IL-20 signaling in activated human neutrophils inhibits neutrophil migration and function

P Gough, S Ganesan, SK Datta - The Journal of Immunology, 2017 - journals.aai.org
The Journal of Immunology, 2017journals.aai.org
Neutrophils possess multiple antimicrobial mechanisms that are critical for protection of the
host against infection with extracellular microbes, such as the bacterial pathogen
Staphylococcus aureus. Recruitment and activation of neutrophils at sites of infection are
driven by cytokine and chemokine signals that directly target neutrophils via specific cell
surface receptors. The IL-20 subfamily of cytokines has been reported to act at epithelial
sites and contribute to psoriasis, wound healing, and anti-inflammatory effects during S …
Abstract
Neutrophils possess multiple antimicrobial mechanisms that are critical for protection of the host against infection with extracellular microbes, such as the bacterial pathogen Staphylococcus aureus. Recruitment and activation of neutrophils at sites of infection are driven by cytokine and chemokine signals that directly target neutrophils via specific cell surface receptors. The IL-20 subfamily of cytokines has been reported to act at epithelial sites and contribute to psoriasis, wound healing, and anti-inflammatory effects during S. aureus infection. However, the ability of these cytokines to directly affect neutrophil function remains incompletely understood. In this article, we show that human neutrophils altered their expression of IL-20R chains upon migration and activation in vivo and in vitro. Such activation of neutrophils under conditions mimicking infection with S. aureus conferred responsiveness to IL-20 that manifested as modification of actin polymerization and inhibition of a broad range of actin-dependent functions, including phagocytosis, granule exocytosis, and migration. Consistent with the previously described homeostatic and anti-inflammatory properties of IL-20 on epithelial cells, the current study provides evidence that IL-20 directly targets and inhibits key inflammatory functions of neutrophils during infection with S. aureus.
journals.aai.org