Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Sci Adv ; 10(11): eadj2802, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38489359

ABSTRACT

Development of T cells is controlled by the signal strength of the TCR. The scaffold protein kinase D-interacting substrate of 220 kilodalton (Kidins220) binds to the TCR; however, its role in T cell development was unknown. Here, we show that T cell-specific Kidins220 knockout (T-KO) mice have strongly reduced invariant natural killer T (iNKT) cell numbers and modest decreases in conventional T cells. Enhanced apoptosis due to increased TCR signaling in T-KO iNKT thymocytes of developmental stages 2 and 3 shows that Kidins220 down-regulates TCR signaling at these stages. scRNA-seq indicated that the transcription factor Aiolos is down-regulated in Kidins220-deficient iNKT cells. Analysis of an Aiolos KO demonstrated that Aiolos is a downstream effector of Kidins220 during iNKT cell development. In the periphery, T-KO iNKT cells show reduced TCR signaling upon stimulation with α-galactosylceramide, suggesting that Kidins220 promotes TCR signaling in peripheral iNKT cells. Thus, Kidins220 reduces or promotes signaling dependent on the iNKT cell developmental stage.


Subject(s)
Ikaros Transcription Factor , Membrane Proteins , Natural Killer T-Cells , Thymus Gland , Animals , Mice , Cell Differentiation , Gene Expression Regulation , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Membrane Proteins/metabolism , Ikaros Transcription Factor/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
2.
Blood ; 139(24): 3505-3518, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35316324

ABSTRACT

Oncogenic alterations underlying B-cell acute lymphoblastic leukemia (B-ALL) in adults remain incompletely elucidated. To uncover novel oncogenic drivers, we performed RNA sequencing and whole-genome analyses in a large cohort of unresolved B-ALL. We identified a novel subtype characterized by a distinct gene expression signature and the unique association of 2 genomic microdeletions. The 17q21.31 microdeletion resulted in a UBTF::ATXN7L3 fusion transcript encoding a chimeric protein. The 13q12.2 deletion resulted in monoallelic ectopic expression of the homeobox transcription factor CDX2, located 138 kb in cis from the deletion. Using 4C-sequencing and CRISPR interference experiments, we elucidated the mechanism of CDX2 cis-deregulation, involving PAN3 enhancer hijacking. CDX2/UBTF ALL (n = 26) harbored a distinct pattern of additional alterations including 1q gain and CXCR4 activating mutations. Within adult patients with Ph- B-ALL enrolled in GRAALL trials, patients with CDX2/UBTF ALL (n = 17/723, 2.4%) were young (median age, 31 years) and dramatically enriched in females (male/female ratio, 0.2, P = .002). They commonly presented with a pro-B phenotype ALL and moderate blast cell infiltration. They had poor response to treatment including a higher risk of failure to first induction course (19% vs 3%, P = .017) and higher post-induction minimal residual disease (MRD) levels (MRD ≥ 10-4, 93% vs 46%, P < .001). This early resistance to treatment translated into a significantly higher cumulative incidence of relapse (75.0% vs 32.4%, P = .004) in univariate and multivariate analyses. In conclusion, we discovered a novel B-ALL entity defined by the unique combination of CDX2 cis-deregulation and UBTF::ATXN7L3 fusion, representing a high-risk disease in young adults.


Subject(s)
CDX2 Transcription Factor , Pol1 Transcription Initiation Complex Proteins , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Transcription Factors , Adult , CDX2 Transcription Factor/genetics , Female , Genes, Homeobox , Humans , Male , Neoplasm, Residual/genetics , Oncogene Proteins, Fusion , Pol1 Transcription Initiation Complex Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Transcription Factors/genetics
4.
PLoS One ; 16(1): e0246570, 2021.
Article in English | MEDLINE | ID: mdl-33513189

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0242211.].

5.
PLoS One ; 15(11): e0242211, 2020.
Article in English | MEDLINE | ID: mdl-33180866

ABSTRACT

The IKZF1 gene, which encodes the Ikaros transcription factor, is frequently deleted or mutated in patients with B-cell precursor acute lymphoblastic leukemias that express oncogenes, like BCR-ABL, which activate the JAK-STAT5 pathway. Ikaros functionally antagonizes the transcriptional programs downstream of IL-7/STAT5 during B cell development, as well as STAT5 activity in leukemic cells. However, the mechanisms by which Ikaros interferes with STAT5 function is unknown. We studied the genomic distribution of Ikaros and STAT5 on chromatin in a murine pre-B cell line, and found that both proteins colocalize on >60% of STAT5 target regions. Strikingly, Ikaros activity leads to widespread loss of STAT5 binding at most of its genomic targets within two hours of Ikaros induction, suggesting a direct mechanism. Ikaros did not alter the level of total or phosphorylated STAT5 proteins, nor did it associate with STAT5. Using sequences from the Cish, Socs2 and Bcl6 genes that Ikaros and STAT5 target, we show that both proteins bind overlapping sequences at GGAA motifs. Our results demonstrate that Ikaros antagonizes STAT5 DNA binding, in part by competing for common target sequences. Our study has implications for understanding the functions of Ikaros and STAT5 in B cell development and transformation.


Subject(s)
DNA/metabolism , Ikaros Transcription Factor/metabolism , Precursor Cells, B-Lymphoid/metabolism , STAT5 Transcription Factor/metabolism , Animals , Base Sequence , Cell Line , Chromatin/metabolism , DNA/chemistry , Ikaros Transcription Factor/deficiency , Ikaros Transcription Factor/genetics , Interleukin-17/pharmacology , Mice , Mice, Knockout , Phosphorylation , Precursor Cells, B-Lymphoid/cytology , Protein Binding , STAT5 Transcription Factor/genetics , Suppressor of Cytokine Signaling Proteins/chemistry , Suppressor of Cytokine Signaling Proteins/metabolism , Up-Regulation/drug effects
7.
Curr Opin Immunol ; 51: 14-23, 2018 04.
Article in English | MEDLINE | ID: mdl-29278858

ABSTRACT

The IKZF family of transcription factors are essential regulators of lymphopoiesis. Ikaros, Helios, Aiolos and Eos function as transcriptional repressors and activators during T and B cell differentiation and in mature cell function, depending on the stage of development and/or cell type. Their potential mechanisms of action are varied. Ikaros family proteins partner with multiple complexes, including NuRD, PRC2 and transcription elongation factors, to modulate gene expression and the chromatin state. In humans, mutations in the IKZF genes are associated with B cell deficiency, leukemias and autoimmunity. In this review, we focus on the function of Ikaros family proteins in early T and B lymphocyte development, and discuss the molecular and physiological activities of this family.


Subject(s)
Cell Differentiation/genetics , Ikaros Transcription Factor/genetics , Lymphocytes/cytology , Lymphocytes/metabolism , Multigene Family , Animals , Cell Lineage , Gene Expression Regulation , Humans , Ikaros Transcription Factor/metabolism , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes/immunology , Lymphopoiesis/genetics , Mice , Signal Transduction
8.
J Biol Chem ; 291(17): 9073-86, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26841869

ABSTRACT

B1 B cells secrete most of the circulating natural antibodies and are considered key effector cells of the innate immune response. However, B1 cell-associated antibodies often cross-react with self-antigens, which leads to autoimmunity, and B1 cells have been implicated in cancer. How B1 cell activity is regulated remains unclear. We show that the Ikaros transcription factor is a major negative regulator of B1 cell development and function. Using conditional knock-out mouse models to delete Ikaros at different locations, we show that Ikaros-deficient mice exhibit specific and significant increases in splenic and bone marrow B1 cell numbers, and that the B1 progenitor cell pool is increased ∼10-fold in the bone marrow. Ikaros-null B1 cells resemble WT B1 cells at the molecular and cellular levels, but show a down-regulation of signaling components important for inhibiting proliferation and immunoglobulin production. Ikaros-null B1 cells hyper-react to TLR4 stimulation and secrete high amounts of IgM autoantibodies. These results indicate that Ikaros is required to limit B1 cell homeostasis in the adult.


Subject(s)
Autoantibodies/immunology , B-Lymphocyte Subsets/immunology , Bone Marrow Cells/immunology , Ikaros Transcription Factor/immunology , Immunoglobulin M/immunology , Precursor Cells, B-Lymphoid/immunology , Animals , Ikaros Transcription Factor/genetics , Mice , Mice, Knockout , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
9.
Biochem Biophys Res Commun ; 470(3): 714-720, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26775846

ABSTRACT

The Ikaros transcription factor is essential for early B cell development, but its effect on mature B cells is debated. We show that Ikaros is required to limit the response of naive splenic B cells to B cell receptor signals. Ikaros deficient follicular B cells grow larger and enter cell cycle faster after anti-IgM stimulation. Unstimulated mutant B cells show deregulation of positive and negative regulators of signal transduction at the mRNA level, and constitutive phosphorylation of ERK, p38, SYK, BTK, AKT and LYN. Stimulation results in enhanced and prolonged ERK and p38 phosphorylation, followed by hyper-proliferation. Pharmacological inhibition of ERK and p38 abrogates the increased proliferative response of Ikaros deficient cells. These results suggest that Ikaros functions as a negative regulator of follicular B cell activation.


Subject(s)
Ikaros Transcription Factor/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/physiology , Spleen/cytology , Animals , B-Lymphocytes , Cell Proliferation/physiology , Cells, Cultured , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL
10.
J Exp Med ; 210(13): 2823-32, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24297995

ABSTRACT

Pre-B cell receptor (pre-BCR) signaling and migration from IL-7-rich environments cooperate to drive pre-B cell differentiation via transcriptional programs that remain unclear. We show that the Ikaros transcription factor is required for the differentiation of large pre-B to small pre-B cells. Mice deleted for Ikaros in pro/pre-B cells show a complete block of differentiation at the fraction C' stage, and Ikaros-null pre-B cells cannot differentiate upon withdrawal of IL-7 in vitro. Restoration of Ikaros function rescues pre-B cell differentiation in vitro and in vivo and depends on DNA binding. Ikaros is required for the down-regulation of the pre-BCR, Igκ germline transcription, and Ig L chain recombination. Furthermore, Ikaros antagonizes the IL-7-dependent regulation of >3,000 genes, many of which are up- or down-regulated between fractions C' and D. Affected genes include those important for survival, metabolism, B cell signaling, and function, as well as transcriptional regulators like Ebf1, Pax5, and the Foxo1 family. Our data thus identify Ikaros as a central regulator of IL-7 signaling and pre-B cell development.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Ikaros Transcription Factor/metabolism , Interleukin-7/metabolism , Animals , Cell Separation , Down-Regulation , Flow Cytometry , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/genetics , Leukemia/genetics , Leukemia/metabolism , Mice , Mice, Knockout , Mutation , Phenotype , Recombination, Genetic , Retroviridae/genetics , Signal Transduction , Transcription, Genetic , Transcriptome , VDJ Recombinases/metabolism
11.
Proc Natl Acad Sci U S A ; 107(43): 18563-8, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20940318

ABSTRACT

Upon B-cell activation, the signaling subunits Ig-α and Ig-ß of the B-cell antigen receptor become phosphorylated not only on tyrosines but also on serine residues. Using a specific antibody, we show that serine 197 (S197) in the cytoplasmic tail of Ig-α is phosphorylated upon B-cell antigen receptor activation, and that this modification inhibits the signal output of the B-cell antigen receptor. Surprisingly, we found that the well-known protein tyrosine kinase Syk (spleen tyrosine kinase) phosphorylates S197 on Ig-α, thus not only activating but also inhibiting signaling from the B-cell antigen receptor. This finding identifies Syk as a dual-specificity kinase and establishes a previously unexplored paradigm for the self-regulation of biological signaling processes.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Amino Acid Sequence , Animals , CD79 Antigens/chemistry , CD79 Antigens/genetics , CD79 Antigens/metabolism , Cell Line , Drosophila , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Mutation , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, B-Cell/chemistry , Receptors, Antigen, B-Cell/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Signal Transduction , Structural Homology, Protein , Syk Kinase
SELECTION OF CITATIONS
SEARCH DETAIL