Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
JCI Insight ; 7(13)2022 07 08.
Article in English | MEDLINE | ID: mdl-35801586

ABSTRACT

IL-1 receptor-activated kinase 1 (IRAK1) is involved in signal transduction downstream of many TLRs and the IL-1R. Its potential as a drug target for chronic inflammatory diseases is underappreciated. To study its functional role in joint inflammation, we generated a mouse model expressing a functionally inactive IRAK1 (IRAK1 kinase deficient, IRAK1KD), which also displayed reduced IRAK1 protein expression and cell type-specific deficiencies of TLR signaling. The serum transfer model of arthritis revealed a potentially novel role of IRAK1 for disease development and neutrophil chemoattraction exclusively via its activity in nonhematopoietic cells. Consistently, IRAK1KD synovial fibroblasts showed reduced secretion of neutrophil chemoattractant chemokines following stimulation with IL-1ß or human synovial fluids from patients with rheumatoid arthritis (RA) and gout. Together with patients with RA showing prominent IRAK1 expression in fibroblasts of the synovial lining, these data suggest that targeting IRAK1 may be therapeutically beneficial. As pharmacological inhibition of IRAK1 kinase activity had only mild effects on synovial fibroblasts from mice and patients with RA, targeted degradation of IRAK1 may be the preferred pharmacologic modality. Collectively, these data position IRAK1 as a central regulator of the IL-1ß-dependent local inflammatory milieu of the joints and a potential therapeutic target for inflammatory arthritis.


Subject(s)
Arthritis, Rheumatoid , Interleukin-1 Receptor-Associated Kinases , Neutrophils , Synovial Membrane , Animals , Arthritis, Rheumatoid/metabolism , Cells, Cultured , Disease Models, Animal , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-8/metabolism , Mice , Neutrophils/metabolism , Synovial Membrane/metabolism
2.
Oncogenesis ; 10(4): 32, 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33824280

ABSTRACT

CARD-CC complexes involving BCL10 and MALT1 are major cellular signaling hubs. They govern NF-κB activation through their scaffolding properties as well as MALT1 paracaspase function, which cleaves substrates involved in NF-κB regulation. In human lymphocytes, gain-of-function defects in this pathway lead to lymphoproliferative disorders. CARD10, the prototypical CARD-CC protein in non-hematopoietic cells, is overexpressed in several cancers and has been associated with poor prognosis. However, regulation of CARD10 remains poorly understood. Here, we identified CARD10 as the first MALT1 substrate in non-hematopoietic cells and showed that CARD10 cleavage by MALT1 at R587 dampens its capacity to activate NF-κB. Preventing CARD10 cleavage in the lung tumor A549 cell line increased basal levels of IL-6 and extracellular matrix components in vitro, and led to increased tumor growth in a mouse xenograft model, suggesting that CARD10 cleavage by MALT1 might be a built-in mechanism controlling tumorigenicity.

3.
J Med Chem ; 63(23): 14594-14608, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33216547

ABSTRACT

The paracaspase MALT1 has gained increasing interest as a target for the treatment of subsets of lymphomas as well as autoimmune diseases, and there is a need for suitable compounds to explore the therapeutic potential of this target. Here, we report the optimization of the in vivo potency of pyrazolopyrimidines, a class of highly selective allosteric MALT1 inhibitors. High doses of the initial lead compound led to tumor stasis in an activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) xenograft model, but this compound suffered from a short in vivo half-life and suboptimal potency in whole blood. Guided by metabolism studies, we identified compounds with reduced metabolic clearance and increased in vivo half-life. In the second optimization step, masking one of the hydrogen-bond donors of the central urea moiety through an intramolecular interaction led to improved potency in whole blood. This was associated with improved in vivo potency in a mechanistic model of B cell activation. The optimized compound led to tumor regression in a CARD11 mutant ABC-DLBCL lymphoma xenograft model.


Subject(s)
Blood/metabolism , Caspase Inhibitors/therapeutic use , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Urea/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Caspase Inhibitors/chemical synthesis , Caspase Inhibitors/metabolism , Caspase Inhibitors/pharmacokinetics , Cell Line, Tumor , Female , Half-Life , Humans , Mice, Inbred BALB C , Mice, SCID , Microsomes, Liver/metabolism , Neoplasms/drug therapy , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Rats, Sprague-Dawley , Sheep , Urea/chemical synthesis , Urea/metabolism , Urea/pharmacokinetics , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 19(10): 2089-2104, 2020 10.
Article in English | MEDLINE | ID: mdl-32847974

ABSTRACT

The sole inhibitory Fcγ receptor CD32b (FcγRIIb) is expressed throughout B and plasma cell development and on their malignant counterparts. CD32b expression on malignant B cells is known to provide a mechanism of resistance to rituximab that can be ameliorated with a CD32b-blocking antibody. CD32b, therefore, represents an attractive tumor antigen for targeting with a monoclonal antibody (mAb). To this end, two anti-CD32b mAbs, NVS32b1 and NVS32b2, were developed. Their complementarity-determining regions (CDR) bind the CD32b Fc binding domain with high specificity and affinity while the Fc region is afucosylated to enhance activation of FcγRIIIa on immune effector cells. The NVS32b mAbs selectively target CD32b+ malignant cells and healthy B cells but not myeloid cells. They mediate potent killing of opsonized CD32b+ cells via antibody-dependent cellular cytotoxicity and phagocytosis (ADCC and ADCP) as well as complement-dependent cytotoxicity (CDC). In addition, NVS32b CDRs block the CD32b Fc-binding domain, thereby minimizing CD32b-mediated resistance to therapeutic mAbs including rituximab, obinutuzumab, and daratumumab. NVS32b mAbs demonstrate robust antitumor activity against CD32b+ xenografts in vivo and immunomodulatory activity including recruitment of macrophages to the tumor and enhancement of dendritic cell maturation in response to immune complexes. Finally, the activity of NVS32b mAbs on CD32b+ primary malignant B and plasma cells was confirmed using samples from patients with B-cell chronic lymphocytic leukemia (CLL) and multiple myeloma. The findings indicate the promising potential of NVS32b mAbs as a single agent or in combination with other mAb therapeutics for patients with CD32b+ malignant cells.


Subject(s)
Lymphoma, B-Cell/genetics , Neoplasms, Plasma Cell/genetics , Receptors, IgG/immunology , Animals , CHO Cells , Cricetulus , Humans
5.
Front Immunol ; 11: 745, 2020.
Article in English | MEDLINE | ID: mdl-32425939

ABSTRACT

Genetic disruption or short-term pharmacological inhibition of MALT1 protease is effective in several preclinical models of autoimmunity and B cell malignancies. Despite these protective effects, the severe reduction in regulatory T cells (Tregs) and the associated IPEX-like pathology occurring upon congenital disruption of the MALT1 protease in mice has raised concerns about the long-term safety of MALT1 inhibition. Here we describe the results of a series of toxicology studies in rat and dog species using MLT-943, a novel potent and selective MALT1 protease inhibitor. While MLT-943 effectively prevented T cell-dependent B cell immune responses and reduced joint inflammation in the collagen-induced arthritis rat pharmacology model, in both preclinical species, pharmacological inhibition of MALT1 was associated with a rapid and dose-dependent reduction in Tregs and resulted in the progressive appearance of immune abnormalities and clinical signs of an IPEX-like pathology. At the 13-week time point, rats displayed severe intestinal inflammation associated with mast cell activation, high serum IgE levels, systemic T cell activation and mononuclear cell infiltration in multiple tissues. Importantly, using thymectomized rats we demonstrated that MALT1 protease inhibition affects peripheral Treg frequency independently of effects on thymic Treg output and development. Our data confirm the therapeutic potential of MALT1 protease inhibitors but highlight the safety risks and challenges to consider before potential application of such inhibitors into the clinic.


Subject(s)
Diabetes Mellitus, Type 1/congenital , Diarrhea/etiology , Genetic Diseases, X-Linked/etiology , Immune System Diseases/congenital , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Animals , Diabetes Mellitus, Type 1/etiology , Dogs , Female , Humans , Immune System Diseases/etiology , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Lew , Rats, Wistar , T-Lymphocytes, Regulatory/immunology
6.
Arthritis Rheumatol ; 72(6): 919-930, 2020 06.
Article in English | MEDLINE | ID: mdl-31943941

ABSTRACT

OBJECTIVE: Fcγ receptors (FcγR) play important roles in both protective and pathogenic immune responses. The assembly of the CBM signalosome encompassing caspase recruitment domain-containing protein 9, B cell CLL/lymphoma 10, and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT-1) is required for optimal FcγR-induced canonical NF-κB activation and proinflammatory cytokine release. This study was undertaken to clarify the relevance of MALT-1 protease activity in FcγR-driven events and evaluate the therapeutic potential of selective MALT-1 protease inhibitors in FcγR-mediated diseases. METHODS: Using genetic and pharmacologic disruption of MALT-1 scaffolding and enzymatic activity, we assessed the relevance of MALT-1 function in murine and human primary myeloid cells upon stimulation with immune complexes (ICs) and in murine models of autoantibody-driven arthritis and immune thrombocytopenic purpura (ITP). RESULTS: MALT-1 protease function is essential for optimal FcγR-induced production of proinflammatory cytokines by various murine and human myeloid cells stimulated with ICs. In contrast, MALT-1 protease inhibition did not affect the Syk-dependent, FcγR-mediated production of reactive oxygen species or leukotriene B4 . Notably, pharmacologic MALT-1 protease inhibition in vivo reduced joint inflammation in the murine K/BxN serum-induced arthritis model (mean area under the curve for paw swelling of 45.42% versus 100% in control mice; P = 0.0007) but did not affect platelet depletion in a passive model of ITP. CONCLUSION: Our findings indicate a specific contribution of MALT-1 protease activity to FcγR-mediated events and suggest that MALT-1 protease inhibitors have therapeutic potential in a subset of FcγR-driven inflammatory disorders.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , Receptors, IgG/immunology , Animals , Antigen-Antibody Complex/metabolism , Blood Platelets/metabolism , Cytokines/immunology , Disease Models, Animal , Humans , Mice , Myeloid Cells/metabolism
7.
J Immunol ; 203(11): 2791-2806, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31659015

ABSTRACT

The paracaspase Malt1 is a key regulator of canonical NF-κB activation downstream of multiple receptors in both immune and nonimmune cells. Genetic disruption of Malt1 protease function in mice and MALT1 mutations in humans results in reduced regulatory T cells and a progressive multiorgan inflammatory pathology. In this study, we evaluated the altered immune homeostasis and autoimmune disease in Malt1 protease-deficient (Malt1PD) mice and the Ags driving disease manifestations. Our data indicate that B cell activation and IgG1/IgE production is triggered by microbial and dietary Ags preferentially in lymphoid organs draining mucosal barriers, likely as a result of dysregulated mucosal immune homeostasis. Conversely, the disease was driven by a polyclonal T cell population directed against self-antigens. Characterization of the Malt1PD T cell compartment revealed expansion of T effector memory cells and concomitant loss of a CD4+ T cell population that phenotypically resembles anergic T cells. Therefore, we propose that the compromised regulatory T cell compartment in Malt1PD animals prevents the efficient maintenance of anergy and supports the progressive expansion of pathogenic, IFN-γ-producing T cells. Overall, our data revealed a crucial role of the Malt1 protease for the maintenance of intestinal and systemic immune homeostasis, which might provide insights into the mechanisms underlying IPEX-related diseases associated with mutations in MALT1.


Subject(s)
Autoimmunity/immunology , Homeostasis/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/deficiency , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics
8.
J Vis Exp ; (143)2019 01 07.
Article in English | MEDLINE | ID: mdl-30663686

ABSTRACT

Besides its function in lymphoid cells, which has been addressed by numerous studies, the paracaspase MALT1 also plays an important role in innate cells downstream of pattern recognition receptors. Best studied are the Dectin-1 and Dectin-2 members of the C-type lectin-like receptor family that induce a SYK- and CARD9-dependent signaling cascade leading to NF-κB activation, in a MALT1-dependent manner. By contrast, Toll-like receptors (TLR), such as TLR-4, propagate NF-κB activation but signal via an MYD88/IRAK-dependent cascade. Nonetheless, whether MALT1 might contribute to TLR-4 signaling has remained unclear. Recent evidence with MLT-827, a potent and selective inhibitor of MALT1 paracaspase activity, indicates that TNF- production downstream of TLR-4 in human myeloid cells is independent of MALT1, as opposed to TNF- production downstream of Dectin-1, which is MALT1 dependent. Here, we addressed the selective involvement of MALT1 in pattern recognition sensing further, using a variety of human and mouse cellular preparations, and stimulation of Dectin-1, MINCLE or TLR-4 pathways. We also provided additional insights by exploring cytokines beyond TNF-, and by comparing MLT-827 to a SYK inhibitor (Cpd11) and to an IKK inhibitor (AFN700). Collectively, the data provided further evidence for the MALT1-dependency of C-type lectin-like receptor -signaling by contrast to TLR-signaling.


Subject(s)
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Animals , Humans , Mice , Signal Transduction
9.
Immunol Cell Biol ; 96(1): 81-99, 2018 01.
Article in English | MEDLINE | ID: mdl-29359407

ABSTRACT

Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is essential for immune responses triggered by antigen receptors but the contribution of its paracaspase activity is not fully understood. Here, we studied how MALT1 proteolytic function regulates T-cell activation and fate after engagement of the T-cell receptor pathway. We show that MLT-827, a potent and selective MALT1 paracaspase inhibitor, does not prevent the initial phase of T-cell activation, in contrast to the pan-protein kinase C inhibitor AEB071. However, MLT-827 strongly impacted cell expansion after activation. We demonstrate this is the consequence of profound inhibition of IL-2 production as well as reduced expression of the IL-2 receptor alpha subunit (CD25), resulting from defective canonical NF-κB activation and accelerated mRNA turnover mechanisms. Accordingly, MLT-827 revealed a unique transcriptional fingerprint of MALT1 protease activity, providing evidence for broad control of T-cell signaling pathways. Altogether, this first report with a potent and selective inhibitor elucidates how MALT1 paracaspase activity integrates several T-cell activation pathways and indirectly controls gamma-chain receptor dependent survival, to impact on T-cell expansion.


Subject(s)
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/metabolism , T-Lymphocytes/immunology , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Expression Regulation , Humans , Immunomodulation , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Proteolysis , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
11.
PLoS One ; 12(3): e0173176, 2017.
Article in English | MEDLINE | ID: mdl-28257518

ABSTRACT

Although the role of T cells in autoimmunity has been explored for many years, the mechanisms leading to the initial priming of an autoimmune T cell response remain enigmatic. The 'hit and run' model suggests that self-antigens released upon cell death can provide the initial signal for a self-sustaining autoimmune response. Using a novel transgenic mouse model where we could induce the release of self-antigens via caspase-dependent apoptosis. We tracked the fate of CD8+ T cells specific for the self-antigen. Our studies demonstrated that antigens released from apoptotic cells were cross-presented by CD11c+ cells in the draining lymph node. This cross-presentation led to proliferation of self-antigen specific T cells, followed by a transient ability to produce IFN-γ, but did not lead to the development of autoimmune diabetes. Using this model we examined the consequences on T cell immunity when apoptosis was combined with dendritic cell maturation signals, an autoimmune susceptible genetic background, and the deletion of Tregs. The results of our study demonstrate that autoimmune diabetes cannot be initiated by the presentation of antigens released from apoptotic cells in vivo even in the presence of factors known to promote autoimmunity.


Subject(s)
Autoantigens/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Apoptosis/genetics , Autoantigens/immunology , Autoimmunity/genetics , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Humans , Immune Tolerance , Interferon-gamma/genetics , Mice , Mice, Transgenic/immunology
12.
PLoS One ; 12(1): e0169026, 2017.
Article in English | MEDLINE | ID: mdl-28052131

ABSTRACT

The paracaspase MALT1 has arginine-directed proteolytic activity triggered by engagement of immune receptors. Recruitment of MALT1 into activation complexes is required for MALT1 proteolytic function. Here, co-expression of MALT1 in HEK293 cells, either with activated CARD11 and BCL10 or with TRAF6, was used to explore the mechanism of MALT1 activation at the molecular level. This work identified a prominent self-cleavage site of MALT1 isoform A (MALT1A) at R781 (R770 in MALT1B) and revealed that TRAF6 can activate MALT1 independently of the CBM. Intramolecular cleavage at R781/R770 removes a C-terminal TRAF6-binding site in both MALT1 isoforms, leaving MALT1B devoid of the two key interaction sites with TRAF6. A previously identified auto-proteolysis site of MALT1 at R149 leads to deletion of the death-domain, thereby abolishing interaction with BCL10. By using MALT1 isoforms and cleaved fragments thereof, as well as TRAF6 WT and mutant forms, this work shows that TRAF6 induces N-terminal auto-proteolytic cleavage of MALT1 at R149 and accelerates MALT1 protein turnover. The MALT1 fragment generated by N-terminal self-cleavage at R149 was labile and displayed enhanced signaling properties that required an intact K644 residue, previously shown to be a site for mono-ubiquitination of MALT1. Conversely, C-terminal self-cleavage at R781/R770 hampered the ability for self-cleavage at R149 and stabilized MALT1 by hindering interaction with TRAF6. C-terminal self-cleavage had limited impact on MALT1A but severely reduced MALT1B proteolytic and signaling functions. It also abrogated NF-κB activation by N-terminally cleaved MALT1A. Altogether, this study provides further insights into mechanisms that regulate the scaffolding and activation cycle of MALT1. It also emphasizes the reduced functional capacity of MALT1B as compared to MALT1A.


Subject(s)
Caspases/metabolism , Neoplasm Proteins/metabolism , Protein Isoforms/metabolism , T-Lymphocytes/metabolism , TNF Receptor-Associated Factor 6/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , B-Cell CLL-Lymphoma 10 Protein , Blotting, Western , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Caspases/genetics , Cell Line , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , HEK293 Cells , Humans , Immunoblotting , Jurkat Cells , Lymphocytes/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Mutagenesis , Neoplasm Proteins/genetics , Protein Isoforms/genetics , Signal Transduction/genetics , Signal Transduction/physiology , TNF Receptor-Associated Factor 6/genetics , Ubiquitination/genetics , Ubiquitination/physiology
13.
Sci Transl Med ; 8(367): 367ra166, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27903862

ABSTRACT

Interleukin-2 (IL-2) immunotherapy is an attractive approach in treating advanced cancer. However, by binding to its IL-2 receptor α (CD25) subunit, IL-2 exerts unwanted effects, including stimulation of immunosuppressive regulatory T cells (Tregs) and contribution to vascular leak syndrome. We used a rational approach to develop a monoclonal antibody to human IL-2, termed NARA1, which acts as a high-affinity CD25 mimic, thereby minimizing association of IL-2 with CD25. The structure of the IL-2-NARA1 complex revealed that NARA1 occupies the CD25 epitope of IL-2 and precisely overlaps with CD25. Association of NARA1 with IL-2 occurs with 10-fold higher affinity compared to CD25 and forms IL-2/NARA1 complexes, which, in vivo, preferentially stimulate CD8+ T cells while disfavoring CD25+ Tregs and improving the benefit-to-adverse effect ratio of IL-2. In two transplantable and one spontaneous metastatic melanoma model, IL-2/NARA1 complex immunotherapy resulted in efficient expansion of tumor-specific and polyclonal CD8+ T cells. These CD8+ T cells showed robust interferon-γ production and expressed low levels of exhaustion markers programmed cell death protein-1, lymphocyte activation gene-3, and T cell immunoglobulin and mucin domain-3. These effects resulted in potent anticancer immune responses and prolonged survival in the tumor models. Collectively, our data demonstrate that NARA1 acts as a CD25-mimobody that confers selectivity and increased potency to IL-2 and warrant further assessment of NARA1 as a therapeutic.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunotherapy/methods , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2/antagonists & inhibitors , Neoplasms/therapy , Animals , Binding Sites , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Epitopes/chemistry , Gene Silencing , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Protein Conformation , Recombination, Genetic , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Regulatory/immunology
14.
J Immunol ; 194(8): 3723-34, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25762782

ABSTRACT

The paracaspase MALT1 plays an important role in immune receptor-driven signaling pathways leading to NF-κB activation. MALT1 promotes signaling by acting as a scaffold, recruiting downstream signaling proteins, as well as by proteolytic cleavage of multiple substrates. However, the relative contributions of these two different activities to T and B cell function are not well understood. To investigate how MALT1 proteolytic activity contributes to overall immune cell regulation, we generated MALT1 protease-deficient mice (Malt1(PD/PD)) and compared their phenotype with that of MALT1 knockout animals (Malt1(-/-)). Malt1(PD/PD) mice displayed defects in multiple cell types including marginal zone B cells, B1 B cells, IL-10-producing B cells, regulatory T cells, and mature T and B cells. In general, immune defects were more pronounced in Malt1(-/-) animals. Both mouse lines showed abrogated B cell responses upon immunization with T-dependent and T-independent Ags. In vitro, inactivation of MALT1 protease activity caused reduced stimulation-induced T cell proliferation, impaired IL-2 and TNF-α production, as well as defective Th17 differentiation. Consequently, Malt1(PD/PD) mice were protected in a Th17-dependent experimental autoimmune encephalomyelitis model. Surprisingly, Malt1(PD/PD) animals developed a multiorgan inflammatory pathology, characterized by Th1 and Th2/0 responses and enhanced IgG1 and IgE levels, which was delayed by wild-type regulatory T cell reconstitution. We therefore propose that the pathology characterizing Malt1(PD/PD) animals arises from an immune imbalance featuring pathogenic Th1- and Th2/0-skewed effector responses and reduced immunosuppressive compartments. These data uncover a previously unappreciated key function of MALT1 protease activity in immune homeostasis and underline its relevance in human health and disease.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Caspases/immunology , Cell Differentiation/immunology , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes, Regulatory/pathology , Caspases/genetics , Cell Differentiation/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Mice , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/genetics , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
15.
PLoS One ; 8(3): e59630, 2013.
Article in English | MEDLINE | ID: mdl-23544080

ABSTRACT

BACKGROUND: Sphingosine-1-phosphate (S1P) regulates the egress of T cells from lymphoid organs; levels of S1P in the tissues are controlled by S1P lyase (Sgpl1). Hence, Sgpl1 offers a target to block T cell-dependent inflammatory processes. However, the involvement of Sgpl1 in models of disease has not been fully elucidated yet, since Sgpl1 KO mice have a short life-span. METHODOLOGY: We generated inducible Sgpl1 KO mice featuring partial reduction of Sgpl1 activity and analyzed them with respect to sphingolipid levels, T-cell distribution, and response in models of inflammation. PRINCIPAL FINDINGS: The partially Sgpl1 deficient mice are viable but feature profound reduction of peripheral T cells, similar to the constitutive KO mice. While thymic T cell development in these mice appears normal, mature T cells are retained in thymus and lymph nodes, leading to reduced T cell numbers in spleen and blood, with a skewing towards increased proportions of memory T cells and T regulatory cells. The therapeutic relevance of Sgpl1 is demonstrated by the fact that the inducible KO mice are protected in experimental autoimmune encephalomyelitis (EAE). T cell immigration into the CNS was found to be profoundly reduced. Since S1P levels in the brain of the animals are unchanged, we conclude that protection in EAE is due to the peripheral effect on T cells, leading to reduced CNS immigration, rather than on local effects in the CNS. SIGNIFICANCE: The data suggest Sgpl1 as a novel therapeutic target for the treatment of multiple sclerosis.


Subject(s)
Aldehyde-Lyases/deficiency , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Aldehyde-Lyases/metabolism , Animals , Brain/metabolism , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/complications , Forkhead Transcription Factors/metabolism , Hypersensitivity, Delayed/blood , Hypersensitivity, Delayed/complications , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Immunologic Memory/immunology , Integrases/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Count , Mice , Mice, Knockout , Sheep , Sphingolipids/metabolism , Spleen/immunology , Spleen/pathology , Survival Analysis , Thymus Gland/immunology , Thymus Gland/pathology
16.
Transpl Immunol ; 26(4): 212-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22456277

ABSTRACT

The GDP exchange factor (GEF) Vav1 is a central signal transducer downstream of the T cell receptor and has been identified as a key factor for T cell activation in the context of allograft rejection. Vav1 has been shown to transduce signals both dependent and independent of its GEF function. The most promising approach to disrupt Vav1 activity by pharmacological inhibition would be to target its GEF function. However, the contribution of Vav1 GEF activity for allogeneic T cell activation has not been clarified yet. To address this question, we used knock-in mice bearing a mutated Vav1 with disrupted GEF activity but intact GEF-independent functions. T cells from these mice showed strongly reduced proliferation and activation in response to allogeneic stimulation. Furthermore, lack of Vav1 GEF activity strongly abrogated the in vivo expansion of T cells in a systemic graft-versus-host model. In a cardiac transplantation model, mice with disrupted Vav1 GEF activity show prolonged allograft survival. These findings demonstrate a strong requirement for Vav1 GEF activity for allogeneic T cell activation and graft rejection suggesting that disruption of Vav1 GEF activity alone is sufficient to induce significant immunosuppression.


Subject(s)
Graft Rejection/immunology , Graft vs Host Disease/immunology , Heart Transplantation , Proto-Oncogene Proteins c-vav/metabolism , T-Lymphocytes/immunology , Animals , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , Graft Rejection/etiology , Immunosuppression Therapy , Isoantigens/immunology , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , Mutation/genetics , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/immunology , Transcription Factors/genetics
17.
J Immunol ; 188(9): 4405-11, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22461693

ABSTRACT

Safety of human therapeutic Abs is generally assessed in nonhuman primates. Whereas IgG1 shows identical FcγR interaction and effector function profile in both species, fundamental differences in the IgG2 and IgG4 Ab subclasses were found between the two species. Granulocytes, the main effector cells against IgG2- and IgG4-opsonized bacteria and parasites, do not express FcγRIIIb, but show higher levels of FcγRII in cynomolgus monkey. In humans, IgG2 and IgG4 adapted a silent Fc region with weak binding to FcγR and effector functions, whereas, in contrast, cynomolgus monkey IgG2 and IgG4 display strong effector function as well as differences in IgG4 Fab arm exchange. To balance this shift toward activation, the cynomolgus inhibitory FcγRIIb shows strongly increased affinity for IgG2. In view of these findings, in vitro and in vivo results for human IgG2 and IgG4 obtained in the cynomolgus monkey have to be cautiously interpreted, whereas effector function-related effects of human IgG1 Abs are expected to be predictable for humans.


Subject(s)
Granulocytes/immunology , Immunoglobulin G/immunology , Receptors, IgG/immunology , Animals , Base Sequence , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Humans , Immunoglobulin G/genetics , Macaca fascicularis , Molecular Sequence Data , Receptors, IgG/genetics , Species Specificity
18.
J Exp Med ; 209(1): 77-91, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22184633

ABSTRACT

The signaling adaptor TNFR-associated factor 1 (TRAF1) is specifically lost from virus-specific CD8 T cells during the chronic phase of infection with HIV in humans or lymphocytic choriomeningitis virus (LCMV) clone 13 in mice. In contrast, TRAF1 is maintained at higher levels in virus-specific T cells of HIV controllers or after acute LCMV infection. TRAF1 expression negatively correlates with programmed death 1 expression and HIV load and knockdown of TRAF1 in CD8 T cells from viral controllers results in decreased HIV suppression ex vivo. Consistent with the desensitization of the TRAF1-binding co-stimulatory receptor 4-1BB, 4-1BBL-deficient mice have defects in viral control early, but not late, in chronic infection. TGFß induces the posttranslational loss of TRAF1, whereas IL-7 restores TRAF1 levels. A combination treatment with IL-7 and agonist anti-4-1BB antibody at 3 wk after LCMV clone 13 infection expands T cells and reduces viral load in a TRAF1-dependent manner. Moreover, transfer of TRAF1(+) but not TRAF1(-) memory T cells at the chronic stage of infection reduces viral load. These findings identify TRAF1 as a potential biomarker of HIV-specific CD8 T cell fitness during the chronic phase of disease and a target for therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Lymphocytic Choriomeningitis/immunology , TNF Receptor-Associated Factor 1/deficiency , 4-1BB Ligand/immunology , 4-1BB Ligand/metabolism , Adoptive Transfer , Animals , Antibodies/immunology , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Chloroquine/pharmacology , Chronic Disease , Down-Regulation/genetics , Gene Expression , HIV Infections/genetics , Humans , Immunologic Memory , Interleukin-7/pharmacology , Lymphocytic Choriomeningitis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 1/genetics , Transforming Growth Factor beta/metabolism , Viral Load/immunology
19.
Cell ; 144(4): 601-13, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21295337

ABSTRACT

Understanding the factors that impede immune responses to persistent viruses is essential in designing therapies for HIV infection. Mice infected with LCMV clone-13 have persistent high-level viremia and a dysfunctional immune response. Interleukin-7, a cytokine that is critical for immune development and homeostasis, was used here to promote immunity toward clone-13, enabling elucidation of the inhibitory pathways underlying impaired antiviral immune response. Mechanistically, IL-7 downregulated a critical repressor of cytokine signaling, Socs3, resulting in amplified cytokine production, increased T cell effector function and numbers, and viral clearance. IL-7 enhanced thymic output to expand the naive T cell pool, including T cells that were not LCMV specific. Additionally, IL-7 promoted production of cytoprotective IL-22 that abrogated liver pathology. The IL-7-mediated effects were dependent on endogenous IL-6. These attributes of IL-7 have profound implications for its use as a therapeutic in the treatment of chronic viral diseases.


Subject(s)
Interleukin-7/therapeutic use , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Animals , Antigens, Differentiation/metabolism , Down-Regulation , Forkhead Transcription Factors/metabolism , Humans , Interleukin-6/immunology , Interleukin-7/immunology , Mice , Programmed Cell Death 1 Receptor , Recombinant Proteins/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes/immunology
20.
Nat Med ; 15(5): 528-36, 2009 May.
Article in English | MEDLINE | ID: mdl-19396174

ABSTRACT

Identifying key factors that enhance immune responses is crucial for manipulating immunity to tumors. We show that after a vaccine-induced immune response, adjuvant interleukin-7 (IL-7) improves antitumor responses and survival in an animal model. The improved immune response is associated with increased IL-6 production and augmented T helper type 17 cell differentiation. Furthermore, IL-7 modulates the expression of two ubiquitin ligases: Casitas B-lineage lymphoma b (Cbl-b), a negative regulator of T cell activation, is repressed, and SMAD-specific E3 ubiquitin protein ligase-2 (Smurf2) is enhanced, which antagonizes transforming growth factor-beta signaling. Notably, we show that although short term IL-7 therapy potently enhances vaccine-mediated immunity, in the absence of vaccination it is inefficient in promoting antitumor immune responses, despite inducing homeostatic proliferation of T cells. The ability of adjuvant IL-7 to antagonize inhibitory networks at the cellular and molecular level has major implications for immunotherapy in the treatment of tumors.


Subject(s)
Arenaviridae Infections/immunology , Cancer Vaccines/therapeutic use , Immunotherapy/methods , Interleukin-7/therapeutic use , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Neoplasms/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Count , Mice , Neoplasms, Experimental/immunology , Survivors , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...