Published in

American Society of Hematology, Blood, 20(122), p. 3473-3481, 2013

DOI: 10.1182/blood-2013-05-502229

Links

Tools

Export citation

Search in Google Scholar

C3a modulates IL-1β secretion in human monocytes by regulating ATP efflux and subsequent NLRP3 inflammasome activation

This paper is made freely available by the publisher.
This paper is made freely available by the publisher.

Full text: Download

Red circle
Preprint: archiving forbidden
Orange circle
Postprint: archiving restricted
Red circle
Published version: archiving forbidden
Data provided by SHERPA/RoMEO

Abstract

IL-1β is a potent pro-inflammatory cytokine and a therapeutic target in several chronic inflammatory and autoimmune states. Monocytes and macrophages are the major sources of IL-1β. IL-1β production by these cells requires toll-like receptor (TLR) and adenosine triphosphate (ATP)-mediated P2X purinoceptor 7 (P2X7) signals, which together activate the inflammasome. However, how TLR signals and ATP availability are regulated during monocyte activation is unclear and the involvement of another danger signal system has long been proposed. Here, we demonstrate that both lipopolysaccharide (LPS) and the anaphylatoxin C3a, a complement cleavage fragment, are needed for IL-1β production in human macrophages and DCs, while in monocytes, C3a enhanced the secretion of LPS-induced IL-1β. C3a and LPS-stimulated monocytes increased Th17 cell induction in vitro, and human rejecting but not non-rejecting kidney transplant biopsies were characterized by local generation of C3a as well as monocyte and Th17 cell infiltration. Mechanistically, C3a drives IL-1β production in monocytes by controlling the release of intracellular ATP into the extracellular space via regulation of, yet unidentified, ATP-releasing channels in an ERK1/2-dependent fashion. These data define a novel function for complement in inflammasome activation in monocytes and suggest that C3aR-mediated signaling is a vital component of the IL-1β-Th17 axis.