[HTML][HTML] Suppression of allograft rejection by CD8+ CD122+ PD-1+ Tregs is dictated by their Fas ligand-initiated killing of effector T cells versus Fas-mediated own …

H Liu, Y Wang, Q Zeng, YQ Zeng, CL Liang, F Qiu… - Oncotarget, 2017 - ncbi.nlm.nih.gov
H Liu, Y Wang, Q Zeng, YQ Zeng, CL Liang, F Qiu, H Nie, Z Dai
Oncotarget, 2017ncbi.nlm.nih.gov
Mounting evidence has shown that naturally occurring CD8+ CD122+ T cells are regulatory
T cells (Tregs) that suppress both autoimmunity and alloimmunity. We have previously
shown that CD8+ CD122+ PD-1+ Tregs not only suppress allograft rejection, but also are
more potent in suppression than conventional CD4+ CD25+ Tregs. However, the
mechanisms underlying their suppression of alloimmunity are not well understood. In an
adoptive T-cell transfer model of mice lacking lymphocytes, we found that suppression of …
Abstract
Mounting evidence has shown that naturally occurring CD8+ CD122+ T cells are regulatory T cells (Tregs) that suppress both autoimmunity and alloimmunity. We have previously shown that CD8+ CD122+ PD-1+ Tregs not only suppress allograft rejection, but also are more potent in suppression than conventional CD4+ CD25+ Tregs. However, the mechanisms underlying their suppression of alloimmunity are not well understood. In an adoptive T-cell transfer model of mice lacking lymphocytes, we found that suppression of skin allograft rejection by CD8+ CD122+ PD-1+ Tregs was mostly dependent on their expression of Fas ligand as either lacking Fas ligand or blocking it with antibodies largely abolished their suppression of allograft rejection mediated by transferred T cells. Their suppression was also mostly reversed when effector T cells lacked Fas receptor. Indeed, these FasL+ Tregs induced T cell apoptosis in vitro in a Fas/FasL-dependent manner. However, their suppression of T cell proliferation in vitro was dependent on IL-10, but not FasL expression. Furthermore, adoptive transfer of CD8+ CD122+ PD-1+ Tregs significantly extended allograft survival even in wild-type mice if Tregs lacked Fas receptor or if recipients received recombinant IL-15, as these two measures synergistically expanded adoptively-transferred Tregs in recipients. Thus, this study may have important implications for Treg therapies in clinical transplantation.
ncbi.nlm.nih.gov