Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(48): e2309205120, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-37988467

ABSTRACT

Constitutive activation of the MALT1 paracaspase in conventional T cells of Malt1TBM/TBM (TRAF6 Binding Mutant = TBM) mice causes fatal inflammation and autoimmunity, but the involved targets and underlying molecular mechanisms are unknown. We genetically rendered a single MALT1 substrate, the RNA-binding protein (RBP) Roquin-1, insensitive to MALT1 cleavage. These Rc3h1Mins/Mins mice showed normal immune homeostasis. Combining Rc3h1Mins/Mins alleles with those encoding for constitutively active MALT1 (TBM) prevented spontaneous T cell activation and restored viability of Malt1TBM/TBM mice. Mechanistically, we show how antigen/MHC recognition is translated by MALT1 into Roquin cleavage and derepression of Roquin targets. Increasing T cell receptor (TCR) signals inactivated Roquin more effectively, and only high TCR strength enabled derepression of high-affinity targets to promote Th17 differentiation. Induction of experimental autoimmune encephalomyelitis (EAE) revealed increased cleavage of Roquin-1 in disease-associated Th17 compared to Th1 cells in the CNS. T cells from Rc3h1Mins/Mins mice did not efficiently induce the high-affinity Roquin-1 target IκBNS in response to TCR stimulation, showed reduced Th17 differentiation, and Rc3h1Mins/Mins mice were protected from EAE. These data demonstrate how TCR signaling and MALT1 activation utilize graded cleavage of Roquin to differentially regulate target mRNAs that control T cell activation and differentiation as well as the development of autoimmunity.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental , Mice , Animals , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Inflammation/metabolism , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/genetics , Receptors, Antigen, T-Cell/genetics , Ubiquitin-Protein Ligases
2.
Front Immunol ; 14: 1111398, 2023.
Article in English | MEDLINE | ID: mdl-36761777

ABSTRACT

MALT1 is a core component of the CARD11-BCL10-MALT1 (CBM) signalosome, in which it acts as a scaffold and a protease to bridge T cell receptor (TCR) ligation to immune activation. As a scaffold, MALT1 binds to TRAF6, and T cell-specific TRAF6 ablation or destruction of MALT1-TRAF6 interaction provokes activation of conventional T (Tconv) effector cells. In contrast, MALT1 protease activity controls the development and suppressive function of regulatory T (Treg) cells in a T cell-intrinsic manner. Thus, complete loss of TRAF6 or selective inactivation of MALT1 catalytic function in mice skews the immune system towards autoimmune inflammation, but distinct mechanisms are responsible for these immune disorders. Here we demonstrate that TRAF6 deletion or MALT1 paracaspase inactivation are highly interdependent in causing the distinct immune pathologies. We crossed mice with T cell-specific TRAF6 ablation (Traf6-ΔT) and mice with a mutation rendering the MALT1 paracaspase dead in T cells (Malt1 PD-T) to yield Traf6-ΔT;Malt1 PD-T double mutant mice. These mice reveal that the autoimmune inflammation caused by TRAF6-ablation relies strictly on the function of the MALT1 protease to drive the activation of Tconv cells. Vice versa, despite the complete loss of Treg cells in Traf6-ΔT;Malt1 PD-T double mutant mice, inactivation of the MALT1 protease is unable to cause autoinflammation, because the Tconv effector cells are not activated in the absence of TRAF6. Consequentially, combined MALT1 paracaspase inactivation and TRAF6 deficiency in T cells mirrors the immunodeficiency seen upon T cell-specific MALT1 ablation.


Subject(s)
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Signal Transduction , TNF Receptor-Associated Factor 6 , Animals , Mice , Endopeptidases/metabolism , Homeostasis , Inflammation , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Peptide Hydrolases/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism
3.
Bio Protoc ; 12(10): e4423, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35813027

ABSTRACT

Although CRISPR-Cas9 genome editing can be performed directly in single-cell mouse zygotes, the targeting efficiency for more complex modifications such as the insertion of two loxP sites, multiple mutations in cis, or the precise insertion or deletion of longer DNA sequences often remains low (Cohen, 2016). Thus, targeting and validation of correct genomic modification in murine embryonic stem cells (ESCs) with subsequent injection into early-stage mouse embryos may still be preferable, allowing for large-scale screening in vitro before transfer of thoroughly characterized and genetically defined ESC clones into the germline. This procedure can result in a reduction of animal numbers with cost effectiveness and compliance with the 3R principle of animal welfare regulations. Here, we demonstrate that after transfection of homology templates and PX458 CRISPR-Cas9 plasmids, EGFP-positive ESCs can be sorted with a flow cytometer for the enrichment of CRISPR-Cas9-expressing cells. Cell sorting obviates antibiotic selection and therefore allows for more gentle culture conditions and faster outgrowth of ESC clones, which are then screened by qPCR for correct genomic modifications. qPCR screening is more convenient and less time-consuming compared to analyzing PCR samples on agarose gels. Positive ESC clones are validated by PCR analysis and sequencing and can serve for injection into early-stage mouse embryos for the generation of chimeric mice with germline transmission. Therefore, we describe here a simple and straightforward protocol for CRISPR-Cas9-directed gene targeting in ESCs. Graphical abstract.

4.
JCI Insight ; 7(4)2022 02 02.
Article in English | MEDLINE | ID: mdl-35108221

ABSTRACT

The molecular mechanisms that drive the acquisition of distinct neural crest cell (NCC) fates is still poorly understood. Here, we identified Prdm6 as an epigenetic modifier that temporally and spatially regulates the expression of NCC specifiers and determines the fate of a subset of migrating cardiac NCCs (CNCCs). Using transcriptomic analysis and genetic and fate mapping approaches in transgenic mice, we showed that disruption of Prdm6 was associated with impaired CNCC differentiation, delamination, and migration and led to patent ductus arteriosus (DA) and ventricular noncompaction. Bulk and single-cell RNA-Seq analyses of the DA and CNCCs identified Prdm6 as a regulator of a network of CNCC specification genes, including Wnt1, Tfap2b, and Sox9. Loss of Prdm6 in CNCCs diminished its expression in the pre-epithelial-mesenchymal transition (pre-EMT) cluster, resulting in the retention of NCCs in the dorsal neural tube. This defect was associated with diminished H4K20 monomethylation and G1-S progression and augmented Wnt1 transcript levels in pre-EMT and neural tube clusters, which we showed was the major driver of the impaired CNCC migration. Altogether, these findings revealed Prdm6 as a key regulator of CNCC differentiation and migration and identified Prdm6 and its regulated network as potential targets for the treatment of congenital heart diseases.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Neural Crest/pathology , Organogenesis/genetics , RNA/genetics , Repressor Proteins/genetics , Animals , Cell Differentiation , Cell Movement , Disease Models, Animal , Female , Heart Defects, Congenital/metabolism , Mice , Mice, Knockout , Neural Crest/metabolism , Repressor Proteins/metabolism
5.
Sci Immunol ; 6(65): eabh2095, 2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34767456

ABSTRACT

Balanced control of T cell signaling is critical for adaptive immunity and protection from autoimmunity. By combining genetically engineered mouse models, biochemical analyses and pharmacological interventions, we describe an unexpected dual role of the tumor necrosis factor receptor­associated factor 6 (TRAF6) E3 ligase as both a positive and negative regulator of mucosa-associated lymphoid tissue 1 (MALT1) paracaspase. Although MALT1-TRAF6 recruitment is indispensable for nuclear factor κB signaling in activated T cells, TRAF6 counteracts basal MALT1 protease activity in resting T cells. In mice, loss of TRAF6-mediated homeostatic suppression of MALT1 protease leads to severe autoimmune inflammation, which is completely reverted by genetic or therapeutic inactivation of MALT1 protease function. Thus, TRAF6 functions as a molecular brake for MALT1 protease in resting T cells and a signaling accelerator for MALT1 scaffolding in activated T cells, revealing that TRAF6 controls T cell activation in a switch-like manner. Our findings have important implications for development and treatment of autoimmune diseases.


Subject(s)
Homeostasis/immunology , Inflammation/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , TNF Receptor-Associated Factor 6/immunology , Animals , Female , Mice , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , TNF Receptor-Associated Factor 6/genetics
6.
Methods Mol Biol ; 2366: 125-143, 2021.
Article in English | MEDLINE | ID: mdl-34236636

ABSTRACT

Jurkat T cells have been of central importance for the discovery of signalling mediators driving NF-κB activation in response to T cell antigen receptor (TCR)/CD28 co-stimulation. The critical function of the key regulators identified in Jurkat T cells has subsequently been verified in primary murine and human T cells. CRISPR/Cas9-mediated genomic editing techniques in combination with viral reconstitution are powerful tools that now enable the investigation of the exact molecular mechanisms that govern T cell signalling, especially the impact of protein-protein interactions, protein modifications, or cancer-associated gain- or loss-of-function mutations. As exemplified by the CARD11 gene encoding a key regulator of NF-κB signalling in T cells, we describe here the detailed workflow for the generation of CRISPR/Cas9 knockout (KO) Jurkat T cells and the subsequent reconstitution using a lentiviral transduction protocol. In addition, we explain the use of a stable NF-κB-dependent EGFP reporter system that enables a reliable quantification of NF-κB transcriptional activation in the reconstituted KO Jurkat T cells.


Subject(s)
Leukemia, T-Cell/metabolism , Animals , Apoptosis Regulatory Proteins , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , HEK293 Cells , Humans , Jurkat Cells , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
8.
Front Pharmacol ; 11: 570939, 2020.
Article in English | MEDLINE | ID: mdl-33071784

ABSTRACT

BACKGROUND: The anticancer potential of pharmacologic ascorbic acid (AA) has been detected in a number of cancer cells. However, in vivo study suggested a strongly reduced cytotoxic activity of AA. It was known that pH could be a critical influencing factor for multiple anticancer treatments. In this study, we explored the influence of pH on the cytotoxicity of ascorbic acid. We employed castration-resistant prostate cancer (CRPC) cell lines PC3 and DU145 to observe the therapeutic effect of AA on PCa cells that were cultured with different pH in vitro. We also analyzed the influence of pH and extracellular oxidation on cytotoxicity of AA in cancer cells using reactive oxygen species (ROS) assay, cellular uptake of AA, and NADPH assay. Male BALB/c nude mice bearing prostate carcinoma xenografts (PC3 or DU145) were used to assess treatment response to AA with or without bicarbonate in vivo. The cellular uptake of AA in PCa xenografts was detected using positron emission tomography (PET). Small animal PET/CT scans were performed on mice after the administration of 6-deoxy-6-[18F] fluoro-L-ascorbic acid (18F-DFA). RESULTS: Our in vitro studies demonstrate that acidic pH attenuates the cytotoxic activity of pharmacologic ascorbic acid by inhibiting AA uptake in PCa cells. Additionally, we found that the cancer cell-selective toxicity of AA depends on ROS. In vivo, combination of AA and bicarbonate could provide a significant better therapeutic outcome in comparison with controls or AA single treated mice. 18F-DFA PET imaging illustrated that the treatment with NaHCO3 could significantly increase the AA uptake in tumor. CONCLUSIONS: The alkalinity of tumor microenvironment plays an important role in anticancer efficiency of AA in CRPC. 18F-DFA PET/CT imaging could predict the therapeutic response of PCa animal model through illustration of tumoral uptake of AA. 18F-DFA might be a potential PET tracer in clinical diagnosis and treatment for CRPC.

9.
Nat Commun ; 10(1): 2352, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138793

ABSTRACT

Regulatory T cells (Tregs) have crucial functions in the inhibition of immune responses. Their development and suppressive functions are controlled by the T cell receptor (TCR), but the TCR signaling mechanisms that mediate these effects remain ill-defined. Here we show that CARD11-BCL10-MALT1 (CBM) signaling mediates TCR-induced NF-κB activation in Tregs and controls the conversion of resting Tregs to effector Tregs under homeostatic conditions. However, in inflammatory milieus, cytokines can bypass the CBM requirement for this differentiation step. By contrast, CBM signaling, in a MALT1 protease-dependent manner, is essential for mediating the suppressive function of Tregs. In malignant melanoma models, acute genetic blockade of BCL10 signaling selectively in Tregs or pharmacological MALT1 inhibition enhances anti-tumor immune responses. Together, our data uncover a segregation of Treg differentiation and suppressive function at the CBM complex level, and provide a rationale to explore MALT1 inhibitors for cancer immunotherapy.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/immunology , CARD Signaling Adaptor Proteins/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Cell CLL-Lymphoma 10 Protein/metabolism , CARD Signaling Adaptor Proteins/metabolism , Cell Differentiation , Cytokines/immunology , Melanoma, Experimental , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
10.
Gut ; 67(1): 146-156, 2018 01.
Article in English | MEDLINE | ID: mdl-27646934

ABSTRACT

OBJECTIVE: The initial steps of pancreatic regeneration versus carcinogenesis are insufficiently understood. Although a combination of oncogenic Kras and inflammation has been shown to induce malignancy, molecular networks of early carcinogenesis remain poorly defined. DESIGN: We compared early events during inflammation, regeneration and carcinogenesis on histological and transcriptional levels with a high temporal resolution using a well-established mouse model of pancreatitis and of inflammation-accelerated KrasG12D-driven pancreatic ductal adenocarcinoma. Quantitative expression data were analysed and extensively modelled in silico. RESULTS: We defined three distinctive phases-termed inflammation, regeneration and refinement-following induction of moderate acute pancreatitis in wild-type mice. These corresponded to different waves of proliferation of mesenchymal, progenitor-like and acinar cells. Pancreas regeneration required a coordinated transition of proliferation between progenitor-like and acinar cells. In mice harbouring an oncogenic Kras mutation and challenged with pancreatitis, there was an extended inflammatory phase and a parallel, continuous proliferation of mesenchymal, progenitor-like and acinar cells. Analysis of high-resolution transcriptional data from wild-type animals revealed that organ regeneration relied on a complex interaction of a gene network that normally governs acinar cell homeostasis, exocrine specification and intercellular signalling. In mice with oncogenic Kras, a specific carcinogenic signature was found, which was preserved in full-blown mouse pancreas cancer. CONCLUSIONS: These data define a transcriptional signature of early pancreatic carcinogenesis and a molecular network driving formation of preneoplastic lesions, which allows for more targeted biomarker development in order to detect cancer earlier in patients with pancreatitis.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Acinar Cells/pathology , Acute Disease , Animals , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/genetics , Disease Models, Animal , Disease Progression , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Mesenchymal Stem Cells/pathology , Mice, Transgenic , Pancreas/physiology , Pancreatic Neoplasms/pathology , Pancreatitis/genetics , Pancreatitis/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Regeneration/genetics
11.
Nat Commun ; 7: 11292, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068814

ABSTRACT

MALT1 channels proximal T-cell receptor (TCR) signalling to downstream signalling pathways. With MALT1A and MALT1B two conserved splice variants exist and we demonstrate here that MALT1 alternative splicing supports optimal T-cell activation. Inclusion of exon7 in MALT1A facilitates the recruitment of TRAF6, which augments MALT1 scaffolding function, but not protease activity. Naive CD4(+) T cells express almost exclusively MALT1B and MALT1A expression is induced by TCR stimulation. We identify hnRNP U as a suppressor of exon7 inclusion. Whereas selective depletion of MALT1A impairs T-cell signalling and activation, downregulation of hnRNP U enhances MALT1A expression and T-cell activation. Thus, TCR-induced alternative splicing augments MALT1 scaffolding to enhance downstream signalling and to promote optimal T-cell activation.


Subject(s)
Alternative Splicing/genetics , CD4-Positive T-Lymphocytes/immunology , Caspases/genetics , Lymphocyte Activation/immunology , Neoplasm Proteins/genetics , Signal Transduction , Animals , Caspases/metabolism , Down-Regulation , Enzyme Activation , Exons/genetics , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Humans , Interleukin-2/biosynthesis , JNK Mitogen-Activated Protein Kinases/metabolism , Jurkat Cells , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , TNF Receptor-Associated Factor 6/metabolism , Th17 Cells/immunology , Up-Regulation
12.
PLoS One ; 10(11): e0143735, 2015.
Article in English | MEDLINE | ID: mdl-26618925

ABSTRACT

Type I interferon constitutes an essential component of the combinational therapy against viral disease. Acute pancreatitis is one side effect of type I interferon-based therapy, implying that activation of type I interferon signaling affects the homeostasis and integrity of pancreatic acinar cells. Here, we investigated the role of type I interferon signaling in pancreatic acinar cells using a caerulein-induced murine model of acute pancreatitis. Pancreas-specific ablation of interferon (alpha and beta) receptor 1 (Ifnar1) partially protected animals from caerulein-induced pancreatitis, as demonstrated by reduced tissue damage. Profiling of infiltrating immune cells revealed that this dampened tissue damage response correlated with the number of macrophages in the pancreas. Pharmacologic depletion of macrophages reversed the protective effect of Ifnar1 deficiency. Furthermore, expression of chemokine (C-C motif) ligand 2 (Ccl2), a potent factor for macrophage recruitment, was significantly increased in the Ifnar1-deficient pancreas. Thus, type I interferon signaling in pancreatic acinar cells controls pancreatic homeostasis by affecting the macrophage-mediated inflammatory response in the pancreas.


Subject(s)
Acinar Cells/metabolism , Pancreatitis, Acute Necrotizing/metabolism , Receptor, Interferon alpha-beta/genetics , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Pancreatitis, Acute Necrotizing/genetics , Pancreatitis, Acute Necrotizing/pathology , Receptor, Interferon alpha-beta/metabolism
13.
Cell Rep ; 9(4): 1292-305, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456129

ABSTRACT

The paracaspase Malt1 is a central regulator of antigen receptor signaling that is frequently mutated in human lymphoma. As a scaffold, it assembles protein complexes for NF-κB activation, and its proteolytic domain cleaves negative NF-κB regulators for signal enforcement. Still, the physiological functions of Malt1-protease are unknown. We demonstrate that targeted Malt1-paracaspase inactivation induces a lethal inflammatory syndrome with lymphocyte-dependent neurodegeneration in vivo. Paracaspase activity is essential for regulatory T cell (Treg) and innate-like B cell development, but it is largely dispensable for overcoming Malt1-dependent thresholds for lymphocyte activation. In addition to NF-κB inhibitors, Malt1 cleaves an entire set of mRNA stability regulators, including Roquin-1, Roquin-2, and Regnase-1, and paracaspase inactivation results in excessive interferon gamma (IFNγ) production by effector lymphocytes that drive pathology. Together, our results reveal distinct threshold and modulatory functions of Malt1 that differentially control lymphocyte differentiation and activation pathways and demonstrate that selective paracaspase blockage skews systemic immunity toward destructive autoinflammation.


Subject(s)
Autoimmunity , Caspases/metabolism , Inflammation/immunology , Inflammation/pathology , Neoplasm Proteins/metabolism , Animals , B-Lymphocytes/immunology , Caspases/deficiency , Cell Differentiation/immunology , Gene Expression Regulation , Homeostasis/immunology , Humans , Immunity, Mucosal/immunology , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , Mice, Mutant Strains , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/deficiency , Nerve Degeneration/immunology , Nerve Degeneration/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology
14.
J Allergy Clin Immunol ; 134(2): 276-84, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25087226

ABSTRACT

Next-generation DNA sequencing has accelerated the genetic characterization of many human primary immunodeficiency diseases (PIDs). These discoveries can be lifesaving for the affected patients and also provide a unique opportunity to study the effect of specific genes on human immune function. In the past 18 months, a number of independent groups have begun to define novel PIDs caused by defects in the caspase recruitment domain family, member 11 (CARD11)-B-cell chronic lymphocytic leukemia/lymphoma 10 (BCL10)-mucosa-associated lymphoid tissue lymphoma translocation gene 1 (MALT1 [CBM]) signalosome complex. The CBM complex forms an essential molecular link between the triggering of cell-surface antigen receptors and nuclear factor κB activation. Germline mutations affecting the CBM complex are now recognized as the cause of novel combined immunodeficiency phenotypes, which all share abnormal nuclear factor κB activation and dysregulated B-cell development as defining features. For this "Current perspectives" article, we have engaged experts in both basic biology and clinical immunology to capture the worldwide experience in recognizing and managing patients with PIDs caused by CBM complex mutations.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , B-Lymphocytes/pathology , CARD Signaling Adaptor Proteins/genetics , Caspases/genetics , Guanylate Cyclase/genetics , Immunologic Deficiency Syndromes/genetics , Neoplasm Proteins/genetics , Adaptor Proteins, Signal Transducing/immunology , B-Cell CLL-Lymphoma 10 Protein , B-Lymphocytes/immunology , CARD Signaling Adaptor Proteins/immunology , Caspases/immunology , Gene Expression Regulation , Germ-Line Mutation , Guanylate Cyclase/immunology , High-Throughput Nucleotide Sequencing , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/genetics , NF-kappa B/immunology , Neoplasm Proteins/immunology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Signal Transduction
15.
Immunity ; 40(3): 389-99, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24631154

ABSTRACT

Recognition of cell death by the innate immune system triggers inflammatory responses. However, how these reactions are regulated is not well understood. Here, we identify the inhibitory C-type lectin receptor Clec12a as a specific receptor for dead cells. Both human and mouse Clec12a could physically sense uric acid crystals (monosodium urate, MSU), which are key danger signals for cell-death-induced immunity. Clec12a inhibited inflammatory responses to MSU in vitro, and Clec12a-deficient mice exhibited hyperinflammatory responses after being challenged with MSU or necrotic cells and after radiation-induced thymocyte killing in vivo. Thus, we identified a negative regulatory MSU receptor that controls noninfectious inflammation in response to cell death that has implications for autoimmunity and inflammatory disease.


Subject(s)
Cell Death , Inflammation/metabolism , Lectins, C-Type/metabolism , Receptors, Mitogen/metabolism , Uric Acid/metabolism , Animals , Cell Death/genetics , Cell Death/immunology , Cell Line , Inflammation/genetics , Inflammation/immunology , Lectins, C-Type/genetics , Mice , Mice, Knockout , Models, Biological , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Mitogen/genetics , Uric Acid/immunology
16.
PLoS One ; 8(11): e81833, 2013.
Article in English | MEDLINE | ID: mdl-24278461

ABSTRACT

Members of the PRDM protein family have been shown to play important roles during embryonic development. Previous in vitro and in situ analyses indicated a function of Prdm6 in cells of the vascular system. To reveal physiological functions of Prdm6, we generated conditional Prdm6-deficient mice. Complete deletion of Prdm6 results in embryonic lethality due to cardiovascular defects associated with aberrations in vascular patterning. However, smooth muscle cells could be regularly differentiated from Prdm6-deficient embryonic stem cells and vascular smooth muscle cells were present and proliferated normally in Prdm6-deficient embryos. Conditional deletion of Prdm6 in the smooth muscle cell lineage using a SM22-Cre driver line resulted in perinatal lethality due to hemorrhage in the lungs. We thus identified Prdm6 as a factor that is essential for the physiological control of cardiovascular development.


Subject(s)
Cardiovascular System/embryology , Repressor Proteins/physiology , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Body Patterning , Cell Differentiation , Cell Proliferation , DNA Primers , Mice , Mice, Knockout , Muscle, Smooth/cytology , Neovascularization, Physiologic , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Repressor Proteins/genetics
17.
J Allergy Clin Immunol ; 132(1): 151-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23727036

ABSTRACT

BACKGROUND: Combined immunodeficiency (CID) is characterized by severe recurrent infections with normal numbers of T and B lymphocytes but with deficient cellular and humoral immunity. Most cases are sporadic, but autosomal recessive inheritance has been described. In most cases, the cause of CID remains unknown. OBJECTIVE: We wanted to identify the genetic cause of CID in 2 siblings, the products of a first-cousin marriage, who experienced recurrent bacterial and candidal infections with bronchiectasis, growth delay, and early death. METHODS: We performed immunologic, genetic, and biochemical studies in the 2 siblings, their family members, and healthy controls. Reconstitution studies were performed with T cells from mucosa-associated lymphoid tissue lymphoma-translocation gene 1-deficient (Malt1(-/-)) mice. RESULTS: The numbers of circulating T and B lymphocytes were normal, but T-cell proliferation to antigens and antibody responses to vaccination were severely impaired in both patients. Whole genome sequencing of 1 patient and her parents, followed by DNA sequencing of family members and healthy controls, showed the presence in both patients of a homozygous missense mutation in MALT1 that resulted in loss of protein expression. Analysis of T cells that were available on one of the patients showed severely impaired IκBα degradation and IL-2 production after activation, 2 events that depend on MALT1. In contrast to wild-type human MALT1, the patients' MALT1 mutant failed to correct defective nuclear factor-κB activation and IL-2 production in MALT1-deficient mouse T cells. CONCLUSIONS: An autosomal recessive form of CID is associated with homozygous mutations in MALT1. If future patients are found to be similarly affected, they should be considered as candidates for allogeneic hematopoietic cell transplantation.


Subject(s)
Caspases/genetics , Mutation , Neoplasm Proteins/genetics , Severe Combined Immunodeficiency/genetics , Amino Acid Sequence , Animals , Caspases/analysis , Cells, Cultured , Child , Child, Preschool , Humans , I-kappa B Kinase/metabolism , Lymphocyte Activation , Molecular Sequence Data , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/analysis , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/metabolism
18.
J Exp Med ; 207(5): 1031-44, 2010 May 10.
Article in English | MEDLINE | ID: mdl-20439541

ABSTRACT

Peripheral T cell lymphomas (PTCLs) are highly aggressive malignancies with poor prognosis. Their molecular pathogenesis is not well understood and small animal models for the disease are lacking. Recently, the chromosomal translocation t(5;9)(q33;q22) generating the interleukin-2 (IL-2)-inducible T cell kinase (ITK)-spleen tyrosine kinase (SYK) fusion tyrosine kinase was identified as a recurrent event in PTCL. We show that ITK-SYK associates constitutively with lipid rafts in T cells and triggers antigen-independent phosphorylation of T cell receptor (TCR)-proximal proteins. These events lead to activation of downstream pathways and acute cellular outcomes that correspond to regular TCR ligation, including up-regulation of CD69 or production of IL-2 in vitro or deletion of thymocytes and activation of peripheral T cells in vivo. Ultimately, conditional expression of patient-derived ITK-SYK in mice induces highly malignant PTCLs with 100% penetrance that resemble the human disease. Our work demonstrates that constitutively enforced antigen receptor signaling can, in principle, act as a powerful oncogenic driver. Moreover, we establish a robust clinically relevant and genetically tractable model of human PTCL.


Subject(s)
Lymphoma, T-Cell, Peripheral/genetics , Protein-Tyrosine Kinases/metabolism , Animals , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 9/genetics , Disease Models, Animal , Embryonic Stem Cells/physiology , Gene Expression Regulation, Neoplastic , Humans , Interleukin-2/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lectins, C-Type/genetics , Lymphoma, T-Cell, Peripheral/pathology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/pathology , Mice , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/metabolism , Signal Transduction , Spleen/enzymology , Syk Kinase , Translocation, Genetic
19.
J Exp Med ; 206(11): 2313-20, 2009 Oct 26.
Article in English | MEDLINE | ID: mdl-19841089

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is the most common type of lymphoma in humans. The aggressive activated B cell-like (ABC) subtype of DLBCL is characterized by constitutive NF-kappaB activity and requires signals from CARD11, BCL10, and the paracaspase MALT1 for survival. CARD11, BCL10, and MALT1 are scaffold proteins that normally associate upon antigen receptor ligation. Signal-induced CARD11-BCL10-MALT1 (CBM) complexes couple upstream events to IkappaB kinase (IKK)/NF-kappaB activation. MALT1 also possesses a recently recognized proteolytic activity that cleaves and inactivates the negative NF-kappaB regulator A20 and BCL10 upon antigen receptor ligation. Yet, the relevance of MALT1 proteolytic activity for malignant cell growth is unknown. Here, we demonstrate preassembled CBM complexes and constitutive proteolysis of the two known MALT1 substrates in ABC-DLBCL, but not in germinal center B cell-like (GCB) DLBCL. ABC-DLBCL cell treatment with a MALT1 protease inhibitor blocks A20 and BCL10 cleavage, reduces NF-kappaB activity, and decreases the expression of NF-kappaB targets genes. Finally, MALT1 paracaspase inhibition results in death and growth retardation selectively in ABC-DLBCL cells. Thus, our results indicate a growth-promoting role for MALT1 paracaspase activity in ABC-DLBCL and suggest that a pharmacological MALT1 protease inhibition could be a promising approach for lymphoma treatment.


Subject(s)
Caspase Inhibitors , Cytotoxicity, Immunologic/drug effects , Lymphocyte Activation/drug effects , Lymphoma, Large B-Cell, Diffuse/enzymology , Lymphoma, Large B-Cell, Diffuse/pathology , Neoplasm Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA, Neoplasm/metabolism , Guanylate Cyclase/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Protein Binding/drug effects
20.
J Exp Med ; 205(6): 1317-29, 2008 Jun 09.
Article in English | MEDLINE | ID: mdl-18490492

ABSTRACT

CD40, a member of the tumor necrosis factor (TNF) receptor family, plays an essential role in T cell-dependent immune responses. Because CD40 is widely expressed on the surface of tumor cells in various B cell malignancies, deregulated CD40 signaling has been suggested to contribute to lymphomagenesis. In this study, we show that B cell-specific expression of a constitutively active CD40 receptor, in the form of a latent membrane protein 1 (LMP1)/CD40 chimeric protein, promoted an increase in the number of follicular and marginal zone B cells in secondary lymphoid organs in transgenic mice. The B cells displayed an activated phenotype, prolonged survival and increased proliferation, but were significantly impaired in T cell-dependent immune responses. Constitutive CD40 signaling in B cells induced selective and constitutive activation of the noncanonical NF-kappaB pathway and the mitogen-activated protein kinases Jnk and extracellular signal-regulated kinase. LMP1/CD40-expressing mice older than 12 mo developed B cell lymphomas of mono- or oligoclonal origin at high incidence, thus showing that the interplay of the signaling pathways induced by constitutive CD40 signaling is sufficient to initiate a tumorigenic process, ultimately leading to the development of B cell lymphomas.


Subject(s)
B-Lymphocytes/immunology , CD40 Antigens/immunology , Lymphoma, B-Cell/immunology , Lymphoma/physiopathology , NF-kappa B/physiology , Animals , CD40 Antigens/deficiency , CD40 Antigens/genetics , Crosses, Genetic , Germinal Center/immunology , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Transgenic , Signal Transduction , Spleen/immunology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...