Beneficial bacteria activate type-I interferon production via the intracellular cytosolic sensors STING and MAVS

Gut Microbes. 2020 Jul 3;11(4):771-788. doi: 10.1080/19490976.2019.1707015. Epub 2020 Jan 15.

Abstract

Type-I interferon (IFN-I) cytokines are produced by immune cells in response to microbial infections, cancer and autoimmune diseases, and subsequently, trigger cytoprotective and antiviral responses through the activation of IFN-I stimulated genes (ISGs). The ability of intestinal microbiota to modulate innate immune responses is well known, but the mechanisms underlying such responses remain elusive. Here we report that the intracellular sensors stimulator of IFN genes (STING) and mitochondrial antiviral signaling (MAVS) are essential for the production of IFN-I in response to lactic acid bacteria (LAB), common gut commensal bacteria with beneficial properties. Using human macrophage cells we show that LAB strains that potently activate the inflammatory transcription factor NF-κB are poor inducers of IFN-I and conversely, those triggering significant amounts of IFN-I fail to activate NF-κB. This IFN-I response is also observed in human primary macrophages, which modulate CD64 and CD40 upon challenge with IFN-I-inducing LAB. Mechanistically, IFN-I inducers interact more intimately with phagocytes as compared to NF-κB-inducers, and fail to activate IFN-I in the presence of phagocytosis inhibitors. These bacteria are then sensed intracellularly by the cytoplasmic sensors STING and, to a lesser extent, MAVS. Accordingly, macrophages deficient for STING showed dramatically reduced phosphorylation of TANK-binding kinase (TBK)-1 and IFN-I activation, which resulted in lower expression of ISGs. Our findings demonstrate a major role for intracellular sensing and STING in the production of IFN-I by beneficial bacteria and the existence of bacteria-specific immune signatures, which can be exploited to promote cytoprotective responses and prevent overreactive NF-κB-dependent inflammation in the gut.

Keywords: Lactic acid bacteria; MAVS; STING; beneficial microbes; interferon.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adaptor Proteins, Signal Transducing / metabolism*
  • Humans
  • Immunity, Innate
  • Interferon Type I / biosynthesis*
  • Lactobacillales / immunology
  • Lactobacillales / physiology*
  • Lactobacillus plantarum / immunology
  • Lactobacillus plantarum / physiology
  • Leukocytes, Mononuclear / immunology
  • Leukocytes, Mononuclear / metabolism
  • Leukocytes, Mononuclear / microbiology
  • Macrophages / immunology
  • Macrophages / metabolism*
  • Macrophages / microbiology
  • Membrane Proteins / metabolism*
  • Monocytes / immunology
  • Monocytes / metabolism
  • Monocytes / microbiology
  • NF-kappa B / metabolism
  • Pediococcus pentosaceus / immunology
  • Pediococcus pentosaceus / physiology
  • Phagocytosis
  • Phosphorylation
  • Protein Serine-Threonine Kinases / metabolism
  • THP-1 Cells

Substances

  • Adaptor Proteins, Signal Transducing
  • Interferon Type I
  • MAVS protein, human
  • Membrane Proteins
  • NF-kappa B
  • STING1 protein, human
  • Protein Serine-Threonine Kinases
  • TBK1 protein, human