1
|
Yu L, Guo Y, Wu JL. Influence of mode of delivery on infant gut microbiota composition: a pilot study. J OBSTET GYNAECOL 2024; 44:2368829. [PMID: 38913773 DOI: 10.1080/01443615.2024.2368829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/09/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND Microbial colonisation in infants is initially dependent on the mother and is affected by the mode of delivery. Understanding these impacts is crucial as the early-life gut microbiota plays a vital role in immune development, metabolism, and overall health. Early-life infant gut microbiota is diverse among populations and geographic origins. However, in this context, only a few studies have explored the impact of the mode of delivery on the intestinal microbiome in children in Guangzhou, China. Therefore, this study aimed to investigate the influence of birth mode on the intestinal microbiota of healthy infants in Guangzhou, China. METHODS Faecal samples were collected once from 20 healthy full-term infants aged 1-6 months, delivered via either caesarean section (CS) or vaginal delivery (VD), post-enrolment. The intestinal microbiota were characterised using full-length 16S rRNA gene sequencing. Bacterial quantity and community composition were compared between the two groups. RESULTS No significant differences in gut bacterial diversity and richness were observed between the CS and VD groups. The Pseudomonadota phylum (44.15 ± 33.05% vs 15.62 ± 15.60%, p = 0.028) and Enterobacteriaceae family (44.00 ± 33.11% vs 15.31 ± 15.47%, p = 0.028) were more abundant in the CS group than in the VD group. The VD group exhibited a higher abundance of the Bacillota phylum (40.51 ± 32.77% vs 75.57 ± 27.83%, p = 0.019). CONCLUSIONS The early stage of intestinal bacterial colonisation was altered in the CS group as compared with the VD group. Our findings provide evidence that CS has the potential to disrupt the maturation of intestinal microbial communities in infants by influencing the colonisation of specific microorganisms. Further comprehensive studies that consider geographical locations are necessary to elucidate the progression of microbiota in infants born via different delivery modes.
Collapse
Affiliation(s)
- Li Yu
- Department of Children's Health Care, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yong Guo
- Department of Children's Health Care, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jie-Ling Wu
- Department of Children's Health Care, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Qu Z, Zhang B, Lin G, Guo M, Tian P, Wang L, Chen W, Zhang H, Wang G. Dietary nucleotides drive changes in infant fecal microbiota in vitro and gut microbiota-gut-brain development in neonatal rats: A potential "nitrogen source" for early microbiota growth. Food Chem 2024; 463:141333. [PMID: 39340921 DOI: 10.1016/j.foodchem.2024.141333] [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: 05/23/2024] [Revised: 08/14/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024]
Abstract
Various dietary factors in human milk are important nutrients for the formation of the infant gut microbiota (GM). While promoting the growth of the GM, some human milk components that are difficult to absorb and utilize will be broken down by the GM, and converted into nutrients that the baby can use, such as breast milk oligosaccharides-the 'carbon source' for infant GM. This study reveals that nucleotides (NTs), significant non-protein nitrogen sources in human milk, can enhance the abundance of beneficial microbial genera such as g_Bifidobacterium, g_Bacteroides, and g_Blautia in in vitro fecal fermentation fluids of infants at low doses (2 mg/mL). Conversely, high doses of NTs (20 mg/mL) increased the abundance of g_Escherichia-Shigella. Furthermore, low-dose NTs fermentation broth significantly enhanced the expression of neurodevelopmental marker genes such as Tuj1, Sox2, Dcx, and NeuN in NE-4C neural stem cells, whereas a single NTs digestion broth did not exhibit significant activity. However, in vivo studies using neonatal rats as a model demonstrated that both low-dose NTs fermentation broth and NTs digestive juices promoted behavioral development in neonatal rats (PND 20) and neuron maturation in the prefrontal cortex and hippocampus. Non-targeted metabolomics results indicate that low-dose dietary NTs promote the production of certain neuroregulatory metabolites in infant fecal fermentation, such as uridine, L-tyrosine, L-glutamic acid, and succinic acid. These findings suggest that NTs may serve as an important "nitrogen source" during GM formation in early life and have a dose effect in driving the development of the microbiota-gut-brain axis in early life.
Collapse
Affiliation(s)
- Zhihao Qu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Bo Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Guopeng Lin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Min Guo
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Linlin Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Heng Zhang
- Department of Child Health Care, Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu 214002, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China.
| |
Collapse
|
3
|
Shi X, Liu Y, Ma T, Jin H, Zhao F, Sun Z. Delivery mode and maternal gestational diabetes are important factors in shaping the neonatal initial gut microbiota. Front Cell Infect Microbiol 2024; 14:1397675. [PMID: 39268487 PMCID: PMC11390658 DOI: 10.3389/fcimb.2024.1397675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/06/2024] [Indexed: 09/15/2024] Open
Abstract
Background The infant gut microbiome's establishment is pivotal for health and immune development. Understanding it unveils insights into growth, development, and maternal microbial interactions. Research often emphasizes gut bacteria, neglecting the phageome. Methods To investigate the influence of geographic or maternal factors (mode of delivery, mode of breastfeeding, gestational diabetes mellitus) on the gut microbiota and phages of newborns, we collected fecal samples from 34 pairs of mothers and their infants within 24 hours of delivery from three regions (9 pairs from Enshi, 7 pairs from Hohhot, and 18 pairs from Hulunbuir) using sterile containers. Gut microbiota analysis by Shotgun sequencing was subsequently performed. Results Our results showed that geographic location affects maternal gut microbiology (P < 0.05), while the effect on infant gut microbiology was not significant (P = 0.184). Among the maternal factors, mode of delivery had a significant (P < 0.05) effect on the newborn. Specific bacteria (e.g., Bacteroides, Escherichia spp., Phocaeicola vulgatus, Escherichia coli, Staphylococcus hominis, Veillonella spp.), predicted active metabolites, and bacteriophage vOTUs varied with delivery mode. Phocaeicola vulgatus significantly correlated with some metabolites and bacteriophages in the early infant gut (P < 0.05). In the GD group, a strong negative correlation of phage diversity between mother and infants was observed (R = -0.58, P=0.04). Conclusion In conclusion, neonatal early gut microbiome (including bacteria and bacteriophages) colonization is profoundly affected by the mode of delivery, and maternal gestational diabetes mellitus. The key bacteria may interact with bacteriophages to influence the levels of specific metabolites. Our study provides new evidence for the study of the infant microbiome, fills a gap in the analysis of the infant gut microbiota regarding the virome, and emphasizes the importance of maternal health for the infant initial gut virome.
Collapse
Affiliation(s)
- Xuan Shi
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Collaborative Innovation Center of Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Yanfang Liu
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Collaborative Innovation Center of Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Teng Ma
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Collaborative Innovation Center of Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Hao Jin
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Collaborative Innovation Center of Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Feiyan Zhao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Collaborative Innovation Center of Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Collaborative Innovation Center of Lactic Acid Bacteria and Fermented Dairy Products, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| |
Collapse
|
4
|
Iorfida D, Valitutti F, Vestri A, D'Adamo G, Passaro T, Crocco M, Malerba F, Monzani A, Rabbone I, Pensabene L, Giancotti L, Graziano F, Citrano M, Ferretti F, Trovato CM, Pacenza C, Iasevoli M, Banzato C, Lubrano R, Montuori M. Prevalence of delivery mode in an Italian nationwide cohort with celiac disease: a SIGENP multicenter retrospective study (the CD-deliver-IT). Ital J Pediatr 2024; 50:129. [PMID: 39061072 PMCID: PMC11282831 DOI: 10.1186/s13052-024-01710-0] [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: 04/17/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Studies have indicated an association between cesarean section (CS), especially elective CS, and an increased risk of celiac disease (CD), but the conclusions of other studies are contradictory. The primary aim of this study (CD-deliver-IT) was to evaluate the rate of CS in a large population of CD patients throughout Italy. METHODS: This national multicenter retrospective study was conducted between December 2020 and November 2021. The coordinating center was the Pediatric Gastroenterology and Liver Unit of Policlinico Umberto I, Sapienza, University of Rome, Lazio, Italy. Eleven other referral centers for CD have participated to the study. Each center has collected data on mode of delivery and perinatal period of all CD patients referring to the center in the last 40 years. RESULTS Out of 3,259 CD patients recruited in different Italian regions, data on the mode of delivery were obtained from 3,234. One thousand nine hundred forty-one (1,941) patients (60%) were born vaginally and 1,293 (40%) by CS (8.3% emergency CS, 30.1% planned CS, 1.5% undefined CS). A statistically significant difference was found comparing median age at time of CD diagnosis of patients who were born by emergency CS (4 years, CI 95% 3.40-4.59), planned CS (7 years, CI 95% 6.02-7.97) and vaginal delivery (6 years, CI 95% 5.62-6.37) (log rank p < 0.0001). CONCLUSIONS This is the first Italian multicenter study aiming at evaluating the rate of CS in a large population of CD patients through Italy. The CS rate found in our CD patients is higher than rates reported in the general population over the last 40 years and emergency CS seems to be associated with an earlier onset of CD compared to vaginal delivery or elective CS in our large nationwide retrospective cohort. This suggests a potential role of the mode of delivery on the risk of developing CD and on its age of onset, but it is more likely that it works in concert with other perinatal factors. Further prospective studies on other perinatal factors potentially influencing gut microbiota are awaited in order to address heavy conflicting evidence reaming in this research field.
Collapse
Affiliation(s)
- Donatella Iorfida
- Department of Maternal and Child Health, Pediatrics and Neonatology Unit, Santa Maria Goretti Hospital, Sapienza - University of Rome, Latina, Italy
| | - Francesco Valitutti
- Department of Surgical and Biomedical Sciences, Pediatric Clinic, University of Perugia, Perugia, Italy
| | - Annarita Vestri
- Department of Public Health and Infectious Disease, Sapienza - University of Rome, Rome, Italy
| | - Grazia D'Adamo
- Pediatric Unit, AOU Salerno, P.O. Cava de' Tirreni, Salerno, Italy
| | - Tiziana Passaro
- Pediatric Unit, AOU Salerno, P.O. Cava de' Tirreni, Salerno, Italy
| | - Marco Crocco
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, Genoa, 16147, Italy
| | - Federica Malerba
- Pediatric Gastroenterology and Endoscopy Unit, IRCCS Istituto Giannina Gaslini, Genoa, 16147, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Alice Monzani
- Department of Health Sciences, Division of Paediatrics, University of Piemonte Orientale, Novara, Italy
| | - Ivana Rabbone
- Department of Health Sciences, Division of Paediatrics, University of Piemonte Orientale, Novara, Italy
| | - Licia Pensabene
- Department of Surgical and Medical Sciences, Pediatric Unit, Magna Graecia University, Catanzaro, Italy
| | - Laura Giancotti
- Department of Surgical and Medical Sciences, Pediatric Unit, Magna Graecia University, Catanzaro, Italy
| | | | - Michele Citrano
- Pediatric Unit, Villa Sofia - Cervello Hospital, Palermo, Italy
| | - Francesca Ferretti
- Hepatology Gastroenterology and Nutrition Unit, Bambino Gesù Children Hospital, Rome, Italy
| | - Chiara Maria Trovato
- Hepatology Gastroenterology and Nutrition Unit, Bambino Gesù Children Hospital, Rome, Italy
| | | | - Mario Iasevoli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Pediatrics Section, University of Salerno, Baronissi, Italy
| | | | - Riccardo Lubrano
- Department of Maternal and Child Health, Pediatrics and Neonatology Unit, Santa Maria Goretti Hospital, Sapienza - University of Rome, Latina, Italy
| | - Monica Montuori
- Maternal and Child Health Department, Pediatric Gastroenterology and Liver Unit, Sapienza - University of Rome, Rome, Italy.
| |
Collapse
|
5
|
Leech SM, Borg DJ, Rae KM, Kumar S, Clifton VL, Dekker Nitert M. Delivery mode is a larger determinant of infant gut microbiome composition at 6 weeks than exposure to peripartum antibiotics. Microb Genom 2024; 10:001269. [PMID: 38995243 PMCID: PMC11316550 DOI: 10.1099/mgen.0.001269] [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: 02/23/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Background. Previous research has shown that delivery mode can shape infant gut microbiome composition. However, mothers delivering by caesarean section routinely receive prophylactic antibiotics prior to delivery, resulting in antibiotic exposure to the infant via the placenta. Previously, only a small number of studies have examined the effect of delivery mode versus antibiotic exposure on the infant gut microbiome with mixed findings.Objective. We aimed to determine the effect of delivery mode compared to antibiotic use during labour and delivery on the infant and maternal gut microbiome at 6 weeks post-partum.Methodology. Twenty-five mother-infant dyads were selected from the longitudinal Queensland Family Cohort Study. The selected dyads comprised nine vaginally delivered infants without antibiotics, seven vaginally delivered infants exposed to antibiotics and nine infants born by caesarean section with routine maternal prophylactic antibiotics. Shotgun-metagenomic sequencing of DNA from stool samples collected at 6 weeks post-partum from mother and infant was used to assess microbiome composition.Results. Caesarean section infants exhibited decreases in Bacteroidetes (ANCOM-BC q<0.0001, MaAsLin 2 q=0.041), changes to several functional pathways and altered beta diversity (R 2=0.056, P=0.029), while minimal differences due to antibiotic exposure were detected. For mothers, caesarean delivery (P=0.0007) and antibiotic use (P=0.016) decreased the evenness of the gut microbiome at 6 weeks post-partum without changing beta diversity. Several taxa in the maternal microbiome were altered in association with antibiotic use, with few differentially abundant taxa associated with delivery mode.Conclusion. For infants, delivery mode appears to have a larger effect on gut microbiome composition at 6 weeks post-partum than intrapartum antibiotic exposure. For mothers, both delivery mode and intrapartum antibiotic use have a small effect on gut microbiome composition at 6 weeks post-partum.
Collapse
Affiliation(s)
- Sophie M. Leech
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Danielle J. Borg
- Pregnancy and Development Group, Mater Research Institute, South Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Kym M. Rae
- Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
- Indigenous Health Group, Mater Research Institute, South Brisbane, QLD, Australia
| | - Sailesh Kumar
- Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
- Mater Mothers’ Hospital, Brisbane, QLD, Australia
| | - Vicki L. Clifton
- Pregnancy and Development Group, Mater Research Institute, South Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Marloes Dekker Nitert
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
6
|
Magalhães MI, Azevedo MJ, Castro F, Oliveira MJ, Costa ÂM, Sampaio Maia B. The link between obesity and the gut microbiota and immune system in early-life. Crit Rev Microbiol 2024:1-21. [PMID: 38651972 DOI: 10.1080/1040841x.2024.2342427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
In early-life, the gut microbiota is highly modifiable, being modulated by external factors such as maternal microbiota, mode of delivery, and feeding strategies. The composition of the child's gut microbiota will deeply impact the development and maturation of its immune system, with consequences for future health. As one of the main sources of microorganisms to the child, the mother represents a crucial factor in the establishment of early-life microbiota, impacting the infant's wellbeing. Recent studies have proposed that dysbiotic maternal gut microbiota could be transmitted to the offspring, influencing the development of its immunity, and leading to the development of diseases such as obesity. This paper aims to review recent findings in gut microbiota and immune system interaction in early-life, highlighting the benefits of a balanced gut microbiota in the regulation of the immune system.
Collapse
Affiliation(s)
- Maria Inês Magalhães
- Doctoral Program in Biomedical Sciences, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| | - Maria João Azevedo
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
- Academic Center for Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Flávia Castro
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria José Oliveira
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ângela M Costa
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Benedita Sampaio Maia
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| |
Collapse
|
7
|
Jones JM, Reinke SN, Mousavi-Derazmahalleh M, Garssen J, Jenmalm MC, Srinivasjois R, Silva D, Keelan J, Prescott SL, Palmer DJ, Christophersen CT. Maternal prebiotic supplementation during pregnancy and lactation modifies the microbiome and short chain fatty acid profile of both mother and infant. Clin Nutr 2024; 43:969-980. [PMID: 38452522 DOI: 10.1016/j.clnu.2024.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND & AIMS Improving maternal gut health in pregnancy and lactation is a potential strategy to improve immune and metabolic health in offspring and curtail the rising rates of inflammatory diseases linked to alterations in gut microbiota. Here, we investigate the effects of a maternal prebiotic supplement (galacto-oligosaccharides and fructo-oligosaccharides), ingested daily from <21 weeks' gestation to six months' post-partum, in a double-blinded, randomised placebo-controlled trial. METHODS Stool samples were collected at multiple timepoints from 74 mother-infant pairs as part of a larger, double-blinded, randomised controlled allergy intervention trial. The participants were randomised to one of two groups; with one group receiving 14.2 g per day of prebiotic powder (galacto-oligosaccharides GOS and fructo-oligosaccharides FOS in ratio 9:1), and the other receiving a placebo powder consisting of 8.7 g per day of maltodextrin. The faecal microbiota of both mother and infants were assessed based on the analysis of bacterial 16S rRNA gene (V4 region) sequences, and short chain fatty acid (SCFA) concentrations in stool. RESULTS Significant differences in the maternal microbiota profiles between baseline and either 28-weeks' or 36-weeks' gestation were found in the prebiotic supplemented women. Infant microbial beta-diversity also significantly differed between prebiotic and placebo groups at 12-months of age. Supplementation was associated with increased abundance of commensal Bifidobacteria in the maternal microbiota, and a reduction in the abundance of Negativicutes in both maternal and infant microbiota. There were also changes in SCFA concentrations with maternal prebiotics supplementation, including significant differences in acetic acid concentration between intervention and control groups from 20 to 28-weeks' gestation. CONCLUSION Maternal prebiotic supplementation of 14.2 g per day GOS/FOS was found to favourably modify both the maternal and the developing infant gut microbiome. These results build on our understanding of the importance of maternal diet during pregnancy, and indicate that it is possible to intervene and modify the development of the infant microbiome by dietary modulation of the maternal gut microbiome.
Collapse
Affiliation(s)
- Jacquelyn M Jones
- Trace and Environmental DNA Laboratory, School of Molecular and Life Sciences, Curtin University, Bentley, WA 6102, Australia; The Western Australian Human Microbiome Collaboration Centre, Curtin University, Bentley, WA 6027, Australia.
| | - Stacey N Reinke
- Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Mahsa Mousavi-Derazmahalleh
- Trace and Environmental DNA Laboratory, School of Molecular and Life Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CS Utrecht, the Netherlands; Nutricia Research, 3584 CT Utrecht, the Netherlands
| | - Maria C Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Ravisha Srinivasjois
- Joondalup Health Campus, Joondalup, WA 6027, Australia; School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia
| | - Desiree Silva
- Joondalup Health Campus, Joondalup, WA 6027, Australia; School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia; Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; School of Medical & Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Jeffrey Keelan
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; School of Biomedical Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Susan L Prescott
- School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia; Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; Department of Immunology and Dermatology, Perth Children's Hospital, Nedlands, WA 6009, Australia; Nova Institute for Health, Baltimore, MD 21231, USA
| | - Debra J Palmer
- School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia; Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Claus T Christophersen
- Trace and Environmental DNA Laboratory, School of Molecular and Life Sciences, Curtin University, Bentley, WA 6102, Australia; The Western Australian Human Microbiome Collaboration Centre, Curtin University, Bentley, WA 6027, Australia; Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Joondalup, WA 6027, Australia.
| |
Collapse
|
8
|
Catassi G, Aloi M, Giorgio V, Gasbarrini A, Cammarota G, Ianiro G. The Role of Diet and Nutritional Interventions for the Infant Gut Microbiome. Nutrients 2024; 16:400. [PMID: 38337684 PMCID: PMC10857663 DOI: 10.3390/nu16030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
The infant gut microbiome plays a key role in the healthy development of the human organism and appears to be influenced by dietary practices through multiple pathways. First, maternal diet during pregnancy and infant nutrition significantly influence the infant gut microbiota. Moreover, breastfeeding fosters the proliferation of beneficial bacteria, while formula feeding increases microbial diversity. The timing of introducing solid foods also influences gut microbiota composition. In preterm infants the gut microbiota development is influenced by multiple factors, including the time since birth and the intake of breast milk, and interventions such as probiotics and prebiotics supplementation show promising results in reducing morbidity and mortality in this population. These findings underscore the need for future research to understand the long-term health impacts of these interventions and for further strategies to enrich the gut microbiome of formula-fed and preterm infants.
Collapse
Affiliation(s)
- Giulia Catassi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Umberto I Hospital, 00161 Rome, Italy;
| | - Valentina Giorgio
- Department of Woman and Child Health and Public Health, UOC Pediatria, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
9
|
Al-Busaidi A, Alabri O, Alomairi J, ElSharaawy A, Al Lawati A, Al Lawati H, Das S. Gut Microbiota and Insulin Resistance: Understanding the Mechanism of Better Treatment of Type 2 Diabetes Mellitus. Curr Diabetes Rev 2024; 21:e170124225723. [PMID: 38243954 DOI: 10.2174/0115733998281910231231051814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
Gut microbiota refers to the population of trillions of microorganisms present in the human intestine. The gut microbiota in the gastrointestinal system is important for an individual's good health and well-being. The possibility of an intrauterine colonization of the placenta further suggests that the fetal environment before birth may also affect early microbiome development. Various factors influence the gut microbiota. Dysbiosis of microbiota may be associated with various diseases. Insulin regulates blood glucose levels, and disruption of the insulin signaling pathway results in insulin resistance. Insulin resistance or hyperinsulinemia is a pathological state in which the insulin-responsive cells have a diminished response to the hormone compared to normal physiological responses, resulting in reduced glucose uptake by the tissue cells. Insulin resistance is an important cause of type 2 diabetes mellitus. While there are various factors responsible for the etiology of insulin resistance, dysbiosis of gut microbiota may be an important contributing cause for metabolic disturbances. We discuss the mechanisms in skeletal muscles, adipose tissue, liver, and intestine by which insulin resistance can occur due to gut microbiota's metabolites. A better understanding of gut microbiota may help in the effective treatment of type 2 diabetes mellitus and metabolic syndrome.
Collapse
Affiliation(s)
- Alsalt Al-Busaidi
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Omer Alabri
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Jaifar Alomairi
- Department of Medicine, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | | | | | - Hanan Al Lawati
- Pharmacy Program, Department of Pharmaceutics, Oman College of Health Sciences, Muscat 113, Oman
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
10
|
Marinho CS, Koch C, Santos MR. Eating habits: what foods do children between 12 and 36 months consume? Rev Bras Enferm 2023; 76:e20220393. [PMID: 37820136 PMCID: PMC10561921 DOI: 10.1590/0034-7167-2022-0393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/26/2022] [Indexed: 10/13/2023] Open
Abstract
OBJECTIVE to identify toddlers' eating habits. METHOD a cross-sectional study of quantitative analysis, with a sample of 808 toddlers who attended day care centers in the district of Viseu, Portugal, between November 2018 and September 2019. Data were collected using a questionnaire directed at parents. RESULTS the prevalence of children who ate six meals a day was 42.8%, and 42.5%, those who ate five meals. It was found that 2.0% of children consumed chocolates, 1.0%, desserts, and 0.4%, carbonated beverages, daily. On average, dairy product (M=5.61; SD=2.62) and meat/fish/egg (M=4.80; SD=3.57) consumption was higher than recommended, while fat (M=0.48; SD=0.40), legume (M=0.49; SD=0.45), vegetable (M=1.18; SD=0.87) and water (M=0 .51; SD=0.29) consumption was lower. CONCLUSIONS there was a higher or lower consumption than recommended for some foods, highlighting the need to implement nursing intervention programs aimed at promoting healthy eating habits in toddlers and families.
Collapse
Affiliation(s)
| | - Cândida Koch
- Escola Superior de Enfermagem do Porto. Porto, Portugal
| | - Margarida Reis Santos
- Universidade do Porto. Porto, Portugal
- Escola Superior de Enfermagem do Porto. Porto, Portugal
| |
Collapse
|
11
|
Dos Santos SJ, Shukla I, Hill JE, Money DM. Birth Mode Does Not Determine the Presence of Shared Bacterial Strains between the Maternal Vaginal Microbiome and the Infant Stool Microbiome. Microbiol Spectr 2023; 11:e0061423. [PMID: 37338388 PMCID: PMC10433807 DOI: 10.1128/spectrum.00614-23] [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: 02/09/2023] [Accepted: 06/04/2023] [Indexed: 06/21/2023] Open
Abstract
Dysbiosis of the neonatal gut microbiome during early life has been suggested as the missing link that may explain higher rates of certain diseases in caesarean section-delivered infants. Many studies report delivery mode-related dysbiosis in infants due to a lack of maternal vaginal microbiome exposure, prompting interventions to correct the neonatal gut microbiome by transferring these missing microbes after caesarean delivery. The maternal vaginal microbiome is among the first microbial exposures that many infants experience, yet little is known about the extent of direct transmission of maternal vaginal microbes. As part of the Maternal Microbiome Legacy Project, we aimed to determine if maternal vaginal bacteria are vertically transmitted to infants. We employed cpn60 microbiome profiling, culture-based screening, molecular strain typing, and whole-genome sequencing to determine whether identical maternal vaginal strains were present in infant stool microbiomes. We identified identical cpn60 sequence variants in both halves of maternal-infant dyads in 204 of 585 Canadian women and their newborn infants (38.9%). The same species of Bifidobacterium and Enterococcus were cultured from maternal and corresponding infant samples in 33 and 13 of these mother-infant dyads, respectively. Pulsed-field gel electrophoresis and whole-genome sequencing determined that near-identical strains were detected in these dyads irrespective of delivery mode, indicating an alternative source in cases of caesarean delivery. Overall, we demonstrated that vertical transmission of maternal vaginal microbiota is likely limited and that transmission from other maternal body sites, such as the gut and breast milk, may compensate for the lack of maternal vaginal microbiome exposure during caesarean delivery. IMPORTANCE The importance of the gut microbiome in human health and disease is widely recognized, and there has been a growing appreciation that alterations in gut microbiome composition during a "critical window" of development may impact health in later life. Attempts to correct gut microbiome dysbiosis related to birth mode are underpinned by the assumption that the lack of exposure to maternal vaginal microbes during caesarean delivery is responsible for dysbiosis. Here, we demonstrate that there is limited transmission of the maternal vaginal microbiome to the neonatal gut, even in cases of vaginal delivery. Furthermore, the presence of identical strains shared between mothers and infants in early life, even in cases of caesarean delivery, highlights compensatory microbial exposures and sources for the neonatal stool microbiome other than the maternal vagina.
Collapse
Affiliation(s)
- Scott J. Dos Santos
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ishika Shukla
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Janet E. Hill
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Deborah M. Money
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
- Women’s Health Research Institute, B.C. Women’s Hospital, Vancouver, British Columbia, Canada
| | - The Maternal Microbiome Legacy Project Team
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
- Women’s Health Research Institute, B.C. Women’s Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
12
|
Liu Y, Ma J, Zhu B, Liu F, Qin S, Lv N, Feng Y, Wang S, Yang H. A health-promoting role of exclusive breastfeeding on infants through restoring delivery mode-induced gut microbiota perturbations. Front Microbiol 2023; 14:1163269. [PMID: 37492252 PMCID: PMC10363731 DOI: 10.3389/fmicb.2023.1163269] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/15/2023] [Indexed: 07/27/2023] Open
Abstract
The establishment of human gut microbiota in early life is closely associated with both short- and long-term infant health. Delivery mode and feeding pattern are two important determinants of infant gut microbiota. In this longitudinal cohort study, we examined the interplay between the delivery mode and feeding pattern on the dynamics of infant gut microbiota from 6 weeks to 6 months post-delivery in 139 infants. We also assessed the relationship between infant respiratory infection susceptibility and gut microbial changes associated with delivery mode and feeding pattern. At 6 weeks postpartum, the composition and structure of gut microbiota of cesarean section-delivered (CSD) infants differed from those of vaginally delivered (VD) infants, with decreased Bacteroides and Escherichia-Shigella and increased Klebsiella, Veillonella, and Enterococcus. At 6 months postpartum, these delivery mode-induced microbial shifts were restored by exclusive breastfeeding, resulting in similar gut microbial profiles between VD and CSD infants who were exclusively breastfed (P = 0.57) and more variable gut microbial profiles between VD and CSD infants who were mixed fed (P < 0.001). We identified that the VD-associated genera were enriched in healthy infants, while the CSD-associated genera were enriched in infants who suffered from respiratory infections. Our findings indicate that exclusive breastfeeding may play a health-promoting role by reducing infant respiratory infection susceptibility through the restoration of gut microbiota perturbations caused by cesarean section.
Collapse
Affiliation(s)
- Yu Liu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Jingmei Ma
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Baoli Zhu
- Key Laboratory of Pathogenic Microbiology and Immunology/Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Fei Liu
- Key Laboratory of Pathogenic Microbiology and Immunology/Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Shengtang Qin
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Na Lv
- Key Laboratory of Pathogenic Microbiology and Immunology/Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Ye Feng
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Shuxian Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| |
Collapse
|
13
|
Carrizales-Sánchez AK, Tamez-Rivera O, García-Gamboa R, García-Cayuela T, Rodríguez-Gutiérrez NA, Elizondo-Montemayor L, García-Rivas G, Pacheco A, Hernández-Brenes C, Senés-Guerrero C. Gut microbial composition and functionality of school-age Mexican population with metabolic syndrome and type-2 diabetes mellitus using shotgun metagenomic sequencing. Front Pediatr 2023; 11:1193832. [PMID: 37342535 PMCID: PMC10277889 DOI: 10.3389/fped.2023.1193832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Gut metagenome in pediatric subjects with metabolic syndrome (MetS) and type-2 diabetes mellitus (T2DM) has been poorly studied, despite an alarming worldwide increase in the prevalence and incidence of obesity and MetS within this population. The objective of this study was to characterize the gut microbiome taxonomic composition of Mexican pediatric subjects with MetS and T2DM using shotgun metagenomics and analyze the potential relationship with metabolic changes and proinflammatory effects. Paired-end reads of fecal DNA samples were obtained through the Illumina HiSeq X Platform. Statistical analyses and correlational studies were conducted using gut microbiome data and metadata from all individuals. Gut microbial dysbiosis was observed in MetS and T2DM children compared to healthy subjects, which was characterized by an increase in facultative anaerobes (i.e., enteric and lactic acid bacteria) and a decrease in strict anaerobes (i.e., Erysipelatoclostridium, Shaalia, and Actinomyces genera). This may cause a loss of gut hypoxic environment, increased gut microbial nitrogen metabolism, and higher production of pathogen-associated molecular patterns. These metabolic changes may trigger the activation of proinflammatory activity and impair the host's intermediate metabolism, leading to a possible progression of the characteristic risk factors of MetS and T2DM, such as insulin resistance, dyslipidemia, and an increased abdominal circumference. Furthermore, specific viruses (Jiaodavirus genus and Inoviridae family) showed positive correlations with proinflammatory cytokines involved in these metabolic diseases. This study provides novel evidence for the characterization of MetS and T2DM pediatric subjects in which the whole gut microbial composition has been characterized. Additionally, it describes specific gut microorganisms with functional changes that may influence the onset of relevant health risk factors.
Collapse
Affiliation(s)
| | - Oscar Tamez-Rivera
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
| | - Ricardo García-Gamboa
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Zapopan, Jalisco, Mexico
- Tecnologico de Monterrey, Escuela de Medicina, Colonia Nuevo México, Zapopan, Jalisco, México
| | - Tomás García-Cayuela
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Zapopan, Jalisco, Mexico
| | - Nora A Rodríguez-Gutiérrez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
- Hospital Regional Materno Infantil de Alta Especialidad, Guadalupe, Nuevo Leon, Mexico
| | | | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, Nuevo Leon, Mexico
| | - Adriana Pacheco
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Monterrey, Nuevo Leon, Mexico
| | - Carmen Hernández-Brenes
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, Nuevo Leon, Mexico
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Monterrey, Nuevo Leon, Mexico
| | | |
Collapse
|
14
|
Ahlberg E, Al-Kaabawi A, Thune R, Simpson MR, Pedersen SA, Cione E, Jenmalm MC, Tingö L. Breast milk microRNAs: Potential players in oral tolerance development. Front Immunol 2023; 14:1154211. [PMID: 36999032 PMCID: PMC10045994 DOI: 10.3389/fimmu.2023.1154211] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Breast milk is an essential source of nutrition and hydration for the infant. In addition, this highly complex biological fluid contains numerous immunologically active factors such as microorganisms, immunoglobulins, cytokines and microRNAs (miRNAs). Here, we set out to predict the function of the top 10 expressed miRNAs in human breast milk, focusing on their relevance in oral tolerance development and allergy prevention in the infant. The top expressed miRNAs in human breast milk were identified on basis of previous peer-reviewed studies gathered from a recent systematic review and an updated literature search. The miRNAs with the highest expression levels in each study were used to identify the 10 most common miRNAs or miRNA families across studies and these were selected for subsequent target prediction. The predictions were performed using TargetScan in combination with the Database for Annotation, Visualization and Integrated Discovery. The ten top expressed miRNAs were: let-7-5p family, miR-148a-3p, miR-30-5p family, miR-200a-3p + miR-141-3p, miR-22-3p, miR-181-5p family, miR-146b-5p, miR-378a-3p, miR-29-3p family, miR-200b/c-3p and miR-429-3p. The target prediction identified 3,588 potential target genes and 127 Kyoto Encyclopedia of Genes and Genomes pathways; several connected to the immune system, including TGF-b and T cell receptor signaling and T-helper cell differentiation. This review highlights the role of breast milk miRNAs and their potential contribution to infant immune maturation. Indeed, breast milk miRNAs seem to be involved in several pathways that influence oral tolerance development.
Collapse
Affiliation(s)
- Emelie Ahlberg
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ahmed Al-Kaabawi
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Rebecka Thune
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sindre Andre Pedersen
- Library Section for Research Support, Data and Analysis, NTNU University Library, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Cosenza, Italy
| | - Maria Christina Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lina Tingö
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
- Food and Health Programme, Örebro University, Örebro, Sweden
- *Correspondence: Lina Tingö,
| |
Collapse
|
15
|
Kapoor B, Gulati M, Gupta R, Singla RK. Microbiota dysbiosis and myasthenia gravis: Do all roads lead to Rome? Autoimmun Rev 2023; 22:103313. [PMID: 36918089 DOI: 10.1016/j.autrev.2023.103313] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
Dysregulated immune system with a failure to recognize self from non-self-antigens is one of the common pathogeneses seen in autoimmune diseases. The complex interplay of genetic and environmental factors is important for the occurrence and development of the disease. Among the environmental factors, disturbed gut microbiota (gut dysbiosis) has recently attracted particular attention, especially with advancement in human microbiome research. Although the alterations in microbiota have been seen in various autoimmune diseases, including those of nervous system, there is paucity of information on neuromuscular system diseases. Myasthenia gravis (MG) is one such rare autoimmune disease of neuromuscular junction, and is caused by generation of pathogenic autoantibodies to components of the postsynaptic muscle endplate. In the recent years, accumulating evidences have endorsed the key role of host microbiota, particularly those of gut, in the pathogenesis of MG. Differential microbiota composition, characterized by increased abundance of Fusobacteria, Bacteroidetes, and Proteobacteria, and decreased abundance of Actinobacteria and Firmicutes, has been seen in MG patients in comparison to healthy subjects. Disturbance of microbiota composition, particularly reduced ratio of Firmicutes/Bacteroidetes, alter the gut permeability, subsequently triggering the immunological response. Resultant reduction in levels of short chain fatty acids (SCFAs) is another factor contributing to the immunological response in MG patients. Modulation of gut microbiota via intervention of probiotics, prebiotics, synbiotics, postbiotics (metabiotics), and fecal microbiota transplantation (FMT) is considered to be the futuristic approach for the management of MG. This review summarizes the role of gut microbiota and their metabolites (postbiotics) in the progression of MG. Also, various bacteriotherapeutic approaches involving gut microbiota are discussed for the prevention of MG progression.
Collapse
Affiliation(s)
- Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, NSW 2007, Australia.
| | - Reena Gupta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rajeev K Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road, 2222, Chengdu, Sichuan, China; iGlobal Research and Publishing Foundation, New Delhi, India
| |
Collapse
|
16
|
Development of the Anaerobic Microbiome in the Infant Gut. Pediatr Infect Dis J 2023:00006454-990000000-00384. [PMID: 36917032 DOI: 10.1097/inf.0000000000003905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Ninety-five percent of gut microbiota are anaerobes and vary according to age and diet. Complex carbohydrates in human milk enhance the growth of Bifidobacterium and Bacteroides in the first year. Complex carbohydrates in solid foods enhance the growth of Bacteroides and Clostridium in the second year. Short-chain fatty acids produced by Akkermansia and Faecalibacterium may reduce obesity, diabetes and IBD.
Collapse
|
17
|
Samarra A, Esteban-Torres M, Cabrera-Rubio R, Bernabeu M, Arboleya S, Gueimonde M, Collado MC. Maternal-infant antibiotic resistance genes transference: what do we know? Gut Microbes 2023; 15:2194797. [PMID: 37020319 PMCID: PMC10078139 DOI: 10.1080/19490976.2023.2194797] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
Resistance to antibiotics is becoming a worldwide threat as infections caused by multidrug-resistant pathogenic microorganisms can overcome antibiotic treatments and spread quickly in the population. In the context of early life, newborns are at increased risk as their immune system is still under development, so infections and acquisition of resistance during childhood have short- and long-term consequences for the health. The moment of birth is the first exposure of infants to possible antibiotic-resistant microorganisms that may colonize their gut and other body sites. Different factors including mode of delivery, previous antibiotic exposure of the mother, gestational age and consumption of antibiotics in early-life have been described to modulate the neonate's microbiota, and thus, the resistome. Other factors, such as lactation, also impact the establishment and development of gut microbiota, but little is known about the role of breastmilk in transferring Antibiotic Resistant Genes (ARG). A deeper understanding of vertical transmission of antibiotic resistance from mothers to their offspring is necessary to determine the most effective strategies for reducing antibiotic resistance in the early life. In this review, we aim to present the current perspective on antibiotic resistances in mother-infant dyads, as well as a new insight on the study of the human gut and breastmilk resistome, and current strategies to overcome this public health problem, toward highlighting the gaps of knowledge that still need to be closed.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
| | - Maria Esteban-Torres
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
| | - Manuel Bernabeu
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
- Vicerectorat de Recerca, Universitat de Barcelona (UB), Barcelona, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry, Dairy Research Institute- National Research Council (IPLA-CSIC), Villaviciosa, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry, Dairy Research Institute- National Research Council (IPLA-CSIC), Villaviciosa, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
18
|
Strobel KM, Del Vecchio G, Devaskar SU, Calkins KL. Gut Microbes and Circulating Cytokines in Preterm Infants with Growth Failure. J Nutr 2023; 153:120-130. [PMID: 36913445 PMCID: PMC10196572 DOI: 10.1016/j.tjnut.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/12/2022] [Accepted: 10/23/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Growth failure (GF) is a multifactorial problem in preterm infants. The intestinal microbiome and inflammation may contribute to GF. OBJECTIVES This study's objective was to compare the gut microbiome and plasma cytokines in preterm infants with and without GF. METHODS This was a prospective cohort study of infants with birth weights of <1750 g. Infants with a weight or length z-score change from birth to discharge or death that was less than or equal to -0.8 (GF group) were compared with infants without GF [control (CON) group]. The primary outcome was the gut microbiome (at weeks 1-4 of age), assessed by 16S rRNA gene sequencing using Deseq2. Secondary outcomes included inferred metagenomic function and plasma cytokines. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States determined metagenomic function, which was compared using ANOVA. Cytokines were measured by 2-multiplexed immunometric assays and compared using Wilcoxon tests and linear mixed models. RESULTS GF (n = 14) and CON group (n = 13) had similar median (IQR) birth weight (1380 [780-1578] g vs. 1275 [1013-1580] g) and gestational age (29 [25-31] weeks vs. 30 [29-32] weeks). Compared with the CON group, the GF group had a greater abundance of Escherichia/Shigella in weeks 2 and 3, Staphylococcus in week 4, and Veillonella in weeks 3 and 4 (P-adjusted < 0.001 for all). Plasma cytokine concentrations did not differ significantly between the cohorts. When all time points are combined, fewer microbes were involved in TCA cycle activity in the GF group compared with the CON group (P = 0.023). CONCLUSIONS In this study, when compared with CON infants, GF infants had a distinct microbial signature with increased Escherichia/Shigella and Firmicutes and fewer microbes associated with energy production at later weeks of hospitalization. These findings may suggest a mechanism for aberrant growth.
Collapse
Affiliation(s)
- Katie M Strobel
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Giorgia Del Vecchio
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Sherin U Devaskar
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Kara L Calkins
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Dualib PM, Fernandes G, Taddei CR, Carvalho CRS, Sparvoli LG, Bittencourt C, Silva IT, Mattar R, Ferreira SRG, Dib SA, de Almeida-Pititto B. The gut microbiome of obese postpartum women with and without previous gestational diabetes mellitus and the gut microbiota of their babies. Diabetol Metab Syndr 2022; 14:194. [PMID: 36566315 PMCID: PMC9790115 DOI: 10.1186/s13098-022-00954-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/14/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The incidence of gestational diabetes mellitus (GDM) is increasing worldwide, and has been associated with some changes in the gut microbiota. Studies have shown that the maternal gut microbiota pattern with hyperglycemia can be transmitted to the offspring. The study aimed to evaluate the gut microbiota of obese postpartum women with and without previous GDM and their offspring. METHODS We evaluated a total of 84 puerperal women who had (n = 40) or not GDM (n = 44), and their infants were also included. Stool samples were obtained 2-6 months after delivery. The molecular profile of the fecal microbiota was obtained by sequencing V4 region of 16S rRNA gene (Illumina® MiSeq). RESULTS We found that the gut microbiota structures of the puerperal women and their infants were similar. Stratifying according to the type of delivery, the relative abundance of Victivallis genus was higher in women who had natural delivery. Exposure to exclusive breastfeeding was associated with a greater abundance of Bacteroides and Staphylococcus. The differential abundance test showed correlations to clinical and laboratory parameters. This work showed no difference in the microbiota of obese puerperal women with and without GDM and their offspring. However, breastfeeding contributed to the ecological succession of the intestinal microbiota of the offspring. CONCLUSION This work can contribute to understanding the potential effects of GDM and early life events on the gut microbiome of mothers and their offspring and its possible role in metabolism later in life.
Collapse
Affiliation(s)
- Patricia Medici Dualib
- Department of Medicine, Escola Paulista Medicina, Universidade Federal de São Paulo, Rua Sena Madureira, 1500, Vila Clementino, São Paulo, SP, CEP 04021-001, Brazil.
| | - Gabriel Fernandes
- DepaBiosystems Informatics and Genomics Group, Instituto René Rachou - Fiocruz Minas, Av Augusto de Lima, 1714, Belo Horizonte, MG, CEP 30190-002, Brazil
| | - Carla R Taddei
- Department of Clinical and Toxicological Analysis, Universidade de São Paulo (USP), Av. Prof. Lineu Prestes 580-Bloco 17, São Paulo, SP, CEP 05508-000, Brazil
| | - Camila R S Carvalho
- Graduate Program in Endocrinology and Metabology, Universidade Federal de São Paulo, Rua Estado de Israel, nº 639, Vila Clementino, São Paulo, SP, CEP 04022-001, Brazil
| | - Luiz Gustavo Sparvoli
- Department of Clinical and Toxicological Analysis, Universidade de São Paulo (USP), Av. Prof. Lineu Prestes 580-Bloco 17, São Paulo, SP, CEP 05508-000, Brazil
| | - Célia Bittencourt
- Graduate Program in Endocrinology and Metabology, Universidade Federal de São Paulo, Rua Estado de Israel, nº 639, Vila Clementino, São Paulo, SP, CEP 04022-001, Brazil
| | - Isis T Silva
- Nutrition Course, Centro Universitário Estácio de Sá, Rua Erê, 207, Belo Horizonte, MG, CEP 30411-052, Brazil
| | - Rosiane Mattar
- Departament of Obstetrics, Escola Paulista de Medicina, Universidade Federal de São Paulo, R. Napoleão de Barros, 875 - Vila Clementino, São Paulo, SP, CEP 04024-002, Brazil
| | - Sandra R G Ferreira
- Department of Epidemiology, School of Public Health, Universidade de São Paulo, Av. Dr. Arnaldo, 715 - Cerqueira César, São Paulo, SP, CEP 01246-904, Brazil
| | - Sergio A Dib
- Department of Medicine, Escola Paulista Medicina, Universidade Federal de São Paulo, Rua Sena Madureira, 1500, Vila Clementino, São Paulo, SP, CEP 04021-001, Brazil
| | - Bianca de Almeida-Pititto
- Department of Preventive Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, Campus São Paulo, Rua Botucatu, n° 740, Vila Clementino, São Paulo, SP, CEP 04023-062, Brazil
| |
Collapse
|
20
|
Tang M, Marroquin E. The role of the gut microbiome in the intergenerational transmission of the obesity phenotype: A narrative review. Front Med (Lausanne) 2022; 9:1057424. [PMID: 36619646 PMCID: PMC9812955 DOI: 10.3389/fmed.2022.1057424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Obesity is considered an epidemic by the World Health Organization. In particular, maternal obesity can affect the development of obesity and other related metabolic disorders in infants. Recently, both animal and human studies have pointed to the importance of the gut microbiome in facilitating the transmission of the obesity phenotype from mother to offspring. The gut microbiome changes significantly during the progression of pregnancy, and the microbiota of the amniotic fluid and placenta have recently been shown to colonize the infant gut in utero. Microbial composition, diversity, and richness are significantly altered by maternal obesity, which in turn affects the infant's acquisition of the gut microbiome and their risk to develop metabolic disorders. C-section has also been shown to affect the colonization of the infant gut and offspring metabolic and immune health. This narrative review seeks to discuss the role of the gut microbiome in the transmission of the obesity phenotype from mother to child, as well as how birth delivery, breastfeeding, and probiotic interventions may modulate this relationship.
Collapse
Affiliation(s)
- Mabel Tang
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Elisa Marroquin
- Department of Nutritional Sciences, Texas Christian University, Fort Worth, TX, United States,*Correspondence: Elisa Marroquin,
| |
Collapse
|
21
|
Exploring the Potential of Human Milk and Formula Milk on Infants’ Gut and Health. Nutrients 2022; 14:nu14173554. [PMID: 36079814 PMCID: PMC9460722 DOI: 10.3390/nu14173554] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Early-life gut microbiota plays a role in determining the health and risk of developing diseases in later life. Various perinatal factors have been shown to contribute to the development and establishment of infant gut microbiota. One of the important factors influencing the infant gut microbial colonization and composition is the mode of infant feeding. While infant formula milk has been designed to resemble human milk as much as possible, the gut microbiome of infants who receive formula milk differs from that of infants who are fed human milk. A diverse microbial population in human milk and the microbes seed the infant gut microbiome. Human milk contains nutritional components that promote infant growth and bioactive components, such as human milk oligosaccharides, lactoferrin, and immunoglobulins, which contribute to immunological development. In an attempt to encourage the formation of a healthy gut microbiome comparable to that of a breastfed infant, manufacturers often supplement infant formula with prebiotics or probiotics, which are known to have a bifidogenic effect and can modulate the immune system. This review aims to elucidate the roles of human milk and formula milk on infants’ gut and health.
Collapse
|
22
|
Pivrncova E, Kotaskova I, Thon V. Neonatal Diet and Gut Microbiome Development After C-Section During the First Three Months After Birth: A Systematic Review. Front Nutr 2022; 9:941549. [PMID: 35967823 PMCID: PMC9364824 DOI: 10.3389/fnut.2022.941549] [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] [Received: 05/11/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background Cesarean section (C-section) delivery imprints fundamentally on the gut microbiota composition with potential health consequences. With the increasing incidence of C-sections worldwide, there is a need for precise characterization of neonatal gut microbiota to understand how to restore microbial imbalance after C-section. After birth, gut microbiota development is shaped by various factors, especially the infant’s diet and antibiotic exposure. Concerning diet, current research has proposed that breastfeeding can restore the characteristic gut microbiome after C-section. Objectives In this systematic review, we provide a comprehensive summary of the current literature on the effect of breastfeeding on gut microbiota development after C-section delivery in the first 3 months of life. Methods The retrieved data from PubMed, Scopus, and Web of Science were evaluated according to the PICO/PECO strategy. Quality assessment was conducted by the Newcastle–Ottawa Scale. Results After critical selection, we identified 14 out of 4,628 studies for the evaluation of the impact of the diet after C-section delivery. The results demonstrate consistent evidence that C-section and affiliated intrapartum antibiotic exposure affect Bacteroidetes abundance and the incapacity of breastfeeding to reverse their reduction. Furthermore, exclusive breastfeeding shows a positive effect on Actinobacteria and Bifidobacteria restoration over the 3 months after birth. None of the included studies detected any significant changes in Lactobacillus abundance in breastfed infants after C-section. Conclusion C-section and intrapartum antibiotic exposure influence an infant’s gut microbiota by depletion of Bacteroides, regardless of the infant’s diet in the first 3 months of life. Even though breastfeeding increases the presence of Bifidobacteria, further research with proper feeding classification is needed to prove the restoration effect on some taxa in infants after C-section. Systematic Review Registration: [www.crd.york.ac.uk/prospero/], identifier [CRD42021287672].
Collapse
Affiliation(s)
- Eliska Pivrncova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Iva Kotaskova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Vojtech Thon
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
23
|
Qin S, Wang Y, Wang S, Ning B, Huai J, Yang H. Gut microbiota in women with gestational diabetes mellitus has potential impact on metabolism in pregnant mice and their offspring. Front Microbiol 2022; 13:870422. [PMID: 35992705 PMCID: PMC9389115 DOI: 10.3389/fmicb.2022.870422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/07/2022] [Indexed: 11/25/2022] Open
Abstract
Studies have shown that gestational diabetes mellitus (GDM) is closely related to abnormalities in the gut microbiota, and the offspring of these women have an increased risk of diabetes. There is no direct evidence of whether bacteria in women with GDM colonize the intestinal tract of offspring and cause hyperglycemia. In this fecal microbiota transplantation (FMT), pregnant mouse model study, two groups of germ-free (GF) mice after FMT showed different colonization patterns of gut microbiota and phenotype. Compared with the control group (healthy-FMT), we found in the GDM-FMT group as a lower relative abundance of Akkermansia and Faecalibacterium; a lower content of short-chain fatty acids and naringenin in feces; an elevated blood glucose; an inflammatory factor expression (TNF-α, CXCL-15, and IL-6), and a hepatic fat deposition. In addition, the influence of the gut microbiota continued in offspring. The gut microbiota of the offspring of GDM-FMT mice was still different from that of the control group as a lower relative abundance of Akkermansia and Parvibacter; and a higher relative abundance of bacteria such as Oscillibacter, Romboutsia, and Harryflintia. In addition, the offspring of GDM-FMT mice had higher body weight and blood glucose levels than the control offspring.
Collapse
Affiliation(s)
- Shengtang Qin
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Yutong Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Shuxian Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Bohan Ning
- Department of Pathology, Peking University First Hospital, Beijing, China
| | - Jing Huai
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
- *Correspondence: Huixia Yang,
| |
Collapse
|
24
|
Time-Specific Factors Influencing the Development of Asthma in Children. Biomedicines 2022; 10:biomedicines10040758. [PMID: 35453508 PMCID: PMC9025817 DOI: 10.3390/biomedicines10040758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 02/01/2023] Open
Abstract
Susceptibility to asthma is complex and heterogeneous, as it involves both genetic and environmental insults (pre- and post-birth) acting in a critical window of development in early life. According to the Developmental Origins of Health and Disease, several factors, both harmful and protective, such as nutrition, diseases, drugs, microbiome, and stressors, interact with genotypic variation to change the capacity of the organism to successfully adapt and grow in later life. In this review, we aim to provide the latest evidence about predictive risk and protective factors for developing asthma in different stages of life, from the fetal period to adolescence, in order to develop strategic preventive and therapeutic interventions to predict and improve health later in life. Our study shows that for some risk factors, such as exposure to cigarette smoke, environmental pollutants, and family history of asthma, the evidence in favor of a strong association of those factors with the development of asthma is solid and widely shared. Similarly, the clear benefits of some protective factors were shown, providing new insights into primary prevention. On the contrary, further longitudinal studies are required, as some points in the literature remain controversial and a source of debate.
Collapse
|
25
|
Cirpanli C, Hicyilmaz BD. Postcesarean Difficulties and their Association with Breastfeeding Success in Postpartum Women. Niger J Clin Pract 2022; 25:69-77. [PMID: 35046198 DOI: 10.4103/njcp.njcp_546_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background The rate of cesarean section (CS) has been gradually increasing throughout the world. The impact of CS on the issue of breastfeeding success therefore merits attention. Aim This study investigates postcesarean difficulties and their association with breastfeeding success. Patients and Methods This study is a cross-sectional study involving the participation of 220 mothers who were receiving care at the Women's Health Training and Research Hospital in Ankara, Turkey between the dates of February 10 and August 10 of 2016. Data were collected using the Socio-demographic and Obstetric Characteristics Form and the LATCH Breastfeeding Assessment Tool (LBAT). Results Women who had breastfeeding problems both previously (OR = 0.59; 95% CI: 0.18-1.76; P = 0.004) and currently (OR = 0.29; 95% CI: 0.14-1.30; P < 0.001) were more likely to have low breastfeeding success scores. There was no statistically significant relationship found between LBAT mean scores and other post-CS difficulties. Women aged 35 and older were more likely to have breastfeeding success compared to that of women of ≤19 years of age (OR = 4.33; 95% CI: 3.50-4.45; P = 0.051). Furthermore, women who had breastfeeding experience were more likely (OR = 1.86; 95% CI: 1.40-2.67; P = 0.010) to have a high breastfeeding success score. Conclusions This study has revealed that breastfeeding success was associated more with breastfeeding problems than with other difficulties experienced after a CS. To prevent breastfeeding problems before they occur, newborns who are without complications should be kept in skin-to-skin contact (SSC) with their mothers immediately after the CS, breastfed within the first 1 hour after birth, and remain in SSC until the end of the first breastfeeding. Most importantly, to increase breastfeeding success after CS, creative staffing solutions need to be employed.
Collapse
Affiliation(s)
- C Cirpanli
- Neonatal Intensive Care Unit, Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | | |
Collapse
|
26
|
Vasilescu IM, Chifiriuc MC, Pircalabioru GG, Filip R, Bolocan A, Lazăr V, Diţu LM, Bleotu C. Gut Dysbiosis and Clostridioides difficile Infection in Neonates and Adults. Front Microbiol 2022; 12:651081. [PMID: 35126320 PMCID: PMC8810811 DOI: 10.3389/fmicb.2021.651081] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
In this review, we focus on gut microbiota profiles in infants and adults colonized (CDC) or infected (CDI) with Clostridioides difficile. After a short update on CDI epidemiology and pathology, we present the gut dysbiosis profiles associated with CDI in adults and infants, as well as the role of dysbiosis in C. difficile spores germination and multiplication. Both molecular and culturomic studies agree on a significant decrease of gut microbiota diversity and resilience in CDI, depletion of Firmicutes, Bacteroidetes, and Actinobacteria phyla and a high abundance of Proteobacteria, associated with low butyrogenic and high lactic acid-bacteria levels. In symptomatic cases, microbiota deviations are associated with high levels of inflammatory markers, such as calprotectin. In infants, colonization with Bifidobacteria that trigger a local anti-inflammatory response and abundance of Ruminococcus, together with lack of receptors for clostridial toxins and immunological factors (e.g., C. difficile toxins neutralizing antibodies) might explain the lack of clinical symptoms. Gut dysbiosis amelioration through administration of “biotics” or non-toxigenic C. difficile preparations and fecal microbiota transplantation proved to be very useful for the management of CDI.
Collapse
Affiliation(s)
- Iulia-Magdalena Vasilescu
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- INBI “Prof. Dr. Matei Balş” – National Institute for Infectious Diseases, Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- The Romanian Academy, Bucharest, Romania
- *Correspondence: Mariana-Carmen Chifiriuc,
| | | | - Roxana Filip
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, Suceava, Romania
- Regional County Emergency Hospital, Suceava, Romania
| | - Alexandra Bolocan
- Department of General Surgery, University Emergency Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Veronica Lazăr
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Lia-Mara Diţu
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Coralia Bleotu
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Research Institute of the University of Bucharest, Bucharest, Romania
- Ştefan S. Nicolau Institute of Virology, Romanian Academy, Bucharest, Romania
| |
Collapse
|
27
|
Yang B, Ding M, Chen Y, Han F, Yang C, Zhao J, Malard P, Stanton C, Ross RP, Zhang H, Chen W. Development of gut microbiota and bifidobacterial communities of neonates in the first 6 weeks and their inheritance from mother. Gut Microbes 2022; 13:1-13. [PMID: 33847206 PMCID: PMC8049200 DOI: 10.1080/19490976.2021.1908100] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Microbiota especially Bifidobacterium play an important role in adjusting and maintaining homeostatic balance within the infant intestine. The aim of this study was to elucidate the relationship between maternal and infant gut microbiota and identify the Bifidobacterium species that may transfer from mother to infant over the first 42 days of the infant's life. Nineteen mother-infant-pair fecal samples were collected and the diversity and composition of the total bacterial and Bifidobacterium communities were analyzed via 16S rDNA and bifidobacterial groEL gene high throughput sequencing. The results revealed that the relative abundance of Bifidobacterium was significantly higher in the infant gut while Parabacteroides, Blautia, Coprococcus, Lachnospira and Faecalibacterium were at lower relative abundance in 7-day and 42-day infant fecal samples compared to the maternal samples. The maternal gut has more B. pseudocatenulatum. In the infant group, B. breve and B. dentium relative abundance increased while B. animalis subsp. lactis decreased from days 7 to 42. Additionally, B. longum subsp. longum isolated from FGZ16 and FGZ35 may have transferred from mother to infant and colonized the infant gut. The results of the current study provide insight toward the infant gut microbiota composition and structure during the first 42 days and may help guide Bifidobacterium supplementation strategies in mothers and infants.
Collapse
Affiliation(s)
- Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China,International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China
| | - Mengfan Ding
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yingqi Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Fengzhen Han
- Department of Gynaecology and Obsterics, Guangdong Province People’s Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Chunyan Yang
- Department of Gynaecology and Obsterics, Guangdong Province People’s Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Patrice Malard
- Biostime (Guangzhou) Health Products Ltd., Guangzhou, China
| | - Catherine Stanton
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China,Food Bioscience, Teagasc Food Research Centre, Fermoy, Ireland,CONTACT Catherine Stanton Teagasc Food Research Centre, Fermoy, Ireland
| | - R. Paul Ross
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China,School of Food Science and Technology, Jiangnan University, Wuxi, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China,Wei Chen School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
28
|
Korpela K, de Vos WM. Infant gut microbiota restoration: state of the art. Gut Microbes 2022; 14:2118811. [PMID: 36093611 PMCID: PMC9467569 DOI: 10.1080/19490976.2022.2118811] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023] Open
Abstract
The gut microbiota has a central role in the programming of the host's metabolism and immune function, with both immediate and long-term health consequences. Recent years have witnessed an accumulation of understanding of the process of the colonization and development of the gut microbiota in infants. The natural gut microbiota colonization during birth is frequently disrupted due to C-section birth or intrapartum or postpartum antibiotic exposure, and consequently aberrant gut microbiota development is common. On a positive note, research has shown that restoration of normal gut microbiota development is feasible. We discuss here the current understanding of the infant microbiota, provide an overview of the sources of disturbances, and critically evaluate the evidence on early life gut microbiota restoration for improved health outcomes by analyzing published data from infant gut microbiota restoration studies.
Collapse
Affiliation(s)
- Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Willem M. de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, WE Wageningen, The Netherlands
| |
Collapse
|
29
|
Vitamin D Supplementation in Exclusively Breastfed Infants Is Associated with Alterations in the Fecal Microbiome. Nutrients 2022; 14:nu14010202. [PMID: 35011077 PMCID: PMC8747039 DOI: 10.3390/nu14010202] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
Breastfeeding and introduction of solid food are the two major components of infant feeding practices that influence gut microbiota composition in early infancy. However, it is unclear whether additional factors influence the microbiota of infants either exclusively breastfed or not breastfed. We obtained 194 fecal samples from infants at 3–9 months of age, extracted DNA, and sequenced the V4 region of the 16S rRNA gene. Feeding practices and clinical information were collected by questionnaire and abstraction of birth certificates. The gut microbiota of infants who were exclusively breastfed displayed significantly lower Shannon diversity (p-adjust < 0.001) and different gut microbiota composition compared to infants who were not breastfed (p-value = 0.001). Among the exclusively breastfed infants, recipients of vitamin D supplements displayed significantly lower Shannon diversity (p-adjust = 0.007), and different gut microbiota composition structure than non-supplemented, breastfed infants (p-value = 0.02). MaAslin analysis identified microbial taxa that associated with breastfeeding and vitamin D supplementation. Breastfeeding and infant vitamin D supplement intake play an important role in shaping infant gut microbiota.
Collapse
|
30
|
Gołębiewski M, Łoś-Rycharska E, Sikora M, Grzybowski T, Gorzkiewicz M, Krogulska A. Mother's Milk Microbiome Shaping Fecal and Skin Microbiota in Infants with Food Allergy and Atopic Dermatitis: A Pilot Analysis. Nutrients 2021; 13:nu13103600. [PMID: 34684601 PMCID: PMC8537811 DOI: 10.3390/nu13103600] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 01/04/2023] Open
Abstract
The child microbiome, including gut and skin communities, is shaped by a multitude of factors, and breastfeeding is one of the most essential. Food allergy (FA) and atopic dermatitis (AD) are among the most common diseases in pediatrics, with the prevalence of each up to 6% and 20%, respectively. Therefore, we aimed at finding differences between the fecal and skin microbiomes of FA and AD patients in the context of breastfeeding, by means of the Illumina sequencing of 16S rRNA gene fragment libraries amplified from the total DNA isolated from samples collected from allergic and healthy infants. We also analyzed milk samples from the mothers of the examined children and searched for patterns of incidence suggesting milk influence on an infant's allergy status. Here we show that a mother's milk influences her child's fecal and skin microbiomes and identify Acinetobacter as the taxon whose abundance is correlated with milk and child-derived samples. We demonstrate that breastfeeding makes allergic children's fecal and skin communities more similar to those of healthy infants than in the case of formula-feeding. We also identify signature taxa that might be important in maintaining health or allergy development.
Collapse
Affiliation(s)
- Marcin Gołębiewski
- Department of Plant Physiology and Biotechnology, Nicolaus Copernicus University, 87-100 Torun, Poland
- Interdisciplinary Centre of Modern Technologies, Nicolaus Copernicus University, 87-100 Torun, Poland;
- Correspondence: (M.G.); (E.Ł.-R.); Tel.: +48-56-611-2512 (M.G.); +48-52-585-4850 (E.Ł.-R.); Fax: +48-56-611-4559 (M.G.); +48-52-585-4086 (E.Ł.-R.)
| | - Ewa Łoś-Rycharska
- Department of Pediatrics, Allergology and Gastroenterology, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland;
- Correspondence: (M.G.); (E.Ł.-R.); Tel.: +48-56-611-2512 (M.G.); +48-52-585-4850 (E.Ł.-R.); Fax: +48-56-611-4559 (M.G.); +48-52-585-4086 (E.Ł.-R.)
| | - Marcin Sikora
- Interdisciplinary Centre of Modern Technologies, Nicolaus Copernicus University, 87-100 Torun, Poland;
| | - Tomasz Grzybowski
- Department of Forensic Medicine, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland; (T.G.); (M.G.)
| | - Marta Gorzkiewicz
- Department of Forensic Medicine, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland; (T.G.); (M.G.)
| | - Aneta Krogulska
- Department of Pediatrics, Allergology and Gastroenterology, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland;
| |
Collapse
|
31
|
Association of Cesarean Delivery and Formula Supplementation with the Stool Metabolome of 6-Week-Old Infants. Metabolites 2021; 11:metabo11100702. [PMID: 34677417 PMCID: PMC8540440 DOI: 10.3390/metabo11100702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/15/2022] Open
Abstract
Cesarean delivery and formula feeding have both been implicated as important factors associated with perturbations to the infant gut microbiome. To investigate the functional metabolic response of the infant gut microbial milieu to these factors, we profiled the stool metabolomes of 121 infants from a US pregnancy cohort study at approximately 6 weeks of life and evaluated associations with delivery mode and feeding method. Multivariate analysis of six-week stool metabolomic profiles indicated discrimination by both delivery mode and diet. For diet, exclusively breast-fed infants exhibited metabolomic profiles that were distinct from both exclusively formula-fed and combination-fed infants, which were relatively more similar to each other in metabolomic profile. We also identified individual metabolites that were important for differentiating delivery mode groups and feeding groups and metabolic pathways related to delivery mode and feeding type. We conclude based on previous work and this current study that the microbial communities colonizing the gastrointestinal tracts of infants are not only taxonomically, but also functionally distinct when compared according to delivery mode and feeding groups. Further, different sets of metabolites and metabolic pathways define delivery mode and diet metabotypes.
Collapse
|
32
|
Yang W, Tian L, Luo J, Yu J. Ongoing Supplementation of Probiotics to Cesarean-Born Neonates during the First Month of Life may Impact the Gut Microbial. Am J Perinatol 2021; 38:1181-1191. [PMID: 32446263 DOI: 10.1055/s-0040-1710559] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE The delivery mode is considered to be a significant influencing factor in the early gut microbiota composition, which is associated with the long-term health of the host. In this study, we tried to explore the effects of probiotics on the intestinal microbiota of C-section neonates. STUDY DESIGN Twenty-six Chinese neonates were enrolled in this study. The neonates were divided into four groups: VD (natural delivery neonates, n = 3), CD (cesarean-born neonates, n = 9), CDL (cesarean-born neonates supplemented with probiotic at a lower dosage, n = 7), and CDH (cesarean-born neonates supplemented with probiotic at a higher dosage, n = 7). Fecal samples were collected on the 3rd, 7th, and 28th day since birth. The V3-V4 region of the 16S ribosomal ribonucleic acid gene was sequenced by next-generation sequencing technology. RESULTS The α-diversity of the intestinal microbiota of cesarean delivery neonates was significantly lower than that of the naturally delivered neonates on the 28th day (p = 0.005). After supplementation with probiotics for 28 days, the α-diversity and the β-diversity of the gut flora in the cesarean-born infants (CDL28 and CDH28) was similar to that in the vaginally delivery infants. Meanwhile, the abundances of Lactobacillus and Bifidobacterium were significantly increased since the 3rd day of probiotic supplementation. Besides, the sustained supplementation of probiotics to neonates would help improve the abundance of the operational taxonomic units in several different Clusters of Orthologous Groups of proteins. CONCLUSION This study showed that probiotics supplementation to cesarean-born neonates since birth might impact the diversity and function of gut microbiota. KEY POINTS · Cesarean-born neonates. · Probiotic supplementation impact gut flora. · Bifidobacterium and Lactobacillus.
Collapse
Affiliation(s)
- Wenqing Yang
- Department of Neonatal Diagnosis and Treatment Center, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing Medical University, Chongqing, People's Republic of China.,Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Liang Tian
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jiao Luo
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jialin Yu
- Department of Neonatal Diagnosis and Treatment Center, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Pediatrics, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
33
|
Korpela K. Impact of Delivery Mode on Infant Gut Microbiota. ANNALS OF NUTRITION & METABOLISM 2021; 77:1-9. [PMID: 34515049 DOI: 10.1159/000518498] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/08/2021] [Indexed: 11/19/2022]
Abstract
Microbial colonization of the neonate is an important feature of normal birth. The gut microbiota has a central role in the programming of the host's metabolism and immune function, with both immediate and long-term health consequences. During vaginal birth, the infant is exposed to diverse maternal microbes, of which specific faecal microbes colonize the infant's gut. C-section eliminates the infant's contact with maternal microbes, preventing vertical transmission of gut microbes. Consequently, infants are colonized by bacteria from the environment, including potential pathogens from the hospital environment. Recent studies have shown that intrapartum antibiotic exposure has a C-section-like effect on the infant gut microbiota. While the composition of the gut microbiota largely normalizes during the first year of life, epidemiological studies suggest that the aberrant early microbial exposures have long-term immunological and metabolic consequences. Because of the high prevalence of procedures that prevent normal gut microbiota development, effective methods to normalize the gut microbiota of neonates are urgently needed. Even more importantly, attention should be paid to the microbiota imbalance in C-section-born and antibiotic-exposed infants in clinical practice. Breastfeeding and probiotics are particularly important for infants with disrupted gut colonization.
Collapse
Affiliation(s)
- Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Brame JE, Liddicoat C, Abbott CA, Breed MF. The potential of outdoor environments to supply beneficial butyrate-producing bacteria to humans. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 777:146063. [PMID: 33684759 DOI: 10.1016/j.scitotenv.2021.146063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/19/2021] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
Butyrate is an important mediator of human health and disease. The mechanisms of action of butyrate are becoming increasingly well-known. Many commensal bacteria that inhabit the human gut can synthesise butyrate, which is then absorbed into the human host. Simultaneously, several immune- and inflammatory-mediated diseases are being linked to insufficient exposure to beneficial microbes from our environment, including butyrate-producing bacteria. However, the role of outdoor environmental exposure to butyrate-producing bacteria remains poorly understood. Here we review the literature on the human exposure pathways to butyrate-producing bacteria, with a particular focus on outdoor environmental sources (e.g. associated with plants, plant-based residues, and soil), and the health implications of exposure to them. Emerging evidence suggests that environmental butyrate-producers may help supplement the human gut microbiota and represent an important component of the Biodiversity and Old Friends hypotheses. Improving our understanding of potential sources, precursors, and exposure pathways of environmental butyrate-producers that influence the gut microbiota and butyrate production offers promise to advance multiple disciplines of health and environmental science. We outline research priorities to address knowledge gaps in the outdoor environment-butyrate-health nexus and build knowledge of the potential pathways to help optimise exposure to human-beneficial butyrate-producing bacteria from the outdoor environment during childhood and adulthood.
Collapse
Affiliation(s)
- Joel E Brame
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia.
| | - Craig Liddicoat
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia; School of Public Health, The University of Adelaide, SA 5005, Australia
| | - Catherine A Abbott
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Martin F Breed
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
35
|
Chang HY, Chiang Chiau JS, Ho YH, Chang JH, Tsai KN, Liu CY, Hsu CH, Lin CY, Ko MHJ, Lee HC. Impact of Early Empiric Antibiotic Regimens on the Gut Microbiota in Very Low Birth Weight Preterm Infants: An Observational Study. Front Pediatr 2021; 9:651713. [PMID: 34136438 PMCID: PMC8200535 DOI: 10.3389/fped.2021.651713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022] Open
Abstract
Frequent use of antibiotics in preterm infants disturbs their gut microbial balance. In this preliminary observational study, we investigated the effect of different antibiotic regimens, administered during the first week of life, on microbial composition and diversity in very low birth weight (VLBW) preterm infants. We performed fecal sampling of breastfed VLBW infants on days 7, 14, and 30. After excluding stool samples from infants who received probiotics or who were administered antibiotics beyond the age of 7 days, we compared gut microbiota profiles between infants receiving a combination of ampicillin and gentamicin for 3 days (AG group, n = 10) and those receiving a combination of ampicillin and cefotaxime for 7 days (AC group, n = 14) using 16S ribosomal DNA community profiling. We also assessed the changes over time in each group. Compared to the AG group, Enterococcus species were significantly more abundant in the AC group (P = 0.002), especially in 7-day samples (12.3 vs. 0.6%, respectively, P = 0.032). No difference was observed at phylum and genus level over time within each group. Species richness in the AC group decreased significantly in the 14-day (P = 0.038) and 30-day (P = 0.03) samples compared to that in the 7-day sample. The same was observed for microbial evenness; in contrast, no significant difference in Shannon index and beta-diversity was detected between the two groups. Controlling for relevant confounding variables did not change the results. In conclusion, different antibiotic regimens affect the early development of gut microbiota in VLBW preterm infants. Prolonged use of ampicillin and cefotaxime might result in overabundance of Enterococcus. However, given that no significant differences were observed in 1-month samples, bacterial genera appear to continue colonizing the gastrointestinal tract despite previous exposure to antibiotics. The clinical relevance of these findings should be elucidated by further studies.
Collapse
Affiliation(s)
- Hung-Yang Chang
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | | | - Yu-Hsuan Ho
- Life Science, Delta Research Center, Delta Electronics Incorporation, Taipei, Taiwan
| | - Jui-Hsing Chang
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Kun-Nan Tsai
- Life Science, Delta Research Center, Delta Electronics Incorporation, Taipei, Taiwan
| | - Chia-Yen Liu
- Life Science, Delta Research Center, Delta Electronics Incorporation, Taipei, Taiwan
| | - Chyong-Hsin Hsu
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
| | - Chia-Ying Lin
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
| | - Mary Hsin-Ju Ko
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
| | - Hung-Chang Lee
- Department of Pediatrics, MacKay Children's Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| |
Collapse
|
36
|
Cortés-Macías E, Selma-Royo M, Martínez-Costa C, Collado MC. Breastfeeding Practices Influence the Breast Milk Microbiota Depending on Pre-Gestational Maternal BMI and Weight Gain over Pregnancy. Nutrients 2021; 13:1518. [PMID: 33946343 PMCID: PMC8146841 DOI: 10.3390/nu13051518] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 01/04/2023] Open
Abstract
Breastfeeding is critical for adequate neonatal microbial and immune system development affecting neonate health outcomes in the short and long term. There is a great interest in ascertaining which are the maternal factors contributing to the milk microbiota and the potential relevance for the developing infant. Thus, our study aimed to characterize the effect of mixed and exclusive breastfeeding practices on the milk microbiota and to determine the impact of pre-pregnancy body mass index (BMI) and weight gain over pregnancy on its composition. Breast milk samples from 136 healthy women were collected within the first month post-partum and milk microbiota profiling was analyzed by 16S rRNA gene sequencing. Information on breastfeeding habits and maternal-infant clinical data were recorded. Breastfeeding practices (exclusive vs. mixed), maternal pre-gestational BMI, and weight gain over pregnancy contributed to the milk microbiota variation. Pre-gestational normal-weight women with exclusive breastfeeding habits harbored a significantly higher abundance of Bifidobacterium genus, and also, higher alpha-diversity compared to the rest of the women. Our results confirm the importance of controlling weight during pregnancy and breastfeeding practices in terms of milk microbiota. Further studies to clarify the potential impact of these maternal factors on milk and infant development and health will be necessary.
Collapse
Affiliation(s)
- Erika Cortés-Macías
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain; (E.C.-M.); (M.S.-R.)
| | - Marta Selma-Royo
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain; (E.C.-M.); (M.S.-R.)
| | - Cecilia Martínez-Costa
- Department of Pediatrics, INCLIVA Research Institute, School of Medicine, University of Valencia, 46003 Valencia, Spain;
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, INCLIVA, 46010 Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain; (E.C.-M.); (M.S.-R.)
| |
Collapse
|
37
|
Han P, Gu JQ, Li LS, Wang XY, Wang HT, Wang Y, Chang C, Sun JL. The Association Between Intestinal Bacteria and Allergic Diseases-Cause or Consequence? Front Cell Infect Microbiol 2021; 11:650893. [PMID: 33937097 PMCID: PMC8083053 DOI: 10.3389/fcimb.2021.650893] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
The incidence of allergic disorders has been increasing over the past few decades, especially in industrialized countries. Allergies can affect people of any age. The pathogenesis of allergic diseases is complex and involves genetic, epigenetic, and environmental factors, and the response to medication is very variable. For some patients, avoidance is the sole effective therapy, and only when the triggers are identifiable. In recent years, the intestinal microbiota has emerged as a significant contributor to the development of allergic diseases. However, the precise mechanisms related to the effects of the microbiome on the pathogenesis of allergic diseases are unknown. This review summarizes the recent association between allergic disorders and intestinal bacterial dysbiosis, describes the function of gut microbes in allergic disease development from both preclinical and clinical studies, discusses the factors that influence gut microbial diversity and advanced techniques used in microbial analysis. Ultimately, more studies are required to define the host-microbial relationship relevant to allergic disorders and amenable to new therapeutic interventions.
Collapse
Affiliation(s)
- Pei Han
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jian-Qing Gu
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Sha Li
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue-Yan Wang
- Department of Allergy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hong-Tian Wang
- Department of Allergy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
- Division of Pediatric Immunology and Allergy, Joe DiMaggio Children’s Hospital, Hollywood, FL, United States
| | - Jin-Lyu Sun
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
38
|
Coker MO, Laue HE, Hoen AG, Hilliard M, Dade E, Li Z, Palys T, Morrison HG, Baker E, Karagas MR, Madan JC. Infant Feeding Alters the Longitudinal Impact of Birth Mode on the Development of the Gut Microbiota in the First Year of Life. Front Microbiol 2021; 12:642197. [PMID: 33897650 PMCID: PMC8059768 DOI: 10.3389/fmicb.2021.642197] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Cesarean-delivered (CD) infants harbor a distinct gut microbiome from vaginally delivered (VD) infants, however, during infancy, the most important driver of infant gut microbial colonization is infant feeding. Earlier studies have shown that breastfeeding is associated with higher levels of health-promoting bacteria such and Bifidobacterium and Bacteroides via modulation of the immune system, and production of metabolites. As the infant gut matures and solid foods are introduced, it is unclear whether longer duration of breast feeding restore loss of beneficial taxa within the intestinal microbiota of operatively delivered infants. Within the New Hampshire Birth Cohort Study, we evaluated the longitudinal effect of delivery mode and infant feeding on the taxonomic composition and functional capacity of developing gut microbiota in the First year of life. Microbiota of 500 stool samples collected between 6 weeks and 12 months of age (from 229 infants) were characterized by 16S ribosomal RNA sequencing. Shotgun metagenomic sequencing was also performed on 350 samples collected at either 6 weeks or 12 months of age. Among infant participants, 28% were cesarean-delivered (CD) infants and most (95%) initiated breastfeeding within the first six months of life, with 26% exclusively breastfed and 69% mixed-fed (breast milk and formula), in addition to complementary foods by age 1. Alpha (within-sample) diversity was significantly lower in CD infants compared to vaginally delivered (VD) infants (P < 0.05) throughout the study period. Bacterial community composition clustering by both delivery mode and feeding duration at 1 year of age revealed that CD infants who were breast fed for < 6 months were more dissimilar to VD infants than CD infants who breast fed for ≥ 6 months. We observed that breastfeeding modified the longitudinal impact of delivery mode on the taxonomic composition of the microbiota by 1 year of age, with an observed increase in abundance of Bacteroides fragilis and Lactobacillus with longer duration of breastfeeding among CD infants while there was an increase in Faecalibacterium for VD infants. Our findings confirm that duration of breastfeeding plays a critical role in restoring a health-promoting microbiome, call for further investigations regarding the association between breast milk exposure and health outcomes in early life.
Collapse
Affiliation(s)
- Modupe O. Coker
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- School of Dental Medicine, School of Public Health at Rutgers, Newark, NJ, United States
| | - Hannah E. Laue
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Anne G. Hoen
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Center for Molecular Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, United States
| | - Margaret Hilliard
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Erika Dade
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Zhigang Li
- Department of Biostatistics, University of Florida, Gainsville, FL, United States
| | - Thomas Palys
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, United States
| | - Hilary G. Morrison
- Marine Biological Laboratory, Josephine Bay Paul Center, Woods Hole, MA, United States
| | - Emily Baker
- Department of Pediatrics, Children’s Hospital at Dartmouth, Lebanon, NH, United States
| | - Margaret R. Karagas
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Center for Molecular Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, United States
| | - Juliette C. Madan
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, United States
- Department of Pediatrics, Children’s Hospital at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
39
|
Ngowi EE, Wang YZ, Khattak S, Khan NH, Mahmoud SSM, Helmy YASH, Jiang QY, Li T, Duan SF, Ji XY, Wu DD. Impact of the factors shaping gut microbiota on obesity. J Appl Microbiol 2021; 131:2131-2147. [PMID: 33570819 DOI: 10.1111/jam.15036] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022]
Abstract
Obesity is considered as a risk factor for chronic health diseases such as heart diseases, cancer and diabetes 2. Reduced physical activities, lifestyle, poor nutritional diet and genetics are among the risk factors associated with the development of obesity. In recent years, several studies have explored the link between the gut microbiome and the progression of diseases including obesity, with the shift in microbiome abundance and composition being the main focus. The alteration of gut microbiome composition affects both nutrients metabolism and specific gene expressions, thereby disturbing body physiology. Specifically, the abundance of fibre-metabolizing microbes is associated with weight loss and that of protein and fat-metabolizing bacteria with weight gain. Various internal and external factors such as genetics, maternal obesity, mode of delivery, breastfeeding, nutrition, antibiotic use and the chemical compounds present in the environment are known to interfere with the richness of the gut microbiota (GM), thus influencing weight gain/loss and ultimately the development of obesity. However, the effectiveness of each factor in potentiating the shift in microbes' abundance to result in significant changes that can lead to obesity is not yet clear. In this review, we will highlight the factors involved in shaping GM, their influence on obesity and possible interventions. Understanding the influence of these factors on the diversity of the GM and how to improve their effectiveness on disease conditions could be keys in the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China.,Department of Biological Sciences, Faculty of Science, Dares Salaam University College of Education, Dares Salaam, Tanzania
| | - Yi-Zhen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Salma Sayed Mohamed Mahmoud
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Yasmeen Ahmed Saleheldin Hassan Helmy
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Tao Li
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan, China
| | - Shao-Feng Duan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China.,School of Stomatology, Henan University, Kaifeng, Henan, China
| |
Collapse
|
40
|
Zhou J, Wang Y, Lei Q. Using Bioinformatics to Quantify the Variability and Diversity of the Microbial Community Structure in Pond Ecosystems of a Subtropical Catchment. Curr Bioinform 2021. [DOI: 10.2174/1574893615999200422120819] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
In rural China, many natural water bodies and farmlands have been
converted into fish farming ponds as an economic developmental strategy. There is still a limited
understanding of how the diversity and structure of microbial communities change in nature and
become managed fish pond ecosystems.
Objective:
We aimed to identify the changes of the diversity and structure of microbial community
and driving mechanism in pond ecosystems.
Methods:
The datasets of 16S rRNA amplicon sequencing and the concentrations of N and P
fractions were achieved in water samples of pond ecosystems. Bioinformatics analysis was used to
analyze the diversity and structure of the microbial communities.
Results:
Our results indicated that the diversity and structure of the microbial communities in the
natural ponds were significantly different from ones in managed fish ponds. The nutrients of N and
P and water environmental factors were responsible for 46.3% and 19.5% of the changes in the
structure and diversity of the microbial community, respectively.
Conclusion:
The N and P fractions and water environmental factors influenced the microbial
community structure and diversity in pond ecosystems. Fish farming indirectly affected the
microbial community by altering the contents of N and P fractions in water bodies of ponds, when
a natural pond was converted into a managed fish pond.
Collapse
Affiliation(s)
- Jiaogen Zhou
- School of Urban and Environmental Science, Huaiyin Normal University, Jiangsu 223300, China
| | - Yang Wang
- School of Urban and Environmental Science, Huaiyin Normal University, Jiangsu 223300, China
| | - Qiuliang Lei
- Key Laboratory of Non-point Source Pollution Control, Ministry of Agriculture, Institute of Agricultural Resources and Regional Planning, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, China
| |
Collapse
|
41
|
Tonon KM, Morais TB, Taddei CR, Araújo-Filho HB, Abrão ACFV, Miranda A, de Morais MB. Gut microbiota comparison of vaginally and cesarean born infants exclusively breastfed by mothers secreting α1-2 fucosylated oligosaccharides in breast milk. PLoS One 2021; 16:e0246839. [PMID: 33556125 PMCID: PMC7870049 DOI: 10.1371/journal.pone.0246839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/26/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Exclusive breastfeeding promotes beneficial modifications on the microbiota of cesarean born infants, but little is known about the role of specific breast milk components in this modulation. Women with an active FUT2 gene (called secretors) secrete α1-2 fucosylated human milk oligosaccharides (HMOs), which promote Bifidobacterium in the infant's gut and may modulate the microbiota of cesarean born infants. OBJECTIVE To compare the microbiota composition of cesarean and vaginally born infants breastfed by secretor mothers. METHODS Maternal secretor status was determined by the occurrence of 4 different α1-2 fucosylated HMOs in breast milk by LC-MS. The fecal microbiota composition from cesarean and vaginally born infants was analyzed by 16S rRNA gene sequencing and qPCR, stratified by the maternal secretor status, and compared. RESULTS Alpha and beta diversity were not significantly different in cesarean born, secretor-fed infants (CSe+) compared to vaginally born, secretor-fed infants (VSe+). There were no significant differences in the fecal relative abundance of Bifidobacterium between CSe+ and VSe+ infants, but the prevalence of the species B. longum was lower in CSe+. The fecal relative abundance of Bacteroides was also lower, while Akkermansia and Kluyvera were higher in CSe+ infants. CONCLUSION Cesarean and vaginally born infants fed with breast milk containing the α1-2 fucosylated HMOs fraction present similar amounts of Bifidobacterium in the feces, but differences are observed in other members of the microbiota.
Collapse
Affiliation(s)
- Karina M. Tonon
- Nutrition Postgraduate Program, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Tania B. Morais
- Food Quality Control Laboratory, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carla R. Taddei
- Department of Clinical and Toxicological Analysis, Universidade de São Paulo, São Paulo, Brazil
- School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, Brazil
| | | | - Ana Cristina F. V. Abrão
- Breastfeeding Incentive and Support Center, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Antonio Miranda
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mauro B. de Morais
- Division of Pediatric Gastroenterology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Carrizales-Sánchez AK, García-Cayuela T, Hernández-Brenes C, Senés-Guerrero C. Gut microbiota associations with metabolic syndrome and relevance of its study in pediatric subjects. Gut Microbes 2021; 13:1960135. [PMID: 34491882 PMCID: PMC8425709 DOI: 10.1080/19490976.2021.1960135] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 02/04/2023] Open
Abstract
Childhood obesity and T2DM have shown a recent alarming increase due to important changes in global lifestyle and dietary habits, highlighting the need for urgent and novel solutions to improve global public health. Gut microbiota has been shown to be relevant in human health and its dysbiosis has been associated with MetS, a health condition linked to the onset of relevant diseases including T2DM. Even though there have been recent improvements in the understanding of gut microbiota-host interactions, pediatric gut microbiota has been poorly studied compared to adults. This review provides an overview of MetS and its relevance in school-age children, discusses gut microbiota and its possible association with this metabolic condition including relevant emerging gut microbiome-based interventions for its prevention and treatment, and outlines future challenges and perspectives in preventing microbiota dysbiosis from the early stages of life.
Collapse
Affiliation(s)
| | - Tomás García-Cayuela
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Zapopan, Jalisco, Mexico
| | | | | |
Collapse
|
43
|
Hoang DM, Levy EI, Vandenplas Y. The impact of Caesarean section on the infant gut microbiome. Acta Paediatr 2021; 110:60-67. [PMID: 33405258 DOI: 10.1111/apa.15501] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
AIM Description of the impact of caesarean section on the infant gut microbiome, infant health and solutions to restore the dysbiosis. METHODS We searched PubMed and Google Scholar for relevant articles. Only articles published in English were selected. Separate searches were performed for each topic. We selected 60 articles published between 1999 and 2019 by extracting subject headings and key words of interest for this topic. RESULTS Caesarean section is an obstetrical procedure that is increasing in prevalence worldwide. On top of the maternal and neonatal risks that this procedure carries, it also induces a dysbiosis of the infant intestinal microbiome possibly challenging the health outcome for the infant. Antibiotics administered during Caesarean deliveries contribute to the development of the gut microbiome. Nonetheless, breastfeeding and several interventions such as vaginal seeding and supplementation with probiotics, prebiotics and synbiotics may contribute to the restoration of the dysbiosis. CONCLUSION Caesarean section is associated with the development of an imbalance of the infant gut microbiome. Long-term consequences of this imbalance are debated. Besides breastfeeding, other strategies to restore this dysbiosis need further studies before they can be recommended.
Collapse
Affiliation(s)
- Delphine M. Hoang
- KidZ Health Castle Universitair Ziekenhuis Brussel Vrije Universiteit Brussel Brussels Belgium
| | - Elvira I. Levy
- KidZ Health Castle Universitair Ziekenhuis Brussel Vrije Universiteit Brussel Brussels Belgium
| | - Yvan Vandenplas
- KidZ Health Castle Universitair Ziekenhuis Brussel Vrije Universiteit Brussel Brussels Belgium
| |
Collapse
|
44
|
Abstract
Aging is characterized by the functional decline of tissues and organs and increased risk of aging-associated disorders, which pose major societal challenges and are a public health priority. Despite extensive human genetics studies, limited progress has been made linking genetics with aging. There is a growing realization that the altered assembly, structure and dynamics of the gut microbiota actively participate in the aging process. Age-related microbial dysbiosis is involved in reshaping immune responses during aging, which manifest as immunosenescence (insufficiency) and inflammaging (over-reaction) that accompany many age-associated enteric and extraenteric diseases. The gut microbiota can be regulated, suggesting a potential target for aging interventions. This review summarizes recent findings on the physiological succession of gut microbiota across the life-cycle, the roles and mechanisms of gut microbiota in healthy aging, alterations of gut microbiota and aging-associated diseases, and the gut microbiota-targeted anti-aging strategies.
Collapse
Affiliation(s)
- Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xia Liu
- Department of Intensive Care Unit, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiumei Yan
- Department of Geriatrics, Lishui Second People's Hospital, Lishui, Zhejiang, China
| | - Shaochang Wu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
45
|
Zachariassen LF, Sørensen DB, Krych L, Hansen AK, Hansen CHF. Effects of delivery mode on behavior in mouse offspring. Physiol Behav 2020; 230:113285. [PMID: 33309952 DOI: 10.1016/j.physbeh.2020.113285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022]
Abstract
Cesarean section (CS) has been associated with an increased risk of mental disorders in the offspring. This could possibly be explained by an inadequate microbial colonization early in life with a consequential disturbed gut-brain interaction. To investigate the link between delivery mode and behavior and develop a suitable animal model for further research of the gut-brain axis, the aim of this study was to characterize the gut microbiota (GM) together with the behavioral response in various behavioral tests in CS-delivered mice. We hypothesized that mice delivered by CS would present with disturbances in normal physiological behavior possibly due to an inadequate microbial colonization. C57BL/6 mice delivered by CS or vaginal delivery (VD) were cross fostered and, as adults, observed for anxiety-related behavior in the open field test, social deficits in a sociability test and compulsive behavior in the marble burying test. GM was analyzed by 16S rRNA gene amplicon sequencing. The open field test showed that CS-delivered mice had a decreased activity and accelerated defecation compared to VD-delivered mice. In addition, CS-delivered female mice spend less time interacting with cage mates in the sociability test, whereas there was no effect of CS delivery on the average number of marbles buried. In conclusion, CS-delivered mice had a more pronounced anxiety-like behavior and showed less preference for sociability in female offspring.
Collapse
Affiliation(s)
- Line Fisker Zachariassen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Dorte Bratbo Sørensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
46
|
Kim G, Bae J, Kim MJ, Kwon H, Park G, Kim SJ, Choe YH, Kim J, Park SH, Choe BH, Shin H, Kang B. Delayed Establishment of Gut Microbiota in Infants Delivered by Cesarean Section. Front Microbiol 2020; 11:2099. [PMID: 33013766 PMCID: PMC7516058 DOI: 10.3389/fmicb.2020.02099] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022] Open
Abstract
The maternal vaginal microbiome is an important source for infant gut microbiome development. However, infants delivered by Cesarean section (CS) do not contact the maternal vaginal microbiome and this delivery method may perturb the early establishment and development of the gut microbiome. The aim of this study was to investigate the early gut microbiota of Korean newborns receiving the same postpartum care services for two weeks after birth by delivery mode using fecal samples collected at days 3, 7, and 14. Early gut microbiota development patterns were examined using 16S rRNA gene-based sequencing from 132 infants either born vaginally (VD, n = 64) or via Cesarean section (CS, n = 68). VD-born neonates showed increased alpha diversity in infant fecal samples collated at days 7 and 14 compared to those from day 3, while those of CS infants did not differ (p < 0.015). Bacterial structures of infants from both groups separated at day 7 (p < 0.001) and day 14 (p < 0.01). The bacterial structure of VD infants gradually changed over time (day 3 vs. day 7, p < 0.012; day 3 vs. day 14, p < 0.001). Day 14 samples of CS infants differed from day 3 and 7 samples (day 3 vs. day 14, p < 0.001). VD infant relative abundance of Bifidobacterium (days 7, 14), Bacteroides (days 7, 14), and Lachnospiraceae (day 7) significantly increased compared to CS infants, with a lower abundance of Enterobacteriaceae (found in all periods of the CS group) (LDA > 3.0). Relative abundances of Bifidobacterium, Lactobacillus, and Staphylococcus were significantly increased in both VD and CS groups at day 14 (LDA > 3.0). Predicted functional analysis showed that VD infants had overrepresented starch/sucrose, amino acid and nucleotide metabolism in gut microbiota with depleted lipopolysaccharide biosynthesis until day 14 compared to CS infants. This study confirmed that delivery mode is the major determinant of neonatal intestinal microbiome establishment and provides a profile of microbiota perturbations in CS infants. Our findings provide preliminary insight for establishing recovery methods to supply the specific microbes missing in CS infants.
Collapse
Affiliation(s)
- Gyungcheon Kim
- R&D Institute, BioEleven Co., Ltd., Seoul, South Korea.,Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul, South Korea
| | - Jaewoong Bae
- R&D Institute, BioEleven Co., Ltd., Seoul, South Korea
| | - Mi Jin Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyeji Kwon
- R&D Institute, BioEleven Co., Ltd., Seoul, South Korea
| | - Gwoncheol Park
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul, South Korea
| | - Seok-Jin Kim
- R&D Institute, BioEleven Co., Ltd., Seoul, South Korea
| | - Yon Ho Choe
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jisook Kim
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Sook-Hyun Park
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Byung-Ho Choe
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Hakdong Shin
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul, South Korea
| | - Ben Kang
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
47
|
The Infant Gut Microbiota and Risk of Asthma: The Effect of Maternal Nutrition during Pregnancy and Lactation. Microorganisms 2020; 8:microorganisms8081119. [PMID: 32722458 PMCID: PMC7466123 DOI: 10.3390/microorganisms8081119] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Research has amply demonstrated that early life dysbiosis of the gut microbiota influences the propensity to develop asthma. The influence of maternal nutrition on infant gut microbiota is therefore of growing interest. However, a handful of prospective studies have examined the role of maternal dietary patterns during pregnancy in influencing the infant gut microbiota but did not assess whether this resulted in an increased risk of asthma later in life. The mechanisms involved in the process are also, thus far, poorly documented. There have also been few studies examining the effect of maternal dietary nutrient intake during lactation on the milk microbiota, the effect on the infant gut microbiota and, furthermore, the consequences for asthma development remain largely unknown. Therefore, the specific aim of this mini review is summarizing the current knowledge regarding the effect of maternal nutrition during pregnancy and lactation on the infant gut microbiota composition, and whether it has implications for asthma development.
Collapse
|
48
|
Abstract
The neonatal developmental window represents a key time for establishment of the gut microbiota. First contact with these microbes within the infant gastrointestinal tract signifies the start of a critical mutualistic relationship, which is central for short- and longer-term health. Recent research has provided insights into the origin of these microbial pioneers, how they are maintained within the gut environment, and how factors such as antibiotics or preterm birth may disrupt the succession of beneficial microbes. The acquisition, colonisation, and maintenance of the early life microbiota, and subsequent interactions with the host is a rapidly developing research area. In this review we explore some of these key topics which have been illuminated by recent research, and we highlight some of the important unresolved questions which currently limit our overall understanding of the neonatal gut microbiome.
Collapse
Affiliation(s)
- Matthew J. Dalby
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Lindsay J. Hall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Intestinal Microbiome, School of Life Sciences, Technical University of Munich, Freising, Germany
- ZIEL – Institute for Food & Health, Technical University of Munich, Freising, Germany
| |
Collapse
|
49
|
Siranosian BA, Tamburini FB, Sherlock G, Bhatt AS. Acquisition, transmission and strain diversity of human gut-colonizing crAss-like phages. Nat Commun 2020; 11:280. [PMID: 31941900 PMCID: PMC6962324 DOI: 10.1038/s41467-019-14103-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/04/2019] [Indexed: 12/28/2022] Open
Abstract
CrAss-like phages are double-stranded DNA viruses that are prevalent in human gut microbiomes. Here, we analyze gut metagenomic data from mother-infant pairs and patients undergoing fecal microbiota transplantation to evaluate the patterns of acquisition, transmission and strain diversity of crAss-like phages. We find that crAss-like phages are rarely detected at birth but are increasingly prevalent in the infant microbiome after one month of life. We observe nearly identical genomes in 50% of cases where the same crAss-like clade is detected in both the mother and the infant, suggesting vertical transmission. In cases of putative transmission of prototypical crAssphage (p-crAssphage), we find that a subset of strains present in the mother are detected in the infant, and that strain diversity in infants increases with time. Putative tail fiber proteins are enriched for nonsynonymous strain variation compared to other genes, suggesting a potential evolutionary benefit to maintaining strain diversity in specific genes. Finally, we show that p-crAssphage can be acquired through fecal microbiota transplantation.
Collapse
Affiliation(s)
| | | | - Gavin Sherlock
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ami S Bhatt
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
50
|
Van Daele E, Knol J, Belzer C. Microbial transmission from mother to child: improving infant intestinal microbiota development by identifying the obstacles. Crit Rev Microbiol 2019; 45:613-648. [DOI: 10.1080/1040841x.2019.1680601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Emmy Van Daele
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Gut Biology and Microbiology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|