Requirement for MyD88 signaling in B cells and dendritic cells for germinal center anti-nuclear antibody production in Lyn-deficient mice

Z Hua, AJ Gross, C Lamagna… - The Journal of …, 2014 - journals.aai.org
Z Hua, AJ Gross, C Lamagna, N Ramos-Hernández, P Scapini, M Ji, H Shao, CA Lowell
The Journal of Immunology, 2014journals.aai.org
The intracellular tyrosine kinase Lyn mediates inhibitory receptor function in B cells and
myeloid cells, and Lyn−/− mice spontaneously develop an autoimmune and inflammatory
disease that closely resembles human systemic lupus erythematosus. TLR-signaling
pathways have been implicated in the production of anti-nuclear Abs in systemic lupus
erythematosus and mouse models of it. We used a conditional allele of Myd88 to determine
whether the autoimmunity of Lyn−/− mice is dependent on TLR/MyD88 signaling in B cells …
Abstract
The intracellular tyrosine kinase Lyn mediates inhibitory receptor function in B cells and myeloid cells, and Lyn−/− mice spontaneously develop an autoimmune and inflammatory disease that closely resembles human systemic lupus erythematosus. TLR-signaling pathways have been implicated in the production of anti-nuclear Abs in systemic lupus erythematosus and mouse models of it. We used a conditional allele of Myd88 to determine whether the autoimmunity of Lyn−/− mice is dependent on TLR/MyD88 signaling in B cells and/or in dendritic cells (DCs). The production of IgG anti-nuclear Abs, as well as the deposition of these Abs in the glomeruli of the kidneys, leading to glomerulonephritis in Lyn−/− mice, were completely abolished by selective deletion of Myd88 in B cells, and autoantibody production and glomerulonephritis were delayed or decreased by deletion of Myd88 in DCs. The reduced autoantibody production in mice lacking MyD88 in B cells or DCs was accompanied by a dramatic decrease in the spontaneous germinal center (GC) response, suggesting that autoantibodies in Lyn−/− mice may depend on GC responses. Consistent with this view, IgG anti-nuclear Abs were absent if T cells were deleted (TCRβ−/− TCRδ−/− mice) or if T cells were unable to contribute to GC responses as the result of mutation of the adaptor molecule SAP. Thus, the autoimmunity of Lyn−/− mice was dependent on T cells and on TLR/MyD88 signaling in B cells and in DCs, supporting a model in which DC hyperactivity combines with defects in tolerance in B cells to lead to a T cell–dependent systemic autoimmunity in Lyn−/− mice.
journals.aai.org