that influence several aspects of our health. This dense microbial community targets almost all dietary polysaccharides and releases multiple metabolites, some of which have physiological effects on the host. A healthy equilibrium between members of the gut microbiota, its microbial diversity, and their metabolites is required for intestinal health, promoting regulatory or anti-inflammatory immune responses. In contrast, the loss of this equilibrium due to antibiotics, low fiber intake, or other conditions results in alterations in gut microbiota composition, a term known as gut dysbiosis. This dysbiosis can be characterized by a reduction in health-associated microorganisms, such as butyrate-producing bacteria, enrichment of a small number of opportunistic pathogens, or a reduction in microbial diversity. Bifidobacterium species are key species in the gut microbiome, serving as primary degraders and contributing to a balanced gut environment in various ways. Colonization resistance is a fundamental property of gut microbiota for the prevention and control of infections. This community competes strongly with foreign microorganisms, such as gastrointestinal pathogens, antibiotic-resistant bacteria, or even probiotics. Resistance to colonization is based on microbial interactions such as metabolic cross-feeding, competition for nutrients, or antimicrobial-based inhibition. These interactions are mediated by metabolites and metabolic pathways, representing the inner workings of the gut microbiota, and play a protective role through colonization resistance. This review presents a rationale for how microbial interactions provide resistance to colonization and gut dysbiosis, highlighting the protective role of Bifidobacterium species.
Collapse
Affiliation(s)
- Alberto J.M. Martin
- Laboratorio de Redes Biológicas, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Santiago 8580702, Chile
| | - Kineret Serebrinsky-Duek
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
| | - Erick Riquelme
- Department of Respiratory Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Pedro A. Saa
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
- Institute for Mathematical and Computational Engineering, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
| |
Collapse
|
13
|
Rosa CD, Altomare A, Terrigno V, Carbone F, Tack J, Cicala M, Guarino MPL. Constipation-Predominant Irritable Bowel Syndrome (IBS-C): Effects of Different Nutritional Patterns on Intestinal Dysbiosis and Symptoms. Nutrients 2023; 15:nu15071647. [PMID: 37049488 PMCID: PMC10096616 DOI: 10.3390/nu15071647] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic functional gastrointestinal disorder characterized by abdominal pain associated with defecation or a change in bowel habits. The pathogenesis of IBS is not completely clear, but it is known to be multifactorial and complex. Endogenous and exogenous factors such as abnormal GI motility, low-grade inflammation, increased epithelial permeability and visceral hypersensitivity, but diet and psychosocial aspects are also recognized as important actors. Furthermore, the interaction between diet and gut microbiota has gained interest as a potential contributor to the pathophysiology of IBS. To date, there is no specific diet for IBS with constipation (IBS-C); however, many studies show that fiber intake, especially soluble fiber such as inulin, could have a positive effect on symptoms. This review aims to evaluate the effects of some nutritional components such as fibers but also functional foods, prebiotics, probiotics and symbiotics on symptoms and microbiota in IBS-C subjects.
Collapse
Affiliation(s)
- Claudia Di Rosa
- Research Unit of Food Science and Human Nutrition, Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy; (C.D.R.)
| | - Annamaria Altomare
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy
- Operative Research Unit of Gastroenterology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200-00128 Roma, Italy
- Correspondence:
| | - Vittoria Terrigno
- Research Unit of Food Science and Human Nutrition, Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy; (C.D.R.)
| | - Florencia Carbone
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Jan Tack
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Michele Cicala
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy
- Operative Research Unit of Gastroenterology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200-00128 Roma, Italy
| | - Michele Pier Luca Guarino
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21-00128 Roma, Italy
- Operative Research Unit of Gastroenterology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200-00128 Roma, Italy
| |
Collapse
|
14
|
A Multiscale Spatiotemporal Model Including a Switch from Aerobic to Anaerobic Metabolism Reproduces Succession in the Early Infant Gut Microbiota. mSystems 2022; 7:e0044622. [PMID: 36047700 PMCID: PMC9600552 DOI: 10.1128/msystems.00446-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The human intestinal microbiota starts to form immediately after birth and is important for the health of the host. During the first days, facultatively anaerobic bacterial species generally dominate, such as Enterobacteriaceae. These are succeeded by strictly anaerobic species, particularly Bifidobacterium species. An early transition to Bifidobacterium species is associated with health benefits; for example, Bifidobacterium species repress growth of pathogenic competitors and modulate the immune response. Succession to Bifidobacterium is thought to be due to consumption of intracolonic oxygen present in newborns by facultative anaerobes, including Enterobacteriaceae. To study if oxygen depletion suffices for the transition to Bifidobacterium species, here we introduced a multiscale mathematical model that considers metabolism, spatial bacterial population dynamics, and cross-feeding. Using publicly available metabolic network data from the AGORA collection, the model simulates ab initio the competition of strictly and facultatively anaerobic species in a gut-like environment under the influence of lactose and oxygen. The model predicts that individual differences in intracolonic oxygen in newborn infants can explain the observed individual variation in succession to anaerobic species, in particular Bifidobacterium species. Bifidobacterium species became dominant in the model by their use of the bifid shunt, which allows Bifidobacterium to switch to suboptimal yield metabolism with fast growth at high lactose concentrations, as predicted here using flux balance analysis. The computational model thus allows us to test the internal plausibility of hypotheses for bacterial colonization and succession in the infant colon. IMPORTANCE The composition of the infant microbiota has a great impact on infant health, but its controlling factors are still incompletely understood. The frequently dominant anaerobic Bifidobacterium species benefit health, e.g., they can keep harmful competitors under control and modulate the intestinal immune response. Controlling factors could include nutritional composition and intestinal mucus composition, as well as environmental factors, such as antibiotics. We introduce a modeling framework of a metabolically realistic intestinal microbial ecology in which hypothetical scenarios can be tested and compared. We present simulations that suggest that greater levels of intraintestinal oxygenation more strongly delay the dominance of Bifidobacterium species, explaining the observed variety of microbial composition and demonstrating the use of the model for hypothesis generation. The framework allowed us to test a variety of controlling factors, including intestinal mixing and transit time. Future versions will also include detailed modeling of oligosaccharide and mucin metabolism.
Collapse
|
15
|
Sivasankari R, Usha B. Reshaping the Gut Microbiota Through Lifestyle Interventions in Women with PCOS: A Review. Indian J Microbiol 2022; 62:351-363. [PMID: 35974920 PMCID: PMC9375820 DOI: 10.1007/s12088-022-01019-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/30/2022] [Indexed: 11/05/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder evolving as a global threat to women's health. However, its multifactorial etiology causes difficulty in eliminating it. The interrelation between the gut microbiota and metabolic disorders has been trending recently, giving rise to new opportunities on the etiology and pathogenesis of PCOS. Lifestyle interventions such as healthy diet, physical exercises, and behavioral interventions such as regulation of stress and sleep cycles have been identified to improve the symptoms of PCOS across the endocrinological, metabolic and psychological scales and are recommended as the first line of treatment for PCOS. The impact of the unhealthy lifestyle factors on intestinal dysbiosis that cause PCOS is summarized in this review. This review also provides an insight on the therapeutic approaches that primarily target the gut microbiota and offers novel gut microflora-associated treatment strategies for PCOS. Further, this survey also highlights the need for the implementation of lifestyle management strategies and strongly recommends a healthy and stress-free lifestyle to promote gut health and manage PCOS.
Collapse
Affiliation(s)
- Ramadurai Sivasankari
- Department of Genetic Engineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Tamil Nadu 603 203 India
| | - Balasundaram Usha
- Department of Genetic Engineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Tamil Nadu 603 203 India
| |
Collapse
|
16
|
Amevor FK, Cui Z, Du X, Feng J, Shu G, Ning Z, Xu D, Deng X, Song W, Wu Y, Cao X, Wei S, He J, Kong F, Du X, Tian Y, Karikari B, Li D, Wang Y, Zhang Y, Zhu Q, Zhao X. Synergy of Dietary Quercetin and Vitamin E Improves Cecal Microbiota and Its Metabolite Profile in Aged Breeder Hens. Front Microbiol 2022; 13:851459. [PMID: 35656004 PMCID: PMC9152675 DOI: 10.3389/fmicb.2022.851459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 04/25/2022] [Indexed: 11/23/2022] Open
Abstract
In the present study, the synergistic effects of quercetin (Q) and vitamin E (E) on cecal microbiota composition and function, as well as the microbial metabolic profile in aged breeder hens were investigated. A total of 400 (65 weeks old) Tianfu breeder hens were randomly allotted to four experimental groups (four replicates per group). The birds were fed diets containing quercetin at 0.4 g/kg, vitamin E (0.2 g/kg), quercetin and vitamin E (QE; 0.4 g/kg and 0.2 g/kg), and a basal diet for a period of 10 wks. After the 10 week experimental period, the cecal contents of 8 aged breeder hens per group were sampled aseptically and subjected to high-throughput 16S rRNA gene sequencing and untargeted metabolomic analysis. The results showed that the relative abundances of phyla Bacteroidota, Firmicutes, and Actinobacteriota were the most prominent among all the dietary groups. Compared to the control group, the relative abundance of the families Bifidobacteriaceae, Lachnospiraceae, Tannerellaceae, Mathonobacteriaceae, Barnesiellaceae, and Prevotellaceae were enriched in the QE group; and Bacteroidaceae, Desulfovibrionaceae, Peptotostretococcaceae, and Fusobacteriaceae were enriched in the Q group, whereas those of Lactobacillaceae, Veillonellaceae, Ruminococcaceae, Akkermansiaceae, and Rikenellaceae were enriched in the E group compared to the control group. Untargeted metabolomics analyses revealed that Q, E, and QE modified the abundance of several metabolites in prominent pathways including ubiquinone and other terpenoid-quinone biosynthesis, regulation of actin cytoskeleton, insulin secretion, pancreatic secretion, nicotine addiction, and metabolism of xenobiotics by cytochrome P450. Furthermore, key cecal microbiota, significantly correlated with important metabolites, for example, (S)-equol positively correlated with Alistipes and Chlamydia in E_vs_C, and negatively correlated with Olsenella, Paraprevotella, and Mucispirillum but, a contrary trend was observed with Parabacteroides in QE_vs_C. This study establishes that the synergy of quercetin and vitamin E alters the cecal microbial composition and metabolite profile in aged breeder hens, which lays a foundation for chicken improvement programs.
Collapse
Affiliation(s)
- Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Zhifu Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xiaxia Du
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jing Feng
- Institute of Animal Husbandry and Veterinary Medicine, College of Agriculture and Animal Husbandry, Tibet Autonomous Region, China
| | - Gang Shu
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zifan Ning
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Dan Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xun Deng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Weizhen Song
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Youhao Wu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xueqing Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Shuo Wei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Juan He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Fanli Kong
- College of Life Science, Sichuan Agricultural University, Ya’an, China
| | - Xiaohui Du
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yaofu Tian
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Benjamin Karikari
- Key Laboratory of Biology and Genetics and Breeding for Soybean, Nanjing Agricultural University, Nanjing, China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Xiaoling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
17
|
The Intestinal Dysbiosis of Mothers with Gestational Diabetes Mellitus (GDM) and Its Impact on the Gut Microbiota of Their Newborns. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2021; 2021:3044534. [PMID: 34603565 PMCID: PMC8481071 DOI: 10.1155/2021/3044534] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/11/2021] [Indexed: 02/06/2023]
Abstract
Gestational diabetes mellitus (GDM) is defined as “diagnosed as impaired glucose tolerance for the first time during pregnancy,” which can lead to adverse pregnancy outcomes and produces divergent effects on mothers and newborns. In recent years, with the continuous expansion of obese people, GDM shows an upward trend. The abundant and diverse members of the human gut microbiota exert critical roles in the maintenance of human health. Studies have shown that GDM may be associated with disordered gut microbiota in both mothers and newborns. Taking into account the potential effects on maternal and consequently neonatal health, in this review, we analyzed the available data and discussed the current knowledge about the potential relationship between GDM and intestinal dysbiosis in mothers and newborns. In addition, we also discussed the influencing factors derived from GDM mothers on the gut microbiome of their newborns, including the vertical transmission of microbiota from mothers, the alteration of milk components of GDM mothers, and using of probiotics. Hoping that new insights into the role of the gut microbiota in GDM could lead to the development of integrated strategies to prevent and treat these metabolic disorders.
Collapse
|
18
|
Dwivedi M, Powali S, Rastogi S, Singh A, Gupta DK. Microbial community in human gut: a therapeutic prospect and implication in health and diseases. Lett Appl Microbiol 2021; 73:553-568. [PMID: 34365651 DOI: 10.1111/lam.13549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
The interest in the working and functionality of the human gut microbiome has increased drastically over the years. Though the existence of gut microbes has long been speculated for long over the last few decades, a lot of research has sprung up in studying and understanding the role of gut microbes in the human digestive tract. The microbes present in the gut are highly instrumental in maintaining the metabolism in the body. Further research is going on in this field to understand how gut microbes can be employed as potential sources of novel therapeutics; moreover, probiotics have also elucidated their significant place in this direction. As regards the clinical perspective, microbes can be engineered to afford defence mechanisms while interacting with foreign pathogenic bodies. More investigations in this field may assist us to evaluate and understand how these cells communicate with human cells and promote immune interactions. Here we elaborate on the possible implication of human gut microbiota into the immune system as well as explore the probiotics in the various human ailments. Comprehensive information on the human gut microbiome at the same platform may contribute effectively to our understanding of the human microbiome and possible mechanisms of associated human diseases.
Collapse
Affiliation(s)
- M Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - S Powali
- Maulana Abdul Kalam Azad University of Technology, Kolkatta, India
| | - S Rastogi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - A Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - D K Gupta
- Department of Biochemistry, University of Allahabad, Prayagraj, India
| |
Collapse
|
19
|
Mukonowenzou NC, Adeshina KA, Donaldson J, Ibrahim KG, Usman D, Erlwanger KH. Medicinal Plants, Phytochemicals, and Their Impacts on the Maturation of the Gastrointestinal Tract. Front Physiol 2021; 12:684464. [PMID: 34393812 PMCID: PMC8363294 DOI: 10.3389/fphys.2021.684464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
The gastrointestinal tract (GIT) is the first point of contact for ingested substances and thus represents a direct interface with the external environment. Apart from food processing, this interface plays a significant role in immunity and contributes to the wellbeing of individuals through the brain-gut-microbiota axis. The transition of life from the in utero environment, to suckling and subsequent weaning has to be matched by phased development and maturation of the GIT; from an amniotic fluid occupancy during gestation, to the milk in the suckling state and ultimately solid food ingestion at weaning. This phased maturation of the GIT can be affected by intrinsic and extrinsic factors, including diet. Despite the increasing dietary inclusion of medicinal plants and phytochemicals for health benefits, a dearth of studies addresses their impact on gut maturation. In this review we focus on some recent findings mainly on the positive impact of medicinal plants and phytochemicals in inducing precocious maturation of the GIT, not only in humans but in pertinent animals. We also discuss Paneth cells as mediators and potential markers of GIT maturation.
Collapse
Affiliation(s)
| | - Kehinde Ahmad Adeshina
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Sokoto, Nigeria
| | - Janine Donaldson
- Faculty of Health Sciences, School of Physiology, University of the Witwatersrand, Johannesburg, Johannesburg, South Africa
| | - Kasimu Ghandi Ibrahim
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Sokoto, Nigeria
| | - Dawoud Usman
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Sokoto, Nigeria
| | - Kennedy Honey Erlwanger
- Faculty of Health Sciences, School of Physiology, University of the Witwatersrand, Johannesburg, Johannesburg, South Africa
| |
Collapse
|
20
|
Zhang S, Li T, Xie J, Zhang D, Pi C, Zhou L, Yang W. Gold standard for nutrition: a review of human milk oligosaccharide and its effects on infant gut microbiota. Microb Cell Fact 2021; 20:108. [PMID: 34049536 PMCID: PMC8162007 DOI: 10.1186/s12934-021-01599-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/21/2021] [Indexed: 02/08/2023] Open
Abstract
Human milk is the gold standard for nutrition of infant growth, whose nutritional value is mainly attributed to human milk oligosaccharides (HMOs). HMOs, the third most abundant component of human milk after lactose and lipids, are complex sugars with unique structural diversity which are indigestible by the infant. Acting as prebiotics, multiple beneficial functions of HMO are believed to be exerted through interactions with the gut microbiota either directly or indirectly, such as supporting beneficial bacteria growth, anti-pathogenic effects, and modulation of intestinal epithelial cell response. Recent studies have highlighted that HMOs can boost infants health and reduce disease risk, revealing potential of HMOs in food additive and therapeutics. The present paper discusses recent research in respect to the impact of HMO on the infant gut microbiome, with emphasis on the molecular basis of mechanism underlying beneficial effects of HMOs.
Collapse
Affiliation(s)
- Shunhao Zhang
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tianle Li
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jing Xie
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Demao Zhang
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Caixia Pi
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lingyun Zhou
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, 610041, China.
| | - Wenbin Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, Department of Medical Affairs, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
21
|
Shaterian N, Abdi F, Ghavidel N, Alidost F. Role of cesarean section in the development of neonatal gut microbiota: A systematic review. Open Med (Wars) 2021; 16:624-639. [PMID: 33869784 PMCID: PMC8035494 DOI: 10.1515/med-2021-0270] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Background The delivery mode is one of the factors affecting the type of colonization of the human gut. Gut colonization affects all stages of the human life cycle, and the type of gut microbiome can contribute to immune system function, the development of some diseases, and brain development; and it has a significant impact on a newborn’s growth and development. Methods Terms defined as MeSH keywords were searched by the databases, and web search engines such as PubMed, ClinicalTrials.gov, Embase, Scopus, ProQuest, Web of Science, and Google Scholar were searched between 2010 and 2020. The quality of each study was assessed according to the Newcastle–Ottawa scale, and seven eligible and high-quality studies were analyzed. Finding The abundances of Bacteroides and Bifidobacterium during the first 3 months of life; Lactobacillus and Bacteroides during the second 3 months of life; Bacteroides and Bifidobacterium during the second 6 months of life; and Bacteroides, Enterobacter, and Streptococcus after the first year of life were higher in vaginal delivery-born infants. While infants born by cesarean section (CS) had higher abundances of Clostridium and Lactobacillus during the first 3 months of life, Enterococcus and Clostridium during the second 3 months of life, and Lactobacillus and Staphylococcus after the first year of life. Discussion Delivery mode can affect the type of the human intestinal microbiota. The CS-born babies had lower colonization rates of Bifidobacterium and Bacteroides, but they had higher colonization rates of Clostridium, Lactobacillus, Enterobacter, Enterococcus, and Staphylococcus. Given the effect of microbiota colonization on neonatal health, it is therefore recommended to conduct further studies in order to investigate the effect of the colonization on the delivery mode and on baby’s growth and development. Application to practice The aim of this study was to investigate the role of CS in the development of the neonatal gut microbiota.
Collapse
Affiliation(s)
- Negin Shaterian
- Student Research Committee, Nursing and Midwifery faculty, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Abdi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Nooshin Ghavidel
- Social Determinants of Health Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farzane Alidost
- Student Research Committee, School of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Escherichia coli Nissle 1917 Enhances Innate and Adaptive Immune Responses in a Ciprofloxacin-Treated Defined-Microbiota Piglet Model of Human Rotavirus Infection. mSphere 2021; 6:6/2/e00074-21. [PMID: 33789939 PMCID: PMC8546683 DOI: 10.1128/msphere.00074-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human rotavirus (HRV) infection is a major cause of gastroenteritis in children worldwide. Broad-spectrum antibiotic-induced intestinal microbial imbalance and the ensuing immune-metabolic dysregulation contribute to the persistence of HRV diarrhea. Escherichia coli Nissle 1917 (EcN), a Gram-negative probiotic, was shown to be a potent immunostimulant and alleviated HRV-induced diarrhea in monocolonized gnotobiotic (Gn) piglets. Our goal was to determine how EcN modulates immune responses in ciprofloxacin (Cipro)-treated Gn piglets colonized with a defined commensal microbiota (DM) and challenged with virulent HRV (VirHRV). Cipro given in therapeutic doses for a short term reduced serum and intestinal total and HRV-specific antibody titers, while EcN treatment alleviated this effect. Similarly, EcN treatment increased the numbers of total immunoglobulin-secreting cells, HRV-specific antibody-secreting cells, activated antibody-forming cells, resting/memory antibody-forming B cells, and naive antibody-forming B cells in systemic and/or intestinal tissues. Decreased levels of proinflammatory but increased levels of immunoregulatory cytokines and increased frequencies of Toll-like receptor-expressing cells were evident in the EcN-treated VirHRV-challenged group. Moreover, EcN treatment increased the frequencies of T helper and T cytotoxic cells in systemic and/or intestinal tissues pre-VirHRV challenge and the frequencies of T helper cells, T cytotoxic cells, effector T cells, and T regulatory cells in systemic and/or intestinal tissues postchallenge. Moreover, EcN treatment increased the frequencies of systemic and mucosal conventional and plasmacytoid dendritic cells, respectively, and the frequencies of systemic natural killer cells. Our findings demonstrated that Cipro use altered immune responses of DM-colonized neonatal Gn pigs, while EcN supplementation rescued these immune parameters partially or completely. IMPORTANCE Rotavirus (RV) is a primary cause of malabsorptive diarrhea in children and is associated with significant morbidity and mortality, especially in developing countries. The use of antibiotics exacerbates intestinal microbial imbalance and results in the persistence of RV-induced diarrhea. Probiotics are now being used to treat enteric infections and ulcerative colitis. We showed previously that probiotics partially protected gnotobiotic (Gn) piglets against human RV (HRV) infection and decreased the severity of diarrhea by modulating immune responses. However, the interactions between antibiotic and probiotic treatments and HRV infection in the context of an established gut microbiota are poorly understood. In this study, we developed a Gn pig model to study antibiotic-probiotic-HRV interactions in the context of a defined commensal microbiota (DM) that mimics aspects of the infant gut microbiota. Our results provide valuable information that will contribute to the treatment of antibiotic- and/or HRV-induced diarrhea and may be applicable to other enteric infections in children.
Collapse
|
23
|
Salvadori M, Tsalouchos A. Microbiota, renal disease and renal transplantation. World J Transplant 2021; 11:16-36. [PMID: 33816144 PMCID: PMC8009061 DOI: 10.5500/wjt.v11.i3.16] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/06/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Aim of this frontier review has been to highlight the role of microbiota in healthy subjects and in patients affected by renal diseases with particular reference to renal transplantation. The microbiota has a relevant role in conditioning the healthy status and the diseases. In particular gut microbiota is essential in the metabolism of food and has a relevant role for its relationship with the immune system. The indigenous microbiota in patients with chronic renal failure is completely different than that of the healthy subjects and pathobionts appear. This abnormality in microbiota composition is called dysbiosis and may cause a rapid deterioration of the renal function both for activating the immune system and producing large quantity of uremic toxins. Similarly, after renal trans-plantation the microbiota changes with the appearance of pathobionts, principally in the first period because of the assumption of immunosuppressive drugs and antibiotics. These changes may deeply interfere with the graft outcome causing acute rejection, renal infections, diarrhea, and renal interstitial fibrosis. In addition, change in the microbiota may modify the metabolism of immuno-suppressive drugs causing in some patients the need of modifying the immunosuppressant dosing. The restoration of the indigenous microbiota after transplantation is important, either to avoiding the complications that impair the normal renal graft, and because recent studies have documented the role of an indigenous microbiota in inducing tolerance towards the graft. The use of prebiotics, probiotics, smart bacteria and diet modification may restore the indigenous microbiota, but these studies are just at their beginning and more data are needed to draw definitive conclusions.
Collapse
Affiliation(s)
- Maurizio Salvadori
- Department of Transplantation Renal Unit, Careggi University Hospital, Florence 50139, Italy
| | - Aris Tsalouchos
- Nephrology and Dialysis Unit, Saints Cosmas and Damian Hospital, Pescia 51017, Italy
| |
Collapse
|
24
|
Liu F, Yan J, Wang X, Wang C, Chen L, Li Y, Chen J, Guo H. Maternal Fucosyltransferase 2 Status Associates with the Profiles of Human Milk Oligosaccharides and the Fecal Microbiota Composition of Breastfed Infants. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:3032-3043. [PMID: 33677972 DOI: 10.1021/acs.jafc.0c04575] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Human milk oligosaccharides (HMOs) play key roles in shaping infant fecal microbiota, and HMOs profiles have been reported to vary according to the mother's glycosyltransferase phenotype. In this study, the profiles of HMOs in human milk from secretor or non-secretor mothers collected at 2 months postpartum were analyzed by liquid chromatography quadrupole time-of-flight tandem mass spectrometry. 16S rRNA sequencing was used to characterize the fecal microbiota of breastfed infants. The amount of total and fucosylated HMOs were higher in secretor than non-secretor mothers, while Bifidobacterium genus were highly enriched in infants fed by non-secretor mothers. Associations between HMOs and infant fecal microbiota showed that the relative abundance of Bifidobacterium-OTU158 was positively associated with 2'-fucosyllactose and 3-fucosyllactose, and Bifidobacterium-OTU90 was negatively associated with lacto-N-difucohexaose. The present study provides the HMO profiles from Chinese mothers and their associations with infant fecal microbiota composition, suggesting that HMO compositions are associated with different Bifidobacterium strains in species-specific manner.
Collapse
Affiliation(s)
- Fan Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jingyu Yan
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116011, China
| | - Xifan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Chenyuan Wang
- Key Laboratory of Functional Dairy, China Agricultural University, Beijing 100083, China
| | - Lingli Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yiran Li
- Key Laboratory of Functional Dairy, China Agricultural University, Beijing 100083, China
| | - Jianliang Chen
- Key Laboratory of Functional Dairy, China Agricultural University, Beijing 100083, China
| | - Huiyuan Guo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Functional Dairy, China Agricultural University, Beijing 100083, China
| |
Collapse
|
25
|
Abstract
Scientists have invested considerable resources in the study of the microbiota of the human body. These microorganisms play pivotal roles in immunity and disease. Of which, probiotics are live beneficial microorganisms that keep your intestinal or lung microbiota healthy, and occupy a special role in combating the infections. Thus, it is critical to understand their contributions to these processes. Technology can facilitate advanced studies of the microbiota, including how it develops and its positive and negatives effects on the immune system. This paper investigates how several factors (e.g. birth delivery mode, metabolic activities, types of microorganisms, and immune system interactions) affect the microbiota, particularly in early life. The paper also discusses how gastrointestinal microbes in particular may be associated with certain disease processes, such as those related to schizophrenia, autism, and diabetes. Clinical studies show that certain probiotic strains, like Lactobacillus rhamnosus GG and Bifidobacterium animalis ssp. lactis help to prevent infection of pathogenic organisms (both bacterial and viral). This research may yield crucial contributions to disease prevention and public health. The dysbiosis may result in changes in the acquired immunity later on. The probiotic strains can prevent viral replication during SARS-CoV-2 or COVID-19 infection by reducing proinflammatory cytokines. There has been much interest into the intestinal flora as proposed by the diversity, volume, and proposed role in disease. Future research in the field of microbiome should be done in order to uncover their association to gut virome by noting both their influence on each other and relevant health and disease.
Collapse
|
26
|
Tay CJX, Ta LDH, Ow Yeong YX, Yap GC, Chu JJH, Lee BW, Tham EH. Role of Upper Respiratory Microbiota and Virome in Childhood Rhinitis and Wheeze: Collegium Internationale Allergologicum Update 2021. Int Arch Allergy Immunol 2021; 182:265-276. [PMID: 33588407 DOI: 10.1159/000513325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 11/19/2022] Open
Abstract
There is emerging evidence that the respiratory microbiota influences airway health, and there has been intense research interest in its role in respiratory infections and allergic airway disorders. This review aims to summarize current knowledge of nasal microbiome and virome and their associations with childhood rhinitis and wheeze. The healthy infant nasal microbiome is dominated by Corynebacteriaceae and Staphylococcaceae. In contrast, infants who subsequently develop respiratory disorders are depleted of these microbes and are instead enriched with Proteobacteria spp. Although human rhinovirus and human respiratory syncytial virus are well-documented major viral pathogens that trigger rhinitis and wheezing disorders in infants, recent limited data indicate that bacteriophages may have a role in respiratory health. Future work investigating the interplay between commensal microbiota, virome, and host immunological responses is an important step toward understanding the dynamics of the nasal community in order to develop a strategical approach to combat these common childhood respiratory disorders.
Collapse
Affiliation(s)
- Carina Jing Xuan Tay
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Le Duc Huy Ta
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yu Xiang Ow Yeong
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gaik Chin Yap
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Bee Wah Lee
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elizabeth Huiwen Tham
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore, .,Khoo Teck Puat- National University Children's Medical Institute, National University Health System, Singapore, Singapore, .,Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore,
| |
Collapse
|
27
|
Kumar H, Collado MC, Wopereis H, Salminen S, Knol J, Roeselers G. The Bifidogenic Effect Revisited-Ecology and Health Perspectives of Bifidobacterial Colonization in Early Life. Microorganisms 2020; 8:E1855. [PMID: 33255636 PMCID: PMC7760687 DOI: 10.3390/microorganisms8121855] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Extensive microbial colonization of the infant gastrointestinal tract starts after parturition. There are several parallel mechanisms by which early life microbiome acquisition may proceed, including early exposure to maternal vaginal and fecal microbiota, transmission of skin associated microbes, and ingestion of microorganisms present in breast milk. The crucial role of vertical transmission from the maternal microbial reservoir during vaginal delivery is supported by the shared microbial strains observed among mothers and their babies and the distinctly different gut microbiome composition of caesarean-section born infants. The healthy infant colon is often dominated by members of the keystone genus Bifidobacterium that have evolved complex genetic pathways to metabolize different glycans present in human milk. In exchange for these host-derived nutrients, bifidobacteria's saccharolytic activity results in an anaerobic and acidic gut environment that is protective against enteropathogenic infection. Interference with early-life microbiota acquisition and development could result in adverse health outcomes. Compromised microbiota development, often characterized by decreased abundance of Bifidobacterium species has been reported in infants delivered prematurely, delivered by caesarean section, early life antibiotic exposure and in the case of early life allergies. Various microbiome modulation strategies such as probiotic, prebiotics, synbiotics and postbiotics have been developed that are able to generate a bifidogenic shift and help to restore the microbiota development. This review explores the evolutionary ecology of early-life type Bifidobacterium strains and their symbiotic relationship with humans and discusses examples of compromised microbiota development in which stimulating the abundance and activity of Bifidobacterium has demonstrated beneficial associations with health.
Collapse
Affiliation(s)
- Himanshu Kumar
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (H.K.); (H.W.); (J.K.)
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-Spanish National Research Council (IATA-CSIC), Paterna, 46980 Valencia, Spain;
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20500 Turku, Finland;
| | - Harm Wopereis
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (H.K.); (H.W.); (J.K.)
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20500 Turku, Finland;
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (H.K.); (H.W.); (J.K.)
- Laboratory for Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Guus Roeselers
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (H.K.); (H.W.); (J.K.)
| |
Collapse
|
28
|
Chia LW, Mank M, Blijenberg B, Aalvink S, Bongers RS, Stahl B, Knol J, Belzer C. Bacteroides thetaiotaomicron Fosters the Growth of Butyrate-Producing Anaerostipes caccae in the Presence of Lactose and Total Human Milk Carbohydrates. Microorganisms 2020; 8:E1513. [PMID: 33019531 PMCID: PMC7601031 DOI: 10.3390/microorganisms8101513] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/20/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022] Open
Abstract
The development of infant gut microbiota is strongly influenced by nutrition. Human milk oligosaccharides (HMOSs) in breast milk selectively promote the growth of glycan-degrading microbes, which lays the basis of the microbial network. In this study, we investigated the trophic interaction between Bacteroides thetaiotaomicron and the butyrate-producing Anaerostipes caccae in the presence of early-life carbohydrates. Anaerobic bioreactors were set up to study the monocultures of B. thetaiotaomicron and the co-cultures of B. thetaiotaomicron with A. caccae in minimal media supplemented with lactose or a total human milk carbohydrate fraction. Bacterial growth (qPCR), metabolites (HPLC), and HMOS utilization (LC-ESI-MS2) were monitored. B. thetaiotaomicron displayed potent glycan catabolic capability with differential preference in degrading specific low molecular weight HMOSs, including the neutral trioses (2'-FL and 3-FL), neutral tetraoses (DFL, LNT, LNnT), neutral pentaoses (LNFP I, II, III, V), and acidic trioses (3'-SL and 6'-SL). In contrast, A. caccae was not able to utilize lactose and HMOSs. However, the signature metabolite of A. caccae, butyrate, was detected in co-culture with B. thetaiotaomicron. As such, A. caccae cross-fed on B. thetaiotaomicron-derived monosaccharides, acetate, and d-lactate for growth and concomitant butyrate production. This study provides a proof of concept that B. thetaiotaomicron could drive the butyrogenic metabolic network in the infant gut.
Collapse
Affiliation(s)
- Loo Wee Chia
- Laboratory of Microbiology, Wageningen University and Research, 6708 WE Wageningen, The Netherlands; (L.W.C.); (S.A.); (J.K.)
| | - Marko Mank
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (M.M.); (B.B.); (R.S.B.); (B.S.)
| | - Bernadet Blijenberg
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (M.M.); (B.B.); (R.S.B.); (B.S.)
| | - Steven Aalvink
- Laboratory of Microbiology, Wageningen University and Research, 6708 WE Wageningen, The Netherlands; (L.W.C.); (S.A.); (J.K.)
| | - Roger S. Bongers
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (M.M.); (B.B.); (R.S.B.); (B.S.)
| | - Bernd Stahl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (M.M.); (B.B.); (R.S.B.); (B.S.)
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University and Research, 6708 WE Wageningen, The Netherlands; (L.W.C.); (S.A.); (J.K.)
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (M.M.); (B.B.); (R.S.B.); (B.S.)
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, 6708 WE Wageningen, The Netherlands; (L.W.C.); (S.A.); (J.K.)
| |
Collapse
|
29
|
Mukherji R, Weinberg BA. The gut microbiome and potential implications for early-onset colorectal cancer. COLORECTAL CANCER 2020. [DOI: 10.2217/crc-2020-0007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recently, there has been an unexpected trend toward increased incidence of colorectal cancer in younger individuals, particularly distal colon and rectal cancer in those under age 50. There is evidence to suggest that the human gut microbiome may play a role in carcinogenesis. The microbiome is dynamic and varies with age, geography, ethnicity and diet. Certain bacteria such as Fusobacterium nucleatum have been implicated in the development of colorectal and other gastrointestinal cancers. Recent data suggest that bacteria can alter the inflammatory and immune environment, influencing carcinogenesis, lack of treatment response and prognosis. Studies to date focus on older patients. Because the microbiome varies with age, it could be a potential explanation for the rise in early-onset colorectal cancer.
Collapse
Affiliation(s)
- Reetu Mukherji
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057 USA
| | - Benjamin A Weinberg
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057 USA
| |
Collapse
|
30
|
Salminen S, Stahl B, Vinderola G, Szajewska H. Infant Formula Supplemented with Biotics: Current Knowledge and Future Perspectives. Nutrients 2020; 12:E1952. [PMID: 32629970 PMCID: PMC7400136 DOI: 10.3390/nu12071952] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/06/2023] Open
Abstract
Breastfeeding is natural and the optimal basis of infant nutrition and development, with many benefits for maternal health. Human milk is a dynamic fluid fulfilling an infant's specific nutritional requirements and guiding the growth, developmental, and physiological processes of the infant. Human milk is considered unique in composition, and it is influenced by several factors, such as maternal diet and health, body composition, and geographic region. Human milk stands as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as oligosaccharides, bacteria, and bacterial metabolites. The objective of this narrative review is to discuss the most recent developments in infant formula with a special focus on human milk oligosaccharides and postbiotics.
Collapse
Affiliation(s)
- Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland;
| | - Bernd Stahl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands;
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santiago del Estero 2829, Santa Fe 3000, Argentina;
| | - Hania Szajewska
- Department of Paediatrics at the Medical University of Warsaw, 02091 Warsaw, Poland
| |
Collapse
|
31
|
Moles L, Otaegui D. The Impact of Diet on Microbiota Evolution and Human Health. Is Diet an Adequate Tool for Microbiota Modulation? Nutrients 2020; 12:nu12061654. [PMID: 32498430 PMCID: PMC7352211 DOI: 10.3390/nu12061654] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 02/08/2023] Open
Abstract
The human microbiome is emerging as an interesting field in research into the prevention of health problems and recovery from illness in humans. The complex ecosystem formed by the microbiota is continuously interacting with its host and the environment. Diet could be assumed to be one of the most prominent factors influencing the microbiota composition. Nevertheless, and in spite of numerous strategies proposed to modulate the human microbiota through dietary means, guidelines to achieve this goal have yet to be established. This review assesses the correlation between social and dietary changes over the course of human evolution and the adaptation of the human microbiota to those changes. In addition, it discusses the main dietary strategies for modulating the microbiota and the difficulties of putting them properly into practice.
Collapse
|
32
|
Development and Functions of the Infant Gut Microflora: Western vs. Indian Infants. Int J Pediatr 2020; 2020:7586264. [PMID: 32454840 PMCID: PMC7229554 DOI: 10.1155/2020/7586264] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/07/2020] [Indexed: 12/29/2022] Open
Abstract
The human gut is colonized by trillions of bacteria as well as other microorganisms, collectively referred to as the “gut microflora.” This microflora plays an important role in metabolism as well as immunity, and alterations in its normal composition and pattern of colonization can disturb the development and functioning of the immune system, predisposing the individual to several diseases. Neonates acquire their gut microflora from the mother as well as the surroundings, and as the infant grows, the gut microflora undergoes several changes, ultimately acquiring an adult-like composition. Characterization of the gut microflora of healthy infants is important to protect infants from infectious diseases. Furthermore, formulation of prebiotics and probiotics for boosting infant immunity in a specific population also requires prior knowledge of the normal gut microflora in a healthy infant in that population. To this end, several studies have been performed on Western infants; however, the gut microflora of Indian infants is as yet insufficiently studied. Moreover, there has been no comparative analysis of the development and characteristics of the infant gut microflora between the two populations. In this review, we discuss the development and maturation of the infant gut microflora and its effect on immunity, as well as the factors affecting the patterns of colonization. In addition, we compare the patterns of colonization of gut microflora between Western and Indian infants based on the available literature in an attempt to identify the extent of similarity or difference between the two populations.
Collapse
|
33
|
Kurath-Koller S, Neumann C, Moissl-Eichinger C, Kraschl R, Kanduth C, Hopfer B, Pausan MR, Urlesberger B, Resch B. Hospital Regimens Including Probiotics Guide the Individual Development of the Gut Microbiome of Very Low Birth Weight Infants in the First Two Weeks of Life. Nutrients 2020; 12:E1256. [PMID: 32354144 PMCID: PMC7281991 DOI: 10.3390/nu12051256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/23/2020] [Accepted: 04/25/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND It is unknown to what extent the microbiome of preterm infants is influenced by hospital regimens including the use of different probiotics when it comes to the prevention of necrotizing enterocolitis (NEC). METHODS Prospective controlled multicenter cohort study including very low birth weight infants from three neonatal intensive care units (NICUs) between October 2015 and March 2017. During this time span, stool was sampled every other day during the first two weeks and samples were subjected to amplicon-based microbiome analyses. Out of these, seventeen negative controls were processed (German Registry of Clinical Trials (No.: DRKS00009290)). RESULTS The groups (3 × 18 infants) showed no statistically significant difference regarding gestational age, birth weight, APGAR scores and oxygen demand. 2029 different taxa were detected, including Enterococcus and Staphylococcus, as well as the probiotic genera Lactobacillus and Bifidobacterium predominating. The bacterial load was found to increase earlier on when probiotics were used. Without probiotics administration, Lactobacillus and Bifidobacterium contributed only marginally to the fecal microbiome. Some infants did not respond to probiotic administration. The samples from all centers participating reached a very similar diversity after two weeks while the microbiome samples from all three centers clustered significantly yet varied from each other. CONCLUSION Probiotics proved to be safe and initiated an earlier increase of bacterial load (with marked individual divergences), which might play a crucial role in the prevention of neonatal morbidities. Meconium was found not to be free of bacterial DNA, and oral antibiotics did not influence the fecal microbiome development negatively, and hospital regimes led to a center-specific, distinct cluster formation.
Collapse
Affiliation(s)
- Stefan Kurath-Koller
- Division of Neonatology, Department of Pediatrics, Medical University of Graz, Austria Auenbruggerplatz 34/2, 8036 Graz, Austria
- Division of Pediatric Cardiology, Department of Pediatrics, Medical University of Graz, 8036 Graz, Austria
| | - Charlotte Neumann
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University Graz, 8010 Graz, Austria
| | - Christine Moissl-Eichinger
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University Graz, 8010 Graz, Austria
- Biotechmed Graz, 8010 Graz, Austria
| | - Raimund Kraschl
- Department of Pediatrics, General Hospital Klagenfurt am Wörthersee, 9020 Klagenfurt, Austria
| | - Claudia Kanduth
- Department of Pediatrics, General Hospital Klagenfurt am Wörthersee, 9020 Klagenfurt, Austria
| | - Barbara Hopfer
- Department of Pediatrics, General Hospital Hochsteiermark, 8700 Leoben, Austria
| | - Manuela-Raluca Pausan
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University Graz, 8010 Graz, Austria
| | - Berndt Urlesberger
- Division of Neonatology, Department of Pediatrics, Medical University of Graz, Austria Auenbruggerplatz 34/2, 8036 Graz, Austria
| | - Bernhard Resch
- Division of Neonatology, Department of Pediatrics, Medical University of Graz, Austria Auenbruggerplatz 34/2, 8036 Graz, Austria
- Research Unit for Neonatal Infectious Diseases and Epidemiology, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
34
|
The association between breastmilk oligosaccharides and faecal microbiota in healthy breastfed infants at two, six, and twelve weeks of age. Sci Rep 2020; 10:4270. [PMID: 32144305 PMCID: PMC7060319 DOI: 10.1038/s41598-020-61024-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/14/2020] [Indexed: 02/07/2023] Open
Abstract
Several factors affect gut microbiota development in early life, among which breastfeeding plays a key role. We followed 24 mother-infant pairs to investigate the associations between concentrations of selected human milk oligosaccharides (HMOs) in breastmilk, infant faeces, and the faecal microbiota composition in healthy, breastfed infants at two, six and 12 weeks of age. Lactation duration had a significant effect on breastmilk HMO content, which decreased with time, except for 3-fucosyllactose (3FL) and Lacto-N-fucopentaose III (LNFP III). We confirmed that microbiota composition was strongly influenced by infant age and was associated with mode of delivery and breastmilk LNFP III concentration at two weeks, with infant sex, delivery mode, and concentrations of 3′sialyllactose (3′SL) in milk at six weeks, and infant sex and Lacto-N-hexaose (LNH) in milk at 12 weeks of age. Correlations between levels of individual breastmilk HMOs and relative abundance of OTUs found in infant faeces, including the most predominant Bifidobacterium OTUs, were weak and varied with age. The faecal concentration of HMOs decreased with age and were strongly and negatively correlated with relative abundance of OTUs within genera Bifidobacterium, Parabacteroides, Escherichia-Shigella, Bacteroides, Actinomyces, Veillonella, Lachnospiraceae Incertae Sedis, and Erysipelotrichaceae Incertae Sedis, indicating the likely importance of these taxa for HMO metabolism in vivo.
Collapse
|
35
|
Zhu S, Jiang Y, Xu K, Cui M, Ye W, Zhao G, Jin L, Chen X. The progress of gut microbiome research related to brain disorders. J Neuroinflammation 2020; 17:25. [PMID: 31952509 PMCID: PMC6969442 DOI: 10.1186/s12974-020-1705-z] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
There is increasing evidence showing that the dynamic changes in the gut microbiota can alter brain physiology and behavior. Cognition was originally thought to be regulated only by the central nervous system. However, it is now becoming clear that many non-nervous system factors, including the gut-resident bacteria of the gastrointestinal tract, regulate and influence cognitive dysfunction as well as the process of neurodegeneration and cerebrovascular diseases. Extrinsic and intrinsic factors including dietary habits can regulate the composition of the microbiota. Microbes release metabolites and microbiota-derived molecules to further trigger host-derived cytokines and inflammation in the central nervous system, which contribute greatly to the pathogenesis of host brain disorders such as pain, depression, anxiety, autism, Alzheimer’s diseases, Parkinson’s disease, and stroke. Change of blood–brain barrier permeability, brain vascular physiology, and brain structure are among the most critical causes of the development of downstream neurological dysfunction. In this review, we will discuss the following parts:
Overview of technical approaches used in gut microbiome studies Microbiota and immunity Gut microbiota and metabolites Microbiota-induced blood–brain barrier dysfunction Neuropsychiatric diseases
■ Stress and depression ■ Pain and migraine ■ Autism spectrum disorders
Neurodegenerative diseases
■ Parkinson’s disease ■ Alzheimer’s disease ■ Amyotrophic lateral sclerosis ■ Multiple sclerosis
Cerebrovascular disease
■ Atherosclerosis ■ Stroke ■ Arteriovenous malformation
Conclusions and perspectives
Collapse
Affiliation(s)
- Sibo Zhu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Kelin Xu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,School of Data Science, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Genming Zhao
- School of Data Science, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Human Phenome Institute, Fudan University, 825 Zhangheng Road, Shanghai, 201203, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China. .,Fudan University Taizhou Institute of Health Sciences, Taizhou, China. .,Human Phenome Institute, Fudan University, 825 Zhangheng Road, Shanghai, 201203, China.
| |
Collapse
|
36
|
Santos SCD, Konstantyner T, Cocco RR. Effects of probiotics in the treatment of food hypersensitivity in children: a systematic review. Allergol Immunopathol (Madr) 2020; 48:95-104. [PMID: 31477401 DOI: 10.1016/j.aller.2019.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Food allergy is considered a public health problem for children. The modulation of the intestinal microbiota seems a promising strategy for the control of allergic reactions. OBJECTIVE To describe the effects of different forms of probiotics in pediatric food hypersensitivity treatment. DATA SOURCE We conducted a systematic review based on clinical trials published in the PubMed and Web of Science databases. The searches were carried out using the MeSH terms "Food Hypersensitivity," "Probiotics," "Lactobacillus," and "Bifidobacterium". DATA SYNTHESIS The final selection resulted in 18 clinical trials, which were predominantly samples of infants and pre-school children. The most-often used strain, either alone or in combination, was Lactobacillus rhamnosus GG; a placebo was mainly used in the control group. As for the vehicle, the most common forms were capsules and infant formulas, and the period of intervention ranged from four weeks to 24 months, with weekly or monthly visits to measure the outcomes. In these 18 trials, 46 analyses were performed with 27 different types of outcomes to evaluate the effects of probiotics (12 laboratory and 15 clinical). Twenty-seven of these analyses demonstrated the benefits of using these microorganisms. The SCORAD (atopic dermatitis index) and IgE levels and cytokines were the outcomes mostly evaluated. CONCLUSION The use of probiotics is beneficial in promoting immunomodulation and reducing clinical symptoms. However, more methodologically based research is needed to clarify the effect from each type, dose, and time of using them for the establishment of definitive care protocols.
Collapse
|
37
|
Kirmiz N, Robinson RC, Shah IM, Barile D, Mills DA. Milk Glycans and Their Interaction with the Infant-Gut Microbiota. Annu Rev Food Sci Technol 2019; 9:429-450. [PMID: 29580136 DOI: 10.1146/annurev-food-030216-030207] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human milk is a unique and complex fluid that provides infant nutrition and delivers an array of bioactive molecules that serve various functions. Glycans, abundant in milk, can be found as free oligosaccharides or as glycoconjugates. Milk glycans are increasingly linked to beneficial outcomes in neonates through protection from pathogens and modulation of the immune system. Indeed, these glycans influence the development of the infant and the infant-gut microbiota. Bifidobacterium species commonly are enriched in breastfed infants and are among a limited group of bacteria that readily consume human milk oligosaccharides (HMOs) and milk glycoconjugates. Given the importance of bifidobacteria in infant health, numerous studies have examined the molecular mechanisms they employ to consume HMOs and milk glycans, thus providing insight into this unique enrichment and shedding light on a range of translational opportunities to benefit at-risk infants.
Collapse
Affiliation(s)
- Nina Kirmiz
- Foods for Health Institute and Department of Food Science and Technology, University of California, Davis, California 95616, USA;
| | - Randall C Robinson
- Foods for Health Institute and Department of Food Science and Technology, University of California, Davis, California 95616, USA;
| | - Ishita M Shah
- Foods for Health Institute and Department of Food Science and Technology, University of California, Davis, California 95616, USA;
| | - Daniela Barile
- Foods for Health Institute and Department of Food Science and Technology, University of California, Davis, California 95616, USA;
| | - David A Mills
- Foods for Health Institute and Department of Food Science and Technology, University of California, Davis, California 95616, USA; .,Department of Viticulture and Enology, University of California, Davis, California 95616, USA
| |
Collapse
|
38
|
Warren MF, Hallowell HA, Higgins KV, Liles MR, Hood WR. Maternal Dietary Protein Intake Influences Milk and Offspring Gut Microbial Diversity in a Rat ( Rattus norvegicus) Model. Nutrients 2019; 11:E2257. [PMID: 31546967 PMCID: PMC6769776 DOI: 10.3390/nu11092257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/11/2019] [Accepted: 09/14/2019] [Indexed: 12/23/2022] Open
Abstract
Historically, investigators have assumed microorganisms identified in mother's milk to be contaminants, but recent data suggest that milk microbiota may contribute to beneficial maternal effects. Microorganisms that colonize the gastrointestinal tracts of newborn mammals are derived, at least in part, from the maternal microbial population. Milk-derived microbiota is an important source of this microbial inocula and we hypothesized that the maternal diet contributes to variation in this microbial community. To evaluate the relationship between a mother's diet and milk microbiome, we fed female rats a low- or high-protein diet and mated all individuals. Milk and cecal contents were collected from dams at peak lactation (14-day post-partum), and the bacterial composition of each community was assessed by 16S rRNA gene amplicon sequencing. Our findings revealed higher dietary protein intake decreased fecal microbial diversity but increased milk microbial and pup cecum diversity. Further, the higher dietary protein intake resulted in a greater abundance of potentially health-promoting bacteria, such as Lactobacillus spp. These data suggest that dietary protein levels contribute to significant shifts in the composition of maternal milk microbiota and that the functional consequences of these changes in microbial inocula might be biologically important and should be further explored.
Collapse
Affiliation(s)
- Matthew F Warren
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA.
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA.
| | - Haley A Hallowell
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Keah V Higgins
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Mark R Liles
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA.
| | - Wendy R Hood
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
39
|
Toghi M, Bitarafan S, Kasmaei HD, Ghafouri-Fard S. Bifidobacteria: A probable missing puzzle piece in the pathogenesis of multiple sclerosis. Mult Scler Relat Disord 2019; 36:101378. [PMID: 31487552 DOI: 10.1016/j.msard.2019.101378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/29/2019] [Accepted: 08/27/2019] [Indexed: 12/27/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disorder in which the immunopathogenesis is not fully understood. In the recent years, the role of gut microbiome in the pathogenesis of this disorder has been highlighted. Bifidobacteria as a component of gut microbiome might also be involved in MS pathogenesis. Being emerged in early days after birth, bifidobacteria have a prominent role in immune system maturation and function. Some factors like mode of delivery, breast feeding, mother's blood group and her secretory state and also environmental factors could influence its level in the early infancy, which may remain throughout lifetime. In this review, we discussed possible immunopathogenic link between the bifidobacteria and MS.
Collapse
Affiliation(s)
- Mehdi Toghi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Bitarafan
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hosein Delavar Kasmaei
- Department of Neurology, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Hassanzad M, Maleki Mostashari K, Ghaffaripour H, Emami H, Rahimi Limouei S, Velayati AA. Synbiotics and Treatment of Asthma: A Double-Blinded, Randomized, Placebo-Controlled Clinical Trial. Galen Med J 2019; 8:e1350. [PMID: 34466498 PMCID: PMC8343960 DOI: 10.31661/gmj.v8i0.1350] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/12/2018] [Accepted: 03/28/2019] [Indexed: 11/24/2022] Open
Abstract
Background: We examined the efficiency and safety of a specific synbiotic compound, brand name Kidilact®, in the treatment of asthma in children 12 years of age or younger. Materials and Methods: This double-blinded, randomized, placebo-controlled clinical trial was conducted in Tehran, Iran, from May 22, 2016, to May 21, 2017. One hundred children, 12 years of age or younger, who suffered from mild to moderate asthma were recruited in this study. The subjects were randomly divided into two groups; the experimental group received a sachet of Kidilact®, and the control group received a sachet of placebo once a day for six months. Both groups were compared in terms of the frequency of asthma attacks that were severe enough to require administration of fast-acting medications, the number of outpatient visits for asthma-related problems, and the frequency of hospitalization due to exacerbated symptoms of asthma. Results: There were fewer complaints of drug-induced side effects, e.g., vomiting, headache, stomachache, and diarrhea, exacerbated cough, and constipation in the experimental group than in the control group. Overall, a significantly greater number of participants in the experimental group were satisfied with the therapeutic intervention than those in the control group, as verified by the participants and their parents/guardians self-report. There was no significant difference between both groups in the frequency of asthma attacks and hospitalization due to exacerbated symptoms of asthma. The only significant difference between both groups was the count of outpatient visits. While the control group made 55 outpatient visits to the hospital, participants in the experimental group visited the hospital only 19 times (P=0.001). Conclusion: Results of our study indicates that synbiotic compound Kidilact® generally alleviates the symptoms of asthma in children of 12 years of age or younger, resulting in less frequent outpatient visits to the hospital due to asthma-related problems while rarely causing any side effects. Due to ease of use, the rarity of side effects, and their indirect positive effects on quality of life of asthmatic patients, we recommend that synbiotics be incorporated in regular treatment and management of children with asthma.
Collapse
Affiliation(s)
- Maryam Hassanzad
- Pediatric Respiratory Diseases Research Center (PRDRC), National Research Institute of Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Keyvan Maleki Mostashari
- Pediatric Respiratory Diseases Research Center (PRDRC), National Research Institute of Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Correspondence to: Keyvan Maleki Mostashari, National Research Institute of Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran Telephone Number: +982126109549 Email Address:
| | - Hosseinali Ghaffaripour
- Pediatric Respiratory Diseases Research Center (PRDRC), National Research Institute of Tuberculosis and Lung Disease (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Habib Emami
- Chronic Respiratory Diseases Research Center (PRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ali Akbar Velayati
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Steimle A, Menz S, Bender A, Ball B, Weber ANR, Hagemann T, Lange A, Maerz JK, Parusel R, Michaelis L, Schäfer A, Yao H, Löw HC, Beier S, Tesfazgi Mebrhatu M, Gronbach K, Wagner S, Voehringer D, Schaller M, Fehrenbacher B, Autenrieth IB, Oelschlaeger TA, Frick JS. Flagellin hypervariable region determines symbiotic properties of commensal Escherichia coli strains. PLoS Biol 2019; 17:e3000334. [PMID: 31206517 PMCID: PMC6597123 DOI: 10.1371/journal.pbio.3000334] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 06/27/2019] [Accepted: 06/05/2019] [Indexed: 12/19/2022] Open
Abstract
Escherichia coli represents a classical intestinal gram-negative commensal. Despite this commensalism, different E. coli strains can mediate disparate immunogenic properties in a given host. Symbiotic E. coli strains such as E. coli Nissle 1917 (EcN) are attributed beneficial properties, e.g., promotion of intestinal homeostasis. Therefore, we aimed to identify molecular features derived from symbiotic bacteria that might help to develop innovative therapeutic alternatives for the treatment of intestinal immune disorders. This study was performed using the dextran sodium sulphate (DSS)-induced colitis mouse model, which is routinely used to evaluate potential therapeutics for the treatment of Inflammatory Bowel Diseases (IBDs). We focused on the analysis of flagellin structures of different E. coli strains. EcN flagellin was found to harbor a substantially longer hypervariable region (HVR) compared to other commensal E. coli strains, and this longer HVR mediated symbiotic properties through stronger activation of Toll-like receptor (TLR)5, thereby resulting in interleukin (IL)-22–mediated protection of mice against DSS-induced colitis. Furthermore, using bone-marrow–chimeric mice (BMCM), CD11c+ cells of the colonic lamina propria (LP) were identified as the main mediators of these flagellin-induced symbiotic effects. We propose flagellin from symbiotic E. coli strains as a potential therapeutic to restore intestinal immune homeostasis, e.g., for the treatment of IBD patients. A flagellum renders bacteria motile, but this study reveals another property important for symbiosis: the hypervariable region of Escherichia coli flagellin strongly determines activation of TLR5, mediating benefits for the host such as protection against colitis.
Collapse
Affiliation(s)
- Alex Steimle
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Sarah Menz
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Annika Bender
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Brianna Ball
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | | | - Thomas Hagemann
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Anna Lange
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Jan K. Maerz
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Raphael Parusel
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Lena Michaelis
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Andrea Schäfer
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Hans Yao
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Hanna-Christine Löw
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Sina Beier
- Chair of Algorithms in Bioinformatics, Faculty of Computer Science, University of Tübingen, Tübingen, Germany
| | - Mehari Tesfazgi Mebrhatu
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Kerstin Gronbach
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Samuel Wagner
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen, Erlangen, Germany
| | - Martin Schaller
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | | | - Ingo B. Autenrieth
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | | | - Julia-Stefanie Frick
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
- * E-mail:
| |
Collapse
|
42
|
Sales-Campos H, Soares SC, Oliveira CJF. An introduction of the role of probiotics in human infections and autoimmune diseases. Crit Rev Microbiol 2019; 45:413-432. [PMID: 31157574 DOI: 10.1080/1040841x.2019.1621261] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During the last decades, studies exploring the role of microorganisms inhabiting human body in different scenarios have demonstrated the great potential of modulating them to treat and prevent diseases. Among the most outstanding applications, probiotics have been used for over a century to treat infections and inflammation. Despite the beneficial role of other probiotics, Lactobacillus and Bifidobacterium species are the most frequently used, and have been effective as a therapeutic option in the treatment/prevention of dental caries, periodontal diseases, urogenital infections, and gastrointestinal infections. Additionally, as gastrointestinal tract harbors a great diversity of microbial species that directly or indirectly modulate host metabolism and immune response, the influence of intestinal microbiota, one of the targets of therapies using probiotics, on the biology of immune cells can be explored to treat inflammatory disorders or immune-mediated diseases. Thus, it is not surprising that probiotics have presented promising results in modulating human inflammatory diseases such as type 1 diabetes, multiple sclerosis, rheumatoid arthritis and inflammatory bowel disease, among others. Hence, the purpose of this review is to discuss the potential of therapeutic approaches using probiotics to constrain infection and development of inflammation on human subjects.
Collapse
Affiliation(s)
- Helioswilton Sales-Campos
- Laboratory of Immunology and Bioinformatics, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro , Uberaba , Minas Gerais , Brazil.,Department of Biosciences and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás , Goiás , Goiânia , Brazil
| | - Siomar Castro Soares
- Laboratory of Immunology and Bioinformatics, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro , Uberaba , Minas Gerais , Brazil
| | - Carlo José Freire Oliveira
- Laboratory of Immunology and Bioinformatics, Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro , Uberaba , Minas Gerais , Brazil
| |
Collapse
|
43
|
Li W, Zhu Y, Li Y, Shu M, Wen Y, Gao X, Wan C. The gut microbiota of hand, foot and mouth disease patients demonstrates down-regulated butyrate-producing bacteria and up-regulated inflammation-inducing bacteria. Acta Paediatr 2019; 108:1133-1139. [PMID: 30427066 DOI: 10.1111/apa.14644] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 10/08/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022]
Abstract
AIM This study explored the gut microbiota of children with hand, foot and mouth disease (HFMD). METHODS We enrolled 15 cases with HFMD admitted to the West China Second Hospital, Sichuan University, China, from July to September 2016 at a median age of three years. The controls were 15 healthy children of a similar age who underwent routine health examinations at the hospital during the same period. Gut microbiota was analysed using high throughput 16S ribosomal deoxyribonucleic acid sequencing. RESULTS The gut microbiota in the HFMD patients was distinct from the controls. Compared with the controls, the composition of gut microbiota in the HFMD cases represented a reduction of two butyrate-producing bacteria, Ruminococcus (0.73 ± 1.28 versus 7.78 ± 20.01, p = 0.026) and Roseburia (0.67 ± 1.69 versus 1.61 ± 3.27, p = 0.024) and an up-regulation of Escherichia (5.26 ± 10.50 versus 1.59 ± 5.90,p < 0.01) and Enterococcus (4.12 ± 12.49 versus 0.12 ± 0.41, p = 0.015). CONCLUSION The dysbiosis of gut microbiota of the HFMD cases included a reduction of butyrate-producing bacteria and an up-regulation of inflammation-inducing bacteria. These may have impaired the intestinal biological mucosal barrier and host immune functions, promoting the invasion of the enterovirus.
Collapse
Affiliation(s)
- Weiran Li
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Yu Zhu
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Yiyuan Li
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Min Shu
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Yang Wen
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Xiaolin Gao
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| | - Chaomin Wan
- Department of Paediatrics West China Second Hospital Sichuan University Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu China
| |
Collapse
|
44
|
|
45
|
Hurley E, Mullins D, Barrett MP, O'Shea CA, Kinirons M, Ryan CA, Stanton C, Whelton H, Harris HMB, O'Toole PW. The microbiota of the mother at birth and its influence on the emerging infant oral microbiota from birth to 1 year of age: a cohort study. J Oral Microbiol 2019; 11:1599652. [PMID: 32128038 PMCID: PMC7034431 DOI: 10.1080/20002297.2019.1599652] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/12/2019] [Accepted: 03/22/2019] [Indexed: 01/11/2023] Open
Abstract
Background: The acquisition of microbial communities and the influence of delivery mode on the oral microbiota of the newborn infant remains poorly characterised. Methods: A cohort of pregnant women were enrolled in the study (n = 84). All infants were born full term, by Spontaneous vaginal delivery (SVD) or by Caesarean section (CS). At delivery a saliva sample along with a vaginal/skin sample from the mother. Saliva samples were the taken from the infant within one week of birth, and at week 4, week 8, 6 months and 1 year of age. We used high-throughput sequencing of V4-V5 region 16S rRNA amplicons to compare the microbiota of all samples. Results: The vaginal microbiota had a lower alpha diversity than the skin microbiota of the mother, while the infant oral microbiota diversity remained relatively stable from birth to 8 weeks of age. The oral microbiota of the neonate differed by birth modality up to 1 week of age (p < 0.05), but birth modality did not have any influence on the infant oral microbiota beyond this age. Conclusions: We conclude thatbirth mode does not have an effect on the infant oral microbiota beyond 4 weeks of age, and the oral microbiota of infants continues to develop until 1 year of age.
Collapse
Affiliation(s)
- Eimear Hurley
- School of Microbiology, University College Cork, Cork, Ireland.,Cork University Dental School & Hospital, Cork University Hospital, Wilton, Cork, Ireland
| | - David Mullins
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Maurice P Barrett
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Carol Anne O'Shea
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Neonatology, Cork University Maternity Hospital, Wilton, Cork, Ireland
| | - Martin Kinirons
- Cork University Dental School & Hospital, Cork University Hospital, Wilton, Cork, Ireland
| | - C Anthony Ryan
- Department of Neonatology, Cork University Maternity Hospital, Wilton, Cork, Ireland
| | | | - Helen Whelton
- Cork University Dental School & Hospital, Cork University Hospital, Wilton, Cork, Ireland.,College of Medicine and Health, UCC, Cork, Ireland
| | - Hugh M B Harris
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul W O'Toole
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
46
|
|
47
|
Henderickx JGE, Zwittink RD, van Lingen RA, Knol J, Belzer C. The Preterm Gut Microbiota: An Inconspicuous Challenge in Nutritional Neonatal Care. Front Cell Infect Microbiol 2019; 9:85. [PMID: 31001489 PMCID: PMC6454191 DOI: 10.3389/fcimb.2019.00085] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/12/2019] [Indexed: 12/16/2022] Open
Abstract
The nutritional requirements of preterm infants are unique and challenging to meet in neonatal care, yet crucial for their growth, development and health. Normally, the gut microbiota has distinct metabolic capacities, making their role in metabolism of dietary components indispensable. In preterm infants, variation in microbiota composition is introduced while facing a unique set of environmental conditions. However, the effect of such variation on the microbiota's metabolic capacity and on the preterm infant's growth and development remains unresolved. In this review, we will provide a holistic overview on the development of the preterm gut microbiota and the unique environmental conditions contributing to this, in addition to maturation of the gastrointestinal tract and immune system in preterm infants. The role of prematurity, as well as the role of human milk, in the developmental processes is emphasized. Current research stresses the early life gut microbiota as cornerstone for simultaneous development of the gastrointestinal tract and immune system. Besides that, literature provides clues that prematurity affects growth and development. As such, this review is concluded with our hypothesis that prematurity of the gut microbiota may be an inconspicuous clinical challenge in achieving optimal feeding besides traditional challenges, such as preterm breast milk composition, high nutritional requirements and immaturity of the gastrointestinal tract and immune system. A better understanding of the metabolic capacity of the gut microbiota and its impact on gut and immune maturation in preterm infants could complement current feeding regimens in future neonatal care and thereby facilitate growth, development and health in preterm infants.
Collapse
Affiliation(s)
- Jannie G E Henderickx
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - Romy D Zwittink
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - Richard A van Lingen
- Department of Neonatology, Isala Women and Children's Hospital, Zwolle, Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, Netherlands.,Danone Nutricia Research, Utrecht, Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
48
|
Nobs SP, Tuganbaev T, Elinav E. Microbiome diurnal rhythmicity and its impact on host physiology and disease risk. EMBO Rep 2019; 20:embr.201847129. [PMID: 30877136 DOI: 10.15252/embr.201847129] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/29/2018] [Accepted: 02/22/2019] [Indexed: 12/29/2022] Open
Abstract
Host-microbiome interactions constitute key determinants of host physiology, while their dysregulation is implicated in a wide range of human diseases. The microbiome undergoes diurnal variation in composition and function, and this in turn drives oscillations in host gene expression and functions. In this review, we discuss the newest developments in understanding circadian host-microbiome interplays, and how they may be relevant in health and disease contexts. We summarize the molecular mechanisms by which the microbiome influences host function in a diurnal manner, and inversely describe how the host orchestrates circadian rhythmicity of the microbiome. Furthermore, we highlight the future perspectives and challenges in studying this new and exciting facet of host-microbiome interactions. Finally, we illustrate how the elucidation of the microbiome chronobiology may pave the way for novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Timur Tuganbaev
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel .,Cancer-Microbiome Division, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
49
|
Vemuri R, Sylvia KE, Klein SL, Forster SC, Plebanski M, Eri R, Flanagan KL. The microgenderome revealed: sex differences in bidirectional interactions between the microbiota, hormones, immunity and disease susceptibility. Semin Immunopathol 2019; 41:265-275. [PMID: 30298433 PMCID: PMC6500089 DOI: 10.1007/s00281-018-0716-7] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023]
Abstract
Sex differences in immunity are well described in the literature and thought to be mainly driven by sex hormones and sex-linked immune response genes. The gastrointestinal tract (GIT) is one of the largest immune organs in the body and contains multiple immune cells in the GIT-associated lymphoid tissue, Peyer's patches and elsewhere, which together have profound effects on local and systemic inflammation. The GIT is colonised with microbial communities composed of bacteria, fungi and viruses, collectively known as the GIT microbiota. The GIT microbiota drives multiple interactions locally with immune cells that regulate the homeostatic environment and systemically in diverse tissues. It is becoming evident that the microbiota differs between the sexes, both in animal models and in humans, and these sex differences often lead to sex-dependent changes in local GIT inflammation, systemic immunity and susceptibility to a range of inflammatory diseases. The sexually dimorphic microbiome has been termed the 'microgenderome'. Herein, we review the evidence for the microgenderome and contemplate the role it plays in driving sex differences in immunity and disease susceptibility. We further consider the impact that biological sex might play in the response to treatments aimed at manipulating the GIT microbiota, such as prebiotics, live biotherapeutics, (probiotics, synbiotics and bacteriotherapies) and faecal microbial transplant. These alternative therapies hold potential in the treatment of both psychological (e.g., anxiety, depression) and physiological (e.g., irritable bowel disease) disorders differentially affecting males and females.
Collapse
Affiliation(s)
- Ravichandra Vemuri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kristyn E Sylvia
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Samuel C Forster
- Microbiota and Systems Biology Laboratory, Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Molecular and Translational Sciences, Monash University, Melbourne, Victoria, Australia
| | - Magdalena Plebanski
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
- School of Health and Biomedical Science, RMIT University, Melbourne, Victoria, Australia
| | - Raj Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Katie L Flanagan
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.
- School of Health and Biomedical Science, RMIT University, Melbourne, Victoria, Australia.
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
50
|
Borewicz K, Suarez-Diez M, Hechler C, Beijers R, de Weerth C, Arts I, Penders J, Thijs C, Nauta A, Lindner C, Van Leusen E, Vaughan EE, Smidt H. The effect of prebiotic fortified infant formulas on microbiota composition and dynamics in early life. Sci Rep 2019; 9:2434. [PMID: 30792412 PMCID: PMC6385197 DOI: 10.1038/s41598-018-38268-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/19/2018] [Indexed: 01/15/2023] Open
Abstract
Gastrointestinal (GI) microbiota composition differs between breastfed and formula-fed infants. Today's infant formulas are often fortified with prebiotics to better mimic properties of human milk with respect to its effect on GI microbiota composition and function. We used Illumina HiSeq sequencing of PCR-amplified 16S rRNA gene fragments to investigate the composition of faecal microbiota in 2-12 week old infants receiving either breastmilk, infant formulas fortified with prebiotics, or mixed feeding. We compared these results with results from infants fed traditional formulas used in the Netherlands in 2002-2003, which contained no added prebiotics. We showed that today's formulas supplemented with either scGOS (0.24-0.50 g/100 ml) or scGOS and lcFOS (at a 9:1 ratio; total 0.6 g/100 ml) had a strong bifidogenic effect as compared to traditional formulas, and they also resulted in altered patterns of microbial colonisation within the developing infant gastrointestinal tract. We identified three microbial states (or developmental stages) in the first 12 weeks of life, with a gradual transition pattern towards a bifidobacteria dominated state. In infants receiving only fortified formulas, this transition towards the bifidobacteria dominated state was accelerated, whereas in infants receiving mixed feeding the transition was delayed, as compared to exclusively breastfed infants.
Collapse
Affiliation(s)
- Klaudyna Borewicz
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| | - Maria Suarez-Diez
- Laboratory of Systems and Synthetic Biology, Wageningen & Research University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Christine Hechler
- Behavioral Science Institute, Radboud University, Montessorilaan 3, 6525 HR, Nijmegen, The Netherlands
| | - Roseriet Beijers
- Behavioral Science Institute, Radboud University, Montessorilaan 3, 6525 HR, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Kapittelweg 29, 6525 EN, Nijmegen, The Netherlands
| | - Carolina de Weerth
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Kapittelweg 29, 6525 EN, Nijmegen, The Netherlands
| | - Ilja Arts
- Department of Epidemiology, Maastricht University, Care and Public Health Research Institute, Minderbroedersberg 4-6, 6211 LK, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Universiteitssingel 50 6229ER Maastricht; and Maastricht Center for Systems Biology (MaCSBio), Universiteitssingel 60, 6229 ER, Maastricht, The Netherlands
| | - John Penders
- Department of Medical Microbiology, Maastricht University Medical Center, P. Debyelaan 25, 6229HX, Maastricht, The Netherlands
- NUTRIM School for Nutrition, Toxicology and Metabolism, Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
| | - Carel Thijs
- Department of Epidemiology, Maastricht University, Care and Public Health Research Institute, Minderbroedersberg 4-6, 6211 LK, Maastricht, The Netherlands
| | - Arjen Nauta
- FrieslandCampina, Stationsplein 4, 3818 LE, Amersfoort, The Netherlands
| | - Cordula Lindner
- FrieslandCampina, Stationsplein 4, 3818 LE, Amersfoort, The Netherlands
| | - Ellen Van Leusen
- FrieslandCampina, Stationsplein 4, 3818 LE, Amersfoort, The Netherlands
| | - Elaine E Vaughan
- Sensus B.V. (Royal Cosun), Borchwerf 3, 4704 RG, Roosendaal, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| |
Collapse
|