Identification of a novel antisepsis pathway: Sectm1a enhances macrophage phagocytosis of bacteria through activating GITR

X Mu, P Wang, X Wang, Y Li, H Zhao, Q Li… - The Journal of …, 2020 - journals.aai.org
X Mu, P Wang, X Wang, Y Li, H Zhao, Q Li, K Essandoh, S Deng, T Peng, GC Fan
The Journal of Immunology, 2020journals.aai.org
The inability to effectively control invading bacteria or other pathogens is a major cause of
multiple organ dysfunction and death in sepsis. As the first-line defense of the immune
system, macrophages play a crucial role in the removal of pathogens during sepsis. In this
study, we define secreted and transmembrane 1A (Sectm1a) as a novel ligand of
glucocorticoid-induced TNFR (GITR) that greatly boosts macrophage phagocytosis and
bactericidal capacity. Using a global Sectm1a knockout (KO) mouse model, we observed …
Abstract
The inability to effectively control invading bacteria or other pathogens is a major cause of multiple organ dysfunction and death in sepsis. As the first-line defense of the immune system, macrophages play a crucial role in the removal of pathogens during sepsis. In this study, we define secreted and transmembrane 1A (Sectm1a) as a novel ligand of glucocorticoid-induced TNFR (GITR) that greatly boosts macrophage phagocytosis and bactericidal capacity. Using a global Sectm1a knockout (KO) mouse model, we observed that Sectm1a deficiency significantly suppressed phagocytosis and bactericidal activity in both recruited macrophages and tissue-resident macrophages, which consequently aggravated bacterial burden in the blood and multiple organs and further increased systemic inflammation, leading to multiple organ injury and increased mortality during polymicrobial sepsis. By contrast, treatment of septic mice with recombinant Sectm1a protein (rSectm1a) not only promoted macrophage phagocytosis and bactericidal activity but also significantly improved survival outcome. Mechanistically, we identified that Sectm1a could bind to GITR in the surface of macrophages and thereby activate its downstream PI3K–Akt pathway. Accordingly, rSectm1a-mediated phagocytosis and bacterial killing were abolished in macrophages by either KO of GITR or pharmacological inhibition of the PI3K–Akt pathway. In addition, rSectm1a-induced therapeutic effects on sepsis injury were negated in GITR KO mice. Taken together, these results uncover that Sectm1a may represent a novel target for drug development to control bacterial dissemination during sepsis or other infectious diseases.
journals.aai.org