Abstract

Ehrlichia chaffeensis (E. chaffeensis) exploits evolutionarily conserved Notch and Wnt host cell signaling pathways to downregulate innate immune host defenses and promote infection. The multifunctional E. chaffeensis TRP120 effector which has HECT E3 ubiquitin ligase activity, interacts with the host nuclear tumor suppressor F-BOX and WD domain repeating-containing 7 (FBW7). FBW7 is the substrate recognition subunit of the Skp1-cullin-1-FBOX E3 ubiquitin (Ub) ligase complex (SCF) known to negatively regulate a network of oncoproteins (Notch, cyclin E, c-Jun, MCL1 and cMYC). In this study, we demonstrate that TRP120 and FBW7 colocalize strongly in the nucleus by confocal immunofluorescent microscopy and interactions between TRP120 and FBW7 FBOX and WD40 domains were demonstrated by ectopic expression and co-immunoprecipitation. Although FBW7 gene expression increased during E. chaffeensis infection, FBW7 levels significantly decreased (>70%) by 72 h post infection. Moreover, an iRNA knockdown of FBW7 coincided with increased E. chaffeensis infection and levels of Notch intracellular domain (NICD), phosphorylated c-Jun, MCL-1 and cMYC, which are negatively regulated by FBW7. An increase in FBW7 K48 ubiquitination was detected during infection by co-IP, and FBW7 degradation was inhibited in infected cells treated with the proteasomal inhibitor bortezomib. Direct TRP120 ubiquitination of native and recombinant FBW7 was demonstrated in vitro and confirmed by ectopic expression of TRP120 HECT Ub ligase catalytic site mutant. This study identifies the tumor suppressor, FBW7, as a TRP120 HECT E3 Ub ligase substrate, and demonstrates that TRP120 ligase activity promotes ehrlichial infection by degrading FBW7 to maintain stability of Notch and other oncoproteins involved in cell survival and apoptosis.

Highlights

  • Ehrlichia chaffeensis (E. chaffeensis) is an obligately intracellular, gram-negative bacterium that exhibits tropism for mononuclear phagocytes and resides in microcolonies within membranebound cytoplasmic vacuoles known as morulae [1,2]

  • E. chaffeensis is an obligately intracellular bacterium that replicates in mononuclear phagocytes by secreting effectors that manipulate host cell processes and exploit evolutionarily conserved pathways

  • We demonstrate that E. chaffeensis TRP120 HECT Ub ligase targets the nuclear tumor suppressor Skp1-cullin-1-FBOX E3 ubiquitin (Ub) ligase complex substrate recognition subunit, F-BOX and WD domain repeating-containing 7 (FBW7) for degradation

Read more

Summary

Introduction

Ehrlichia chaffeensis (E. chaffeensis) is an obligately intracellular, gram-negative bacterium that exhibits tropism for mononuclear phagocytes and resides in microcolonies within membranebound cytoplasmic vacuoles known as morulae [1,2]. E. chaffeensis survival in the mononuclear phagocyte is dependent in part on pathogen-host interactions involving tandem repeat protein (TRP) effectors that are secreted via the type-1 secretion system and interact with a diverse array of host targets [3,4,5]. TRPs translocate across the morula membrane via an unknown mechanism and enter the host cell cytosol and nucleus where they function to reprogram the cell through direct interactions with well-defined and lesser known host cell targets [3]. One of the most studied E. chaffeensis effectors is TRP120, a moonlighting effector that has several defined functions. TRP120 is a functional HECT E3 ligase that ubiquitinates host cell substrates including a known interacting partner, polycomb group ring finger protein 5 (PCGF5), a component of the nuclear polycomb repressive complex [10]. TRP120 itself exploits host cell post-translational machinery and is SUMOylated at a canonical motif, which is known to affect TRP120-host target interactions [7,11]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call