Orientia tsutsugamushi uses two Ank effectors to modulate NF-κB p65 nuclear transport and inhibit NF-κB transcriptional activation

PLoS Pathog. 2018 May 7;14(5):e1007023. doi: 10.1371/journal.ppat.1007023. eCollection 2018 May.

Abstract

Orientia tsutsugamushi causes scrub typhus, a potentially fatal infection that threatens over one billion people. Nuclear translocation of the transcription factor, NF-κB, is the central initiating cellular event in the antimicrobial response. Here, we report that NF-κB p65 nuclear accumulation and NF-κB-dependent transcription are inhibited in O. tsutsugamushi infected HeLa cells and/or primary macrophages, even in the presence of TNFα. The bacterium modulates p65 subcellular localization by neither degrading it nor inhibiting IκBα degradation. Rather, it exploits host exportin 1 to mediate p65 nuclear export, as this phenomenon is leptomycin B-sensitive. O. tsutsugamushi antagonizes NF-κB-activated transcription even when exportin 1 is inhibited and NF-κB consequently remains in the nucleus. Two ankyrin repeat-containing effectors (Anks), Ank1 and Ank6, each of which possess a C-terminal F-box and exhibit 58.5% amino acid identity, are linked to the pathogen's ability to modulate NF-κB. When ectopically expressed, both translocate to the nucleus, abrogate NF-κB-activated transcription in an exportin 1-independent manner, and pronouncedly reduce TNFα-induced p65 nuclear levels by exportin 1-dependent means. Flag-tagged Ank 1 and Ank6 co-immunoprecipitate p65 and exportin 1. Both also bind importin β1, a host protein that is essential for the classical nuclear import pathway. Importazole, which blocks importin β1 activity, abrogates Ank1 and Ank6 nuclear translocation. The Ank1 and Ank6 regions that bind importin β1 also mediate their transport into the nucleus. Yet, these regions are distinct from those that bind p65/exportin 1. The Ank1 and Ank6 F-box and the region that lies between it and the ankyrin repeat domain are essential for blocking p65 nuclear accumulation. These data reveal a novel mechanism by which O. tsutsugamushi modulates the activity and nuclear transport of NF-κB p65 and identify the first microbial proteins that co-opt both importin β1 and exportin 1 to antagonize a critical arm of the antimicrobial response.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Active Transport, Cell Nucleus
  • Ankyrin Repeat
  • Bacterial Proteins / chemistry
  • Bacterial Proteins / genetics
  • Bacterial Proteins / metabolism*
  • Exportin 1 Protein
  • HeLa Cells
  • Host-Pathogen Interactions / genetics
  • Host-Pathogen Interactions / immunology
  • Host-Pathogen Interactions / physiology
  • Humans
  • Karyopherins / metabolism
  • NF-KappaB Inhibitor alpha / metabolism
  • NF-kappa B / genetics*
  • Orientia tsutsugamushi / immunology
  • Orientia tsutsugamushi / metabolism*
  • Orientia tsutsugamushi / pathogenicity*
  • Receptors, Cytoplasmic and Nuclear / metabolism
  • Scrub Typhus / immunology
  • Scrub Typhus / microbiology
  • Transcription Factor RelA / metabolism*
  • Transcriptional Activation
  • Virulence / genetics
  • Virulence / immunology
  • Virulence / physiology
  • beta Karyopherins / metabolism

Substances

  • Bacterial Proteins
  • KPNB1 protein, human
  • Karyopherins
  • NF-kappa B
  • RELA protein, human
  • Receptors, Cytoplasmic and Nuclear
  • Transcription Factor RelA
  • beta Karyopherins
  • NF-KappaB Inhibitor alpha