Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 535039, 2021.
Article in English | MEDLINE | ID: mdl-33815354

ABSTRACT

The BTB zinc finger transcription factor MAZR (also known as PATZ1) controls, partially in synergy with the transcription factor Runx3, the development of CD8 lineage T cells. Here we explored the role of MAZR as well as combined activities of MAZR/Runx3 during cytotoxic T lymphocyte (CTL) and memory CD8+ T cell differentiation. In contrast to the essential role of Runx3 for CTL effector function, the deletion of MAZR had a mild effect on the generation of CTLs in vitro. However, a transcriptome analysis demonstrated that the combined deletion of MAZR and Runx3 resulted in much more widespread downregulation of CTL signature genes compared to single Runx3 deletion, indicating that MAZR partially compensates for loss of Runx3 in CTLs. Moreover, in line with the findings made in vitro, the analysis of CTL responses to LCMV infection revealed that MAZR and Runx3 cooperatively regulate the expression of CD8α, Granzyme B and perforin in vivo. Interestingly, while memory T cell differentiation is severely impaired in Runx3-deficient mice, the deletion of MAZR leads to an enlargement of the long-lived memory subset and also partially restored the differentiation defect caused by loss of Runx3. This indicates distinct functions of MAZR and Runx3 in the generation of memory T cell subsets, which is in contrast to their cooperative roles in CTLs. Together, our study demonstrates complex interplay between MAZR and Runx3 during CTL and memory T cell differentiation, and provides further insight into the molecular mechanisms underlying the establishment of CTL and memory T cell pools.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Core Binding Factor Alpha 3 Subunit/immunology , Immunologic Memory/immunology , Neoplasm Proteins/immunology , Repressor Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cell Differentiation/immunology , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation/immunology , Host-Pathogen Interactions/immunology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology
2.
Cell Rep ; 29(13): 4447-4459.e6, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875552

ABSTRACT

Forkhead box protein P3+ (FOXP3+) regulatory T cells (Treg cells) play a key role in maintaining tolerance and immune homeostasis. Here, we report that a T cell-specific deletion of the transcription factor MAZR (also known as PATZ1) leads to an increased frequency of Treg cells, while enforced MAZR expression impairs Treg cell differentiation. Further, MAZR expression levels are progressively downregulated during thymic Treg cell development and during in-vitro-induced human Treg cell differentiation, suggesting that MAZR protein levels are critical for controlling Treg cell development. However, MAZR-deficient Treg cells show only minor transcriptional changes ex vivo, indicating that MAZR is not essential for establishing the transcriptional program of peripheral Treg cells. Finally, the loss of MAZR reduces the clinical score in dextran-sodium sulfate (DSS)-induced colitis, suggesting that MAZR activity in T cells controls the extent of intestinal inflammation. Together, these data indicate that MAZR is part of a Treg cell-intrinsic transcriptional network that modulates Treg cell development.


Subject(s)
Forkhead Transcription Factors/metabolism , Kruppel-Like Transcription Factors/metabolism , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Differentiation , Colitis/immunology , Dextran Sulfate , Humans , Mice, Knockout , Thymus Gland/cytology , Transcription, Genetic
3.
Cell Mol Life Sci ; 76(21): 4391-4404, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31065747

ABSTRACT

Invariant natural killer T (iNKT) cells represent a subgroup of innate-like T cells and play an important role in immune responses against certain pathogens. In addition, they have been linked to autoimmunity and antitumor immunity. iNKT cells consist of several subsets with distinct functions; however, the transcriptional networks controlling iNKT subset differentiation are still not fully characterized. Myc-associated zinc-finger-related factor (MAZR, also known as PATZ1) is an essential transcription factor for CD8+ lineage differentiation of conventional T cells. Here, we show that MAZR plays an important role in iNKT cells. T-cell lineage-specific deletion of MAZR resulted in an iNKT cell-intrinsic defect that led to an increase in iNKT2 cell numbers, concurrent with a reduction in iNKT1 and iNKT17 cells. Consistent with the alteration in the subset distribution, deletion of MAZR also resulted in an increase in the percentage of IL-4-producing cells. Moreover, MAZR-deficient iNKT cells displayed an enhanced expression of Erg2 and ThPOK, key factors for iNKT cell generation and subset differentiation, indicating that MAZR controls iNKT cell development through fine-tuning of their expression levels. Taken together, our study identified MAZR as an essential transcription factor regulating iNKT cell subset differentiation and effector function.


Subject(s)
Cell Differentiation/genetics , Natural Killer T-Cells/physiology , Neoplasm Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Regulation , Lymphocyte Subsets/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Natural Killer T-Cells/classification , Transcription Factors/physiology , Zinc Fingers/physiology
4.
Front Immunol ; 10: 409, 2019.
Article in English | MEDLINE | ID: mdl-30915074

ABSTRACT

CD8 expression in T lymphocytes is tightly regulated by the activity of at least six Cd8 enhancers (E8I-E8VI), however their complex developmental stage-, subset-, and lineage-specific interplays are incompletely understood. Here we analyzed ATAC-seq data on the Immunological Genome Project database and identified a similar developmental regulation of chromatin accessibility of a subregion of E8I, designated E8I-core, and of E8VI. Loss of E8I-core led to a similar reduction in CD8 expression in naïve CD8+ T cells and in IELs as observed in E8I-/- mice, demonstrating that we identified the core enhancer region of E8I. While E8VI-/- mice displayed a mild reduction in CD8 expression levels on CD8SP thymocytes and peripheral CD8+ T cells, CD8 levels were further reduced upon combined deletion of E8I-core and E8VI. Moreover, activated E8I-core-/-E8VI-/- CD8+ T cells lost CD8 expression to a greater degree than E8I-core-/- and E8VI-/- CD8+ T cells, suggesting that the combined activity of both enhancers is required for establishment and maintenance of CD8 expression before and after TCR activation. Finally, we observed a severe reduction of CD4 CTLs among the TCRß+CD4+ IEL population in E8I-core-/- but not E8VI-/- mice. Such a reduction was not observed in Cd8a-/- mice, indicating that E8I-core controls the generation of CD4 CTLs independently of its role in Cd8a gene regulation. Further, the combined deletion of E8I-core and E8VI restored CD4 CTL subsets, suggesting an antagonistic function of E8VI in the generation of CD4 CTLs. Together, our study demonstrates a complex utilization and interplay of E8I-core and E8VI in regulating CD8 expression in cytotoxic lineage T cells and in IELs. Moreover, we revealed a novel E8I-mediated regulatory mechanism controlling the generation of intestinal CD4 CTLs.


Subject(s)
CD8 Antigens/biosynthesis , Gene Expression Regulation/immunology , Intestinal Mucosa/immunology , Intraepithelial Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Intraepithelial Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Cytotoxic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...