[HTML][HTML] Lactobacillus reuteri tryptophan metabolism promotes host susceptibility to CNS autoimmunity

TL Montgomery, K Eckstrom, KH Lile, S Caldwell… - Microbiome, 2022 - Springer
TL Montgomery, K Eckstrom, KH Lile, S Caldwell, ER Heney, KG Lahue, A D'Alessandro
Microbiome, 2022Springer
Background Dysregulation of gut microbiota-associated tryptophan metabolism has been
observed in patients with multiple sclerosis. However, defining direct mechanistic links
between this apparent metabolic rewiring and individual constituents of the gut microbiota
remains challenging. We and others have previously shown that colonization with the gut
commensal and putative probiotic species, Lactobacillus reuteri, unexpectedly enhances
host susceptibility to experimental autoimmune encephalomyelitis (EAE), a murine model of …
Background
Dysregulation of gut microbiota-associated tryptophan metabolism has been observed in patients with multiple sclerosis. However, defining direct mechanistic links between this apparent metabolic rewiring and individual constituents of the gut microbiota remains challenging. We and others have previously shown that colonization with the gut commensal and putative probiotic species, Lactobacillus reuteri, unexpectedly enhances host susceptibility to experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis. To identify underlying mechanisms, we characterized the genome of commensal L. reuteri isolates, coupled with in vitro and in vivo metabolomic profiling, modulation of dietary substrates, and gut microbiota manipulation.
Results
The enzymes necessary to metabolize dietary tryptophan into immunomodulatory indole derivatives were enriched in the L. reuteri genomes, including araT, fldH, and amiE. Moreover, metabolite profiling of L. reuteri monocultures and serum of L. reuteri-colonized mice revealed a depletion of kynurenines and production of a wide array of known and novel tryptophan-derived aryl hydrocarbon receptor (AhR) agonists and antagonists, including indole acetate, indole-3-glyoxylic acid, tryptamine, p-cresol, and diverse imidazole derivatives. Functionally, dietary tryptophan was required for L. reuteri-dependent EAE exacerbation, while depletion of dietary tryptophan suppressed disease activity and inflammatory T cell responses in the CNS. Mechanistically, L. reuteri tryptophan-derived metabolites activated the AhR and enhanced T cell production of IL-17.
Conclusions
Our data suggests that tryptophan metabolism by gut commensals, such as the putative probiotic species L. reuteri, can unexpectedly enhance autoimmunity, inducing broad shifts in the metabolome and immunological repertoire.
Video Abstract
Springer