Serum amyloid A activates the NLRP3 inflammasome and promotes Th17 allergic asthma in mice

JL Ather, K Ckless, R Martin, KL Foley… - The Journal of …, 2011 - journals.aai.org
JL Ather, K Ckless, R Martin, KL Foley, BT Suratt, JE Boyson, KA Fitzgerald, RA Flavell
The Journal of Immunology, 2011journals.aai.org
IL-1β is a cytokine critical to several inflammatory diseases in which pathogenic Th17
responses are implicated. Activation of the NLRP3 inflammasome by microbial and
environmental stimuli can enable the caspase-1–dependent processing and secretion of IL-
1β. The acute-phase protein serum amyloid A (SAA) is highly induced during inflammatory
responses, wherein it participates in systemic modulation of innate and adaptive immune
responses. Elevated levels of IL-1β, SAA, and IL-17 are present in subjects with severe …
Abstract
IL-1β is a cytokine critical to several inflammatory diseases in which pathogenic Th17 responses are implicated. Activation of the NLRP3 inflammasome by microbial and environmental stimuli can enable the caspase-1–dependent processing and secretion of IL-1β. The acute-phase protein serum amyloid A (SAA) is highly induced during inflammatory responses, wherein it participates in systemic modulation of innate and adaptive immune responses. Elevated levels of IL-1β, SAA, and IL-17 are present in subjects with severe allergic asthma, yet the mechanistic relationship among these mediators has yet to be identified. In this study, we demonstrate that Saa3 is expressed in the lungs of mice exposed to several mixed Th2/Th17-polarizing allergic sensitization regimens. SAA instillation into the lungs elicits robust TLR2-, MyD88-, and IL-1–dependent pulmonary neutrophilic inflammation. Furthermore, SAA drives production of IL-1α, IL-1β, IL-6, IL-23, and PGE 2, causes dendritic cell (DC) maturation, and requires TLR2, MyD88, and the NLRP3 inflammasome for secretion of IL-1β by DCs and macrophages. CD4+ T cells polyclonally stimulated in the presence of conditioned media from SAA-exposed DCs produced IL-17, and the capacity of polyclonally stimulated splenocytes to secrete IL-17 is dependent upon IL-1, TLR2, and the NLRP3 inflammasome. Additionally, in a model of allergic airway inflammation, administration of SAA to the lungs functions as an adjuvant to sensitize mice to inhaled OVA, resulting in leukocyte influx after Ag challenge and a predominance of IL-17 production from restimulated splenocytes that is dependent upon IL-1R signaling.
journals.aai.org