Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nat Immunol ; 19(8): 828-837, 2018 08.
Article in English | MEDLINE | ID: mdl-29988089

ABSTRACT

Memory T cells are critical for the immune response to recurring infections. Their instantaneous reactivity to pathogens is empowered by the persistent expression of cytokine-encoding mRNAs. How the translation of proteins from pre-formed cytokine-encoding mRNAs is prevented in the absence of infection has remained unclear. Here we found that protein production in memory T cells was blocked via a 3' untranslated region (3' UTR)-mediated process. Germline deletion of AU-rich elements (AREs) in the Ifng-3' UTR led to chronic cytokine production in memory T cells. This aberrant protein production did not result from increased expression and/or half-life of the mRNA. Instead, AREs blocked the recruitment of cytokine-encoding mRNA to ribosomes; this block depended on the ARE-binding protein ZFP36L2. Thus, AREs mediate repression of translation in mouse and human memory T cells by preventing undesirable protein production from pre-formed cytokine-encoding mRNAs in the absence of infection.


Subject(s)
3' Untranslated Regions/genetics , AU Rich Elements/genetics , Interferon-gamma/genetics , RNA, Messenger/genetics , T-Lymphocytes/immunology , Animals , Cells, Cultured , Epigenetic Repression , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Chain Elongation, Translational , Ribosomes/metabolism , Tristetraprolin/genetics , Tristetraprolin/metabolism
2.
Eur J Immunol ; : e2451018, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980256

ABSTRACT

CD8+ T cells kill target cells by releasing cytotoxic molecules and proinflammatory cytokines, such as TNF and IFN-γ. The magnitude and duration of cytokine production are defined by posttranscriptional regulation, and critical regulator herein are RNA-binding proteins (RBPs). Although the functional importance of RBPs in regulating cytokine production is established, the kinetics and mode of action through which RBPs control cytokine production are not well understood. Previously, we showed that the RBP ZFP36L2 blocks the translation of preformed cytokine encoding mRNA in quiescent memory T cells. Here, we uncover that ZFP36L2 regulates cytokine production in a time-dependent manner. T cell-specific deletion of ZFP36L2 (CD4-cre) had no effect on T-cell development or cytokine production during early time points (2-6 h) of T-cell activation. In contrast, ZFP36L2 specifically dampened the production of IFN-γ during prolonged T-cell activation (20-48 h). ZFP36L2 deficiency also resulted in increased production of IFN-γ production in tumor-infiltrating T cells that are chronically exposed to antigens. Mechanistically, ZFP36L2 regulates IFN-γ production at late time points of activation by destabilizing Ifng mRNA in an AU-rich element-dependent manner. Together, our results reveal that ZFP36L2 employs different regulatory nodules in effector and memory T cells to regulate cytokine production.

3.
Nucleic Acids Res ; 46(16): 8168-8180, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30124921

ABSTRACT

Hematopoietic stem cells differentiate into a broad range of specialized blood cells. This process is tightly regulated and depends on transcription factors, micro-RNAs, and long non-coding RNAs. Recently, also circular RNA (circRNA) were found to regulate cellular processes. Their expression pattern and their identity is however less well defined. Here, we provide the first comprehensive analysis of circRNA expression in human hematopoietic progenitors, and in differentiated lymphoid and myeloid cells. We here show that the expression of circRNA is cell-type specific, and increases upon maturation. CircRNA splicing variants can also be cell-type specific. Furthermore, nucleated hematopoietic cells contain circRNA that have higher expression levels than the corresponding linear RNA. Enucleated blood cells, i.e. platelets and erythrocytes, were suggested to use RNA to maintain their function, respond to environmental factors or to transmit signals to other cells via microvesicles. Here we show that platelets and erythrocytes contain the highest number of circRNA of all hematopoietic cells, and that the type and numbers of circRNA changes during maturation. This cell-type specific expression pattern of circRNA in hematopoietic cells suggests a hithero unappreciated role in differentiation and cellular function.


Subject(s)
Gene Expression Regulation/genetics , Hematopoietic Stem Cells/metabolism , RNA/genetics , Blood Platelets/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Erythrocytes/cytology , Hematopoietic Stem Cell Transplantation , Humans , RNA/biosynthesis , RNA/blood , RNA, Circular
4.
Eur J Immunol ; 42(11): 3019-27, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22806638

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are key players in antiviral immunity. In addition to massive type I interferon production, activated pDCs express the apoptosis-inducing molecule TRAIL, which enables them to clear infected cells that express the TRAIL receptors TRAIL-R1 and TRAIL-R2. In this study, we examined the molecular mechanisms that govern TRAIL expression in human pDCs. We identify NGFI-A-binding protein 2 (NAB2) as a novel transcriptional regulator that governs TRAIL induction in stimulated pDCs. We show with the pDC-like cell line CAL-1 that NAB2 is exclusively induced downstream of TLR7 and TLR9 signaling, and not upon type I IFN-R signaling. Furthermore, PI3K signaling is required for NAB2-mediated TRAIL expression. Finally, we show that TRAIL induction in CpG-activated human pDCs occurs through two independent signaling pathways: the first is initiated through TLR9 signaling upon recognition of nucleic acids, followed by type I IFN-R-mediated signaling. In conclusion, our data suggest that these two pathways are downstream of different activation signals, but act in concert to allow for full TRAIL expression in pDCs.


Subject(s)
Dendritic Cells/immunology , Lymphocyte Activation/immunology , Repressor Proteins/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Cell Line , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Leukocytes, Mononuclear/immunology , Phosphatidylinositol 3-Kinase/immunology , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology
5.
Cell Rep ; 42(5): 112419, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37074914

ABSTRACT

Potent T cell responses against infections and malignancies require a rapid yet tightly regulated production of toxic effector molecules. Their production level is defined by post-transcriptional events at 3' untranslated regions (3' UTRs). RNA binding proteins (RBPs) are key regulators in this process. With an RNA aptamer-based capture assay, we identify >130 RBPs interacting with IFNG, TNF, and IL2 3' UTRs in human T cells. RBP-RNA interactions show plasticity upon T cell activation. Furthermore, we uncover the intricate and time-dependent regulation of cytokine production by RBPs: whereas HuR supports early cytokine production, ZFP36L1, ATXN2L, and ZC3HAV1 dampen and shorten the production duration, each at different time points. Strikingly, even though ZFP36L1 deletion does not rescue the dysfunctional phenotype, tumor-infiltrating T cells produce more cytokines and cytotoxic molecules, resulting in superior anti-tumoral T cell responses. Our findings thus show that identifying RBP-RNA interactions reveals key modulators of T cell responses in health and disease.


Subject(s)
Cytokines , T-Lymphocytes , Humans , T-Lymphocytes/metabolism , 3' Untranslated Regions , Cytokines/metabolism , RNA-Binding Proteins/metabolism , Butyrate Response Factor 1/genetics , Butyrate Response Factor 1/metabolism
6.
Proc Natl Acad Sci U S A ; 106(40): 17019-24, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19805154

ABSTRACT

Plasma cells daily secrete their own mass in antibodies, which fold and assemble in the endoplasmic reticulum (ER). To reach these levels, cells require pERp1, a novel lymphocyte-specific small ER-resident protein, which attains expression levels as high as BiP when B cells differentiate into plasma cells. Although pERp1 has no homology with known ER proteins, it does contain a CXXC motif typical for oxidoreductases. In steady state, the CXXC cysteines are locked by two parallel disulfide bonds with a downstream C(X)(6)C motif, and pERp1 displays only modest oxidoreductase activity. pERp1 emerged as a dedicated folding factor for IgM, associating with both heavy and light chains and promoting assembly and secretion of mature IgM.


Subject(s)
Endoplasmic Reticulum/metabolism , Immunoglobulin M/metabolism , Molecular Chaperones/metabolism , Plasma Cells/metabolism , Amino Acid Sequence , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/ultrastructure , Cell Differentiation , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum Chaperone BiP , HeLa Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Mass Spectrometry , Mice , Microscopy, Fluorescence , Microscopy, Immunoelectron , Molecular Chaperones/genetics , Oxidoreductases/metabolism , Plasma Cells/cytology , RNA Interference , Sulfhydryl Compounds/metabolism
7.
Open Biol ; 11(3): 210030, 2021 03.
Article in English | MEDLINE | ID: mdl-33715389

ABSTRACT

Nucleic acid sensing through pattern recognition receptors is critical for immune recognition of microbial infections. Microbial DNA is frequently methylated at the N6 position of adenines (m6A), a modification that is rare in mammalian host DNA. We show here how that m6A methylation of 5'-GATC-3' motifs augments the immunogenicity of synthetic double-stranded (ds)DNA in murine macrophages and dendritic cells. Transfection with m6A-methylated DNA increased the expression of the activation markers CD69 and CD86, and of Ifnß, iNos and Cxcl10 mRNA. Similar to unmethylated cytosolic dsDNA, recognition of m6A DNA occurs independently of TLR and RIG-I signalling, but requires the two key mediators of cytosolic DNA sensing, STING and cGAS. Intriguingly, the response to m6A DNA is sequence-specific. m6A is immunostimulatory in some motifs, but immunosuppressive in others, a feature that is conserved between mouse and human macrophages. In conclusion, epigenetic alterations of DNA depend on the context of the sequence and are differentially perceived by innate cells, a feature that could potentially be used for the design of immune-modulating therapeutics.


Subject(s)
Adenine/analogs & derivatives , DNA Methylation , Immunity, Innate , Oligodeoxyribonucleotides/immunology , Adenine/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , B7-2 Antigen/metabolism , Cells, Cultured , Chemokine CXCL10/metabolism , Cytoplasm/metabolism , Dendritic Cells/immunology , Humans , Interferon-beta/metabolism , Lectins, C-Type/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Oligodeoxyribonucleotides/chemistry , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL