Heart immunoengineering by lentiviral vector-mediated genetic modification during normothermic ex vivo perfusion.

Autor: Schmalkuche, Katharina, Rother, Tamina, Burgmann, Jonathan M., Voß, Henrike, Höffler, Klaus, Dogan, Günes, Ruhparwar, Arjang, Schmitto, Jan D., Blasczyk, Rainer, Figueiredo, Constanca
Předmět:
Zdroj: Frontiers in Immunology; 2024, p1-17, 17p
Abstrakt: Heart transplantation is associated with major hurdles, including the limited number of available organs for transplantation, the risk of rejection due to genetic discrepancies, and the burden of immunosuppression. In this study, we demonstrated the feasibility of permanent genetic engineering of the heart during ex vivo perfusion. Lentiviral vectors encoding for short hairpin RNAs targeting beta2-microglobulin (shb2m) and class II transactivator (shCIITA) were delivered to the graft during two hours of normothermic EVHP. Highly efficient genetic engineering was indicated by stable reporter gene expression in endothelial cells and cardiomyocytes. Remarkably, swine leucocyte antigen (SLA) class I and SLA class II expression levels were decreased by 66% and 76%, respectively, in the vascular endothelium. Evaluation of lactate, troponin T, and LDH levels in the perfusate and histological analysis showed no additional cell injury or tissue damage caused by lentiviral vectors. Moreover, cytokine secretion profiles (IL-6, IL-8, and TNF-α) of non-transduced and lentiviral vectortransduced hearts were comparable. This study demonstrated the ex vivo generation of genetically engineered hearts without compromising tissue integrity. Downregulation of SLA expression may contribute to reduce the immunogenicity of the heart and support graft survival after allogeneic or xenogeneic transplantation. [ABSTRACT FROM AUTHOR]
Databáze: Complementary Index