4.8 Article

Lactobacillus reuteri tryptophan metabolism promotes host susceptibility to CNS autoimmunity

Journal

MICROBIOME
Volume 10, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s40168-022-01408-7

Keywords

L. reuteri; Microbiome; Microbiota; Tryptophan; Multiple sclerosis; Experimental autoimmune encephalomyelitis (EAE); Aryl hydrocarbon receptor (AhR)

Categories

Funding

  1. NIH/NINDS [F31NS120381-01A1, R01 NS097596]
  2. Vermont Center for Immunology and Infectious Diseases grant [P30GM118228-05S3]
  3. [T32AI055402-16A1]
  4. [S10OD026843-01]
  5. [P01CA098993-15]

Ask authors/readers for more resources

The study found that gut commensal, such as the potential probiotic species Lactobacillus reuteri, can unexpectedly enhance autoimmunity through tryptophan metabolism, leading to broad shifts in the metabolome and immunological repertoire.
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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available