The Ccr7, Slfn1, and Mapk11 genes were weakly induced in mature c

The Ccr7, Slfn1, and Mapk11 genes were weakly induced in mature cell populations from only one of the mutant mice, but remained at background levels across all populations in the samples derived from the second mouse. This observation suggests that some gene-specific variability exists across mutants in their ability to activate genes induced during positive Erlotinib selection, and is in agreement

with the previous results demonstrating impaired expression of genes associated with positive selection in DP cells from Bcl11b-deficient thymocytes 26. Collectively, these analyses indicate that the premature expression of SP-associated genes in Bcl11bdp−/− DP cells reflects gene-specific dysregulation in cells that have not undergone positive selection. To determine if Bcl11b directly controls the expression of some dysregulated genes, we mapped Bcl11b binding BAY 57-1293 cost to regulatory sequences by performing ChIP-seq experiments on chromatin immunoprecipitated from WT

thymocytes (a full bioinfomatic analysis of these data will be published elsewhere). We found that Bcl11b was present at multiple, specific regions in most of the genes that were dysregulated in our transcriptomic analyses (see Fig. 8 for a representative selection of binding profiles). Of particular interest, Bcl11b bound to several regions within the Zbtb7b locus, including the distal regulatory element, which has been reported to be the target of TCR signal(s) responsible for CD4 lineage commitment Ribonucleotide reductase 42. These data indicate that Bcl11b likely acts directly in DP cells to prevent the premature expression of genes encoding critical regulators of the SP differentiation programs. The results presented herein further establish Bcl11b as a key regulator of cellular differentiation in the αβ T-cell lineage. Bcl11b plays a critical role in at least two stages of T-cell development: progression of DN to DP cells, and differentiation of

DP cells into CD4+ and CD8+ SP cells, and NKT cells. Although our results confirm the previous results with respect to the early T-cell block resulting from the germline deletion of Bcl11b 25, and the block of SP T-cell differentiation of CD4-Cre-deleted mice 26, our studies also bring new and important insights. Specifically, we show that Bcl11b is: (i) absolutely and intrinsically required in DP thymocytes for canonical NKT cell development, (ii) required for the correct expression of approximately 1000 genes in DP cells, acting as a bifunctional transcriptional regulatory protein, and (iii) required in CD3lo DP cells to prevent the premature expression of a large number of SP-specific genes, including the key regulators Zbtb7b and Runx3.

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>