[HTML][HTML] IL-27 receptor signalling restricts the formation of pathogenic, terminally differentiated Th1 cells during malaria infection by repressing IL-12 dependent …

A Villegas-Mendez, JB de Souza, SW Lavelle… - PLoS …, 2013 - journals.plos.org
A Villegas-Mendez, JB de Souza, SW Lavelle, E Gwyer Findlay, TN Shaw, N van Rooijen
PLoS pathogens, 2013journals.plos.org
The IL-27R, WSX-1, is required to limit IFN-γ production by effector CD4+ T cells in a number
of different inflammatory conditions but the molecular basis of WSX-1-mediated regulation of
Th1 responses in vivo during infection has not been investigated in detail. In this study we
demonstrate that WSX-1 signalling suppresses the development of pathogenic, terminally
differentiated (KLRG-1+) Th1 cells during malaria infection and establishes a restrictive
threshold to constrain the emergent Th1 response. Importantly, we show that WSX-1 …
The IL-27R, WSX-1, is required to limit IFN-γ production by effector CD4+ T cells in a number of different inflammatory conditions but the molecular basis of WSX-1-mediated regulation of Th1 responses in vivo during infection has not been investigated in detail. In this study we demonstrate that WSX-1 signalling suppresses the development of pathogenic, terminally differentiated (KLRG-1+) Th1 cells during malaria infection and establishes a restrictive threshold to constrain the emergent Th1 response. Importantly, we show that WSX-1 regulates cell-intrinsic responsiveness to IL-12 and IL-2, but the fate of the effector CD4+ T cell pool during malaria infection is controlled primarily through IL-12 dependent signals. Finally, we show that WSX-1 regulates Th1 cell terminal differentiation during malaria infection through IL-10 and Foxp3 independent mechanisms; the kinetics and magnitude of the Th1 response, and the degree of Th1 cell terminal differentiation, were comparable in WT, IL-10R1−/− and IL-10−/− mice and the numbers and phenotype of Foxp3+ cells were largely unaltered in WSX-1−/− mice during infection. As expected, depletion of Foxp3+ cells did not enhance Th1 cell polarisation or terminal differentiation during malaria infection. Our results significantly expand our understanding of how IL-27 regulates Th1 responses in vivo during inflammatory conditions and establishes WSX-1 as a critical and non-redundant regulator of the emergent Th1 effector response during malaria infection.
PLOS