Ethylenecarbodiimide-fixed donor splenocyte infusions differentially target direct and indirect pathways of allorecognition for induction of transplant tolerance

T Kheradmand, S Wang, J Bryant, JJ Tasch… - The Journal of …, 2012 - journals.aai.org
T Kheradmand, S Wang, J Bryant, JJ Tasch, N Lerret, KL Pothoven, JL Houlihan, SD Miller
The Journal of Immunology, 2012journals.aai.org
Strategic exposure to donor Ags prior to transplantation can be an effective way for inducting
donor-specific tolerance in allogeneic recipients. We have recently shown that pretransplant
infusion of donor splenocytes treated with the chemical cross-linker ethylenecarbodiimide
(ECDI-SPs) induces indefinite islet allograft survival in a full MHC-mismatched model
without the need for any immunosuppression. Mechanisms of allograft protection by this
strategy remain elusive. In this study, we show that the infused donor ECDI-SPs differentially …
Abstract
Strategic exposure to donor Ags prior to transplantation can be an effective way for inducting donor-specific tolerance in allogeneic recipients. We have recently shown that pretransplant infusion of donor splenocytes treated with the chemical cross-linker ethylenecarbodiimide (ECDI-SPs) induces indefinite islet allograft survival in a full MHC-mismatched model without the need for any immunosuppression. Mechanisms of allograft protection by this strategy remain elusive. In this study, we show that the infused donor ECDI-SPs differentially target T cells with indirect versus direct allospecificities. To target indirect allospecific T cells, ECDI-SPs induce upregulation of negative, but not positive, costimulatory molecules on recipient splenic CD11c+ dendritic cells phagocytosing the injected ECDI-SPs. Indirect allospecific T cells activated by such CD11c+ dendritic cells undergo robust initial proliferation followed by rapid clonal depletion. The remaining T cells are sequestered in the spleen without homing to the graft site or the graft draining lymph node. In contrast, direct allospecific T cells interacting with intact donor ECDI-SPs not yet phagocytosed undergo limited proliferation and are subsequently anergized. Furthermore, CD4+ CD25+ Foxp3+ T cells are induced in lymphoid organs and at the graft site by ECDI-SPs. We conclude that donor ECDI-SP infusions target host allogeneic responses via a multitude of mechanisms, including clonal depletion, anergy, and immunoregulation, which act in a synergistic fashion to induce robust transplant tolerance. This simple form of negative vaccination has significant potential for clinical translation in human transplantation.
journals.aai.org