2′-O-methylation within prokaryotic and eukaryotic tRNA inhibits innate immune activation by endosomal Toll-like receptors but does not affect recognition of whole organisms

  1. Tatjana Eigenbrod1
  1. 1Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120 Heidelberg, Germany
  2. 2Institute of Pharmacy and Biochemistry, Johannes Gutenberg-University Mainz, 55128 Mainz, Germany
  3. 3IMoPA UMR7365 CNRS-Lorraine University, BioPole Lorraine University, 54500 Vandoeuvre-les-Nancy, France
  4. 4UMS2008 IBSLor, CNRS-Lorraine University-INSERM, BioPole Lorraine University, 54500 Vandoeuvre-les-Nancy, France
  5. 5Division of Innate Immunity, Research Center Borstel, 23845 Borstel, Germany
  6. 6Institute of Medical Microbiology and Hygiene, Technical University Dresden, 01307 Dresden, Germany
  1. Corresponding author: tatjana.eigenbrod{at}med.uni-heidelberg.de

Abstract

Bacterial RNA has emerged as an important activator of innate immune responses by stimulating Toll-like receptors TLR7 and TLR8 in humans. Guanosine 2′-O-methylation at position 18 (Gm18) in bacterial tRNA was shown to antagonize tRNA-induced TLR7/8 activation, suggesting a potential role of Gm18 as an immune escape mechanism. This modification also occurs in eukaryotic tRNA, yet a physiological immune function remained to be tested. We therefore set out to investigate the immune modulatory role of Gm18 in both prokaryotic and eukaryotic microorganisms, Escherichia coli and Saccharomyces cerevisiae, and in human cells. Using RiboMethSeq analysis we show that mutation of trmH in E. coli, trm3 in S. cereviase, and CRISPR/Cas9-induced knockout of TARBP1 in H. sapiens results in loss of Gm18 within tRNA. Lack of Gm18 across the kingdoms resulted in increased immunostimulation of peripheral blood mononuclear cells when activated by tRNA preparations. In E. coli, lack of 2′-O-methyltransferase trmH also enhanced immune stimulatory properties by whole cellular RNA. In contrast, lack of Gm18 in yeasts and human cells did not affect immunostimulation by whole RNA preparations. When using live E. coli bacteria, lack of trmH did not affect overall immune stimulation although we detected a defined TLR8/RNA-dependent gene expression signature upon E. coli infection. Together, these results demonstrate that Gm18 is a global immune inhibitory tRNA modification across the kingdoms and contributes to tRNA recognition by innate immune cells, but as an individual modification has insufficient potency to modulate recognition of the investigated microorganisms.

Keywords

Footnotes

  • Abbreviations: MOI, multiplicity of infection; PBMC, peripheral blood mononuclear cells; E. coli, Escherichia coli; S. cerevisiae, Saccharomyces cerevisiae; S. aureus, Staphylococcus aureus; PAMP, pathogen-associated molecular pattern; PRR, pattern recognition receptor; TLR, Toll-like receptor

  • Article is online at http://www.rnajournal.org/cgi/doi/10.1261/rna.070243.118.

  • Received January 8, 2019.
  • Accepted April 20, 2019.

This article is distributed exclusively by the RNA Society for the first 12 months after the full-issue publication date (see http://rnajournal.cshlp.org/site/misc/terms.xhtml). After 12 months, it is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), as described at http://creativecommons.org/licenses/by-nc/4.0/.

| Table of Contents