Hepatocyte-intrinsic type I interferon signaling reprograms metabolism and reveals a novel compensatory mechanism of the tryptophan-kynurenine pathway in viral hepatitis

PLoS Pathog. 2020 Oct 12;16(10):e1008973. doi: 10.1371/journal.ppat.1008973. eCollection 2020 Oct.

Abstract

The liver is a central regulator of metabolic homeostasis and serum metabolite levels. Hepatocytes are the functional units of the liver parenchyma and not only responsible for turnover of biomolecules but also act as central immune signaling platforms. Hepatotropic viruses infect liver tissue, resulting in inflammatory responses, tissue damage and hepatitis. Combining well-established in vitro and in vivo model systems with transcriptomic analyses, we show that type I interferon signaling initiates a robust antiviral immune response in hepatocytes. Strikingly, we also identify IFN-I as both, sufficient and necessary, to induce wide-spread metabolic reprogramming in hepatocytes. IFN-I specifically rewired tryptophan metabolism and induced hepatic tryptophan oxidation to kynurenine via Tdo2, correlating with altered concentrations of serum metabolites upon viral infection. Infected Tdo2-deficient animals displayed elevated serum levels of tryptophan and, unexpectedly, also vast increases in the downstream immune-suppressive metabolite kynurenine. Thus, Tdo2-deficiency did not result in altered serum homeostasis of the tryptophan to kynurenine ratio during infection, which seemed to be independent of hepatocyte-intrinsic compensation via the IDO-axis. These data highlight that inflammation-induced reprogramming of systemic tryptophan metabolism is tightly regulated in viral hepatitis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antiviral Agents / metabolism*
  • Female
  • Hepatitis Viruses / isolation & purification
  • Hepatitis, Viral, Animal / immunology*
  • Hepatitis, Viral, Animal / metabolism
  • Hepatitis, Viral, Animal / virology
  • Hepatocytes / immunology*
  • Hepatocytes / metabolism
  • Hepatocytes / virology
  • Humans
  • Immunity, Innate / immunology
  • Inflammation / immunology*
  • Inflammation / metabolism
  • Inflammation / pathology
  • Inflammation / virology
  • Interferon Regulatory Factor-7 / physiology
  • Kynurenine / metabolism*
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Receptor, Interferon alpha-beta / physiology*
  • STAT1 Transcription Factor / physiology
  • Tryptophan / metabolism*
  • Tryptophan Oxygenase / physiology

Substances

  • Antiviral Agents
  • Ifnar1 protein, mouse
  • Interferon Regulatory Factor-7
  • Irf7 protein, mouse
  • STAT1 Transcription Factor
  • Stat1 protein, mouse
  • Receptor, Interferon alpha-beta
  • Kynurenine
  • Tryptophan
  • Tryptophan Oxygenase

Grants and funding

This project received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (grant agreement 677006, “CMIL” awarded to A.B.). B.A. was supported by the Austrian Science Fund (FWF) and the Medical University of Vienna’s joint PhD program in Inflammation and Immunity (FWF1212). A.L. was supported by a DOC fellowship of the Austrian Academy of Sciences. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.