3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Early development of infant gut microbiota in relation to breastfeeding and human milk oligosaccharides

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Infant gut microbiota composition is influenced by various factors early in life. Here, we investigate associations between infant gut microbiome development, infant age, breastfeeding duration, and human milk oligosaccharides (HMO) composition in breastmilk.

          Methods

          A total of 94 mother-infant pairs were recruited as part of the Cambridge Baby Growth and Breastfeeding Study (CBGS-BF) (Cambridge, UK). Infant stool samples ( n = 337) were collected at 2 week, 6 week, 3 month, and 6 month of age. The 16S rRNA V3-V4 rRNA region was sequenced using MiSeq Illumina to determine microbiota composition and diversity. Mother’s hindmilk samples were collected at birth, 2 week, 6 week, 3 month, and 6 month postpartum. Concentrations of five neutral [2′FL, 3′FL, lacto-N-fucopentaose 1 (LNFP1), LNnT, LNT] and two acidic (3′SL, and 6′SL) HMOs were measured in all milk samples using High-Performance Anion-Exchange Chromatography with Pulsed Amperometric Detection (HPAEC-PAD). We explored the associations between infant gut microbiome parameters and age, duration of exclusive breastfeeding (EBF), and levels of individual HMOs.

          Results

          Bifidobacterium was the most abundant genus in infant stool at all-time points, irrespective of breastfeeding duration, with an overall mean relative abundance of 70%. The relative abundance of B. bifidum in stool from infants who were breastfed for longer than 6 months was significantly higher compared to the infant breastfed up to 3 months ( p = 0.0285). Alpha-diversity (both Shannon and ASV-level Richness) of infant gut microbiota showed a biphasic change with infant age, decreasing from 2 weeks until 3 months and then increasing until 6 months of age. Bifidobacterium relative abundance was associated with higher concentrations of 2′FL and LNFP1 in breastmilk across all time-points ( p = 0.049 and 0.017, respectively), with trends toward a higher abundance of B. longum species. No significant association with Bifidobacterium was found for breastmilk LNnT, 3′SL, and 6′SL levels.

          Conclusion

          Our study is in line with previous data demonstrating that EBF duration in the first months of life impacts infant gut microbiota composition. The observed links between specific HMOs in breastmilk and bacteria in infant stool provide evidence of how mother’s milk affects infant microbiome development.

          Related collections

          Most cited references36

          • Record: found
          • Abstract: not found
          • Article: not found

          Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            ape 5.0: an environment for modern phylogenetics and evolutionary analyses in R

            After more than fifteen years of existence, the R package ape has continuously grown its contents, and has been used by a growing community of users. The release of version 5.0 has marked a leap towards a modern software for evolutionary analyses. Efforts have been put to improve efficiency, flexibility, support for 'big data' (R's long vectors), ease of use and quality check before a new release. These changes will hopefully make ape a useful software for the study of biodiversity and evolution in a context of increasing data quantity.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Human milk oligosaccharides: every baby needs a sugar mama.

              L Bode (2012)
              Human milk oligosaccharides (HMOs) are a family of structurally diverse unconjugated glycans that are highly abundant in and unique to human milk. Originally, HMOs were discovered as a prebiotic "bifidus factor" that serves as a metabolic substrate for desired bacteria and shapes an intestinal microbiota composition with health benefits for the breast-fed neonate. Today, HMOs are known to be more than just "food for bugs". An accumulating body of evidence suggests that HMOs are antiadhesive antimicrobials that serve as soluble decoy receptors, prevent pathogen attachment to infant mucosal surfaces and lower the risk for viral, bacterial and protozoan parasite infections. In addition, HMOs may modulate epithelial and immune cell responses, reduce excessive mucosal leukocyte infiltration and activation, lower the risk for necrotizing enterocolitis and provide the infant with sialic acid as a potentially essential nutrient for brain development and cognition. Most data, however, stem from in vitro, ex vivo or animal studies and occasionally from association studies in mother-infant cohorts. Powered, randomized and controlled intervention studies will be needed to confirm relevance for human neonates. The first part of this review introduces the pioneers in HMO research, outlines HMO structural diversity and describes what is known about HMO biosynthesis in the mother's mammary gland and their metabolism in the breast-fed infant. The second part highlights the postulated beneficial effects of HMO for the breast-fed neonate, compares HMOs with oligosaccharides in the milk of other mammals and in infant formula and summarizes the current roadblocks and future opportunities for HMO research.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Nutr
                Front Nutr
                Front. Nutr.
                Frontiers in Nutrition
                Frontiers Media S.A.
                2296-861X
                09 March 2023
                2023
                : 10
                : 1003032
                Affiliations
                [1] 1Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute , Evansville, IN, United States
                [2] 2Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute , Nijmegen, Netherlands
                [3] 3NIZO Food Research BV , Ede, Netherlands
                [4] 4Department of Paediatrics, University of Cambridge , Cambridge, United Kingdom
                [5] 5MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Cambridge, United Kingdom
                [6] 6Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Cambridge, United Kingdom
                Author notes

                Edited by: Defu Ma, Peking University, China

                Reviewed by: Ana Griselda Binetti, CONICET Instituto de Lactología Industrial (INLAIN), Argentina; Kieuske Yoshida, Morinaga Milk Industry Co., Ltd., Japan; Takahiro Matsuki, Yakult Central Institute for Microbiological Research, Japan

                *Correspondence: Janna A. van Diepen, Janna.vanDiepen@ 123456reckitt.com

                Deceased

                This article was submitted to Nutrition and Metabolism, a section of the journal Frontiers in Nutrition

                Article
                10.3389/fnut.2023.1003032
                10034312
                36969811
                d3f36962-a3c6-4f25-8f8f-86c1349c5b99
                Copyright © 2023 Chichlowski, van Diepen, Prodan, Olga, Ong, Kortman, Dunger and Gross.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 25 July 2022
                : 09 January 2023
                Page count
                Figures: 7, Tables: 0, Equations: 0, References: 36, Pages: 10, Words: 6049
                Funding
                This research was supported by the National Institute for Health Research/Wellcome Trust Clinical Research Facility at Cambridge University Hospitals NHS Foundation Trust and the NIHR Cambridge Comprehensive Biomedical Research Centre. KO was supported by the Medical Research Council (Unit programme: MC_UU_00006/2).
                Categories
                Nutrition
                Original Research

                microbiome,human milk oligosaccharides (hmo),bifidobacteria,fucosyllactose,clinical

                Comments

                Comment on this article