Dendritic cell-specific disruption of TGF-β receptor II leads to altered regulatory T cell phenotype and spontaneous multiorgan autoimmunity

R Ramalingam, CB Larmonier, RD Thurston… - The Journal of …, 2012 - journals.aai.org
R Ramalingam, CB Larmonier, RD Thurston, MT Midura-Kiela, SG Zheng, FK Ghishan…
The Journal of Immunology, 2012journals.aai.org
In vitro data and transgenic mouse models suggest a role for TGF-β signaling in dendritic
cells (DCs) to prevent autoimmunity primarily through maintenance of DCs in their immature
and tolerogenic state characterized by low expression of MHC class II (MHCII) and
costimulatory molecules and increased expression of IDO, among others. To test whether a
complete lack of TGF-β signaling in DCs predisposes mice to spontaneous autoimmunity
and to verify the mechanisms implicated previously in vitro, we generated conditional …
Abstract
In vitro data and transgenic mouse models suggest a role for TGF-β signaling in dendritic cells (DCs) to prevent autoimmunity primarily through maintenance of DCs in their immature and tolerogenic state characterized by low expression of MHC class II (MHCII) and costimulatory molecules and increased expression of IDO, among others. To test whether a complete lack of TGF-β signaling in DCs predisposes mice to spontaneous autoimmunity and to verify the mechanisms implicated previously in vitro, we generated conditional knockout (KO) mice with Cre-mediated DC-specific deletion of Tgfbr2 (DC-Tgfbr2 KO). DC-Tgfbr2 KO mice die before 15 wk of age with multiorgan autoimmune inflammation and spontaneous activation of T and B cells. Interestingly, there were no significant differences in the expression of MHCII, costimulatory molecules, or IDO in secondary lymphoid organ DCs, although Tgfbr2-deficient DCs were more proinflammatory in vitro and in vivo. DC-Tgfbr2 KO showed attenuated Foxp3 expression in regulatory T cells (Tregs) and abnormal expansion of CD25− Foxp3+ Tregs in vivo. Tgfbr2-deficient DCs secreted elevated levels of IFN-γ and were not capable of directing Ag-specific Treg conversion unless in the presence of anti–IFN-γ blocking Ab. Adoptive transfer of induced Tregs into DC-Tgfbr2 KO mice partially rescued the phenotype. Therefore, in vivo, TGF-β signaling in DCs is critical in the control of autoimmunity through both Treg-dependent and-independent mechanisms, but it does not affect MHCII and costimulatory molecule expression.
journals.aai.org