[HTML][HTML] Complement serves as a switch between CD4+ T cell–independent and–dependent RBC antibody responses

A Mener, SR Patel, CM Arthur, S Chonat, A Wieland… - JCI insight, 2018 - ncbi.nlm.nih.gov
A Mener, SR Patel, CM Arthur, S Chonat, A Wieland, M Santhanakrishnan, J Liu, CL Maier…
JCI insight, 2018ncbi.nlm.nih.gov
RBC alloimmunization represents a significant immunological challenge for patients
requiring lifelong transfusion support. The majority of clinically relevant non-ABO (H) blood
group antigens have been thought to drive antibody formation through T cell–dependent
immune pathways. Thus, we initially sought to define the role of CD4+ T cells in formation of
alloantibodies to KEL, one of the leading causes of hemolytic transfusion reactions.
Unexpectedly, our findings demonstrated that KEL RBCs actually possess the ability to …
Abstract
RBC alloimmunization represents a significant immunological challenge for patients requiring lifelong transfusion support. The majority of clinically relevant non-ABO (H) blood group antigens have been thought to drive antibody formation through T cell–dependent immune pathways. Thus, we initially sought to define the role of CD4+ T cells in formation of alloantibodies to KEL, one of the leading causes of hemolytic transfusion reactions. Unexpectedly, our findings demonstrated that KEL RBCs actually possess the ability to induce antibody formation independent of CD4+ T cells or complement component 3 (C3), two common regulators of antibody formation. However, despite the ability of KEL RBCs to induce anti-KEL antibodies in the absence of complement, removal of C3 or complement receptors 1 and 2 (CR1/2) rendered recipients completely reliant on CD4+ T cells for IgG anti-KEL antibody formation. Together, these findings suggest that C3 may serve as a novel molecular switch that regulates the type of immunological pathway engaged following RBC transfusion.
ncbi.nlm.nih.gov