The parasitic worm product ES-62 up-regulates IL-22 production by γδ T cells in the murine model of collagen-induced arthritis

Harnett, Margaret M. and Harnett, William and Pineda, Miguel A. (2014) The parasitic worm product ES-62 up-regulates IL-22 production by γδ T cells in the murine model of collagen-induced arthritis. Inflammation and Cell Signaling, 1 (3). pp. 1-19. ISSN 2330-7803 (https://doi.org/10.14800/ics.308)

[thumbnail of Harnett-etal-ICS-2014-The-parasitic-worm-product-ES-62-up-regulates-IL-22-production]
Preview
Text. Filename: Harnett_etal_ICS_2014_The_parasitic_worm_product_ES_62_up_regulates_IL_22_production.pdf
Final Published Version
License: Creative Commons Attribution 4.0 logo

Download (1MB)| Preview

Abstract

ES-62 is a phosphorylcholine (PC)-containing glycoprotein secreted by the filarial nematode Acanthocheilonema viteae that acts to modulate the host immune response to promote the establishment of chronic helminth infection. Reflecting its anti-inflammatory actions, we have previously reported that ES-62 protects mice from developing Collagen-Induced Arthritis (CIA): thus, as this helminth-derived product may exhibit therapeutic potential in Rheumatoid Arthritis (RA), it is important to understand the protective immunoregulatory mechanisms triggered by ES-62 in this model in vivo. We have established to date that ES-62 acts by downregulating pathogenic Th17/IL-17-mediated responses and upregulating the regulatory cytokine IL-10. In addition, our studies have identified that IL-22, another member of the IL-10 family of cytokines, exerts dual pathogenic and protective roles in this model of RA with ES-62 harnessing the cytokine's inflammation-resolving and tissue repair properties in the joint during the established phase of disease. Here, we discuss the counter-regulatory roles of IL-22 in the murine model of CIA and present additional novel data showing that ES-62 selectively induces γδ T cells with the capacity to induce IL-22 production and that γδ T cells with the capacity to produce IL-22, but not IL-17, induced during CIA can be identified by their expression of TLR4. Moreover, we also show that treatment of mice undergoing CIA with the active PC moiety of ES-62, in the form of PC conjugated to BSA, is not only sufficient to mimic the ES-62-dependent suppression of pathogenic IL-17 responses shown previously but also that of the IL-22 and IL-10 up-regulation observed with the parasitic worm product during CIA. These findings not only reinforce the potential of IL-22, firstly described as a Th17-related pro-inflammatory cytokine, as a protective factor in arthritis but also suggest that drugs based on the PC moiety found in ES-62 may be able to harness the joint-protecting activities of IL-22 therapeutically.