Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Immunol ; 68(2 Pt B): 484-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26490636

ABSTRACT

Natural killer T (NKT) cells develop from common CD4(+) CD8(+) thymocyte precursors. Transcriptional programs that regulate the development of NKT cells in the thymus development remain to be fully delineated. Here, we demonstrate a cell-intrinsic requirement for transcription factors TCF1 and LEF1 for the development of all subsets of NKT cells. Conditional deletion of TCF1 alone results in a substantial reduction in NKT cells. The remaining NKT cells are eliminated when TCF1 and LEF1 are both deleted. These data reveal an essential role for TCF1 and LEF1 in development of NKT cells.


Subject(s)
Cell Differentiation/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Natural Killer T-Cells/cytology , Animals , Cell Differentiation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Thymocytes/cytology
2.
BMC Immunol ; 16: 62, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26482437

ABSTRACT

BACKGROUND: Invariant Natural Killer T (iNKT) cells have been implicated in lung inflammation in humans and also shown to be a key cell type in inducing allergic lung inflammation in mouse models. iNKT cells differentiate and acquire functional characteristics during development in the thymus. However, the correlation between development of iNKT cells in the thymus and role in lung inflammation remains unknown. In addition, transcriptional control of differentiation of iNKT cells into iNKT cell effector subsets in the thymus during development is also unclear. In this report we show that ß-catenin dependent mechanisms direct differentiation of iNKT2 and iNKT17 subsets but not iNKT1 cells. METHODS: To study the role for ß-catenin in lung inflammation we utilize mice with conditional deletion and enforced expression of ß-catenin in a well-established mouse model for IL-25-dependen lung inflammation. RESULTS: Specifically, we demonstrate that conditional deletion of ß-catenin permitted development of mature iNKT1 cells while impeding maturation of iNKT2 and 17 cells. A role for ß-catenin expression in promoting iNKT2 and iNKT17 subsets was confirmed when we noted that enforced transgenic expression of ß-catenin in iNKT cell precursors enhanced the frequency and number of iNKT2 and iNKT17 cells at the cost of iNKT1 cells. This effect of expression of ß-catenin in iNKT cell precursors was cell autonomous. Furthermore, iNKT2 cells acquired greater capability to produce type-2 cytokines when ß-catenin expression was enhanced. DISCUSSION: This report shows that ß-catenin deficiency resulted in a profound decrease in iNKT2 and iNKT17 subsets of iNKT cells whereas iNKT1 cells developed normally. By contrast, enforced expression of ß-catenin promoted the development of iNKT2 and iNKT17 cells. It was important to note that the majority of iNKT cells in the thymus of C57BL/6 mice were iNKT1 cells and enforced expression of ß-catenin altered the pattern to iNKT2 and iNKT17 cells suggesting that ß-catenin may be a major factor in the distinct pathways that critically direct differentiation of iNKT effector subsets. CONCLUSIONS: Thus, we demonstrate that ß-catenin expression in iNKT cell precursors promotes differentiation toward iNKT2 and iNKT17 effector subsets and supports enhanced capacity to produce type 2 and 17 cytokines which in turn augment lung inflammation in mice.


Subject(s)
Cell Differentiation , Interleukin-17/metabolism , Natural Killer T-Cells/immunology , Pneumonia/immunology , Pneumonia/pathology , beta Catenin/metabolism , Animals , Bronchial Hyperreactivity/complications , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/complications
3.
Nat Immunol ; 15(7): 646-656, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24836425

ABSTRACT

The transcription factors TCF-1 and LEF-1 are essential for early T cell development, but their roles beyond the CD4(+)CD8(+) double-positive (DP) stage are unknown. By specific ablation of these factors in DP thymocytes, we demonstrated that deficiency in TCF-1 and LEF-1 diminished the output of CD4(+) T cells and redirected CD4(+) T cells to a CD8(+) T cell fate. The role of TCF-1 and LEF-1 in the CD4-versus-CD8 lineage 'choice' was mediated in part by direct positive regulation of the transcription factor Th-POK. Furthermore, loss of TCF-1 and LEF-1 unexpectedly caused derepression of CD4 expression in T cells committed to the CD8(+) lineage without affecting the expression of Runx transcription factors. Instead, TCF-1 physically interacted with Runx3 to cooperatively silence Cd4. Thus, TCF-1 and LEF-1 adopted distinct genetic 'wiring' to promote the CD4(+) T cell fate and establish CD8(+) T cell identity.


Subject(s)
CD4 Antigens/physiology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Core Binding Factor Alpha 3 Subunit/physiology , Lymphoid Enhancer-Binding Factor 1/physiology , T Cell Transcription Factor 1/physiology , Transcription Factors/physiology , Animals , Cell Lineage , Female , Hepatocyte Nuclear Factor 1-alpha , Male , Mice
4.
Immunol Res ; 59(1-3): 45-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24847765

ABSTRACT

Transcription factors have recurring roles during T cell development and activation. Tcf1 and Lef1 are known to be essential for early stages of thymocyte maturation. Recent research has revealed several novel aspects of their functionality. Tcf1 is induced at the very earliest step of specifying hematopoietic progenitors to the T cell lineage as a key target gene downstream of Notch activation. In addition to promoting maturation of T-lineage-committed thymocytes, Tcf1 functions as a tumor suppressor in developing thymocytes, and this is mediated, paradoxically, by restraining Lef1 expression. After positive selection, Tcf1 and Lef1 act together to direct CD4(+)CD8(+) double positive thymocytes to a CD4(+) T cell fate. Although not required for CD8(+) T cell differentiation, Tcf1 and Lef1 cooperate with Runx factors to achieve stable silencing of the Cd4 gene in CD8(+) T cells. Tcf1 is also found to have versatile roles in innate immune cells, which partly mirror its functions in mature T helper cells. Discrepancy in requirements of Tcf1/Lef1 and ß-catenin in T cells has been a long-standing enigma. We will review other protein factors interacting with Tcf1 and Lef1 and discuss their regulatory roles independent of ß-catenin.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Immunity, Innate/physiology , Lymphoid Enhancer-Binding Factor 1/immunology , Animals , CD4 Antigens/immunology , Core Binding Factor alpha Subunits/immunology , Humans , Portraits as Topic , Thymocytes/immunology , beta Catenin/immunology
5.
Immunity ; 37(5): 813-26, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23103132

ABSTRACT

The TCF-1 and LEF-1 transcription factors are known to play critical roles in normal thymocyte development. Unexpectedly, we found that TCF-1-deficient (Tcf7(-/-)) mice developed aggressive T cell malignancy, resembling human T cell acute lymphoblastic leukemia (T-ALL). LEF-1 was aberrantly upregulated in premalignant Tcf7(-/-) early thymocytes and lymphoma cells. We further demonstrated that TCF-1 directly repressed LEF-1 expression in early thymocytes and that conditional inactivation of Lef1 greatly delayed or prevented T cell malignancy in Tcf7(-/-) mice. In human T-ALLs, an early thymic progenitor (ETP) subtype was associated with diminished TCF7 expression, and two of the ETP-ALL cases harbored TCF7 gene deletions. We also showed that TCF-1 and LEF-1 were dispensable for T cell lineage commitment but instead were required for early thymocytes to mature beyond the CD4(-)CD8(-) stage. TCF-1 thus has dual roles, i.e., acting cooperatively with LEF-1 to promote thymocyte maturation while restraining LEF-1 expression to prevent malignant transformation of developing thymocytes.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Animals , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Hepatocyte Nuclear Factor 1-alpha , Humans , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/immunology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Mice , Mice, Inbred C57BL , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Notch/genetics , Receptors, Notch/metabolism , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/immunology , T-Lymphocytes/pathology , Thymocytes/metabolism , Thymocytes/pathology , Transcription Factors/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...