IL-17 promotes neutrophil entry into tumor-draining lymph nodes following induction of sterile inflammation

CM Brackett, JB Muhitch, SS Evans… - The Journal of …, 2013 - journals.aai.org
CM Brackett, JB Muhitch, SS Evans, SO Gollnick
The Journal of Immunology, 2013journals.aai.org
Blood-borne neutrophils are excluded from entering lymph nodes across vascular portals
termed high endothelial venules (HEVs) because of lack of expression of the CCR7
homeostatic chemokine receptor. Induction of sterile inflammation increases neutrophil entry
into tumor-draining lymph nodes (TDLNs), which is critical for induction of antitumor adaptive
immunity following treatments such as photodynamic therapy (PDT). However, the
mechanisms controlling neutrophil entry into TDLNs remain unclear. Prior evidence that IL …
Abstract
Blood-borne neutrophils are excluded from entering lymph nodes across vascular portals termed high endothelial venules (HEVs) because of lack of expression of the CCR7 homeostatic chemokine receptor. Induction of sterile inflammation increases neutrophil entry into tumor-draining lymph nodes (TDLNs), which is critical for induction of antitumor adaptive immunity following treatments such as photodynamic therapy (PDT). However, the mechanisms controlling neutrophil entry into TDLNs remain unclear. Prior evidence that IL-17 promotes neutrophil emigration to sites of infection via induction of CXCL2 and CXCL1 inflammatory chemokines raised the question of whether IL-17 contributes to chemokine-dependent trafficking in TDLNs. In this article, we demonstrate rapid accumulation of IL-17–producing Th17 cells in the TDLNs following induction of sterile inflammation by PDT. We further report that nonhematopoietic expression of IL-17RA regulates neutrophil accumulation in TDLNs following induction of sterile inflammation by PDT. We show that HEVs are the major route of entry of blood-borne neutrophils into TDLNs through interactions of l-selectin with HEV-expressed peripheral lymph node addressin and by preferential interactions between CXCR2 and CXCL2 but not CXCL1. CXCL2 induction in TDLNs was mapped in a linear pathway downstream of IL-17RA–dependent induction of IL-1β. These results define a novel IL-17–dependent mechanism promoting neutrophil delivery across HEVs in TDLNs during acute inflammatory responses.
journals.aai.org