Alveolar epithelial cell–derived prostaglandin E2 serves as a request signal for macrophage secretion of suppressor of cytokine signaling 3 during innate inflammation

JM Speth, E Bourdonnay, LRK Penke… - The Journal of …, 2016 - journals.aai.org
JM Speth, E Bourdonnay, LRK Penke, P Mancuso, BB Moore, JB Weinberg…
The Journal of Immunology, 2016journals.aai.org
Preservation of gas exchange mandates that the pulmonary alveolar surface restrain
unnecessarily harmful inflammatory responses to the many challenges to which it is
exposed. These responses reflect the cross-talk between alveolar epithelial cells (AECs)
and resident alveolar macrophages (AMs). We recently determined that AMs can secrete
suppressor of cytokine signaling (SOCS) proteins within microparticles. Uptake of these
SOCS-containing vesicles by epithelial cells inhibits cytokine-induced STAT activation …
Abstract
Preservation of gas exchange mandates that the pulmonary alveolar surface restrain unnecessarily harmful inflammatory responses to the many challenges to which it is exposed. These responses reflect the cross-talk between alveolar epithelial cells (AECs) and resident alveolar macrophages (AMs). We recently determined that AMs can secrete suppressor of cytokine signaling (SOCS) proteins within microparticles. Uptake of these SOCS-containing vesicles by epithelial cells inhibits cytokine-induced STAT activation. However, the ability of epithelial cells to direct AM release of SOCS-containing vesicles in response to inflammatory insults has not been studied. In this study, we report that SOCS3 protein was elevated in bronchoalveolar lavage fluid of both virus-and bacteria-infected mice, as well as in an in vivo LPS model of acute inflammation. In vitro studies revealed that AEC-conditioned medium (AEC-CM) enhanced AM SOCS3 secretion above basal levels. Increased amounts of PGE 2 were present in AEC-CM after LPS challenge, and both pharmacologic inhibition of PGE 2 synthesis in AECs and neutralization of PGE 2 in AEC-CM implicated this prostanoid as the major AEC-derived factor mediating enhanced AM SOCS3 secretion. Moreover, pharmacologic blockade of PGE 2 synthesis or genetic deletion of a PGE 2 synthase similarly attenuated the increase in bronchoalveolar lavage fluid SOCS3 noted in lungs of mice challenged with LPS in vivo. These results demonstrate a novel tunable form of cross-talk in which AECs use PGE 2 as a signal to request SOCS3 from AMs to dampen their endogenous inflammatory responses during infection.
journals.aai.org