Your browser doesn't support javascript.
loading
The Endoplasmic Reticulum Stress Sensor Inositol-Requiring Enzyme 1α Augments Bacterial Killing through Sustained Oxidant Production.
Abuaita, Basel H; Burkholder, Kristin M; Boles, Blaise R; O'Riordan, Mary X.
Afiliação
  • Abuaita BH; Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA.
  • Burkholder KM; Department of Biological Sciences, University of New England, Biddeford, Maine, USA.
  • Boles BR; Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, USA.
  • O'Riordan MX; Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA oriordan@umich.edu.
mBio ; 6(4): e00705, 2015 Jul 14.
Article em En | MEDLINE | ID: mdl-26173697
ABSTRACT
UNLABELLED Bacterial infection can trigger cellular stress programs, such as the unfolded protein response (UPR), which occurs when misfolded proteins accumulate within the endoplasmic reticulum (ER). Here, we used the human pathogen methicillin-resistant Staphylococcus aureus (MRSA) as an infection model to probe how ER stress promotes antimicrobial function. MRSA infection activated the most highly conserved unfolded protein response sensor, inositol-requiring enzyme 1α (IRE1α), which was necessary for robust bacterial killing in vitro and in vivo. The macrophage IRE1-dependent bactericidal activity required reactive oxygen species (ROS). Viable MRSA cells excluded ROS from the nascent phagosome and strongly triggered IRE1 activation, leading to sustained generation of ROS that were largely Nox2 independent. In contrast, dead MRSA showed early colocalization with ROS but was a poor activator of IRE1 and did not trigger sustained ROS generation. The global ROS stimulated by IRE1 signaling was necessary, but not sufficient, for MRSA killing, which also required the ER resident SNARE Sec22B for accumulation of ROS in the phagosomal compartment. Taken together, these results suggest that IRE1-mediated persistent ROS generation might act as a fail-safe mechanism to kill bacterial pathogens that evade the initial macrophage oxidative burst. IMPORTANCE Cellular stress programs have been implicated as important components of the innate immune response to infection. The role of the IRE1 pathway of the ER stress response in immune secretory functions, such as antibody production, is well established, but its contribution to innate immunity is less well defined. Here, we show that infection of macrophages with viable MRSA induces IRE1 activation, leading to bacterial killing. IRE1-dependent bactericidal activity required generation of reactive oxygen species in a sustained manner over hours of infection. The SNARE protein Sec22B, which was previously demonstrated to control ER-phagosome trafficking, was dispensable for IRE1-driven global ROS production but necessary for late ROS accumulation in bacteria-containing phagosomes. Our study highlights a key role for IRE1 in promoting macrophage bactericidal capacity and reveals a fail-safe mechanism that leads to the concentration of antimicrobial effector molecules in the macrophage phagosome.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Oxidantes / Proteínas Serina-Treonina Quinases / Endorribonucleases / Viabilidade Microbiana / Interações Hospedeiro-Patógeno / Staphylococcus aureus Resistente à Meticilina / Macrófagos Idioma: En Ano de publicação: 2015 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Oxidantes / Proteínas Serina-Treonina Quinases / Endorribonucleases / Viabilidade Microbiana / Interações Hospedeiro-Patógeno / Staphylococcus aureus Resistente à Meticilina / Macrófagos Idioma: En Ano de publicação: 2015 Tipo de documento: Article