[HTML][HTML] Reducing FLI1 levels in the MRL/lpr lupus mouse model impacts T cell function by modulating glycosphingolipid metabolism

EM Richard, T Thiyagarajan, MA Bunni, F Basher… - PloS one, 2013 - journals.plos.org
EM Richard, T Thiyagarajan, MA Bunni, F Basher, PO Roddy, LJ Siskind, PJ Nietert…
PloS one, 2013journals.plos.org
Systemic Lupus erythematosus (SLE) is an autoimmune disease caused, in part, by
abnormalities in cells of the immune system including B and T cells. Genetically reducing
globally the expression of the ETS transcription factor FLI1 by 50% in two lupus mouse
models significantly improves disease measures and survival through an unknown
mechanism. In this study we analyze the effects of reducing FLI1 in the MRL/lpr lupus prone
model on T cell function. We demonstrate that adoptive transfer of MRL/lpr Fli1+/+ or Fli1+/-T …
Systemic Lupus erythematosus (SLE) is an autoimmune disease caused, in part, by abnormalities in cells of the immune system including B and T cells. Genetically reducing globally the expression of the ETS transcription factor FLI1 by 50% in two lupus mouse models significantly improves disease measures and survival through an unknown mechanism. In this study we analyze the effects of reducing FLI1 in the MRL/lpr lupus prone model on T cell function. We demonstrate that adoptive transfer of MRL/lpr Fli1+/+ or Fli1+/- T cells and B cells into Rag1-deficient mice results in significantly decreased serum immunoglobulin levels in animals receiving Fli1+/- lupus T cells compared to animals receiving Fli1+/+ lupus T cells regardless of the genotype of co-transferred lupus B cells. Ex vivo analyses of MRL/lpr T cells demonstrated that Fli1+/- T cells produce significantly less IL-4 during early and late disease and exhibited significantly decreased TCR-specific activation during early disease compared to Fli1+/+ T cells. Moreover, the Fli1+/- T cells expressed significantly less neuraminidase 1 (Neu1) message and decreased NEU activity during early disease and significantly decreased levels of glycosphingolipids during late disease compared to Fli1+/+ T cells. FLI1 dose-dependently activated the Neu1 promoter in mouse and human T cell lines. Together, our results suggest reducing FLI1 in lupus decreases the pathogenicity of T cells by decreasing TCR-specific activation and IL-4 production in part through the modulation of glycosphingolipid metabolism. Reducing the expression of FLI1 or targeting the glycosphingolipid metabolic pathway in lupus may serve as a therapeutic approach to treating lupus.
PLOS