[HTML][HTML] IL-1β–dependent extravasation of preexisting lung-restricted autoantibodies during lung transplantation activates complement and mediates primary graft …

W Yang, EJ Cerier, FL Núñez-Santana… - The Journal of …, 2022 - Am Soc Clin Investig
W Yang, EJ Cerier, FL Núñez-Santana, Q Wu, Y Yan, C Kurihara, X Liu, A Yeldandi…
The Journal of Clinical Investigation, 2022Am Soc Clin Investig
Preexisting lung-restricted autoantibodies (LRAs) are associated with a higher incidence of
primary graft dysfunction (PGD), although it remains unclear whether LRAs can drive its
pathogenesis. In syngeneic murine left lung transplant recipients, preexisting LRAs
worsened graft dysfunction, which was evident by impaired gas exchange, increased
pulmonary edema, and activation of damage-associated pathways in lung epithelial cells.
LRA-mediated injury was distinct from ischemia-reperfusion injury since deletion of donor …
Preexisting lung-restricted autoantibodies (LRAs) are associated with a higher incidence of primary graft dysfunction (PGD), although it remains unclear whether LRAs can drive its pathogenesis. In syngeneic murine left lung transplant recipients, preexisting LRAs worsened graft dysfunction, which was evident by impaired gas exchange, increased pulmonary edema, and activation of damage-associated pathways in lung epithelial cells. LRA-mediated injury was distinct from ischemia-reperfusion injury since deletion of donor nonclassical monocytes and host neutrophils could not prevent graft dysfunction in LRA-pretreated recipients. Whole LRA IgG molecules were necessary for lung injury, which was mediated by the classical and alternative complement pathways and reversed by complement inhibition. However, deletion of Fc receptors in donor macrophages or mannose-binding lectin in recipient mice failed to rescue lung function. LRA-mediated injury was localized to the transplanted lung and dependent on IL-1β–mediated permeabilization of pulmonary vascular endothelium, which allowed extravasation of antibodies. Genetic deletion or pharmacological inhibition of IL-1R in the donor lungs prevented LRA-induced graft injury. In humans, preexisting LRAs were an independent risk factor for severe PGD and could be treated with plasmapheresis and complement blockade. We conclude that preexisting LRAs can compound ischemia-reperfusion injury to worsen PGD for which complement inhibition may be effective.
The Journal of Clinical Investigation