1
|
Yukawa M, Jagannathan S, Vallabh S, Kartashov AV, Chen X, Weirauch MT, Barski A. AP-1 activity induced by co-stimulation is required for chromatin opening during T cell activation. J Exp Med 2020; 217:jem.20182009. [PMID: 31653690 PMCID: PMC7037242 DOI: 10.1084/jem.20182009] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/06/2019] [Accepted: 09/23/2019] [Indexed: 12/24/2022] Open
Abstract
Activation of T cells is dependent on the organized and timely opening and closing of chromatin. Herein, we identify AP-1 as the transcription factor that directs most of this remodeling. Chromatin accessibility profiling showed quick opening of closed chromatin in naive T cells within 5 h of activation. These newly opened regions were strongly enriched for the AP-1 motif, and indeed, ChIP-seq demonstrated AP-1 binding at >70% of them. Broad inhibition of AP-1 activity prevented chromatin opening at AP-1 sites and reduced the expression of nearby genes. Similarly, induction of anergy in the absence of co-stimulation during activation was associated with reduced induction of AP-1 and a failure of proper chromatin remodeling. The translational relevance of these findings was highlighted by the substantial overlap of AP-1-dependent elements with risk loci for multiple immune diseases, including multiple sclerosis, inflammatory bowel disease, and allergic disease. Our findings define AP-1 as the key link between T cell activation and chromatin remodeling.
Collapse
Affiliation(s)
- Masashi Yukawa
- Division of Allergy & Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sajjeev Jagannathan
- Division of Allergy & Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sushmitha Vallabh
- Division of Allergy & Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Andrey V Kartashov
- Division of Allergy & Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH.,Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Artem Barski
- Division of Allergy & Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH.,Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
2
|
Abstract
The immune system is remarkably responsive to a myriad of invading microorganisms and provides continuous surveillance against tissue damage and developing tumor cells. To achieve these diverse functions, multiple soluble and cellular components must react in an orchestrated cascade of events to control the specificity, magnitude and persistence of the immune response. Numerous catabolic and anabolic processes are involved in this process, and prominent roles for l-arginine and l-glutamine catabolism have been described, as these amino acids serve as precursors of nitric oxide, creatine, agmatine, tricarboxylic acid cycle intermediates, nucleotides and other amino acids, as well as for ornithine, which is used to synthesize putrescine and the polyamines spermidine and spermine. Polyamines have several purported roles and high levels of polyamines are manifest in tumor cells as well in autoreactive B- and T-cells in autoimmune diseases. In the tumor microenvironment, l-arginine catabolism by both tumor cells and suppressive myeloid cells is known to dampen cytotoxic T-cell functions suggesting there might be links between polyamines and T-cell suppression. Here, we review studies suggesting roles of polyamines in normal immune cell function and highlight their connections to autoimmunity and anti-tumor immune cell function.
Collapse
Affiliation(s)
- Rebecca S Hesterberg
- University of South Florida Cancer Biology Graduate Program, University of South Florida, 4202 East Fowler Ave, Tampa, FL 33620, USA.
- Department Immunology, PharmD, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, 23033 SRB, Tampa, FL 33612, USA.
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | - Pearlie K Epling-Burnette
- Department Immunology, PharmD, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, 23033 SRB, Tampa, FL 33612, USA.
| |
Collapse
|
3
|
Prandi S, Voigt A, Meyerhof W, Behrens M. Expression profiling of Tas2r genes reveals a complex pattern along the mouse GI tract and the presence of Tas2r131 in a subset of intestinal Paneth cells. Cell Mol Life Sci 2018; 75:49-65. [PMID: 28801754 PMCID: PMC11105753 DOI: 10.1007/s00018-017-2621-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/02/2017] [Accepted: 08/04/2017] [Indexed: 12/18/2022]
Abstract
The chemical variability of the intestinal lumen requires the presence of molecular receptors detecting the various substances naturally occurring in the diet and as a result of the activity of the microbiota. Despite their early discovery, intestinal bitter taste receptors (Tas2r) have not yet been assigned an unambiguous physiological function. Recently, using a CRE-recombinant approach we showed that the Tas2r131 gene is expressed in a subset of mucin-producing goblet cells in the colon of mice. Moreover, we also demonstrated that the expression of the Tas2r131 locus is not restricted to this region. In the present study we aimed at characterizing the presence of positive cells also in other gastrointestinal regions. Our results show that Tas2r131+ cells appear in the jejunum and the ileum, and are absent from the stomach and the duodenum. We identified the positive cells as a subpopulation of deep-crypt Paneth cells in the ileum, strengthening the notion of a defensive role for Tas2rs in the gut. To get a broader perspective on the expression of bitter taste receptors in the alimentary canal, we quantified the expression of all 35 Tas2r genes along the gastrointestinal tract by qRT-PCR. We discovered that the number and expression level of Tas2r genes profoundly vary along the alimentary canal, with the stomach and the colon expressing the largest subsets.
Collapse
Affiliation(s)
- Simone Prandi
- Department of Molecular Genetics, German Institute for Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Anja Voigt
- Department of Molecular Genetics, German Institute for Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Wolfgang Meyerhof
- Department of Molecular Genetics, German Institute for Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Maik Behrens
- Department of Molecular Genetics, German Institute for Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
| |
Collapse
|
4
|
Wang X, Wong CW, Urak R, Taus E, Aguilar B, Chang WC, Mardiros A, Budde LE, Brown CE, Berger C, Forman SJ, Jensen MC. Comparison of naïve and central memory derived CD8 + effector cell engraftment fitness and function following adoptive transfer. Oncoimmunology 2015; 5:e1072671. [PMID: 26942092 DOI: 10.1080/2162402x.2015.1072671] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 07/07/2015] [Accepted: 07/08/2015] [Indexed: 01/01/2023] Open
Abstract
Human CD8+ effector T cells derived from CD45RO+CD62L+ precursors enriched for central memory (TCM) precursors retain the capacity to engraft and reconstitute functional memory upon adoptive transfer, whereas effectors derived from CD45RO+CD62L- precursors enriched for effector memory precursors do not. Here we sought to compare the engraftment fitness and function of CD8+ effector T cells derived from CD45RA+CD62L+ precursors enriched for naïve and stem cell memory precursors (TN/SCM) with that of TCM. We found that cytotoxic T cells (CTLs) derived from TCM transcribed higher levels of CD28, FOS, INFγ, Eomesodermin (Eomes), and lower levels of BCL2L11, maintained higher levels of phosphorylated AKT, and displayed enhanced sensitivity to the proliferative and anti-apoptotic effects of γ-chain cytokines compared to CTLs derived from TN/SCM. Higher frequencies of CTLs derived from TCM retained CD28 expression and upon activation secreted higher levels of IL-2. In NOD/Scid IL-2RγCnull mice, CD8+ TCM derived CTLs engrafted to higher frequencies in response to human IL-15 and mounted robust proliferative responses to an immunostimulatory vaccine. Similarly, CD8+ TCM derived CD19CAR+ CTLs exhibited superior antitumor potency following adoptive transfer compared to their CD8+ TN/SCM derived counterparts. These studies support the use of TCM enriched cell products for adoptive therapy of cancer.
Collapse
Affiliation(s)
- Xiuli Wang
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - ChingLam W Wong
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Ryan Urak
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Ellie Taus
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Brenda Aguilar
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Wen-Chung Chang
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Armen Mardiros
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Lihua E Budde
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Christine E Brown
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Carolina Berger
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Stephen J Forman
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center , Duarte, CA, USA
| | - Michael C Jensen
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
5
|
Bendfeldt H, Benary M, Scheel T, Frischbutter S, Abajyan A, Radbruch A, Herzel H, Baumgrass R. Stable IL-2 decision making by endogenous c-Fos amounts in peripheral memory T-helper cells. J Biol Chem 2012; 287:18386-97. [PMID: 22474330 DOI: 10.1074/jbc.m112.358853] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The cytokine IL-2 performs opposite functions supporting efficient immune responses and playing a key role in peripheral tolerance. Therefore, precise fine-tuning of IL-2 expression is crucial for adjusting the immune response. Combining transcription factor analysis at the single cell and the single nucleus level using flow cytometry with statistical analysis, we showed that physiological differences in the expression levels of c-Fos and NFATc2, but not of c-Jun and NF-κBp65, are limiting for the decision whether IL-2 is expressed in a strongly activated human memory T-helper (Th) cell. Variation in the expression of c-Fos leads to substantial diversity of IL-2 expression in ∼40% of the memory Th cells. The remaining cells exhibit an equally high c-Fos expression level, thereby ensuring robustness in IL-2 response within the population. These findings reveal how memory Th cells benefit from regulated variation in transcription factor expression to achieve a certain stability and variability of cytokine expression in a controlled manner.
Collapse
Affiliation(s)
- Hanna Bendfeldt
- Deutsches Rheuma-Forschungszentrum Berlin, A. Leibniz Institute, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Padhan K, Varma R. Immunological synapse: a multi-protein signalling cellular apparatus for controlling gene expression. Immunology 2010; 129:322-8. [PMID: 20409153 DOI: 10.1111/j.1365-2567.2009.03241.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The interaction of T cells with antigen-presenting cells is the hallmark of adaptive immunity. In vitro studies have described the formation of an immunological synapse between these cells, and intra-vital imaging has described in great detail the dynamics of these interactions. The immunological synapse has become a paradigm to study signals exchanged between the two cells. A wealth of information has been amassed regarding the localization of signalling molecules, their kinetics and the transcription factors they activate. We continue to discover mechanisms that cause receptors and signalling molecules to compartmentalize in the cell; however, the emerging challenge lies in understanding how the immunological synapse contributes to differentiation. Here, we review some of the transcription factors activated downstream of T-cell receptor signalling and discuss mechanisms by which antigen dose and affinity may influence differentiation. Antigen affinity might change the kind of transcription factors that are activated whereas antigen dose is likely to influence the temporal dynamics of the transcription factors. The immunological synapse is therefore likely to influence differentiation by modulating the trafficking of transcription factors and by promoting asymmetric cell division, an emerging concept.
Collapse
Affiliation(s)
- Kartika Padhan
- Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
7
|
Martins GA, Cimmino L, Liao J, Magnusdottir E, Calame K. Blimp-1 directly represses Il2 and the Il2 activator Fos, attenuating T cell proliferation and survival. ACTA ACUST UNITED AC 2008; 205:1959-65. [PMID: 18725523 PMCID: PMC2526191 DOI: 10.1084/jem.20080526] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mice with a T cell–specific deletion of Prdm1, encoding Blimp-1, have aberrant T cell homeostasis and develop fatal colitis. In this study, we show that one critical activity of Blimp-1 in T cells is to repress IL-2, and that it does so by direct repression of Il2 transcription, and also by repression of Fos transcription. Using these mechanisms Blimp-1 participates in an autoregulatory loop by which IL-2 induces Prdm1 expression and thus represses its own expression after T cell activation, ensuring that the immune response is appropriately controlled. This activity of Blimp-1 is important for cytokine deprivation–induced T cell death and for attenuating T cell proliferation in antigen-specific responses both in vitro and in vivo.
Collapse
Affiliation(s)
- Gislâine A Martins
- Department of Microbiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | | | | | | | |
Collapse
|
8
|
Abstract
Genetically modified mice and cells have provided important insights into the biological functions of the dimeric transcription factor complex AP1, in particular into its role in skeletal development. Data obtained from knockout mice revealed that some components, such as c-Fos are key regulators of bone cell differentiation, whereas others, like c-Jun, JunB and Fra-1 are essential in embryonic and/or postnatal development. Apart from identifying the specific roles of AP1 proteins in developmental processes, researchers are beginning to obtain a better molecular understanding of their cell-context dependent functions, their downstream target genes and how they regulate bone cell proliferation, differentiation, and apoptosis.
Collapse
Affiliation(s)
- E F Wagner
- Research Institute of Molecular Pathology (IMP), Dr Bohr-Gasse 7, A-1030 Vienna, Austria.
| |
Collapse
|
9
|
Aune TM, Mora AL, Kim S, Boothby M, Lichtman AH. Costimulation Reverses the Defect in IL-2 But Not Effector Cytokine Production by T Cells with Impaired IκBα Degradation. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.10.5805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Although the transcriptional basis for states of unresponsiveness in primary T cells is unclear, tolerant B lymphocytes exhibit inhibition of both c-Jun N-terminal kinase induction and IκBα (inhibitor of NF-κBα) degradation, leading to lower levels of both nuclear AP-1 and NF-κB. Expression of an IκBα mutant resistant to signal-induced degradation in transgenic T cells caused markedly deficient effector cytokine (IL-4, IFN-γ) production after primary TCR stimulation despite a detectable level of nuclear NF-κB. A TCR response element from the IFN-γ promoter, despite lacking detectable NF-κB/Rel sites, was also unresponsive to TCR ligation. Nuclear induction of AP-1 proteins in response to T cell activation was diminished in transgenic T cells. Costimulation induced by anti-CD28 mAb increased IL-2 production, but failed to reverse the defects in effector cytokine production. Taken together, these data indicate that impaired NF-κB/Rel signaling in T cells interferes with the signal transduction pathways required for efficient induction of effector cytokine production.
Collapse
Affiliation(s)
| | - Ana L. Mora
- †Microbiology/Immunology, Vanderbilt University Medical School, Nashville, TN 37232; and
| | - Somee Kim
- ‡Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Mark Boothby
- †Microbiology/Immunology, Vanderbilt University Medical School, Nashville, TN 37232; and
| | - Andrew H. Lichtman
- ‡Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
10
|
Shapiro VS, Mollenauer MN, Greene WC, Weiss A. c-rel regulation of IL-2 gene expression may be mediated through activation of AP-1. J Exp Med 1996; 184:1663-9. [PMID: 8920856 PMCID: PMC2192877 DOI: 10.1084/jem.184.5.1663] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
T cell activation by antigen/MHC induces the expression of several genes critical to the immune response, including interleukin-2. T cells from mice deficient for the NF-kappa B family member c-rel cannot activate IL-2 gene expression. However, mutating the NF-kappa B site in the IL-2 promoter has only moderate effects. To investigate additional ways c-Rel could regulate IL-2 gene expression, we determined whether c-rel overexpression could increase the activity of other transcription factors involved in IL-2 promoter regulation: NF-AT, Oct/OAP (ARRE-1), and AP-1. In Jurkat TAg cells, overexpression of c-Rel increased AP-1 activation approximately 17-fold. Moreover, AP-1 activity stimulated by anti-TCR Abs or PMA/ionomycin was further increased by c-Rel overexpression. c-Rel overexpression did not affect NF-AT or ARRE-1 activity. Additionally, overexpression of c-Rel activated the nonconsensus AP-1 site from the IL-2 promoter (NF-IL-2B), although to a lesser extent, approximately sixfold. AP-1 activation required both the DNA binding and transactivation domains of c-Rel. Our results may provide an explanation for the effect on IL-2 gene activation in c-rel-deficient mice.
Collapse
Affiliation(s)
- V S Shapiro
- Department of Medicine, University of California San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
11
|
Lee SL, Wang Y, Milbrandt J. Unimpaired macrophage differentiation and activation in mice lacking the zinc finger transplantation factor NGFI-A (EGR1). Mol Cell Biol 1996; 16:4566-72. [PMID: 8754857 PMCID: PMC231455 DOI: 10.1128/mcb.16.8.4566] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The zinc finger protein NGFI-A (also called EGR1, Krox24, or zif268) is a candidate regulator of myeloid cell differentiation. Evidence supporting this hypothesis is twofold. First, NGFI-A antisense oligonucleotides prevent macrophage differentiation in HL-60 and U937 myeloid leukemia cell lines and in normal bone marrow cells. Second, enforced expression of NGFI-A blocks granulocytic differentiation and promotes macrophage differentiation in HL-60 cells and in the hematopoietic progenitor cell line 32D. We sought to determine the effect of NGFI-A deficiency on macrophage differentiation and function in vivo by examining native bone marrow cells from mice homozygous for a disrupted allele of NGFI-A derived from gene-targeted ES cells. Macrophages were observed in peripheral blood and several tissues, indicating that NGFI-A was not required for the formation of a variety of macrophage compartments. No differences in myeloid cell differentiation were observed between wild-type and NGFI-A-/- bone marrow cells cultured in the presence of macrophage, granulocyte-macrophage, or granulocyte colony-stimulating factor (M-CSF, GM-CSF, or G-CSF). Activation of NGFI-A-/- macrophages was comparable to that of wild-type macrophages as determined by nitric oxide production and increased cell surface expression of class II major histocompatibility complex molecules. Moreover, NGFI-A-/- mice showed no increased mortality or bacteria] burden when challenged with Listeria monocytogenes. Together, these results indicate that NGFI-A is not required for macrophage differentiation or activation.
Collapse
Affiliation(s)
- S L Lee
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
12
|
Shaw KT, Ho AM, Raghavan A, Kim J, Jain J, Park J, Sharma S, Rao A, Hogan PG. Immunosuppressive drugs prevent a rapid dephosphorylation of transcription factor NFAT1 in stimulated immune cells. Proc Natl Acad Sci U S A 1995; 92:11205-9. [PMID: 7479966 PMCID: PMC40600 DOI: 10.1073/pnas.92.24.11205] [Citation(s) in RCA: 274] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The immunosuppressive drugs cyclosporin A and FK506 interfere with the inducible transcription of cytokine genes in T cells and in other immune cells, in part by preventing the activation of NF-AT (nuclear factor of activated T cells). We show that transcription factor NFAT1 in T cells is rapidly dephosphorylated on stimulation, that dephosphorylation occurs before translocation of NFAT1 into the cell nucleus, and that dephosphorylation increases the affinity of NFAT1 for its specific sites in DNA. Cyclosporin A prevents the dephosphorylation and the nuclear translocation of NFAT1 in T cells, B cells, macrophages, and mast cells, delineating at least one mechanism that contributes to the profound immunosuppressive effects of this compound.
Collapse
Affiliation(s)
- K T Shaw
- Division of Cellular and Molecular Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Aramburu J, Azzoni L, Rao A, Perussia B. Activation and expression of the nuclear factors of activated T cells, NFATp and NFATc, in human natural killer cells: regulation upon CD16 ligand binding. J Exp Med 1995; 182:801-10. [PMID: 7650486 PMCID: PMC2192167 DOI: 10.1084/jem.182.3.801] [Citation(s) in RCA: 116] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The putative factors that couple the signal transduction from surface receptors to the activation of cytokine synthesis in natural killer (NK) cells have not been elucidated. We report here that the nuclear factor of activated T cells (NFATp), a cyclosporin A (CsA)-sensitive factor that regulates the transcription of several cytokines, mediates CD16-induced activation of cytokine genes in human NK cells. CD16 (Fc gamma RIIIA)-induced expression of cytokine mRNA in NK cells occurs via a CsA-sensitive and Ca(2+)-dependent mechanism. Stimulation of NK cells with CD16 ligands induces NFAT-like DNA binding activity in the nuclear extracts from these cells, as detected in electrophoretic mobility shift assays. This occurs with fast kinetics after stimulation, via a CsA-sensitive and Ca(2+)-dependent mechanism that does not require de novo protein synthesis. NK cell NFAT is present in the cytosol of nonstimulated cells, migrates to the nucleus upon stimulation, and can associate with AP-1. Two distinct molecules, NFATp and NFATc, have been reported to mediate NFAT activity. The results of supershift assays using NFATp- and NFATc- specific antibodies indicate that NK cell activation early after CD16 ligand binding involves primarily, if not exclusively, NFATp, and Western blot analysis shows that this has the same electrophoretic mobility (approximately 120 kD) as that of T lymphocytes. NK cells do not express NFATc constitutively, but NFATc mRNA accumulation is induced in these cells within 2 h of stimulation with CD16 ligands. However, supershift assays using the available mAb recognizing the T cell NFATc revealed no detectable NFATc protein in nuclear and cytoplasmic extracts from CD16- or phorbol ester-stimulated cells at any time tested, up to 4 h. These results provide the first direct evidence that both CsA-sensitive transcription factors, NFATp and NFATc, are expressed in human NK cells, and that their activation and/or expression can be regulated in primary cells by a single stimulus, that, in the case of CD16 in NK cells, results in early activation of NFATp and subsequently induced expression of NFATc mRNA.
Collapse
Affiliation(s)
- J Aramburu
- Department of Microbiology and Immunology, Jefferson Cancer Institute, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | |
Collapse
|
14
|
A central role for Fos in human B- and T-cell NFAT (nuclear factor of activated T cells): an acidic region is required for in vitro assembly. Mol Cell Biol 1994. [PMID: 7935406 DOI: 10.1128/mcb.14.10.6886] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nuclear factor of activated T cells (NFAT) is a multicomponent transcription factor that contains Fos and Jun family proteins in addition to a constitutively expressed factor(s). It is important for the production of interleukin 2 (IL-2) by T cells and is also expressed in B cells. Here we show that NFAT complexes in B- and T-cell nuclear extracts can be supershifted prominently with Fos antibodies and to a variable extent with Jun family protein antibodies. Fos and Jun proteins appear to participate in NFAT complexes as heterodimers, since efficient in vitro reconstitution of NFAT in unstimulated B- or T-cell nuclear extracts required both Fos and Jun. Using Fos and Jun deletion derivatives, we found that an acidic Fos region (amino acids 118 to 138), outside the DNA binding and dimerization domains, was necessary for the in vitro reconstitution of the NFAT complex in both B- and T-lymphocyte extracts although it was not required for binding to an AP-1 site. Fos-Jun heterodimers exhibited low-affinity direct binding to the NFAT site in the absence of nuclear extracts. This binding also required the Fos acidic region, amino acids 118 to 138. Mutating a variant AP-1 site in the NFAT oligonucleotide abolished both direct binding of Fos-Jun heterodimers and in vitro reconstitution of NFAT. These results demonstrate a central role of Fos in NFAT complex formation in both B and T lymphocytes and show that NFAT assembly involves direct binding of Fos-Jun heterodimers to a variant AP-1 site within the human NFAT recognition site.
Collapse
|
15
|
Yaseen NR, Park J, Kerppola T, Curran T, Sharma S. A central role for Fos in human B- and T-cell NFAT (nuclear factor of activated T cells): an acidic region is required for in vitro assembly. Mol Cell Biol 1994; 14:6886-95. [PMID: 7935406 PMCID: PMC359219 DOI: 10.1128/mcb.14.10.6886-6895.1994] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Nuclear factor of activated T cells (NFAT) is a multicomponent transcription factor that contains Fos and Jun family proteins in addition to a constitutively expressed factor(s). It is important for the production of interleukin 2 (IL-2) by T cells and is also expressed in B cells. Here we show that NFAT complexes in B- and T-cell nuclear extracts can be supershifted prominently with Fos antibodies and to a variable extent with Jun family protein antibodies. Fos and Jun proteins appear to participate in NFAT complexes as heterodimers, since efficient in vitro reconstitution of NFAT in unstimulated B- or T-cell nuclear extracts required both Fos and Jun. Using Fos and Jun deletion derivatives, we found that an acidic Fos region (amino acids 118 to 138), outside the DNA binding and dimerization domains, was necessary for the in vitro reconstitution of the NFAT complex in both B- and T-lymphocyte extracts although it was not required for binding to an AP-1 site. Fos-Jun heterodimers exhibited low-affinity direct binding to the NFAT site in the absence of nuclear extracts. This binding also required the Fos acidic region, amino acids 118 to 138. Mutating a variant AP-1 site in the NFAT oligonucleotide abolished both direct binding of Fos-Jun heterodimers and in vitro reconstitution of NFAT. These results demonstrate a central role of Fos in NFAT complex formation in both B and T lymphocytes and show that NFAT assembly involves direct binding of Fos-Jun heterodimers to a variant AP-1 site within the human NFAT recognition site.
Collapse
Affiliation(s)
- N R Yaseen
- Department of Pathology, Roger Williams Cancer/Medical Center, Brown University, Providence, Rhode Island 02908
| | | | | | | | | |
Collapse
|