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.
iScience ; 26(3): 106234, 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36926655

ABSTRACT

In thymus, the ablation of T cell receptor (TCR)-activated transcription factor NFATc1 or its inducible isoforms during the double-negative (DN) stages of thymocyte development leads to a marked increase in γδ thymocytes whereas the development of αß thymocytes remains mostly unaffected. These γδ thymocytes are characterized by the upregulation of the promyelocytic leukemia zinc-finger factor (PLZF), the "master regulator" of natural killer T (NKT) cell development, and the acquisition of an NKT γδ cell phenotype with higher cell survival rates. The suppressive function of NFATc1 in NKT γδ cell formation critically depends on the remote enhancer E2, which is essential for the inducible expression of NFATc1 directed by its distal promoter P1. Thus, the enhancer deciphers a strong γδ TCR signal into the expression of inducible NFATc1 isoforms resulting in high levels of NFATc1 protein that are essential to control the numbers of NKT γδ cells.

2.
Cell Mol Immunol ; 16(5): 508-520, 2019 05.
Article in English | MEDLINE | ID: mdl-29907883

ABSTRACT

B cell development in bone marrow is a precisely regulated complex process. Through successive stages of differentiation, which are regulated by a multitude of signaling pathways and an array of lineage-specific transcription factors, the common lymphoid progenitors ultimately give rise to mature B cells. Similar to early thymocyte development in the thymus, early B cell development in bone marrow is critically dependent on IL-7 signaling. During this IL-7-dependent stage of differentiation, several transcription factors, such as E2A, EBF1, and Pax5, among others, play indispensable roles in B lineage specification and maintenance. Although recent studies have implicated several other transcription factors in B cell development, the role of NFATc1 in early B cell developmental stages is not known. Here, using multiple gene-manipulated mouse models and applying various experimental methods, we show that NFATc1 activity is vital for early B cell differentiation. Lack of NFATc1 activity in pro-B cells suppresses EBF1 expression, impairs immunoglobulin gene rearrangement, and thereby preBCR formation, resulting in defective B cell development. Overall, deficiency in NFATc1 activity arrested the pro-B cell transition to the pre-B cell stage, leading to severe B cell lymphopenia. Our findings suggest that, along with other transcription factors, NFATc1 is a critical component of the signaling mechanism that facilitates early B cell differentiation.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/immunology , NFATC Transcription Factors/metabolism , Precursor Cells, B-Lymphoid/immunology , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Interleukin-7/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/genetics , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism
3.
Oncotarget ; 9(11): 9632-9644, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29515759

ABSTRACT

The role of NFAT family transcription factors in erythropoiesis is so far unknown, although their involvement has been suggested previously. We have shown recently that Il2-/- mice develop severe anemia due to defects in KLF1 activity during BM erythropoiesis. Although, KLF1 activity is indispensable for erythropoiesis, the molecular details of Klf1 expression have not yet been elucidated. Here we show that an enhanced NFATc1 activity induced by increased integrin-cAMP signaling plays a critical role in the dysregulation of Klf1 expression and thereby cause anemia in Il2-/- mice. Interestingly, enhanced NFATc1 activity augmented apoptosis of immature erythrocytes in Il2-/- mice. On the other hand, ablation of NFATc1 activity enhanced differentiation of Ter119+ cells in BM. Restoring IL-2 signaling in Il2-/- mice reversed the increase in cAMP-NFAT signaling and facilitated normal erythropoiesis. Altogether, our study identified an NFAT-mediated negative signaling axis, manipulation of which could facilitate erythropoiesis and prevent anemia development.

4.
Oncotarget ; 8(18): 29625-29642, 2017 May 02.
Article in English | MEDLINE | ID: mdl-28415569

ABSTRACT

The role of IL-2 in HSC maintenance is unknown. Here we show that Il2-/- mice develop severe anomalies in HSC maintenance leading to defective hematopoiesis. Whereas, lack of IL-2 signaling was detrimental for lympho- and erythropoiesis, myelopoiesis was enhanced in Il2-/- mice. Investigation of the underlying mechanisms of dysregulated hematopoiesis in Il2-/- mice shows that the IL-2-Treg cell axis is indispensable for HSC maintenance and normal hematopoiesis. Lack of Treg activity resulted in increased IFN-γ production by activated T cells and an expansion of the HSCs in the bone marrow (BM). Though, restoring Treg population successfully rescued HSC maintenance in Il2-/- mice, preventing IFN-γ activity could do the same even in the absence of Treg cells. Our study suggests that equilibrium in IL-2 and IFN-γ activity is critical for steady state hematopoiesis, and in clinical conditions of BM failure, IL-2 or anti-IFN-γ treatment might help to restore hematopoiesis.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interleukin-2/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cell Self Renewal/genetics , Hematopoiesis/genetics , Hematopoiesis/immunology , Immunophenotyping , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Signal Transduction , T-Lymphocytes, Regulatory/immunology
5.
Oncotarget ; 7(21): 31014-28, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27105536

ABSTRACT

Small cell lung cancer (SCLC) is the most aggressive type of lung cancer with high mortality. One of the MYC family genes, MYC, MYCL or MYCN, is amplified in ~20% of the SCLCs; therefore, MYC proteins are potential therapeutic targets in SCLC patients. We investigated the therapeutic impact of Omomyc, a MYC dominant negative, in a panel of SCLC cell lines. Strikingly, Omomyc suppressed the growth of all tested cell lines by inducing cell cycle arrest and/or apoptosis. Induction of G1 arrest by Omomyc was found to be dependent on the activation of CDKN1A, in part, through the TP73 pathway. Our results strongly indicate that SCLC cells carrying amplification of MYC, MYCL or MYCN are addicted to MYC function, suggesting that MYC targeting would be an efficient therapeutic option for SCLC patients.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/therapy , Peptide Fragments/biosynthesis , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/biosynthesis , Retinoblastoma Binding Proteins/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/therapy , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Cycle Checkpoints/genetics , Cell Death/genetics , Cell Growth Processes/genetics , Gene Amplification , Gene Silencing , Genes, p53 , Genetic Therapy/methods , HEK293 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Targeted Therapy , Peptide Fragments/genetics , Proto-Oncogene Proteins c-myc/genetics , Retinoblastoma Binding Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...