Back to Search Start Over

A Bcl11bN797K variant isolated from an immunodeficient patient inhibits early thymocyte development in mice.

Authors :
Kazuaki Matsumoto
Kazuki Okuyama
Sidwell, Tom
Motoi Yamashita
Takaho Endo
Naoko Satoh-Takayama
Hiroshi Ohno
Tomohiro Morio
Rothenberg, Ellen V.
Ichiro Taniuchi
Source :
Frontiers in Immunology; 2024, p1-14, 14p
Publication Year :
2024

Abstract

BCL11B is a transcription factor with six C<subscript>2</subscript>H<subscript>2</subscript>-type zinc-finger domains. Studies in mice have shown that Bcl11b plays essential roles in T cell development. Several germline heterozygous BCL11B variants have been identified in human patients with inborn errors of immunity (IEI) patients. Among these, two de novo mis-sense variants cause asparagine (N) to lysine (K) replacement in distinct zincfinger domains, BCL11B<superscript>N441K</superscript> and BCL11B<superscript>N807K</superscript>. To elucidate the pathogenesis of the BCL11B<superscript>N807K</superscript> variant, we generated a mouse model of BCL11B<superscript>N807K</superscript> by inserting the corresponding mutation, Bcl11b<superscript>N797K</superscript>, into the mouse genome. In Bcl11b<superscript>+/N797K</superscript> mice, the proportion of immature CD4<superscript>-</superscript>CD8<superscript>+</superscript> single-positive thymocytes was increased, and the development of invariant natural killer cells was severely inhibited in a T-cell-intrinsic manner. Under competitive conditions, γδT cell development was outcompeted by control cells. Bcl11b<superscript>N797K/N797K</superscript> mice died within one day of birth. Recipient mice reconstituted with Bcl11b<superscript>N797K/N797K</superscript> fetal liver cells nearly lacked CD4<superscript>+</superscript>CD8<superscript>+</superscript> double-positive thymocytes, which was consistent with the lack of their emergence in culture from Bcl11b<superscript>N797K/N797K</superscript> fetal liver progenitors. Interestingly, Bcl11b<superscript>N797K/N797K</superscript> progenitors gave rise to aberrant c-Kit<superscript>+</superscript> and CD44<superscript>+</superscript> cells both in vivo and in vitro. The increase in the proportion of immature CD8 single-positive thymocytes in the Bcl11b<superscript>N797K</superscript> mutants is caused, in part, by the inefficient activation of the Cd4 gene due to the attenuated function of the two Cd4 enhancers via distinct mechanisms. Therefore, we conclude that immunodeficient patient-derived Bcl11b<superscript>N797K</superscript> mutant mice elucidated a novel role for Bcl11b in driving the appropriate transition of CD4<superscript>-</superscript>CD8<superscript>-</superscript> into CD4<superscript>+</superscript>CD8<superscript>+</superscript> thymocytes. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
16643224
Database :
Complementary Index
Journal :
Frontiers in Immunology
Publication Type :
Academic Journal
Accession number :
176113817
Full Text :
https://doi.org/10.3389/fimmu.2024.1363704