Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
EMBO Rep ; 21(6): e48927, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32363653

ABSTRACT

CD1d-restricted invariant natural killer T (iNKT) cells constitute a common glycolipid-reactive innate-like T-cell subset with a broad impact on innate and adaptive immunity. While several microbial glycolipids are known to activate iNKT cells, the cellular mechanisms leading to endogenous CD1d-dependent glycolipid responses remain largely unclear. Here, we show that endoplasmic reticulum (ER) stress in APCs is a potent inducer of CD1d-dependent iNKT cell autoreactivity. This pathway relies on the presence of two transducers of the unfolded protein response: inositol-requiring enzyme-1a (IRE1α) and protein kinase R-like ER kinase (PERK). Surprisingly, the neutral but not the polar lipids generated within APCs undergoing ER stress are capable of activating iNKT cells. These data reveal that ER stress is an important mechanism to elicit endogenous CD1d-restricted iNKT cell responses through induction of distinct classes of neutral lipids.


Subject(s)
Natural Killer T-Cells , Antigen-Presenting Cells , Antigens, CD1d/genetics , Endoribonucleases , Lipids , Lymphocyte Activation , Protein Serine-Threonine Kinases
2.
Nature ; 512(7512): 69-73, 2014 08 07.
Article in English | MEDLINE | ID: mdl-25043000

ABSTRACT

Rheumatoid arthritis is a chronic autoinflammatory disease that affects 1-2% of the world's population and is characterized by widespread joint inflammation. Interleukin-1 is an important mediator of cartilage destruction in rheumatic diseases, but our understanding of the upstream mechanisms leading to production of interleukin-1ß in rheumatoid arthritis is limited by the absence of suitable mouse models of the disease in which inflammasomes contribute to pathology. Myeloid-cell-specific deletion of the rheumatoid arthritis susceptibility gene A20/Tnfaip3 in mice (A20(myel-KO) mice) triggers a spontaneous erosive polyarthritis that resembles rheumatoid arthritis in patients. Rheumatoid arthritis in A20(myel-KO) mice is not rescued by deletion of tumour necrosis factor receptor 1 (ref. 2). Here we show, however, that it crucially relies on the Nlrp3 inflammasome and interleukin-1 receptor signalling. Macrophages lacking A20 have increased basal and lipopolysaccharide-induced expression levels of the inflammasome adaptor Nlrp3 and proIL-1ß. As a result, A20-deficiency in macrophages significantly enhances Nlrp3 inflammasome-mediated caspase-1 activation, pyroptosis and interleukin-1ß secretion by soluble and crystalline Nlrp3 stimuli. In contrast, activation of the Nlrc4 and AIM2 inflammasomes is not altered. Importantly, increased Nlrp3 inflammasome activation contributes to the pathology of rheumatoid arthritis in vivo, because deletion of Nlrp3, caspase-1 and the interleukin-1 receptor markedly protects against rheumatoid-arthritis-associated inflammation and cartilage destruction in A20(myel-KO) mice. These results reveal A20 as a novel negative regulator of Nlrp3 inflammasome activation, and describe A20(myel-KO) mice as the first experimental model to study the role of inflammasomes in the pathology of rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/metabolism , Carrier Proteins/metabolism , Cysteine Endopeptidases/metabolism , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/prevention & control , Calcium-Binding Proteins/metabolism , Caspase 1/deficiency , Caspase 1/metabolism , Cysteine Endopeptidases/deficiency , DNA-Binding Proteins , Disease Models, Animal , Female , Interleukin-1/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Macrophages/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/metabolism , Phenotype , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3
3.
Ann Rheum Dis ; 76(3): 585-592, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27551052

ABSTRACT

OBJECTIVES: A20 is an important endogenous regulator of inflammation. Single nucleotide polymorphisms in A20 have been associated with various immune-mediated inflammatory diseases, and cell-specific deletion of A20 results in diverse inflammatory phenotypes. Our goal was to delineate the underlying mechanisms of joint inflammation in myeloid-specific A20-deficient mice (A20myelKO mice). METHODS: Inflammation in A20myelKO mice was assessed in a time-dependent manner. Western blot analysis and quantitative PCR analysis were performed on bone marrow-derived macrophages from A20myelKO and littermate control mice to study the effect of A20 on STAT1/STAT3 expression and STAT1/STAT3-dependent gene transcription in myeloid cells. The in vivo role of Janus kinase-Signal Transducer and Activator of Transcription (JAK-STAT) signalling in the development of enthesitis in A20myelKO mice was assessed following administration of a JAK inhibitor versus placebo control. RESULTS: Enthesitis was found to be an early inflammatory lesion in A20myelKO mice. A20 negatively modulated STAT1-dependent, but generally not STAT3-dependent gene transcription in myeloid cells by suppressing STAT1 but not STAT3 expression, both in unstimulated conditions and after interferon-γ or interleukin-6 stimulation. The increase in STAT1 gene transcription in the absence of A20 was shown to be JAK-STAT-dependent. Moreover, JAK inhibition in vivo resulted in significant reduction of enthesitis, both clinically and histopathologically. CONCLUSIONS: Our data reveal an important and novel interplay between myeloid cells and tissue resident cells at entheseal sites that is regulated by A20. In the absence of A20, STAT1 but not STAT3 expression is enhanced leading to STAT1-dependent inflammation. Therefore, A20 acts as a novel endogenous regulator of STAT1 that prevents onset of enthesitis.


Subject(s)
Enthesopathy/genetics , Enthesopathy/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , Cells, Cultured , Enthesopathy/etiology , Enthesopathy/pathology , Inflammation/complications , Inflammation/genetics , Inflammation/metabolism , Interferon-gamma/pharmacology , Interleukin-6/pharmacology , Janus Kinases/metabolism , Macrophages , Mice , Mice, Knockout , Piperidines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
4.
J Hepatol ; 60(1): 175-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23973929

ABSTRACT

BACKGROUND & AIMS: Immunometabolism is an emerging field of clinical investigation due to the obesity epidemic worldwide. A reciprocal involvement of immune mediators in the body energy metabolism has been recognized for years, but is only partially understood. We hypothesized that the adipokine leptin could provide an important modulator of iNKT cells. METHODS: The expression of leptin receptor (LR) on resting and activated iNKT cells was measured by flow cytometry. FACS-sorted hepatic iNKT cells were stimulated with anti-CD3/CD28Ab coated beads in the absence or presence of a neutralizing anti-leptin Ab. Furthermore, we evaluated the outcome of LR blocking nanobody treatment in ConA induced hepatitis and towards metabolic parameters in WT and iNKT cell deficient mice. RESULTS: The LR is expressed on iNKT cells and leptin suppresses iNKT cell proliferation and cytokine production in vitro. LR deficient iNKT cells are hyper-responsive further enforcing the role of leptin as an important inhibitor of iNKT cell function. Consistently, in vivo blockade of LR signaling exacerbated ConA hepatitis in wild-type but not in iNKT cell deficient mice, through both Janus kinase (JAK)2 and mitogen-activated protein kinase (MAPK) dependent mechanisms. Moreover, LR inhibition altered fat pad features and was accompanied by insulin resistance, only in wild-type mice. Curiously, this interaction was strictly dependent on MAPK mediated LR signaling in iNKT cells and uncoupled from the more central effects of leptin. CONCLUSIONS: Our data support a new concept of immune regulation by which leptin protects towards T cell mediated hepatitis via modulation of iNKT cells.


Subject(s)
Adipocytes/physiology , Cell Communication , Hepatitis/etiology , Leptin/physiology , Natural Killer T-Cells/physiology , T-Lymphocytes/immunology , Adipose Tissue/metabolism , Animals , Disease Susceptibility , Lymphocyte Activation , MAP Kinase Signaling System , Mice , Receptors, Antigen, T-Cell/physiology , Receptors, Leptin/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/physiology
5.
Ann Rheum Dis ; 73(2): 437-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23921997

ABSTRACT

OBJECTIVES: Spondyloarthritides (SpA) are characterised by both peripheral and axial arthritis. The hallmarks of peripheral SpA are the development of enthesitis, most typically of the Achilles tendon and plantar fascia, and new bone formation. This study was undertaken to unravel the mechanisms leading towards enthesitis and new bone formation in preclinical models of SpA. RESULTS: First, we demonstrated that TNF(ΔARE) mice show typical inflammatory features highly reminiscent of SpA. The first signs of inflammation were found at the entheses. Importantly, enthesitis occurred equally in the presence or absence of mature T and B cells, underscoring the importance of stromal cells. Hind limb unloading in TNF(ΔARE) mice significantly suppressed inflammation of the Achilles tendon compared with weight bearing controls. Erk1/2 signalling plays a crucial role in mechanotransduction-associated inflammation. Furthermore, new bone formation is strongly promoted at entheseal sites by biomechanical stress and correlates with the degree of inflammation. CONCLUSIONS: These findings provide a formal proof of the concept that mechanical strain drives both entheseal inflammation and new bone formation in SpA.


Subject(s)
Achilles Tendon/pathology , Arthritis, Experimental/complications , Osteogenesis/physiology , Spondylarthritis/complications , Tendinopathy/etiology , Achilles Tendon/physiopathology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , B-Lymphocytes/immunology , MAP Kinase Signaling System/physiology , Magnetic Resonance Imaging , Mechanotransduction, Cellular/physiology , Mice , Sacroiliitis/etiology , Sacroiliitis/pathology , Spondylarthritis/immunology , Spondylarthritis/pathology , Spondylarthritis/physiopathology , Stress, Mechanical , Stromal Cells/physiology , T-Lymphocytes/immunology , Tendinopathy/immunology , Tendinopathy/pathology , Tendinopathy/physiopathology , Tumor Necrosis Factor-alpha/immunology , Weight-Bearing/physiology , X-Ray Microtomography
6.
Arthritis Rheumatol ; 75(11): 1969-1982, 2023 11.
Article in English | MEDLINE | ID: mdl-37293832

ABSTRACT

OBJECTIVE: Patients with spondyloarthritis (SpA) often present with microscopic signs of gut inflammation, a risk factor for progressive disease. We investigated whether mucosal innate-like T cells are involved in dysregulated interleukin-23 (IL-23)/IL-17 responses in the gut-joint axis in SpA. METHODS: Ileal and colonic intraepithelial lymphocytes (IELs), lamina propria lymphocytes (LPLs), and paired peripheral blood mononuclear cells (PBMCs) were isolated from treatment-naive patients with nonradiographic axial SpA with (n = 11) and without (n = 14) microscopic gut inflammation and healthy controls (n = 15) undergoing ileocolonoscopy. The presence of gut inflammation was assessed histopathologically. Immunophenotyping of innate-like T cells and conventional T cells was performed using intracellular flow cytometry. Unsupervised clustering analysis was done by FlowSOM technology. Serum IL-17A levels were measured via Luminex. RESULTS: Microscopic gut inflammation in nonradiographic axial SpA was characterized by increased ileal intraepithelial γδ-hi T cells, a γδ-T cell subset with elevated γδ-T cell receptor expression. γδ-hi T cells were also increased in PBMCs of patients with nonradiographic axial SpA versus healthy controls and were strongly associated with Ankylosing Spondylitis Disease Activity Score. The abundance of mucosal-associated invariant T cells and invariant natural killer T cells was unaltered. Innate-like T cells in the inflamed gut showed increased RORγt, IL-17A, and IL-22 levels with loss of T-bet, a signature that was less pronounced in conventional T cells. Presence of gut inflammation was associated with higher serum IL-17A levels. In patients treated with tumor necrosis factor blockade, the proportion of γδ-hi cells and RORγt expression in blood was completely restored. CONCLUSION: Intestinal innate-like T cells display marked type 17 skewing in the inflamed gut mucosa of patients with nonradiographic axial SpA. γδ-hi T cells are linked to intestinal inflammation and disease activity in SpA.


Subject(s)
Spondylarthritis , Spondylitis, Ankylosing , Humans , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Leukocytes, Mononuclear/metabolism , Inflammation/metabolism , Spondylarthritis/metabolism , Mucous Membrane/metabolism
7.
Nat Commun ; 10(1): 9, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30602780

ABSTRACT

Dysregulated IL-23/IL-17 responses have been linked to psoriatic arthritis and other forms of spondyloarthritides (SpA). RORγt, the key Thelper17 (Th17) cell transcriptional regulator, is also expressed by subsets of innate-like T cells, including invariant natural killer T (iNKT) and γδ-T cells, but their contribution to SpA is still unclear. Here we describe the presence of particular RORγt+T-betloPLZF- iNKT and γδ-hi T cell subsets in healthy peripheral blood. RORγt+ iNKT and γδ-hi T cells show IL-23 mediated Th17-like immune responses and were clearly enriched within inflamed joints of SpA patients where they act as major IL-17 secretors. SpA derived iNKT and γδ-T cells showed unique and Th17-skewed phenotype and gene expression profiles. Strikingly, RORγt inhibition blocked γδ17 and iNKT17 cell function while selectively sparing IL-22+ subsets. Overall, our findings highlight a unique diversity of human RORγt+ T cells and underscore the potential of RORγt antagonism to modulate aberrant type 17 responses.


Subject(s)
Natural Killer T-Cells/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Spondylarthritis/immunology , T-Lymphocyte Subsets/metabolism , Case-Control Studies , Humans , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Receptors, Interleukin/metabolism
8.
Nat Commun ; 9(1): 5340, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30559399

ABSTRACT

Activated invariant natural killer T (iNKT) cells rapidly produce large amounts of cytokines, but how cytokine mRNAs are induced, stabilized and mobilized following iNKT activation is still unclear. Here we show that an endoplasmic reticulum stress sensor, inositol-requiring enzyme 1α (IRE1α), links key cellular processes required for iNKT cell effector functions in specific iNKT subsets, in which TCR-dependent activation of IRE1α is associated with downstream activation of p38 MAPK and the stabilization of preformed cytokine mRNAs. Importantly, genetic deletion of IRE1α in iNKT cells reduces cytokine production and protects mice from oxazolone colitis. We therefore propose that an IRE1α-dependent signaling cascade couples constitutive cytokine mRNA expression to the rapid induction of cytokine secretion and effector functions in activated iNKT cells.


Subject(s)
Cytokines/genetics , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/genetics , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Protein Serine-Threonine Kinases/genetics , Animals , Cells, Cultured , Colitis/genetics , Gene Deletion , Mice , Mice, Knockout , Oxazolone/toxicity , RNA, Messenger/genetics , Signal Transduction , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Exp Med ; 213(10): 1973-81, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27551157

ABSTRACT

Natural killer T (NKT) cells are innate lymphocytes that differentiate into NKT1, NKT2, and NKT17 sublineages during development. However, the signaling events that control NKT sublineage specification and differentiation remain poorly understood. Here, we demonstrate that the ubiquitin-modifying enzyme TNFAIP3/A20, an upstream regulator of T cell receptor (TCR) signaling in T cells, is an essential cell-intrinsic regulator of NKT differentiation. A20 is differentially expressed during NKT cell development, regulates NKT cell maturation, and specifically controls the differentiation and survival of NKT1 and NKT2, but not NKT17, sublineages. Remaining A20-deficient NKT1 and NKT2 thymocytes are hyperactivated in vivo and secrete elevated levels of Th1 and Th2 cytokines after TCR ligation in vitro. Defective NKT development was restored by compound deficiency of MALT1, a key downstream component of TCR signaling in T cells. These findings therefore show that negative regulation of TCR signaling during NKT development controls the differentiation and survival of NKT1 and NKT2 cells.


Subject(s)
Cell Lineage , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Ubiquitin/metabolism , Animals , CD4 Antigens/metabolism , Caspases/deficiency , Caspases/metabolism , Cell Survival , Integrases/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Transgenic , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Up-Regulation/genetics
10.
Nat Genet ; 43(9): 908-12, 2011 Aug 14.
Article in English | MEDLINE | ID: mdl-21841782

ABSTRACT

A20 (TNFAIP3) is a protein that is involved in the negative feedback regulation of NF-κB signaling in response to specific proinflammatory stimuli in different cell types and has been suggested as a susceptibility gene for rheumatoid arthritis. To define the contribution of A20 to rheumatoid arthritis pathology, we generated myeloid-specific A20-deficient mice and show that specific ablation of Tnfaip3 in myeloid cells results in spontaneous development of a severe destructive polyarthritis with many features of rheumatoid arthritis. Myeloid-A20-deficient mice have high levels of inflammatory cytokines in their serum, consistent with a sustained NF-κB activation and higher TNF production by macrophages. Destructive polyarthritis in myeloid A20 knockout mice was TLR4-MyD88 and IL-6 dependent but was TNF independent. Myeloid A20 deficiency also promoted osteoclastogenesis in mice. Together, these observations indicate a critical and cell-specific function for A20 in the etiology of rheumatoid arthritis, supporting the idea of developing A20 modulatory drugs as cell-targeted therapies.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cysteine Endopeptidases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Myeloid Cells/enzymology , Animals , Arthritis, Rheumatoid/blood , Cytokines/blood , Macrophages/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL