Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 6(65): eabi4425, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34826258

ABSTRACT

Psoriasis is a chronic inflammatory skin disease arising from poorly defined pathological cross-talk between keratinocytes and the immune system. BCL10 (B cell lymphoma/leukemia 10) and MALT1 (mucosa-associated lymphoid tissue lymphoma translocation protein 1) are ubiquitously expressed inflammatory signaling proteins that can interact with the psoriasis susceptibility factor CARD14, but their functions in psoriasis are insufficiently understood. We report that although keratinocyte-intrinsic BCL10/MALT1 deletions completely rescue inflammatory skin pathology triggered by germline Card14 gain-of-function mutation in mice, the BCL10/MALT1 signalosome is unexpectedly not involved in the CARD14-dependent interleukin-17 receptor (IL-17R) proximal pathway. Instead, it plays a more pleiotropic role by amplifying keratinocyte responses to a series of inflammatory cytokines, including IL-17A, IL-1ß, and TNF. Moreover, selective keratinocyte-intrinsic activation of BCL10/MALT1 signaling with an artificial engager molecule is sufficient to initiate lymphocyte-mediated psoriasiform skin inflammation, and aberrant BCL10/MALT1 activity is frequently detected in the skin of human sporadic psoriasis. Together, these results establish that BCL10/MALT1 signalosomes can act as initiators and crucial amplifiers of psoriatic skin inflammation and indicate a critical function for this complex in sporadic psoriasis.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/immunology , Inflammation/immunology , Keratinocytes/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , Psoriasis/immunology , Skin/immunology , Animals , B-Cell CLL-Lymphoma 10 Protein/deficiency , B-Cell CLL-Lymphoma 10 Protein/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/deficiency , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics
2.
JCI Insight ; 4(15)2019 08 08.
Article in English | MEDLINE | ID: mdl-31391340

ABSTRACT

Aberrant activity of the glycoprotein 130 130/JAK/STAT3 (gp130/JAK/STAT3) signaling axis is a recurrent event in inflammation and cancer. In particular, it is associated with a wide range of hematological malignancies, including multiple myeloma and leukemia. Novel targeted therapies have only been successful for some subtypes of these malignancies, underlining the need for developing robust mouse models to better dissect the role of this pathway in specific tumorigenic processes. Here, we investigated the role of selective gp130/JAK/STAT3 activation by generating a conditional mouse model. This model targeted constitutively active, cell-autonomous gp130 activity to B cells, as well as to the entire hematopoietic system. We found that regardless of the timing of activation in B cells, constitutively active gp130 signaling resulted in the formation specifically of mature B cell lymphomas and plasma cell disorders with full penetrance, only with different latencies, where infiltrating CD138+ cells were a dominant feature in every tumor. Furthermore, constitutively active gp130 signaling in all adult hematopoietic cells also led to the development specifically of largely mature, aggressive B cell cancers, again with a high penetrance of CD138+ tumors. Importantly, gp130 activity abrogated the differentiation block induced by a B cell-targeted Myc transgene and resulted in a complete penetrance of the gp130-associated, CD138+, mature B cell lymphoma phenotype. Thus, gp130 signaling selectively provides a strong growth and differentiation advantage for mature B cells and directs lymphomagenesis specifically toward terminally differentiated B cell cancers.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Cytokine Receptor gp130/metabolism , Lymphoma/immunology , Plasmacytoma/immunology , Signal Transduction/immunology , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Differentiation/genetics , Cytokine Receptor gp130/genetics , Disease Models, Animal , Female , Humans , Janus Kinases/metabolism , Lymphocyte Activation/genetics , Lymphoma/genetics , Lymphoma/pathology , Male , Mice , Plasmacytoma/genetics , Plasmacytoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , STAT3 Transcription Factor/metabolism
3.
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
5.
Nature ; 552(7683): 121-125, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29143824

ABSTRACT

T cell non-Hodgkin lymphomas are a heterogeneous group of highly aggressive malignancies with poor clinical outcomes. T cell lymphomas originate from peripheral T cells and are frequently characterized by genetic gain-of-function variants in T cell receptor (TCR) signalling molecules. Although these oncogenic alterations are thought to drive TCR pathways to induce chronic proliferation and cell survival programmes, it remains unclear whether T cells contain tumour suppressors that can counteract these events. Here we show that the acute enforcement of oncogenic TCR signalling in lymphocytes in a mouse model of human T cell lymphoma drives the strong expansion of these cells in vivo. However, this response is short-lived and robustly counteracted by cell-intrinsic mechanisms. A subsequent genome-wide in vivo screen using T cell-specific transposon mutagenesis identified PDCD1, which encodes the inhibitory receptor programmed death-1 (PD-1), as a master gene that suppresses oncogenic T cell signalling. Mono- and bi-allelic deletions of PDCD1 are also recurrently observed in human T cell lymphomas with frequencies that can exceed 30%, indicating high clinical relevance. Mechanistically, the activity of PD-1 enhances levels of the tumour suppressor PTEN and attenuates signalling by the kinases AKT and PKC in pre-malignant cells. By contrast, a homo- or heterozygous deletion of PD-1 allows unrestricted T cell growth after an oncogenic insult and leads to the rapid development of highly aggressive lymphomas in vivo that are readily transplantable to recipients. Thus, the inhibitory PD-1 receptor is a potent haploinsufficient tumour suppressor in T cell lymphomas that is frequently altered in human disease. These findings extend the known physiological functions of PD-1 beyond the prevention of immunopathology after antigen-induced T cell activation, and have implications for T cell lymphoma therapies and for current strategies that target PD-1 in the broader context of immuno-oncology.


Subject(s)
Carcinogenesis/genetics , Genes, Tumor Suppressor , Haploinsufficiency/genetics , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Animals , Cells, Cultured , Female , Humans , Lymphoma, T-Cell/metabolism , Male , Mice , Mutation , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
6.
Eur J Immunol ; 47(8): 1342-1353, 2017 08.
Article in English | MEDLINE | ID: mdl-28586167

ABSTRACT

Inflammatory bowel diseases (IBD) are key risk factors for the development of colorectal cancer, but the mechanisms that link intestinal inflammation with carcinogenesis are insufficiently understood. Card9 is a myeloid cell-specific signaling protein that regulates inflammatory responses downstream of various pattern recognition receptors and which cooperates with the inflammasomes for IL-1ß production. Because polymorphisms in Card9 were recurrently associated with human IBD, we investigated the function of Card9 in a colitis-associated cancer (CAC) model. Card9-/- mice develop smaller, less proliferative and less dysplastic tumors compared to their littermates and in the regenerating mucosa we detected dramatically impaired IL-1ß generation and defective IL-1ß controlled IL-22 production from group 3 innate lymphoid cells. Consistent with the key role of immune-derived IL-22 in activating STAT3 signaling during normal and pathological intestinal epithelial cell (IEC) proliferation, Card9-/- mice also exhibit impaired tumor cell intrinsic STAT3 activation. Our results imply a Card9-controlled, ILC3-mediated mechanism regulating healthy and malignant IEC proliferation and demonstrates a role of Card9-mediated innate immunity in inflammation-associated carcinogenesis.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Carcinogenesis , Colitis/immunology , Colorectal Neoplasms/etiology , Interleukin-1beta/immunology , Interleukins/biosynthesis , Lymphocyte Subsets/immunology , Animals , CARD Signaling Adaptor Proteins/deficiency , CARD Signaling Adaptor Proteins/genetics , Cell Proliferation , Colitis/complications , Colitis/physiopathology , Colorectal Neoplasms/immunology , Immunity, Innate , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammatory Bowel Diseases/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukins/genetics , Interleukins/immunology , Intestines/cytology , Intestines/pathology , Mice , STAT3 Transcription Factor/metabolism , Signal Transduction , Interleukin-22
7.
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
8.
J Clin Invest ; 124(12): 5263-74, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25384216

ABSTRACT

Multiple myeloma (MM) is a plasma cell neoplasm that results from clonal expansion of an Ig-secreting terminally differentiated B cell. Advanced MM is characterized by tissue damage that involves bone, kidney, and other organs and is typically associated with recurrent genetic abnormalities. IL-6 signaling via the IL-6 signal transducer GP130 has been implicated as an important driver of MM pathogenesis. Here, we demonstrated that ectopic expression of constitutively active GP130 (L-GP130) in a murine retroviral transduction-transplantation model induces rapid MM development of high penetrance. L-GP130-expressing mice recapitulated all of the characteristics of human disease, including monoclonal gammopathy, BM infiltration with lytic bone lesions, and protein deposition in the kidney. Moreover, the disease was easily transplantable and allowed different therapeutic options to be evaluated in vitro and in vivo. Using this model, we determined that GP130 signaling collaborated with MYC to induce MM and was responsible and sufficient for directing the plasma cell phenotype. Accordingly, we identified Myc aberrations in the L-GP130 MM model. Evaluation of human MM samples revealed recurrent activation of STAT3, a downstream target of GP130 signaling. Together, our results indicate that deregulated GP130 activity contributes to MM pathogenesis and that pathways downstream of GP130 activity have potential as therapeutic targets in MM.


Subject(s)
Cytokine Receptor gp130/metabolism , Multiple Myeloma/metabolism , Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line, Tumor , Cytokine Receptor gp130/genetics , Female , Humans , Male , Mice , Mice, Inbred BALB C , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Plasma Cells/metabolism , Plasma Cells/pathology , Proto-Oncogene Proteins c-myc/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics
9.
J Immunol ; 192(3): 1024-33, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24376268

ABSTRACT

The development of hematopoietic neoplasms is often associated with mutations, altered gene expression or chromosomal translocations. Recently, the t(5, 9)(q33;q22) translocation was found in a subset of peripheral T cell lymphomas and was shown to result in an IL-2-inducible kinase-spleen tyrosine kinase (ITK-Syk) fusion transcript. In this study, we show that T cell-specific expression of the ITK-Syk oncogene in mice leads to an early onset and aggressive polyclonal T cell lymphoproliferation with concomitant B cell expansion and systemic inflammation by 7-9 wk of age. Because this phenotype is strikingly different from previous work showing that ITK-Syk expression causes clonal T cell lymphoma by 20-27 wk of age, we investigated the underlying molecular mechanism in more detail. We show that the reason for the severe phenotype is the lack of B-lymphocyte-induced maturation protein-1 (Blimp-1) induction by low ITK-Syk expression. In contrast, high ITK-Syk oncogene expression induces terminal T cell differentiation in the thymus by activating Blimp-1, thereby leading to elimination of oncogene-expressing cells early in development. Our data suggest that terminal differentiation is an important mechanism to prevent oncogene-expressing cells from malignant transformation, as high ITK-Syk oncogene activity induces cell elimination. Accordingly, for transformation, a specific amount of oncogene is required, or alternatively, the induction of terminal differentiation is defective.


Subject(s)
Inflammation/etiology , Intracellular Signaling Peptides and Proteins/physiology , Lymphocyte Activation/immunology , Lymphopoiesis/immunology , Lymphoproliferative Disorders/etiology , Oncogene Proteins, Fusion/physiology , Protein-Tyrosine Kinases/physiology , T-Lymphocyte Subsets/pathology , Age Factors , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Cells, Cultured , Chimera , Cytokines/blood , DNA, Complementary/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hypergammaglobulinemia/etiology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/genetics , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Phosphorylation , Positive Regulatory Domain I-Binding Factor 1 , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins , STAT3 Transcription Factor/metabolism , Syk Kinase , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/immunology , Thymus Gland/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transduction, Genetic , Translocation, Genetic
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...