Abstract

Abstract T cell development depends on coordinated gene expression changes in order to progress through developmental checkpoints. The loss of transcription factor Runx1 in CD4-cre Runx1 conditional knockout (cKO) mice leads to decreased numbers of CD4 single positive (SP) thymocytes, CD4 peripheral T cells and lower expression of IL-7Rα, a key player of T cell homeostasis, by CD4+ Runx1 deficient T cells . Low IL-7Rα is thought to cause decreased CD4 T cell survival. However, we show that constitutive expression of an IL-7Rα transgene in CD4-cre Runx1 cKO mice does not restore thymic or peripheral CD4 cellularity. We also see similar expression of Bcl-2 and Bcl-xL in WT and Runx1-deficient CD4 T cells, indicating that a Runx1-mediated IL-7Rα-independent mechanism might be responsible. As IL-7Rα expression increases over the course of T cell maturation, we asked if Runx1 is needed for maturation. During thymic maturation, post-positive selection CD4 SP thymocytes migrate from cortex to medulla, changing from CCR7lowCD24high to CCR7highCD24high and finally CCR7highCD24low before thymic egress. We saw a block at the CCR7lowCD24high stage in CD4-cre Runx1 cKO CD4 SP thymocytes. Additionally, other maturation markers including CD55 and Qa2 were also downregulated. Thus, Runx1 is required for thymic maturation of CD4 SP thymocytes.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call