Affiliations 

  • 1 Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
  • 2 The Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore 637551
  • 3 Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth WA 6150, Australia
  • 4 Department of Chemical Engineering, Pennsylvania State University, University Park, PA 16802
  • 5 Department of Neurobiology, Care and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
  • 6 The School of Biological Sciences, Nanyang Technological University, Singapore 637551
  • 7 Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
  • 8 Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
  • 9 National Neuroscience Institute, Singapore 169857
  • 10 Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921; sven.pettersson@ki.se
Proc Natl Acad Sci U S A, 2021 Jul 06;118(27).
PMID: 34210797 DOI: 10.1073/pnas.2021091118

Abstract

While modulatory effects of gut microbes on neurological phenotypes have been reported, the mechanisms remain largely unknown. Here, we demonstrate that indole, a tryptophan metabolite produced by tryptophanase-expressing gut microbes, elicits neurogenic effects in the adult mouse hippocampus. Neurogenesis is reduced in germ-free (GF) mice and in GF mice monocolonized with a single-gene tnaA knockout (KO) mutant Escherichia coli unable to produce indole. External administration of systemic indole increases adult neurogenesis in the dentate gyrus in these mouse models and in specific pathogen-free (SPF) control mice. Indole-treated mice display elevated synaptic markers postsynaptic density protein 95 and synaptophysin, suggesting synaptic maturation effects in vivo. By contrast, neurogenesis is not induced by indole in aryl hydrocarbon receptor KO (AhR-/-) mice or in ex vivo neurospheres derived from them. Neural progenitor cells exposed to indole exit the cell cycle, terminally differentiate, and mature into neurons that display longer and more branched neurites. These effects are not observed with kynurenine, another AhR ligand. The indole-AhR-mediated signaling pathway elevated the expression of β-catenin, Neurog2, and VEGF-α genes, thus identifying a molecular pathway connecting gut microbiota composition and their metabolic function to neurogenesis in the adult hippocampus. Our data have implications for the understanding of mechanisms of brain aging and for potential next-generation therapeutic opportunities.

* Title and MeSH Headings from MEDLINE®/PubMed®, a database of the U.S. National Library of Medicine.