Your browser doesn't support javascript.
loading
Host-microbial co-metabolites modulated by human milk oligosaccharides relate to reduced risk of respiratory tract infections.
Martin, François-Pierre; Tytgat, Hanne L P; Krogh Pedersen, Helle; Moine, Deborah; Eklund, Aron C; Berger, Bernard; Sprenger, Norbert.
Afiliación
  • Martin FP; Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland.
  • Tytgat HLP; Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland.
  • Krogh Pedersen H; Clinical Microbiomics, Copenhagen, Denmark.
  • Moine D; Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland.
  • Eklund AC; Clinical Microbiomics, Copenhagen, Denmark.
  • Berger B; Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland.
  • Sprenger N; Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland.
Front Nutr ; 9: 935711, 2022.
Article en En | MEDLINE | ID: mdl-35990340
ABSTRACT
Human milk oligosaccharides (HMOs) are structurally diverse oligosaccharides present in breast milk, supporting the development of the gut microbiota and immune system. Previously, 2-HMO (2'fucosyllactose, lacto-N-neotetraose) compared to control formula feeding was associated with reduced risk of lower respiratory tract infections (LRTIs), in part linked to lower acetate and higher bifidobacteria proportions. Here, our objective was to gain further insight into additional molecular pathways linking the 2-HMO formula feeding and LRTI mitigation. From the same trial, we measured the microbiota composition and 743 known biochemical species in infant stool at 3 months of age using shotgun metagenomic sequencing and untargeted mass spectrometry metabolomics. We used multivariate analysis to identify biochemicals associated to 2-HMO formula feeding and LRTI and integrated those findings with the microbiota compositional data. Three molecular pathways stood out increased gamma-glutamylation and N-acetylation of amino acids and decreased inflammatory signaling lipids. Integration of stool metagenomic data revealed some Bifidobacterium and Bacteroides species to be implicated. These findings deepen our understanding of the infant gut/microbiome co-metabolism in early life and provide evidence for how such metabolic changes may influence immune competence at distant mucosal sites such as the airways.
Palabras clave

Texto completo: 1 Base de datos: MEDLINE Tipo de estudio: Etiology_studies / Prognostic_studies / Risk_factors_studies Idioma: En Revista: Front Nutr Año: 2022 Tipo del documento: Article

Texto completo: 1 Base de datos: MEDLINE Tipo de estudio: Etiology_studies / Prognostic_studies / Risk_factors_studies Idioma: En Revista: Front Nutr Año: 2022 Tipo del documento: Article