Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nat Immunol ; 23(11): 1644-1652, 2022 11.
Article in English | MEDLINE | ID: mdl-36271145

ABSTRACT

Interleukin-17A (IL-17A) is a key mediator of protective immunity to yeast and bacterial infections but also drives the pathogenesis of several autoimmune diseases, such as psoriasis or psoriatic arthritis. Here we show that the tetra-transmembrane protein CMTM4 is a subunit of the IL-17 receptor (IL-17R). CMTM4 constitutively associated with IL-17R subunit C to mediate its stability, glycosylation and plasma membrane localization. Both mouse and human cell lines deficient in CMTM4 were largely unresponsive to IL-17A, due to their inability to assemble the IL-17R signaling complex. Accordingly, CMTM4-deficient mice had a severe defect in the recruitment of immune cells following IL-17A administration and were largely resistant to experimental psoriasis, but not to experimental autoimmune encephalomyelitis. Collectively, our data identified CMTM4 as an essential component of IL-17R and a potential therapeutic target for treating IL-17-mediated autoimmune diseases.


Subject(s)
Arthritis, Psoriatic , Encephalomyelitis, Autoimmune, Experimental , Psoriasis , Humans , Mice , Animals , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Interleukin-17/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , MARVEL Domain-Containing Proteins/genetics
2.
EMBO Rep ; 25(8): 3456-3485, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38877170

ABSTRACT

T cells are pivotal in the adaptive immune defense, necessitating a delicate balance between robust response against infections and self-tolerance. Their activation involves intricate cross-talk among signaling pathways triggered by the T-cell antigen receptors (TCR) and co-stimulatory or inhibitory receptors. The molecular regulation of these complex signaling networks is still incompletely understood. Here, we identify the adaptor protein ABIN1 as a component of the signaling complexes of GITR and OX40 co-stimulation receptors. T cells lacking ABIN1 are hyper-responsive ex vivo, exhibit enhanced responses to cognate infections, and superior ability to induce experimental autoimmune diabetes in mice. ABIN1 negatively regulates p38 kinase activation and late NF-κB target genes. P38 is at least partially responsible for the upregulation of the key effector proteins IFNG and GZMB in ABIN1-deficient T cells after TCR stimulation. Our findings reveal the intricate role of ABIN1 in T-cell regulation.


Subject(s)
Adaptor Proteins, Signal Transducing , Signal Transduction , T-Lymphocytes, Cytotoxic , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Glucocorticoid-Induced TNFR-Related Protein , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Lymphocyte Activation/genetics , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, OX40/metabolism , Receptors, OX40/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
3.
Mol Cell ; 65(4): 730-742.e5, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-28212753

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is known for specifically killing cancer cells, whereas in resistant cancers, TRAIL/TRAIL-R can promote metastasis via Rac1 and PI3K. It remains unknown, however, whether and to what extent TRAIL/TRAIL-R signaling in cancer cells can affect the immune microenvironment. Here we show that TRAIL-triggered cytokine secretion from TRAIL-resistant cancer cells is FADD dependent and identify the TRAIL-induced secretome to drive monocyte polarization to myeloid-derived suppressor cells (MDSCs) and M2-like macrophages. TRAIL-R suppression in tumor cells impaired CCL2 production and diminished both lung MDSC presence and tumor growth. In accordance, the receptor of CCL2, CCR2, is required to facilitate increased MDSC presence and tumor growth. Finally, TRAIL and CCL2 are co-regulated with MDSC/M2 markers in lung adenocarcinoma patients. Collectively, endogenous TRAIL/TRAIL-R-mediated CCL2 secretion promotes accumulation of tumor-supportive immune cells in the cancer microenvironment, thereby revealing a tumor-supportive immune-modulatory role of the TRAIL/TRAIL-R system in cancer biology.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cytokines/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Receptors, CCR2/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Microenvironment , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 8/genetics , Caspase 8/metabolism , Cell Proliferation , Chemokine CCL2/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Female , HCT116 Cells , HeLa Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophages/immunology , Macrophages/pathology , Mice, Inbred C57BL , Mice, SCID , Phenotype , RNA Interference , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Burden
4.
EMBO J ; 39(17): e104202, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32696476

ABSTRACT

IL-17 mediates immune protection from fungi and bacteria, as well as it promotes autoimmune pathologies. However, the regulation of the signal transduction from the IL-17 receptor (IL-17R) remained elusive. We developed a novel mass spectrometry-based approach to identify components of the IL-17R complex followed by analysis of their roles using reverse genetics. Besides the identification of linear ubiquitin chain assembly complex (LUBAC) as an important signal transducing component of IL-17R, we established that IL-17 signaling is regulated by a robust negative feedback loop mediated by TBK1 and IKKε. These kinases terminate IL-17 signaling by phosphorylating the adaptor ACT1 leading to the release of the essential ubiquitin ligase TRAF6 from the complex. NEMO recruits both kinases to the IL-17R complex, documenting that NEMO has an unprecedented negative function in IL-17 signaling, distinct from its role in NF-κB activation. Our study provides a comprehensive view of the molecular events of the IL-17 signal transduction and its regulation.


Subject(s)
Feedback, Physiological , Receptors, Interleukin-17/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-17/genetics
5.
Nature ; 557(7703): 112-117, 2018 05.
Article in English | MEDLINE | ID: mdl-29695863

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC) is required for optimal gene activation and prevention of cell death upon activation of immune receptors, including TNFR1 1 . Deficiency in the LUBAC components SHARPIN or HOIP in mice results in severe inflammation in adulthood or embryonic lethality, respectively, owing to deregulation of TNFR1-mediated cell death2-8. In humans, deficiency in the third LUBAC component HOIL-1 causes autoimmunity and inflammatory disease, similar to HOIP deficiency, whereas HOIL-1 deficiency in mice was reported to cause no overt phenotype9-11. Here we show, by creating HOIL-1-deficient mice, that HOIL-1 is as essential for LUBAC function as HOIP, albeit for different reasons: whereas HOIP is the catalytically active component of LUBAC, HOIL-1 is required for LUBAC assembly, stability and optimal retention in the TNFR1 signalling complex, thereby preventing aberrant cell death. Both HOIL-1 and HOIP prevent embryonic lethality at mid-gestation by interfering with aberrant TNFR1-mediated endothelial cell death, which only partially depends on RIPK1 kinase activity. Co-deletion of caspase-8 with RIPK3 or MLKL prevents cell death in Hoil-1-/- (also known as Rbck1-/-) embryos, yet only the combined loss of caspase-8 with MLKL results in viable HOIL-1-deficient mice. Notably, triple-knockout Ripk3-/-Casp8-/-Hoil-1-/- embryos die at late gestation owing to haematopoietic defects that are rescued by co-deletion of RIPK1 but not MLKL. Collectively, these results demonstrate that both HOIP and HOIL-1 are essential LUBAC components and are required for embryogenesis by preventing aberrant cell death. Furthermore, they reveal that when LUBAC and caspase-8 are absent, RIPK3 prevents RIPK1 from inducing embryonic lethality by causing defects in fetal haematopoiesis.


Subject(s)
Carrier Proteins/metabolism , Cell Death , Embryonic Development , Hematopoiesis , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Caspase 8/genetics , Caspase 8/metabolism , Cell Death/genetics , Embryo Loss/genetics , Embryonic Development/genetics , Endothelial Cells/cytology , Female , Hematopoiesis/genetics , Mice , Mice, Inbred C57BL , Protein Domains , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
6.
EMBO J ; 37(14)2018 07 13.
Article in English | MEDLINE | ID: mdl-29752423

ABSTRACT

Virtual memory T cells are foreign antigen-inexperienced T cells that have acquired memory-like phenotype and constitute 10-20% of all peripheral CD8+ T cells in mice. Their origin, biological roles, and relationship to naïve and foreign antigen-experienced memory T cells are incompletely understood. By analyzing T-cell receptor repertoires and using retrogenic monoclonal T-cell populations, we demonstrate that the virtual memory T-cell formation is a so far unappreciated cell fate decision checkpoint. We describe two molecular mechanisms driving the formation of virtual memory T cells. First, virtual memory T cells originate exclusively from strongly self-reactive T cells. Second, the stoichiometry of the CD8 interaction with Lck regulates the size of the virtual memory T-cell compartment via modulating the self-reactivity of individual T cells. Although virtual memory T cells descend from the highly self-reactive clones and acquire a partial memory program, they are not more potent in inducing experimental autoimmune diabetes than naïve T cells. These data underline the importance of the variable level of self-reactivity in polyclonal T cells for the generation of functional T-cell diversity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Immunologic Memory , Receptors, Antigen, T-Cell/analysis , Animals , Homeostasis , Mice
7.
J Biol Chem ; 295(42): 14279-14290, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32759308

ABSTRACT

Bardet-Biedl syndrome (BBS) is a pleiotropic ciliopathy caused by dysfunction of primary cilia. More than half of BBS patients carry mutations in one of eight genes encoding for subunits of a protein complex, the BBSome, which mediates trafficking of ciliary cargoes. In this study, we elucidated the mechanisms of the BBSome assembly in living cells and how this process is spatially regulated. We generated a large library of human cell lines deficient in a particular BBSome subunit and expressing another subunit tagged with a fluorescent protein. We analyzed these cell lines utilizing biochemical assays, conventional and expansion microscopy, and quantitative fluorescence microscopy techniques: fluorescence recovery after photobleaching and fluorescence correlation spectroscopy. Our data revealed that the BBSome formation is a sequential process. We show that the pre-BBSome is nucleated by BBS4 and assembled at pericentriolar satellites, followed by the translocation of the BBSome into the ciliary base mediated by BBS1. Our results provide a framework for elucidating how BBS-causative mutations interfere with the biogenesis of the BBSome.


Subject(s)
Microtubule-Associated Proteins/metabolism , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/metabolism , Bardet-Biedl Syndrome/pathology , CRISPR-Cas Systems/genetics , Cell Line , Cilia/metabolism , Cytoplasm/metabolism , Fluorescence Recovery After Photobleaching , Gene Editing , Humans , Microscopy, Fluorescence , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Mutation , Protein Subunits/genetics , Protein Subunits/metabolism
8.
EMBO J ; 36(9): 1147-1166, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28258062

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC) is the only known E3 ubiquitin ligase which catalyses the generation of linear ubiquitin linkages de novo LUBAC is a crucial component of various immune receptor signalling pathways. Here, we show that LUBAC forms part of the TRAIL-R-associated complex I as well as of the cytoplasmic TRAIL-induced complex II In both of these complexes, HOIP limits caspase-8 activity and, consequently, apoptosis whilst being itself cleaved in a caspase-8-dependent manner. Yet, by limiting the formation of a RIPK1/RIPK3/MLKL-containing complex, LUBAC also restricts TRAIL-induced necroptosis. We identify RIPK1 and caspase-8 as linearly ubiquitinated targets of LUBAC following TRAIL stimulation. Contrary to its role in preventing TRAIL-induced RIPK1-independent apoptosis, HOIP presence, but not its activity, is required for preventing necroptosis. By promoting recruitment of the IKK complex to complex I, LUBAC also promotes TRAIL-induced activation of NF-κB and, consequently, the production of cytokines, downstream of FADD, caspase-8 and cIAP1/2. Hence, LUBAC controls the TRAIL signalling outcome from complex I and II, two platforms which both trigger cell death and gene activation.


Subject(s)
Cell Death , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcriptional Activation , Ubiquitin-Protein Ligases/metabolism , Cell Line , Humans
9.
J Immunol ; 190(4): 1807-18, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23335753

ABSTRACT

When a BCR on a mature B cell is engaged by its ligand, the cell becomes activated, and the Ab-mediated immune response can be triggered. The initiation of BCR signaling is orchestrated by kinases of the Src and Syk families. However, the proximal BCR-induced phosphorylation remains incompletely understood. According to a model of sequential activation of kinases, Syk acts downstream of Src family kinases (SFKs). In addition, signaling independent of SFKs and initiated by Syk has been proposed. Both hypotheses lack sufficient evidence from relevant B cell models, mainly because of the redundancy of Src family members and the importance of BCR signaling for B cell development. We addressed this issue by analyzing controlled BCR triggering ex vivo on primary murine B cells and on murine and chicken B cell lines. Chemical and Csk-based genetic inhibitor treatments revealed that SFKs are required for signal initiation and Syk activation. In addition, ligand and anti-BCR Ab-induced signaling differ in their sensitivity to the inhibition of SFKs.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Protein-Tyrosine Kinases/physiology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/immunology , src-Family Kinases/physiology , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Chickens , Enzyme Activation/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Pre-B Cell Receptors/metabolism , Pre-B Cell Receptors/physiology , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/physiology , Syk Kinase , src-Family Kinases/metabolism
10.
J Biol Chem ; 287(27): 22812-21, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22589543

ABSTRACT

Transmembrane adaptor proteins are membrane-anchored proteins consisting of a short extracellular part, a transmembrane domain, and a cytoplasmic part with various protein-protein interaction motifs but lacking any enzymatic activity. They participate in the regulation of various signaling pathways by recruiting other proteins to the proximity of cellular membranes where the signaling is often initiated and propagated. In this work, we show that LST1/A, an incompletely characterized protein encoded by MHCIII locus, is a palmitoylated transmembrane adaptor protein. It is expressed specifically in leukocytes of the myeloid lineage, where it localizes to the tetraspanin-enriched microdomains. In addition, it binds SHP-1 and SHP-2 phosphatases in a phosphotyrosine-dependent manner, facilitating their recruitment to the plasma membrane. These data suggest a role for LST1/A in negative regulation of signal propagation.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Myeloid Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Amino Acid Sequence , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Major Histocompatibility Complex/physiology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Myeloid Cells/cytology , Plakins/metabolism , Primary Cell Culture , Protein Structure, Tertiary/physiology , Protein Transport/physiology , Pseudopodia/metabolism , Signal Transduction/physiology , U937 Cells
11.
J Biol Chem ; 286(25): 22101-12, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21543337

ABSTRACT

CD148 is a receptor-like protein-tyrosine phosphatase known to inhibit transduction of mitogenic signals in non-hematopoietic cells. Similarly, in the hematopoietic lineage, CD148 inhibited signal transduction downstream of T cell receptor. However, it also augmented immunoreceptor signaling in B cells and macrophages via dephosphorylating C-terminal tyrosine of Src family kinases (SFK). Accordingly, endogenous CD148 compensated for the loss of the main SFK activator CD45 in murine B cells and macrophages but not in T cells. Hypothetical explanations for the difference between T cells and other leukocyte lineages include the inability of CD148 to dephosphorylate a specific set of SFKs involved in T cell activation or the lack of CD148 expression during critical stages of T cell development. Here we describe striking differences in CD148 expression between human and murine thymocyte subsets, the only unifying feature being the absence of CD148 during the positive selection when the major developmental block occurs under CD45 deficiency. Moreover, we demonstrate that similar to CD45, CD148 has both activating and inhibitory effects on the SFKs involved in TCR signaling. However, in the absence of CD45, activating effects prevail, resulting in functional complementation of CD45 deficiency in human T cell lines. Importantly, this is independent of the tyrosines in the CD148 C-terminal tail, contradicting the recently proposed phosphotyrosine displacement model as a mechanism of SFK activation by CD148. Collectively, our data suggest that differential effects of CD148 in T cells and other leukocyte subsets cannot be explained by the CD148 inability to activate T cell SFKs but rather by its dual inhibitory/activatory function and specific expression pattern.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , Signal Transduction , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Enzyme Activation , Gene Expression Regulation, Enzymologic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Humans , Jurkat Cells , Leukocyte Common Antigens/deficiency , Membrane Proteins/metabolism , Mice , Phospholipase C gamma/metabolism , Phosphorylation , Protein Structure, Tertiary , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Tyrosine/metabolism , src-Family Kinases/chemistry
12.
J Biol Chem ; 286(22): 19617-29, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21460222

ABSTRACT

Transmembrane adaptor proteins (TRAPs) are important organizers and regulators of immunoreceptor-mediated signaling. A bioinformatic search revealed several potential novel TRAPs, including a highly conserved protein, proline rich 7 (PRR7), previously described as a component of the PSD-95/N-methyl-d-aspartate receptor protein complex in postsynaptic densities (PSD) of rat neurons. Our data demonstrate that PRR7 is weakly expressed in other tissues but is readily up-regulated in activated human peripheral blood lymphocytes. Transient overexpression of PRR7 in Jurkat T cell line led to gradual apoptotic death dependent on the WW domain binding motif surrounding Tyr-166 in the intracellular part of PRR7. To circumvent the pro-apoptotic effect of PRR7, we generated Jurkat clones with inducible expression of PRR7 (J-iPRR7). In these cells acute induction of PRR7 expression had a dual effect. It resulted in up-regulation of the transcription factor c-Jun and the activation marker CD69 as well as enhanced production of IL-2 after phorbol 12-myristate 13-acetate (PMA) and ionomycin treatment. On the other hand, expression of PRR7 inhibited general tyrosine phosphorylation and calcium influx after T cell receptor cross-linking by antibodies. Moreover, we found PRR7 constitutively tyrosine-phosphorylated and associated with Src. Collectively, these data indicate that PRR7 is a potential regulator of signaling and apoptosis in activated T cells.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Apoptosis/physiology , Calcium Signaling/physiology , Gene Expression Regulation/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Motifs , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Apoptosis/drug effects , Caco-2 Cells , Calcium Signaling/drug effects , Carcinogens/pharmacology , HEK293 Cells , Humans , Interleukin-2/biosynthesis , Interleukin-2/genetics , Interleukin-2/immunology , Ionomycin/pharmacology , Ionophores/pharmacology , Jurkat Cells , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/immunology , Proto-Oncogene Proteins c-jun/metabolism , Rats , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Tetradecanoylphorbol Acetate/pharmacology , U937 Cells
13.
Nucleic Acids Res ; 36(15): e93, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18606615

ABSTRACT

Tetraalkylammonium (TAA) derivatives have been reported to serve as stabilizers of asymmetrical cyanine dyes in aqueous solutions and to increase the yield and efficiency of polymerase chain reaction (PCR) detected by end-point analysis. In this study, we compared the ability of various TAA derivatives (with alkyl chain ranging from 1 to 5 carbons) and some other compounds to serve as enhancers of real-time PCR based on fluorescence detection from intercalating dye SYBR Green I (SGI). Our data indicate that TAA chlorides and some other TAA derivatives serve as potent enhancers of SGI-monitored real-time PCR. Optimal results were obtained with 10-16 mM tetrapropylammonium chloride. The effect of TAA compounds was dependent on the nature of counter ions present and composition of the reaction mixtures used. Based on measurements of SGI-generated fluorescence signal in the presence of PCR-amplified DNA fragments, oligonucleotide primers and/or various additives, we propose that TAA-derivatives reduce the binding of SGI to oligonucleotide primers and thus enhance primer-template interactions during annealing phase. Furthermore, these compounds serve as stabilizers of SGI-containing PCR mixtures. The combined data indicate that TAA derivatives might be a new class of additives contributing to robustness of real-time PCR monitored by asymmetrical cyanine dye SGI.


Subject(s)
Fluorescent Dyes/analysis , Organic Chemicals/analysis , Polymerase Chain Reaction/methods , Quaternary Ammonium Compounds/chemistry , Animals , Benzothiazoles , Diamines , Mice , Mice, Inbred C57BL , Quinolines , Temperature
14.
Cell Signal ; 19(11): 2400-12, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17716862

ABSTRACT

Interleukin 6 (IL-6) is a pleiotropic cytokine that mediates a variety of functions, including induction of the acute-phase response in hepatocytes. IL-6 initiates its action by binding to its cell surface receptor, followed by activation of Janus kinases and tyrosine phosphorylation of the signal transducer and transcription factor (STAT) 3. Although it has been suggested that cholesterol- and sphingolipid-enriched membrane domains, called lipid rafts, and caveolin are involved in this process, their roles in the earliest stages of IL-6-mediated signaling are far from being understood. Here we show that pretreatment of HepG2 hepatoma cells with methyl-beta-cyclodextrin (MbetaCD), which removes cholesterol and destroys lipid rafts, inhibited tyrosine phosphorylation of STAT3 in IL-6-activated, but not PV-activated cells. Furthermore, when the cells were lysed under conditions preserving lipid rafts, no IL-6- or PV-induced phosphorylation of STAT3 was observed. Although most of the STAT3 was found in large MbetaCD-resistant assemblies in both non-activated and IL-6-activated cells, its association with lipid rafts was weak or undetectable. The extent of IL-6-induced tyrosine phosphorylation of STAT3 was comparable in cells expressing low or high levels of caveolin. Similar STAT3 transducer complexes were observed in freshly isolated rat hepatocytes. The combined data suggest that STAT3 tyrosine phosphorylation occurs in preformed transducer complexes that can be activated in the absence of intact lipid rafts or caveolin.


Subject(s)
Hepatocytes/metabolism , STAT3 Transcription Factor/metabolism , Actins/metabolism , Animals , Caveolins/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Separation , Cholesterol/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Detergents , Hepatocytes/drug effects , Humans , Interleukin-6/pharmacology , Male , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Phosphotyrosine/metabolism , Rats , Signal Transduction/drug effects , Solubility/drug effects , Subcellular Fractions/metabolism , Tubulin/metabolism , Vanadates/pharmacology
15.
Nat Cell Biol ; 20(12): 1389-1399, 2018 12.
Article in English | MEDLINE | ID: mdl-30420664

ABSTRACT

The linear-ubiquitin chain assembly complex (LUBAC) modulates signalling via various immune receptors. In tumour necrosis factor (TNF) signalling, linear (also known as M1) ubiquitin enables full gene activation and prevents cell death. However, the mechanisms underlying cell death prevention remain ill-defined. Here, we show that LUBAC activity enables TBK1 and IKKε recruitment to and activation at the TNF receptor 1 signalling complex (TNFR1-SC). While exerting only limited effects on TNF-induced gene activation, TBK1 and IKKε are essential to prevent TNF-induced cell death. Mechanistically, TBK1 and IKKε phosphorylate the kinase RIPK1 in the TNFR1-SC, thereby preventing RIPK1-dependent cell death. This activity is essential in vivo, as it prevents TNF-induced lethal shock. Strikingly, NEMO (also known as IKKγ), which mostly, but not exclusively, binds the TNFR1-SC via M1 ubiquitin, mediates the recruitment of the adaptors TANK and NAP1 (also known as AZI2). TANK is constitutively associated with both TBK1 and IKKε, while NAP1 is associated with TBK1. We discovered a previously unrecognized cell death checkpoint that is mediated by TBK1 and IKKε, and uncovered an essential survival function for NEMO, whereby it enables the recruitment and activation of these non-canonical IKKs to prevent TNF-induced cell death.


Subject(s)
I-kappa B Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , A549 Cells , Animals , Cell Death/drug effects , Cells, Cultured , HeLa Cells , Humans , Mice, Knockout , Phosphorylation/drug effects , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Ubiquitination/drug effects
16.
Nat Commun ; 9(1): 3910, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254289

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC), composed of HOIP, HOIL-1 and SHARPIN, is required for optimal TNF-mediated gene activation and to prevent cell death induced by TNF. Here, we demonstrate that keratinocyte-specific deletion of HOIP or HOIL-1 (E-KO) results in severe dermatitis causing postnatal lethality. We provide genetic and pharmacological evidence that the postnatal lethal dermatitis in HoipE-KO and Hoil-1E-KO mice is caused by TNFR1-induced, caspase-8-mediated apoptosis that occurs independently of the kinase activity of RIPK1. In the absence of TNFR1, however, dermatitis develops in adulthood, triggered by RIPK1-kinase-activity-dependent apoptosis and necroptosis. Strikingly, TRAIL or CD95L can redundantly induce this disease-causing cell death, as combined loss of their respective receptors is required to prevent TNFR1-independent dermatitis. These findings may have implications for the treatment of patients with mutations that perturb linear ubiquitination and potentially also for patients with inflammation-associated disorders that are refractory to inhibition of TNF alone.


Subject(s)
Carrier Proteins/metabolism , Dermatitis/metabolism , Fas Ligand Protein/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Newborn , Carrier Proteins/genetics , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Dermatitis/genetics , Intracellular Signaling Peptides and Proteins , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Skin/drug effects , Skin/metabolism , Skin/pathology , Ubiquitin-Protein Ligases/genetics
17.
FEBS J ; 283(14): 2626-39, 2016 07.
Article in English | MEDLINE | ID: mdl-26749412

ABSTRACT

Tumor necrosis factor (TNF) is a potent cytokine known for its involvement in inflammation, repression of tumorigenesis and activation of immune cells. Consequently, accurate regulation of the TNF signaling pathway is crucial for preventing the potent noxious effects of TNF. These pathological conditions include chronic inflammation, septic shock, cachexia and cancer. The TNF signaling cascade utilizes a complex network of post-translational modifications to control the cellular response following its activation. Next to phosphorylation, the ubiquitination of signaling complex components is probably the most important modification. This process is mediated by a specialist class of enzymes, the ubiquitin ligases. Equally important is the class of dedicated ubiquitin-specific proteases, the deubiquitinases. Together with ubiquitin binding proteins, this ubiquitination-deubiquitination system enables the dynamics of signaling complexes. In TNF signaling, these dynamics translate into the precise regulation of the induction of gene activation or cell death. Here, we review and discuss current knowledge of TNF signaling regulation by the ubiquitin system.


Subject(s)
Cell Death/genetics , Cell Death/physiology , Polyubiquitin/metabolism , Tumor Necrosis Factors/metabolism , Animals , Deubiquitinating Enzyme CYLD , Endopeptidases/metabolism , Humans , Models, Biological , Protein Multimerization , Receptors, Tumor Necrosis Factor, Type I/chemistry , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Transcriptional Activation , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitination
18.
Cell Death Dis ; 7(8): e2346, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27560715

ABSTRACT

Incontinentia Pigmenti (IP) is a rare X-linked disease characterized by early male lethality and multiple abnormalities in heterozygous females. IP is caused by NF-κB essential modulator (NEMO) mutations. The current mechanistic model suggests that NEMO functions as a crucial component mediating the recruitment of the IκB-kinase (IKK) complex to tumor necrosis factor receptor 1 (TNF-R1), thus allowing activation of the pro-survival NF-κB response. However, recent studies have suggested that gene activation and cell death inhibition are two independent activities of NEMO. Here we describe that cells expressing the IP-associated NEMO-A323P mutant had completely abrogated TNF-induced NF-κB activation, but retained partial antiapoptotic activity and exhibited high sensitivity to death by necroptosis. We found that robust caspase activation in NEMO-deficient cells is concomitant with RIPK3 recruitment to the apoptosis-mediating complex. In contrast, cells expressing the ubiquitin-binding mutant NEMO-A323P did not recruit RIPK3 to complex II, an event that prevented caspase activation. Hence NEMO, independently from NF-κB activation, represents per se a key component in the structural and functional dynamics of the different TNF-R1-induced complexes. Alteration of this process may result in differing cellular outcomes and, consequently, also pathological effects in IP patients with different NEMO mutations.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , Caspases/metabolism , Cell Death , Enzyme Activation , Fas-Associated Death Domain Protein/metabolism , Gene Knockout Techniques , I-kappa B Kinase/metabolism , Mice , Models, Biological , Mutant Proteins/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Kinases/metabolism
19.
Cell Rep ; 16(9): 2271-80, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27545878

ABSTRACT

Recruitment of the deubiquitinase CYLD to signaling complexes is mediated by its interaction with HOIP, the catalytically active component of the linear ubiquitin chain assembly complex (LUBAC). Here, we identify SPATA2 as a constitutive direct binding partner of HOIP that bridges the interaction between CYLD and HOIP. SPATA2 recruitment to TNFR1- and NOD2-signaling complexes is dependent on HOIP, and loss of SPATA2 abolishes CYLD recruitment. Deficiency in SPATA2 exerts limited effects on gene activation pathways but diminishes necroptosis induced by tumor necrosis factor (TNF), resembling loss of CYLD. In summary, we describe SPATA2 as a previously unrecognized factor in LUBAC-dependent signaling pathways that serves as an adaptor between HOIP and CYLD, thereby enabling recruitment of CYLD to signaling complexes.


Subject(s)
Macrophages/metabolism , Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Binding Sites , Cloning, Molecular , Deubiquitinating Enzyme CYLD , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Macrophages/cytology , Macrophages/drug effects , Mice , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Plasmids , Primary Cell Culture , Protein Binding , Proteins/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
20.
J Exp Med ; 213(12): 2671-2689, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27810922

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC), consisting of SHANK-associated RH-domain-interacting protein (SHARPIN), heme-oxidized IRP2 ubiquitin ligase-1 (HOIL-1), and HOIL-1-interacting protein (HOIP), is a critical regulator of inflammation and immunity. This is highlighted by the fact that patients with perturbed linear ubiquitination caused by mutations in the Hoip or Hoil-1 genes, resulting in knockouts of these proteins, may simultaneously suffer from immunodeficiency and autoinflammation. TLR3 plays a crucial, albeit controversial, role in viral infection and tissue damage. We identify a pivotal role of LUBAC in TLR3 signaling and discover a functional interaction between LUBAC components and TLR3 as crucial for immunity to influenza A virus infection. On the biochemical level, we identify LUBAC components as interacting with the TLR3-signaling complex (SC), thereby enabling TLR3-mediated gene activation. Absence of LUBAC components increases formation of a previously unrecognized TLR3-induced death-inducing SC, leading to enhanced cell death. Intriguingly, excessive TLR3-mediated cell death, induced by double-stranded RNA present in the skin of SHARPIN-deficient chronic proliferative dermatitis mice (cpdm), is a major contributor to their autoinflammatory skin phenotype, as genetic coablation of Tlr3 substantially ameliorated cpdm dermatitis. Thus, LUBAC components control TLR3-mediated innate immunity, thereby preventing development of immunodeficiency and autoinflammation.


Subject(s)
Immunologic Deficiency Syndromes/metabolism , Inflammation/pathology , Nerve Tissue Proteins/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Death/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Dermatitis/pathology , Gene Silencing/drug effects , Host-Pathogen Interactions/immunology , Humans , Inflammation/immunology , Influenza A virus/drug effects , Influenza A virus/physiology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Poly I-C/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 3/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL