The nuclear receptor PPARγ selectively inhibits Th17 differentiation in a T cell–intrinsic fashion and suppresses CNS autoimmunity

L Klotz, S Burgdorf, I Dani, K Saijo, J Flossdorf… - Journal of Experimental …, 2009 - rupress.org
L Klotz, S Burgdorf, I Dani, K Saijo, J Flossdorf, S Hucke, J Alferink, N Novak, M Beyer
Journal of Experimental Medicine, 2009rupress.org
T helper cells secreting interleukin (IL)-17 (Th17 cells) play a crucial role in autoimmune
diseases like multiple sclerosis (MS). Th17 differentiation, which is induced by a
combination of transforming growth factor (TGF)-β/IL-6 or IL-21, requires expression of the
transcription factor retinoic acid receptor–related orphan receptor γt (RORγt). We identify the
nuclear receptor peroxisome proliferator–activated receptor γ (PPARγ) as a key negative
regulator of human and mouse Th17 differentiation. PPARγ activation in CD4+ T cells …
T helper cells secreting interleukin (IL)-17 (Th17 cells) play a crucial role in autoimmune diseases like multiple sclerosis (MS). Th17 differentiation, which is induced by a combination of transforming growth factor (TGF)-β/IL-6 or IL-21, requires expression of the transcription factor retinoic acid receptor–related orphan receptor γt (RORγt). We identify the nuclear receptor peroxisome proliferator–activated receptor γ (PPARγ) as a key negative regulator of human and mouse Th17 differentiation. PPARγ activation in CD4+ T cells selectively suppressed Th17 differentiation, but not differentiation into Th1, Th2, or regulatory T cells. Control of Th17 differentiation by PPARγ involved inhibition of TGF-β/IL-6–induced expression of RORγt in T cells. Pharmacologic activation of PPARγ prevented removal of the silencing mediator for retinoid and thyroid hormone receptors corepressor from the RORγt promoter in T cells, thus interfering with RORγt transcription. Both T cell–specific PPARγ knockout and endogenous ligand activation revealed the physiological role of PPARγ for continuous T cell–intrinsic control of Th17 differentiation and development of autoimmunity. Importantly, human CD4+ T cells from healthy controls and MS patients were strongly susceptible to PPARγ-mediated suppression of Th17 differentiation. In summary, we report a PPARγ-mediated T cell–intrinsic molecular mechanism that selectively controls Th17 differentiation in mice and in humans and that is amenable to pharmacologic modulation. We therefore propose that PPARγ represents a promising molecular target for specific immunointervention in Th17-mediated autoimmune diseases such as MS.
rupress.org