Molecular mechanisms of biogenesis of apoptotic exosome-like vesicles and their roles as damage-associated molecular patterns

SJ Park, JM Kim, J Kim, J Hur, S Park… - Proceedings of the …, 2018 - National Acad Sciences
SJ Park, JM Kim, J Kim, J Hur, S Park, K Kim, HJ Shin, YJ Chwae
Proceedings of the National Academy of Sciences, 2018National Acad Sciences
Recent research has led to contradictory notions regarding the conventional theory that
apoptotic cell death can evoke inflammatory or immunogenic responses orchestrated by
released damage-associated patterns (DAMPs). By inducing IL-1β from bone marrow-
derived macrophages in an effort to determine the inflammatory mediators released from
apoptotic cells, we found that exosomal fractions called “apoptotic exosome-like
vesicles”(AEVs) prepared from apoptotic-conditioned medium were the main inflammatory …
Recent research has led to contradictory notions regarding the conventional theory that apoptotic cell death can evoke inflammatory or immunogenic responses orchestrated by released damage-associated patterns (DAMPs). By inducing IL-1β from bone marrow-derived macrophages in an effort to determine the inflammatory mediators released from apoptotic cells, we found that exosomal fractions called “apoptotic exosome-like vesicles” (AEVs) prepared from apoptotic-conditioned medium were the main inflammatory factors. These AEVs showed characteristics of exosomes in their size, density, morphology, and protein expression but had unique marker proteins, sphingosine-1-phosphate receptors 1 and 3 (S1PR1 and 3). Their biogenesis was completely dependent on cellular sphingosine-1-phosphate (S1P)/S1PRs signaling from multiple fine spindles of plasma membrane accompanied by F-actin, S1PR1, S1PR3, and CD63 at the early apoptotic phase and progressing to the maturation of F-actin–guided multivesicular endosomes mediated by Gβγ subunits of S1PRs downstream. S1P-loaded S1PRs on AEVs were critical factors for inducing IL-1β via NF-κB transcriptional factor and p38 MAPK, possibly through the RHOA/NOD2 axis, in differentiating macrophages. The AEVs induced genes of proinflammatory cytokines, chemokines, and mediators in both in vitro and in vivo models. In conclusion, AEVs could be key inflammatory mediators, acting as DAMPs that could explain the pathogeneses of various chronic inflammations, autoimmune diseases, or cancers in the future.
National Acad Sciences