Skip to main content

Restriction of Nonpermissive RUNX3 Protein Expression in T Lymphocytes by the Kozak Sequence.


AUTHORS

Kim BByungil , Sasaki Y Yo , Egawa T Takeshi . Journal of immunology (Baltimore, Md. : 1950). 2015 8 15; 195(4). 1517-23

ABSTRACT

The transcription factor Runx3 promotes differentiation of naive CD4(+) T cells into type-1 effector T (TH1) cells at the expense of TH2. TH1 cells as well as CD8(+) T cells express a subset-specific Runx3 transcript from a distal promoter, which is necessary for high protein expression. However, all T cell subsets, including naive CD4(+) T cells and TH2 cells, express a distinct transcript of Runx3 that is derived from a proximal promoter and that produces functional protein in neurons. Therefore, accumulation of RUNX3 protein generated from the proximal transcript needs to be repressed at the posttranscriptional level to preserve CD4(+) T cell capability of differentiating into TH2 cells. In this article, we show that expression of RUNX3 protein from the proximal Runx3 transcript is blocked at the level of translational initiation in T cells. A coding sequence for the proximal Runx3 mRNA is preceded by a nonoptimal context sequence for translational initiation, known as the Kozak sequence, and thus generates protein at low efficiencies and with multiple alternative translational initiations. Editing the endogenous initiation context to an "optimal" Kozak sequence in a human T cell line resulted in enhanced translation of a single RUNX3 protein derived from the proximal transcript. Furthermore, RUNX3 protein represses transcription from the proximal promoter in T cells. These results suggest that nonpermissive expression of RUNX3 protein is restricted at the translational level, and that the repression is further enforced by a transcriptional regulation for maintenance of diverse developmental plasticity of T cells for different effector subsets.


The transcription factor Runx3 promotes differentiation of naive CD4(+) T cells into type-1 effector T (TH1) cells at the expense of TH2. TH1 cells as well as CD8(+) T cells express a subset-specific Runx3 transcript from a distal promoter, which is necessary for high protein expression. However, all T cell subsets, including naive CD4(+) T cells and TH2 cells, express a distinct transcript of Runx3 that is derived from a proximal promoter and that produces functional protein in neurons. Therefore, accumulation of RUNX3 protein generated from the proximal transcript needs to be repressed at the posttranscriptional level to preserve CD4(+) T cell capability of differentiating into TH2 cells. In this article, we show that expression of RUNX3 protein from the proximal Runx3 transcript is blocked at the level of translational initiation in T cells. A coding sequence for the proximal Runx3 mRNA is preceded by a nonoptimal context sequence for translational initiation, known as the Kozak sequence, and thus generates protein at low efficiencies and with multiple alternative translational initiations. Editing the endogenous initiation context to an "optimal" Kozak sequence in a human T cell line resulted in enhanced translation of a single RUNX3 protein derived from the proximal transcript. Furthermore, RUNX3 protein represses transcription from the proximal promoter in T cells. These results suggest that nonpermissive expression of RUNX3 protein is restricted at the translational level, and that the repression is further enforced by a transcriptional regulation for maintenance of diverse developmental plasticity of T cells for different effector subsets.


Leave a Response