Precision modulation of dysbiotic adult microbiomes with a human-milk-derived synbiotic reshapes gut microbial composition and metabolites

Cell Host Microbe. 2023 Sep 13;31(9):1523-1538.e10. doi: 10.1016/j.chom.2023.08.004. Epub 2023 Aug 31.

Abstract

Manipulation of the gut microbiome using live biotherapeutic products shows promise for clinical applications but remains challenging to achieve. Here, we induced dysbiosis in 56 healthy volunteers using antibiotics to test a synbiotic comprising the infant gut microbe, Bifidobacterium longum subspecies infantis (B. infantis), and human milk oligosaccharides (HMOs). B. infantis engrafted in 76% of subjects in an HMO-dependent manner, reaching a relative abundance of up to 81%. Changes in microbiome composition and gut metabolites reflect altered recovery of engrafted subjects compared with controls. Engraftment associates with increases in lactate-consuming Veillonella, faster acetate recovery, and changes in indolelactate and p-cresol sulfate, metabolites that impact host inflammatory status. Furthermore, Veillonella co-cultured in vitro and in vivo with B. infantis and HMO converts lactate produced by B. infantis to propionate, an important mediator of host physiology. These results suggest that the synbiotic reproducibly and predictably modulates recovery of a dysbiotic microbiome.

Keywords: B. infantis; Bifidobacterium; HMO; LBP; Veillonella; gut engraftment; gut microbiome; gut microbiota; human milk oligosaccharides; live biotherapeutic product; microbiome modulation; propionate.

MeSH terms

  • Adult
  • Dysbiosis
  • Gastrointestinal Microbiome*
  • Humans
  • Infant
  • Lactic Acid
  • Microbiota*
  • Milk, Human
  • Synbiotics*
  • Veillonella

Substances

  • Lactic Acid