Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










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.
Cells ; 11(13)2022 06 23.
Article in English | MEDLINE | ID: mdl-35805096

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) maintenance and the differentiation of various lineages is a highly complex but precisely regulated process. Multiple signaling pathways and an array of transcription factors influence HSPC maintenance and the differentiation of individual lineages to constitute a functional hematopoietic system. Nuclear factor of activated T cell (NFAT) family transcription factors have been studied in the context of development and function of multiple mature hematopoietic lineage cells. However, until now their contribution in HSPC physiology and HSPC differentiation to multiple hematopoietic lineages has remained poorly understood. Here, we show that NFAT proteins, specifically NFATc1, play an indispensable role in the maintenance of HSPCs. In the absence of NFATc1, very few HSPCs develop in the bone marrow, which are functionally defective. In addition to HSPC maintenance, NFATc1 also critically regulates differentiation of lymphoid, myeloid, and erythroid lineage cells from HSPCs. Deficiency of NFATc1 strongly impaired, while enhanced NFATc1 activity augmented, the differentiation of these lineages, which further attested to the vital involvement of NFATc1 in regulating hematopoiesis. Hematopoietic defects due to lack of NFATc1 activity can lead to severe pathologies such as lymphopenia, myelopenia, and a drastically reduced lifespan underlining the critical role NFATc1 plays in HSPC maintenance and in the differentaion of various lineages. Our findings suggest that NFATc1 is a critical component of the myriad signaling and transcriptional regulators that are essential to maintain normal hematopoiesis.


Subject(s)
Bone Marrow Cells , Hematopoietic Stem Cells , Cell Differentiation , Cell Proliferation , Hematopoietic Stem Cells/metabolism , Transcription Factors/metabolism
3.
Front Immunol ; 12: 780727, 2021.
Article in English | MEDLINE | ID: mdl-34956208

ABSTRACT

The skin protects the human body against dehydration and harmful challenges. Keratinocytes (KCs) are the most abundant epidermal cells, and it is anticipated that KC-mediated transport of Na+ ions creates a physiological barrier of high osmolality against the external environment. Here, we studied the role of NFAT5, a transcription factor whose activity is controlled by osmotic stress in KCs. Cultured KCs from adult mice were found to secrete more than 300 proteins, and upon NFAT5 ablation, the secretion of several matrix proteinases, including metalloproteinase-3 (Mmp3) and kallikrein-related peptidase 7 (Klk7), was markedly enhanced. An increase in Mmp3 and Klk7 RNA levels was also detected in transcriptomes of Nfat5-/- KCs, along with increases of numerous members of the 'Epidermal Differentiation Complex' (EDC), such as small proline-rich (Sprr) and S100 proteins. NFAT5 and Mmp3 as well as NFAT5 and Klk7 are co-expressed in the basal KCs of fetal and adult epidermis but not in basal KCs of newborn (NB) mice. The poor NFAT5 expression in NB KCs is correlated with a strong increase in Mmp3 and Klk7 expression in KCs of NB mice. These data suggests that, along with the fragile epidermis of adult Nfat5-/- mice, NFAT5 keeps in check the expression of matrix proteases in epidermis. The NFAT5-mediated control of matrix proteases in epidermis contributes to the manifold changes in skin development in embryos before and during birth, and to the integrity of epidermis in adults.


Subject(s)
Epidermis/anatomy & histology , Epidermis/metabolism , Keratinocytes/metabolism , NFATC Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Front Immunol ; 12: 791100, 2021.
Article in English | MEDLINE | ID: mdl-35069572

ABSTRACT

CD4+CXCR5+Foxp3+ T-follicular regulatory (TFR) cells control the germinal center responses. Like T-follicular helper cells, they express high levels of Nuclear Factor of Activated T-cells c1, predominantly its short isoform NFATc1/αA. Ablation of NFATc1 in Tregs prevents upregulation of CXCR5 and migration of TFR cells into B-cell follicles. By contrast, constitutive active NFATc1/αA defines the surface density of CXCR5, whose level determines how deep a TFR migrates into the GC and how effectively it controls antibody production. As one type of effector Treg, TFR cells express B lymphocyte-induced maturation protein-1 (Blimp-1). Blimp-1 can directly repress Cxcr5 and NFATc1/αA is necessary to overcome this Blimp-1-mediated repression. Interestingly, Blimp-1 even reinforces the recruitment of NFATc1 to Cxcr5 by protein-protein interaction and by those means cooperates with NFATc1 for Cxcr5 transactivation. On the contrary, Blimp-1 is necessary to counterbalance NFATc1/αA and preserve the Treg identity. This is because although NFATc1/αA strengthens the follicular development of Tregs, it bears the inherent risk of causing an ex-Treg phenotype.


Subject(s)
Cell Movement/immunology , Germinal Center/immunology , NFATC Transcription Factors/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Animals , Cell Movement/genetics , Mice , Mice, Transgenic , NFATC Transcription Factors/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics
5.
J Exp Med ; 218(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32986812

ABSTRACT

Posttranslational modification with SUMO is known to regulate the activity of transcription factors, but how SUMOylation of individual proteins might influence immunity is largely unexplored. The NFAT transcription factors play an essential role in antigen receptor-mediated gene regulation. SUMOylation of NFATc1 represses IL-2 in vitro, but its role in T cell-mediated immune responses in vivo is unclear. To this end, we generated a novel transgenic mouse in which SUMO modification of NFATc1 is prevented. Avoidance of NFATc1 SUMOylation ameliorated experimental autoimmune encephalomyelitis as well as graft-versus-host disease. Elevated IL-2 production in T cells promoted T reg expansion and suppressed autoreactive or alloreactive immune responses. Mechanistically, increased IL-2 secretion counteracted IL-17 and IFN-γ expression through STAT5 and Blimp-1 induction. Then, Blimp-1 repressed IL-2 itself, as well as the induced, proliferation-associated survival factor Bcl2A1. Collectively, these data demonstrate that prevention of NFATc1 SUMOylation fine-tunes T cell responses toward lasting tolerance. Thus, targeting NFATc1 SUMOylation presents a novel and promising strategy to treat T cell-mediated inflammatory diseases.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental/immunology , NFATC Transcription Factors/immunology , Sumoylation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Mice , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , NFATC Transcription Factors/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Sumoylation/genetics
6.
Front Immunol ; 10: 1917, 2019.
Article in English | MEDLINE | ID: mdl-31447864

ABSTRACT

The massive infiltration of lymphocytes into the skin is a hallmark of numerous human skin disorders. By co-culturing murine keratinocytes with splenic T cells we demonstrate here that T cells affect and control the synthesis and secretion of chemokines by keratinocytes. While pre-activated CD8+T cells induce the synthesis of CXCL9 and CXCL10 in keratinocytes and keep in check the synthesis of CXCL1, CXCL5, and CCL20, keratinocytes dampen the synthesis of CCL3 and CCL4 in pre-activated CD8+T cells. One key molecule is IFN-γ that is synthesized by CD8+T cells under the control of NFATc1 and NFATc2. CD8+T cells deficient for both NFAT factors are unable to induce CXCL9 and CXCL10 expression. In addition, CD8+T cells induced numerous type I IFN-inducible "defense genes" in keratinocytes encoding the PD1 and CD40 ligands, TNF-α and caspase-1. The enhanced expression of type I IFN-inducible genes resembles the gene expression pattern at the dermal/epidermal interface in lichen planus, an inflammatory T lymphocyte-driven skin disease, in which we detected the expression of CXCL10 in keratinocytes in close vicinity to the infiltration front of T cells. These data reflect the multifaceted interplay of lymphocytes with keratinocytes at the molecular level.


Subject(s)
Chemokines/immunology , Keratinocytes/immunology , T-Lymphocytes/immunology , Animals , CD40 Ligand/genetics , Cells, Cultured , Coculture Techniques , Female , Gene Expression Profiling , Male , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , Skin/immunology
7.
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
8.
Front Immunol ; 9: 1338, 2018.
Article in English | MEDLINE | ID: mdl-29946322

ABSTRACT

The immune suppressants cyclosporin A (CsA) and tacrolimus (FK506) are used worldwide in transplantation medicine to suppress graft rejection. Both CsA and FK506 inhibit the phosphatase calcineurin (CN) whose activity controls the immune receptor-mediated activation of lymphocytes. Downstream targets of CN in lymphocytes are the nuclear factors of activated T cells (NFATs). We show here that the activity of NFATc1, the most prominent NFAT factor in activated lymphocytes supports the acute rejection of heterotopic heart allografts. While ablation of NFATc1 in T cells prevented graft rejection, ectopic expression of inducible NFATc1/αA isoform led to rejection of heart allografts in recipient mice. Acceptance of transplanted hearts in mice bearing NFATc1-deficient T cells was accompanied by a reduction in number and cytotoxicity of graft infiltrating cells. In CD8+ T cells, NFATc1 controls numerous intracellular signaling pathways that lead to the metabolic switch to aerobic glycolysis and the expression of numerous lymphokines, chemokines, and their receptors, including Cxcr3 that supports the rejection of allogeneic heart transplants. These findings favors NFATc1 as a molecular target for the development of new strategies to control the cytotoxicity of T cells upon organ transplantation.

9.
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.

10.
Front Immunol ; 9: 32, 2018.
Article in English | MEDLINE | ID: mdl-29416540

ABSTRACT

In lymphocytes, immune receptor signals induce the rapid nuclear translocation of preformed cytosolic NFAT proteins. Along with co-stimulatory signals, persistent immune receptor signals lead to high levels of NFATc1/αA, a short NFATc1 isoform, in effector lymphocytes. Whereas NFATc1 is not expressed in plasma cells, in germinal centers numerous centrocytic B cells express nuclear NFATc1/αA. When overexpressed in chicken DT40 B cells or murine WEHI 231 B cells, NFATc1/αA suppressed their cell death induced by B cell receptor signals and affected the expression of genes controlling the germinal center reaction and plasma cell formation. Among those is the Prdm1 gene encoding Blimp-1, a key factor of plasma cell formation. By binding to a regulatory DNA element within exon 1 of the Prdm1 gene, NFATc1/αA suppresses Blimp-1 expression. Since expression of a constitutive active version of NFATc1/αA interfered with Prdm1 RNA expression, LPS-mediated differentiation of splenic B cells to plasmablasts in vitro and reduced immunoglobulin production in vivo, one may conclude that NFATc1/αA plays an important role in controlling plasmablast/plasma cell formation.


Subject(s)
B-Lymphocytes/cytology , NFATC Transcription Factors/physiology , Positive Regulatory Domain I-Binding Factor 1/physiology , Animals , Antibody Formation , B-Lymphocytes/physiology , Cell Differentiation , Cell Line , Chickens , Humans , Mice, Inbred C57BL , Protein Isoforms/physiology
11.
Immunity ; 47(4): 664-679.e6, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29030115

ABSTRACT

Store-operated Ca2+ entry (SOCE) is the main Ca2+ influx pathway in lymphocytes and is essential for T cell function and adaptive immunity. SOCE is mediated by Ca2+ release-activated Ca2+ (CRAC) channels that are activated by stromal interaction molecule (STIM) 1 and STIM2. SOCE regulates many Ca2+-dependent signaling molecules, including calcineurin, and inhibition of SOCE or calcineurin impairs antigen-dependent T cell proliferation. We here report that SOCE and calcineurin regulate cell cycle entry of quiescent T cells by controlling glycolysis and oxidative phosphorylation. SOCE directs the metabolic reprogramming of naive T cells by regulating the expression of glucose transporters, glycolytic enzymes, and metabolic regulators through the activation of nuclear factor of activated T cells (NFAT) and the PI3K-AKT kinase-mTOR nutrient-sensing pathway. We propose that SOCE controls a critical "metabolic checkpoint" at which T cells assess adequate nutrient supply to support clonal expansion and adaptive immune responses.


Subject(s)
Calcium Channels/immunology , Calcium Signaling/immunology , Calcium/immunology , T-Lymphocytes/immunology , Animals , Calcineurin/immunology , Calcineurin/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Cell Division/immunology , Cells, Cultured , Female , Glycolysis/immunology , HEK293 Cells , Humans , Immunoblotting , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/immunology , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/genetics , Stromal Interaction Molecule 2/immunology , Stromal Interaction Molecule 2/metabolism , T-Lymphocytes/metabolism
12.
Nat Commun ; 8(1): 511, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28894104

ABSTRACT

Cytotoxic T lymphocytes are effector CD8+ T cells that eradicate infected and malignant cells. Here we show that the transcription factor NFATc1 controls the cytotoxicity of mouse cytotoxic T lymphocytes. Activation of Nfatc1 -/- cytotoxic T lymphocytes showed a defective cytoskeleton organization and recruitment of cytosolic organelles to immunological synapses. These cells have reduced cytotoxicity against tumor cells, and mice with NFATc1-deficient T cells are defective in controlling Listeria infection. Transcriptome analysis shows diminished RNA levels of numerous genes in Nfatc1 -/- CD8+ T cells, including Tbx21, Gzmb and genes encoding cytokines and chemokines, and genes controlling glycolysis. Nfatc1 -/- , but not Nfatc2 -/- CD8+ T cells have an impaired metabolic switch to glycolysis, which can be restored by IL-2. Genome-wide ChIP-seq shows that NFATc1 binds many genes that control cytotoxic T lymphocyte activity. Together these data indicate that NFATc1 is an important regulator of cytotoxic T lymphocyte effector functions.NFAT nuclear translocation has been shown to be required for CD8+ T cell cytokine production in response to viral infection. Here the authors show NFATc1 controls the cytotoxicity and metabolic switching of activated CD8+ T cells required for optimal response to bacteria and tumor cells.


Subject(s)
Immunological Synapses/immunology , Lymphocyte Activation/genetics , NFATC Transcription Factors/genetics , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/genetics , Cytoskeleton/metabolism , Gene Expression Profiling , Gene Expression Regulation , Glycolysis/genetics , Granzymes/genetics , Immunological Synapses/metabolism , Listeriosis/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , NFATC Transcription Factors/immunology , Organelles/metabolism , T-Box Domain Proteins/genetics , T-Lymphocytes, Cytotoxic/metabolism
14.
Nat Commun ; 7: 11841, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27312418

ABSTRACT

NFATc1 plays a critical role in double-negative thymocyte survival and differentiation. However, the signals that regulate Nfatc1 expression are incompletely characterized. Here we show a developmental stage-specific differential expression pattern of Nfatc1 driven by the distal (P1) or proximal (P2) promoters in thymocytes. Whereas, preTCR-negative thymocytes exhibit only P2 promoter-derived Nfatc1ß expression, preTCR-positive thymocytes express both Nfatc1ß and P1 promoter-derived Nfatc1α transcripts. Inducing NFATc1α activity from P1 promoter in preTCR-negative thymocytes, in addition to the NFATc1ß from P2 promoter impairs thymocyte development resulting in severe T-cell lymphopenia. In addition, we show that NFATc1 activity suppresses the B-lineage potential of immature thymocytes, and consolidates their differentiation to T cells. Further, in the pTCR-positive DN3 cells, a threshold level of NFATc1 activity is vital in facilitating T-cell differentiation and to prevent Notch3-induced T-acute lymphoblastic leukaemia. Altogether, our results show NFATc1 activity is crucial in determining the T-cell fate of thymocytes.


Subject(s)
Lymphopenia/immunology , NFATC Transcription Factors/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptor, Notch3/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Cell Survival , Gene Expression Regulation, Developmental/immunology , Lymphopenia/genetics , Lymphopenia/pathology , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptor, Notch3/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction , T-Lymphocytes/cytology , Thymocytes/cytology , Thymus Gland/cytology , Thymus Gland/immunology
15.
Nat Commun ; 7: 11724, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27222343

ABSTRACT

Epicutaneous application of Aldara cream containing the TLR7 agonist imiquimod (IMQ) to mice induces skin inflammation that exhibits many aspects of psoriasis, an inflammatory human skin disease. Here we show that mice depleted of B cells or bearing interleukin (IL)-10-deficient B cells show a fulminant inflammation upon IMQ exposure, whereas ablation of NFATc1 in B cells results in a suppression of Aldara-induced inflammation. In vitro, IMQ induces the proliferation and IL-10 expression by B cells that is blocked by BCR signals inducing NFATc1. By binding to HDAC1, a transcriptional repressor, and to an intronic site of the Il10 gene, NFATc1 suppresses IL-10 expression that dampens the production of tumour necrosis factor-α and IL-17 by T cells. These data indicate a close link between NFATc1 and IL-10 expression in B cells and suggest NFATc1 and, in particular, its inducible short isoform, NFATc1/αA, as a potential target to treat human psoriasis.


Subject(s)
B-Lymphocytes/immunology , Interleukin-10/metabolism , NFATC Transcription Factors/physiology , Aminoquinolines/pharmacology , Animals , B-Lymphocytes/metabolism , Disease Models, Animal , Imiquimod , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Psoriasis/chemically induced , Psoriasis/genetics , Psoriasis/metabolism , Signal Transduction
16.
Eur J Immunol ; 44(11): 3392-402, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25179582

ABSTRACT

In peripheral lymphocytes, the transcription factors (TFs) NF-κB, NFAT, and AP-1 are the prime targets of signals that emerge from immune receptors. Upon activation, these TFs induce gene networks that orchestrate the growth, expansion, and effector function of peripheral lymphocytes. NFAT and NF-κB factors share several properties, such as a similar mode of induction and architecture in their DNA-binding domain, and there is a subgroup of κB-like DNA promoter motifs that are bound by both types of TFs. However, unlike NFAT and AP-1 factors that interact and collaborate in binding to DNA, NFAT, and NF-κB seem neither to interact nor to collaborate. We show here that NF-κB1/p50 and c-Rel, the most prominent NF-κB proteins in BCR-induced splenic B cells, control the induction of NFATc1/αA, a prominent short NFATc1 isoform. In part, this is mediated through two composite κB/NFAT-binding sites in the inducible Nfatc1 P1 promoter that directs the induction of NFATc1/αA by BCR signals. In concert with coreceptor signals that induce NF-κB factors, BCR signaling induces a persistent generation of NFATc1/αA. These data suggest a tight connection between NFATc1 and NF-κB induction in B lymphocytes contributing to the effector function of peripheral B cells.


Subject(s)
B-Lymphocytes/immunology , NF-kappa B p50 Subunit/metabolism , NFATC Transcription Factors/genetics , Proto-Oncogene Proteins c-rel/metabolism , Animals , Binding Sites , Chickens , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B p50 Subunit/genetics , NFATC Transcription Factors/biosynthesis , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Transcription Factor RelA/genetics , Transcription Factor RelB/genetics
17.
J Exp Med ; 211(3): 545-61, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24590764

ABSTRACT

Maturation of high-affinity B lymphocytes is precisely controlled during the germinal center reaction. This is dependent on CD4(+)CXCR5(+) follicular helper T cells (TFH) and inhibited by CD4(+)CXCR5(+)Foxp3(+) follicular regulatory T cells (TFR). Because NFAT2 was found to be highly expressed and activated in follicular T cells, we addressed its function herein. Unexpectedly, ablation of NFAT2 in T cells caused an augmented GC reaction upon immunization. Consistently, however, TFR cells were clearly reduced in the follicular T cell population due to impaired homing to B cell follicles. This was TFR-intrinsic because only in these cells NFAT2 was essential to up-regulate CXCR5. The physiological relevance for humoral (auto-)immunity was corroborated by exacerbated lupuslike disease in the presence of NFAT2-deficient TFR cells.


Subject(s)
Autoimmunity/immunology , Germinal Center/immunology , Immunity, Humoral/immunology , NFATC Transcription Factors/immunology , Receptors, CXCR5/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Analysis of Variance , Animals , Chromatin Immunoprecipitation , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Immunohistochemistry , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Real-Time Polymerase Chain Reaction , Receptors, CXCR5/metabolism
18.
Front Immunol ; 5: 21, 2014.
Article in English | MEDLINE | ID: mdl-24550910

ABSTRACT

In lymphocytes, the three NFAT factors NFATc1 (also designated as NFAT2), NFATc2 (NFAT1), and NFATc3 (NFAT4 or NFATx) are expressed and are the targets of immune receptor signals, which lead to a rapid rise of intracellular Ca(++), the activation of phosphatase calcineurin, and to the activation of cytosolic NFATc proteins. In addition to rapid activation of NFAT factors, immune receptor signals lead to accumulation of the short NFATc1/αA isoform in lymphocytes which controls their proliferation and survival. In this mini-review, we summarize our current knowledge on the structure and transcription of the Nfatc1 gene in lymphocytes, which is controlled by two promoters, two poly A addition sites and a remote downstream enhancer. The Nfatc1 gene resembles numerous primary response genes (PRGs) induced by LPS in macrophages. Similar to the PRG promoters, the Nfatc1 promoter region is organized in CpG islands, forms DNase I hypersensitive sites, and is marked by histone tail modifications before induction. By studying gene induction in lymphocytes in detail, it will be important to elucidate whether the properties of the Nfatc1 induction are not only typical for the Nfatc1 gene but also for other transcription factor genes expressed in lymphocytes.

19.
Mol Cell Biol ; 34(5): 820-31, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24344200

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) are ligand-gated ion channels that play an important role in neuronal development, plasticity, and excitotoxicity. NMDAR antagonists are neuroprotective in animal models of neuronal diseases, and the NMDAR open-channel blocker memantine is used to treat Alzheimer's disease. In view of the clinical application of these pharmaceuticals and the reported expression of NMDARs in immune cells, we analyzed the drug's effects on T-cell function. NMDAR antagonists inhibited antigen-specific T-cell proliferation and cytotoxicity of T cells and the migration of the cells toward chemokines. These activities correlated with a reduction in T-cell receptor (TCR)-induced Ca(2+) mobilization and nuclear localization of NFATc1, and they attenuated the activation of Erk1/2 and Akt. In the presence of antagonists, Th1 effector cells produced less interleukin-2 (IL-2) and gamma interferon (IFN-γ), whereas Th2 cells produced more IL-10 and IL-13. However, in NMDAR knockout mice, the presumptive expression of functional NMDARs in wild-type T cells was inconclusive. Instead, inhibition of NMDAR antagonists on the conductivity of Kv1.3 and KCa3.1 potassium channels was found. Hence, NMDAR antagonists are potent immunosuppressants with therapeutic potential in the treatment of immune diseases, but their effects on T cells have to be considered in that Kv1.3 and KCa3.1 channels are their major effectors.


Subject(s)
Immune Tolerance/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Kv1.3 Potassium Channel/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Interferon-gamma/metabolism , Interleukins/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Kv1.3 Potassium Channel/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
20.
J Immunol ; 190(5): 2345-53, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23365084

ABSTRACT

NFAT transcription factors control the proliferation and survival of peripheral lymphocytes. We have reported previously that the short isoform NFATc1/αA whose generation is induced by immune receptor stimulation supports the proliferation and inhibits the activation-induced cell death of peripheral T and B cells. We will show in this study that in novel bacterial artificial chromosome transgenic mice that express EGFP under the control of entire Nfatc1 locus the Nfatc1/Egfp transgene is expressed as early as in double-negative thymocytes and in nonstimulated peripheral T and B cells. Upon immune receptor stimulation, Nfatc1/Egfp expression is elevated in B, Th1, and Th2 cells, but only weakly in T regulatory, Th9, and Th17 cells in vitro whose generation is affected by TGFß. In naive lymphocytes, persistent immune receptor signals led to a 3-5 increase in NFATc1/αA RNA levels during primary and secondary stimulation, but a much stronger induction was observed at the protein level. Whereas anti-CD3(+)CD28 stimulation of primary T cells induces both NFATc1/αA and their proliferation and survival, anti-IgM stimulation of B cells induces NFATc1/αA and proliferation, but activation-induced cell death after 3-d incubation in vitro. The anti-IgM-mediated activation-induced cell death induction of B cells in vitro is suppressed by anti-CD40-, LPS-, and CpG-mediated signals. In addition to inducing NF-κB factors, together with anti-IgM, these signals also support the generation of NFATc1/αA. According to these data and the architecture of its promoter region, the Nfatc1 gene resembles a primary response gene whose induction is affected at the posttranscriptional level.


Subject(s)
B-Lymphocytes/drug effects , NFATC Transcription Factors/genetics , Th1 Cells/drug effects , Th2 Cells/drug effects , Animals , Antibodies/pharmacology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Proliferation/drug effects , Chromosomes, Artificial, Bacterial/genetics , Gene Expression/drug effects , Gene Expression/immunology , Genes, Reporter , Green Fluorescent Proteins , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , NFATC Transcription Factors/agonists , NFATC Transcription Factors/immunology , Promoter Regions, Genetic , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...