Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Eur Respir J ; 61(3)2023 03.
Article in English | MEDLINE | ID: mdl-36328358

ABSTRACT

BACKGROUND: GLPG1205 is a selective functional antagonist of G-protein-coupled receptor 84, which plays an important role in fibrotic processes. This study assessed the efficacy, safety and tolerability of GLPG1205 for treatment of idiopathic pulmonary fibrosis (IPF). METHODS: PINTA (ClinicalTrials.gov: NCT03725852) was a phase 2, randomised, double-blind, placebo-controlled, proof-of-concept trial. Patients with IPF were randomised 2:1 to once-daily oral GLPG1205 100 mg or placebo for 26 weeks and stratified to receive GLPG1205 alone or with local standard of care (nintedanib or pirfenidone). The primary end-point was change from baseline in forced vital capacity (FVC); other end-points were safety and tolerability, and lung volumes measured by imaging (high-resolution computed tomography). The study was not powered for statistical significance. RESULTS: In total, 68 patients received study medication. Least squares mean change from baseline in FVC at week 26 was -33.68 (95% CI -112.0-44.68) mL with GLPG1205 and -76.00 (95% CI -170.7-18.71) mL with placebo (least squares mean difference 42.33 (95% CI -81.84-166.5) mL; p=0.50). Lung volumes by imaging declined -58.30 versus -262.72 mL (whole lung) and -33.68 versus -135.48 mL (lower lobes) with GLPG1205 versus placebo, respectively. Treatment with GLPG1205 versus placebo resulted in higher proportions of serious and severe treatment-emergent adverse events and treatment-emergent discontinuations, most apparent with nintedanib. CONCLUSIONS: Treatment with GLPG1205 did not result in a significant difference in FVC decline versus placebo. GLPG1205 demonstrated a poorer safety and tolerability profile than placebo.


Subject(s)
Idiopathic Pulmonary Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Lung/diagnostic imaging , Vital Capacity , Double-Blind Method , Treatment Outcome
2.
Cell Rep ; 28(1): 21-29.e6, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31269441

ABSTRACT

Regulatory T cells (Tregs) play a crucial role in controlling autoimmune and inflammatory responses. Recent studies have demonstrated that dendritic cells (DCs) contribute to the homeostasis of peripheral Tregs. Autophagy, a critical pathway for cellular homeostasis, is active in DCs and is upregulated in different inflammatory conditions. We have shown that Tregs are expanded and have phenotypic alterations and impaired suppressive functions in mice with autophagy-deficient DCs. RNA profiling of Tregs revealed that autophagy in DCs is required to stabilize Treg expression signatures. This phenotype is linked to the downregulation of ICOS-Ligand expression in autophagy-deficient DCs, a consequence of the accumulation of ADAM10, the metalloproteinase responsible for its cleavage. Upon inflammation, in antigen-induced arthritis, mice with autophagy-deficient DCs exhibit increased synovial inflammation and cartilage and bone erosion correlating with Treg-to-Th17 conversion. Our data reveal a mechanism that couples autophagy deficiency in DCs to the function, homeostasis, and stability of Tregs.


Subject(s)
Autophagy-Related Protein 5/metabolism , Dendritic Cells/immunology , Macroautophagy/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Autophagy-Related Protein 5/genetics , Dendritic Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeostasis/immunology , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology
3.
J Control Release ; 276: 102-112, 2018 04 28.
Article in English | MEDLINE | ID: mdl-29524442

ABSTRACT

This study aimed to formulate nanocrystal-polymer particles (NPPs) containing the potent p38α/ß MAPK inhibitor PH-797804 (PH-NPPs) and to test their extended-release properties over months in comparison to those of conventional PH microparticles for the intra-articular treatment of inflammatory and mechanistic murine models mirroring aspects of human osteoarthritis (OA). The steps of the study were (i) to formulate PH nanocrystals (wet milling), (ii) to encapsulate a high payload of PH nanocrystals in fluorescent particles (spray drying), (iii) to assess in vitro drug release, (iv) to evaluate PH-NPP toxicity to human OA synoviocytes (MTT test), (v) to investigate the in vivo bioactivity of the particles in mice in an inflammatory antigen-induced arthritis (AIA) model (using histology and RT-qPCR) and (vi) to investigate the in vivo bioactivity of the particles in the OA model obtained by mechanistic surgical destabilization of the medial meniscus (DMM) (using histology, micro-CT, and multiplex ELISA). The PH nanocrystals stabilized with vitamin E TPGS had a monomodal size distribution. The PH-NPPs had a mean diameter of 14.2 µm and drug loading of ~31.5% (w/w), and ~20% of the PH was released over 3 months. The NPPs did not exhibit toxicity to cultured human OA synoviocytes at 100 × IC50. Finally, in vivo studies showed good retention of PH-NPPs in the joint and adjacent tissues for up to 2 months, and the PH-NPPs exhibited good functional relevance by significantly reducing inflammation and joint destruction and by inhibiting several biomarkers (e.g., IL-1ß). In conclusion, local treatment with PH-NPPs, used as an extended-release drug delivery system, improved inflammation and joint degradation in two distinct mouse models, indicating treatment potential for human OA.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Benzamides/administration & dosage , Nanoparticles/administration & dosage , Osteoarthritis/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyridones/administration & dosage , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzamides/chemistry , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Disease Models, Animal , Drug Liberation , Humans , Male , Mice, Inbred C57BL , Nanoparticles/chemistry , Protein Kinase Inhibitors/chemistry , Pyridones/chemistry
4.
Nanoscale ; 10(4): 1845-1854, 2018 Jan 25.
Article in English | MEDLINE | ID: mdl-29308811

ABSTRACT

Under pathological conditions, joints and skin are often affected by an imbalance in the breakdown and production of hyaluronic acid (HA). The unique biochemical and biomechanical properties provided by HA must be restored for the long-term lubrication and cushioning effects. To overcome the inconvenience of repeated injections and the rapid degradation of exogenous HA treatments, HA is conjugated to a thermosensitive polymer, enabling the spontaneous formation of nanoparticles (HA Nano) at body temperature. Three HA Nano preparations are tested for their injectability, sensitivity to enzymatic degradation and cytocompatibility. One of them is delivered via subcutaneous and intra-articular injections to healthy mice and tested in a murine osteoarthritis (OA) model. It is found to be biocompatible, to offer a prolonged residence time at the injection site, have the ability to protect cartilage, to reduce pro-inflammatory cytokines and to preserve epiphysis thickness. In this study, HA Nano spontaneously forms nanoparticles at body temperature in vivo and is a promising candidate for the next generation of the sustainable/long-lasting treatment of OA and potentially also dermatological conditions.


Subject(s)
Drug Carriers/chemistry , Hyaluronic Acid/administration & dosage , Nanostructures/chemistry , Osteoarthritis/drug therapy , Animals , Cartilage , Cells, Cultured , Fibroblasts , Humans , Injections, Intra-Articular , Male , Mice , Mice, Inbred C57BL , Synovial Membrane/cytology
5.
Small ; 14(8)2018 02.
Article in English | MEDLINE | ID: mdl-29327460

ABSTRACT

An efficient treatment for osteoarthritis (OA) can benefit from the local release of a high therapeutic dose over an extended period of time. Such a treatment will minimize systemic side effects and avoid the inconvenience of frequent injections. To this aim, nanocrystal-polymer particles (NPPs) are developed by combining the advantages of nanotechnology and microparticles. Nanocrystals are produced by wet milling kartogenin (KGN), which is known to promote chondrogenesis and to foster chondroprotection. A fluorescent biodegradable polymer is synthesized for intravital particle tracking. Polymer microparticles with 320 nm embedded KGN nanocrystals (KGN-NPPs) show a high drug loading of 31.5% (w/w) and an extended drug release of 62% over 3 months. In vitro, these particles do not alter mitochondrial activity in cultured human OA synoviocytes. In vivo, KGN-NPPs demonstrate higher bioactivity than a KGN solution in a murine mechanistic OA model based on histological assessment (Osteoarthritis Research Society International score), epiphyseal thickness (microcomputed tomography), OA biomarkers (e.g., vascular endothelial growth factor, Adamts5), and prolonged intra-articular persistence (fluorescence analysis). This work provides proof-of-concept of a novel and innovative extended drug delivery system with the potential to treat human OA.


Subject(s)
Anilides/therapeutic use , Nanoparticles/chemistry , Osteoarthritis/drug therapy , Phthalic Acids/therapeutic use , Polymers/chemistry , Anilides/chemistry , Animals , Cells, Cultured , Chondrogenesis/drug effects , Drug Delivery Systems , Humans , Injections, Intra-Articular , Mice , Nanotechnology/methods , Phthalic Acids/chemistry
6.
J Immunol ; 198(7): 2916-2926, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28235865

ABSTRACT

The biological activity of IL-1 is tightly regulated by the specific receptor antagonist (IL-1Ra) and the decoy receptor IL-1 receptor type 2 (IL-1R2). The role of IL-1Ra has been well demonstrated in IL-1Ra-deficient mice. In contrast, the role of endogenous IL-1R2 remains widely unknown. To define the functional role of endogenous IL-1R2 in the K/BxN serum transfer arthritis model and in IL-1ß- or LPS-induced systemic inflammation in vivo, IL-1R2-/- mice were created and compared with wild type mice. IL-1R2-/- mice bred habitually and exhibited a normal phenotype. IL-1R2 deficiency aggravated arthritis severity and increased mRNA levels for key cytokines and chemokines such as IL-6, IL-1ß, Cxcl-1, and Cxcl-2 significantly in ankles. There was no effect of IL-1R2 deficiency on the cell-autonomous cytokine response to IL-1ß in the tested cell types, i.e., neutrophils, macrophages, and fibroblasts, but IL-1R2 deficiency on neutrophils increased the IL-1-induced response of fibroblasts in trans. Furthermore, IL-1ß induced shedding of IL-1R2 in vivo. Inflammatory responses to IL-1ß and LPS-induced mortality were not different in IL-1R2-/- compared with wild type mice. Our data demonstrate that the decoy receptor IL-1R2 plays an important inhibitory role in local IL-1- and neutrophil-dependent tissue inflammation as shown in the K/BxN serum transfer arthritis model. In contrast to IL-1Ra, IL-1R2 appears to be less crucial for systemic responses to acute administration of IL-1 or LPS.


Subject(s)
Arthritis, Experimental/immunology , Inflammation/immunology , Receptors, Interleukin-1 Type II/immunology , Animals , Immunohistochemistry , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction
7.
Pharm Res ; 34(3): 599-609, 2017 03.
Article in English | MEDLINE | ID: mdl-28028771

ABSTRACT

PURPOSE: This analysis aimed at describing the effect of the selective sphingosine-1-phosphate receptor 1 modulator ponesimod on lymphocyte subsets in peripheral blood. As the involvement of different lymphocyte subsets varies among different autoimmune diseases, characterizing the effect of ponesimod on these may be beneficial in better understanding treatment effects. METHODS: Three phase 1 clinical studies in healthy human subjects were pooled. Non-linear mixed-effects modeling techniques were used to study the effect of ponesimod on lymphocyte subsets such as B cells, T helper cells, T cytotoxic cells, and natural killer cells in a qualitative and quantitative manner. RESULTS: Indirect-response Imax models including circadian variation best described the effect of ponesimod on lymphocyte subsets. B cells and T helper cells were shown to be more affected compared to T cytotoxic cells with respect to the maximum possible reduction (100% for B and T helper cells, 95% for T cytotoxic cells) and the concentration required to reach half the maximum effect. Inter-individual variability was found to be larger for T cytotoxic compared to T helper, and B cells. CONCLUSION: These first models for ponesimod on the level of lymphocyte subsets offer a valuable tool for the analysis and interpretation of results from ponesimod trials in autoimmune diseases.


Subject(s)
Lymphocyte Subsets/drug effects , Receptors, Lysosphingolipid/metabolism , Thiazoles/pharmacology , Adolescent , Adult , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Circadian Rhythm , Computer Simulation , Dose-Response Relationship, Drug , Female , Humans , Lymphocyte Count , Lymphocyte Subsets/metabolism , Male , Middle Aged , Models, Biological , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Thiazoles/chemistry , Young Adult
8.
Int J Pharm ; 498(1-2): 119-29, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26685724

ABSTRACT

Intra-articular (IA) injection of extended drug release forms based on biodegradable microparticles holds promise for the treatment of joint diseases. However, the fate of microparticles following intra-articular injection is controversial and has not been thoroughly investigated. The aim of this work was therefore to evaluate the biodistribution of fluorescent poly(lactic acid) particles of different sizes after IA injection in arthritic or healthy mice. Regardless of the inflammatory status of the joint, 300 nm-nanoparticles leaked from the joint. Due to inflammation and related increase of vascular permeability, 3 µm-microparticles that were retained in the non-inflamed synovial membrane leaked from the inflamed joint. Complete retention of 10 µm-microparticles was observed independently of the joint inflammatory status. Embedding particles in a hyaluronic acid gel prolonged the retention of the formulations only in inflamed joints. Depending on particle's size, formulations were preferentially eliminated by blood vessels or lymphatic pathways. Poly(lactic acid) particles of 3 µm were biocompatible and retained in knee joints at least for 6 weeks. This work highlights the need to deliver hyaluronic acid-embedded particles of at least 3 µm to guarantee their retention in inflamed joints. These results will contribute to the rational design of long-lasting formulations to treat acute and chronic joint diseases.


Subject(s)
Knee Joint/metabolism , Microspheres , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Particle Size , Aged , Animals , Humans , Injections, Intra-Articular , Knee Joint/drug effects , Male , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Tissue Distribution/drug effects , Tissue Distribution/physiology
9.
Eur J Pharm Biopharm ; 93: 110-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25836052

ABSTRACT

In the treatment of arthritic diseases, oral or systemic administration of anti-inflammatory substances, such as p38 MAPK inhibitors, is hampered by numerous side effects. To overcome them, formulations of rapid and extended drug delivery systems were studied in intra-articular administration. For the first time, VX-745, a highly selective p38 MAPK inhibitor, demonstrated in vivo bioactivity, similar to dexamethasone activity, following intra-articular administration in an antigen-induced arthritic (AIA) mouse model. The in vitro bioactivity of VX-745 was also shown on synoviocytes, reducing the IL-6 concentration. Process and formulation parameters (i.e., polymer concentration, aqueous/organic phase ratio, emulsification speed and process, and evaporation pressure) and particle characterisation (i.e., drug loading, size of particle, and surface aspect) were extensively examined to produce optimised formulations. Indeed, a burst release provides a rapid saturation of intracellular p38 MAPK to relieve patients from pain and inflammation. Then, drug diffusion would be sufficient to maintain an effective dose over 2-3 months. This study confirms the effectiveness of encapsulated p38 MAPK inhibitors in extended drug delivery systems and seems to be a promising strategy for intra-articular treatment.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Arthritis, Experimental/drug therapy , Joints/drug effects , Protein Kinase Inhibitors/administration & dosage , Pyridazines/administration & dosage , Pyrimidines/administration & dosage , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemistry , Arthritis, Experimental/enzymology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cells, Cultured , Chemistry, Pharmaceutical , Delayed-Action Preparations , Diffusion , Drug Carriers , Humans , Injections, Intra-Articular , Joints/enzymology , Joints/immunology , Joints/pathology , Kinetics , Male , Mice, Inbred C57BL , Particle Size , Polymers/chemistry , Protein Kinase Inhibitors/chemistry , Pyridazines/chemistry , Pyrimidines/chemistry , Solubility , Synovial Membrane/drug effects , Synovial Membrane/enzymology , Synovial Membrane/pathology , Technology, Pharmaceutical/methods , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Immunol ; 194(2): 750-60, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25505285

ABSTRACT

IL-33 is a cytokine of the IL-1 family, which signals through the ST2 receptor. Previous studies emphasized a role for IL-33 in shaping innate and adaptive immune responses. IL-33 was also reported to modulate myelopoiesis and myeloid cell activity. In this article, we describe IL-33-overexpressing CMV/IL33 and LysM/IL33 mice, which display an inflammatory phenotype associated with growth retardation and paw swelling. The phenotype of CMV/IL33 mice is dependent on activation of the ST2 receptor and is characterized by extensive neutrophil infiltration into different organs, including the paws. Local or systemic levels of proinflammatory mediators such as IL-1ß, Cxcl-1, G-CSF, and IL-6 are increased. CMV/IL-33 mice also suffer from anemia, thrombocytosis, and a marked dysregulation of myelopoiesis, leading to an important increase in myeloid cell production or accumulation in bone marrow (BM), spleen, and peripheral blood. Consistently, recombinant IL-33 induced proliferation of myeloid lineage cells in BM-derived granulocyte cultures, whereas IL-33 knockout mice exhibited minor deficiencies in spleen and BM myeloid cell populations. Our observations reveal a neutrophil-dominated inflammatory phenotype in IL-33-overexpressing CMV/IL33 and LysM/IL33 mice, and highlight important regulatory effects of IL-33 on myelopoiesis in vitro and in vivo, where excessive IL-33 signaling can translate into the occurrence of a myeloproliferative disorder.


Subject(s)
Interleukins/immunology , Myelopoiesis/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Signal Transduction/immunology , Anemia/genetics , Anemia/immunology , Anemia/pathology , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Gene Expression , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-33 , Interleukin-6/genetics , Interleukin-6/immunology , Interleukins/genetics , Mice , Mice, Knockout , Myelopoiesis/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/pathology , Neutrophil Infiltration/genetics , Neutrophils/pathology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Signal Transduction/genetics , Thrombocytosis/genetics , Thrombocytosis/immunology , Thrombocytosis/pathology
11.
Cytokine ; 69(1): 68-74, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25022964

ABSTRACT

Previous work suggested implication of the interleukin (IL)-1 family cytokine IL-33, signaling through its receptor ST2, in the pathogenesis of human and mouse arthritis. In this study, we directly investigated the role of endogenous IL-33 in antigen-induced arthritis (AIA) and collagen-induced arthritis (CIA) using IL-33 KO mice. AIA was induced by injection of methylated bovine serum albumin (mBSA) into knee joints of previously immunized mice. CIA was induced by immunization with bovine type II collagen. Disease severity was evaluated by clinical and histological scoring and cellular immune responses were assessed in cultured draining lymph node cells. Our results indicate that the development of AIA or CIA, as assessed by clinical or histological evaluation, is not impaired in IL-33 deficient mice. We did not observe any consistent modifications in humoral or cellular immune responses in IL-33 KO mice, although IL-33 deficiency enhanced antigen-specific IFN-γ production, proliferation or IgG2a titers in some experiments, suggesting that endogenous IL-33 may contribute to shaping the adaptive immune response. In conclusion, our data suggest that IL-33 plays a modifying rather than a pivotal role in disease development in two models of immune-mediated arthritis.


Subject(s)
Adaptive Immunity/immunology , Arthritis, Experimental/pathology , Interleukins/genetics , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Cell Proliferation , Collagen Type II , Disease Models, Animal , Disease Progression , Immunoglobulin G/blood , Interferon-gamma/biosynthesis , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Receptors, Interleukin/genetics , T-Lymphocytes/immunology
12.
Arthritis Res Ther ; 15(2): R38, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23452551

ABSTRACT

INTRODUCTION: Interleukin (IL)-36 refers to three related IL-1 family cytokines, IL-36α, IL-36ß, and IL-36γ, that bind to the IL-36 receptor (IL-36R). IL-36 exerts proinflammatory effects in skin and lung and stimulates T cell responses. In the present study, we examined the expression and function of IL-36R and its ligands in experimental arthritis. METHODS: Collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), and K/BxN serum transfer-induced arthritis were induced according to standard protocols. Messenger RNA levels for IL-36R and its ligands in the joints of mice with CIA were determined by RT-qPCR. Mice with CIA were injected with a blocking monoclonal anti-IL-36R, a blocking anti-IL-1RI, or their isotype-matched control antibodies at the time of arthritis onset. Anti-IL-36R or control antibodies were also injected at the time of AIA induction. Finally, IL-36R-deficient mice were examined in AIA and serum transfer-induced arthritis. The development and severity of arthritis were assessed by clinical and histological scoring. RESULTS: IL-36R, IL-36Ra and IL-36γ mRNA were detected in the joints of mice with CIA, but their levels did not correlate with arthritis severity. As opposed to anti-IL-1RI antibody treatment, the injection of an anti-IL-36R antibody was devoid of effect on the development and severity of CIA. The severity of joint inflammation and structural damage in AIA was also unaltered by anti-IL-36R antibody treatment. Finally, the severity of AIA and K/BxN serum transfer-induced arthritis was similar in IL-36R-deficient and wild-type mice. CONCLUSIONS: The development and severity of experimental arthritis are independent of IL-36R signaling.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Receptors, Interleukin-1/immunology , Signal Transduction , Animals , Arthritis, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-1/metabolism , Signal Transduction/immunology
13.
Arthritis Res Ther ; 15(1): R13, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23324173

ABSTRACT

INTRODUCTION: Interleukin (IL)-33 is a cytokine of the IL-1 family, which signals through the ST2 receptor. Previous work suggested implication of the IL-33/ST2 axis in the pathogenesis of human and mouse arthritis. Here, we directly investigated the role of endogenous IL-33 in K/BxN serum transfer-induced arthritis by using IL-33 knockout (KO) mice. METHODS: Arthritis was induced by injection of complete K/BxN serum or purified IgG. Disease severity was monitored by clinical and histological scoring. RESULTS: K/BxN serum transfer induced pronounced arthritis with similar incidence and severity in IL-33 KO and wild-type (WT) mice. In contrast, disease development was significantly reduced in ST2 KO mice. IL-33 expression in synovial tissue was comparable in arthritic WT and ST2 KO mice, and absent in IL-33 KO mice. Transfer of purified arthritogenic IgG instead of complete K/BxN serum also resulted in similar arthritis severity in IL-33 KO and WT mice, excluding a contribution of IL-33 contained in the serum of donor mice to explain this result. We investigated additional potential confounding factors, including purity of genetic background, but the mechanisms underlying reduced arthritis in ST2 KO mice remained unclear. CONCLUSIONS: The data obtained with IL-33 KO mice indicate that endogenous IL-33 is not required for the development of joint inflammation in K/BxN serum transfer-induced arthritis. On the contrary, arthritis severity was reduced in ST2 KO mice. This observation might relate to IL-33 independent effects of ST2, and/or reveal the existence of confounding variables affecting the severity of joint inflammation in these KO strains.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Interleukins/immunology , Animals , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Genotype , Immunohistochemistry , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Real-Time Polymerase Chain Reaction , Receptors, Interleukin/deficiency , Receptors, Interleukin/immunology
14.
Eur J Immunol ; 42(5): 1294-303, 2012 May.
Article in English | MEDLINE | ID: mdl-22539301

ABSTRACT

Interleukin-1 receptor antagonist (IL-1Ra) is a specific IL-1 inhibitor that possesses anti-inflammatory activities. Several studies in human and mouse suggested a protective role for IL-1Ra in liver inflammation, and we previously demonstrated that hepatocytes produce high levels of IL-1Ra in response to inflammatory challenge in vitro and in vivo. In the present study, we investigated the production and the biological function of hepatocyte-derived IL-1Ra in concanavalin A (ConA)-induced hepatitis in mice. We show that the injured liver produces large amounts of IL-1Ra and that secreted and intracellular IL-1Ra isoforms are produced with different kinetics during the course of hepatitis. By using hepatocyte-specific IL-1Ra-deficient mice (IL-1Ra(ΔH)), we demonstrate that hepatocytes represent the major cellular source of local IL-1Ra. Most interestingly, hepatic necrosis and inflammation were increased in IL-1Ra(ΔH) as compared with wild-type mice during the late phase of the disease, leading to a delayed resolution of hepatitis in IL-1Ra(ΔH) mice. In conclusion, our results show that the local production of IL-1Ra by hepatocytes contributes to the resolution of hepatitis.


Subject(s)
Hepatitis/immunology , Hepatocytes/immunology , Interleukin 1 Receptor Antagonist Protein/immunology , Animals , Concanavalin A/toxicity , Cytokines/analysis , Hepatitis/genetics , Hepatitis/pathology , Hepatocytes/drug effects , Interleukin 1 Receptor Antagonist Protein/genetics , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Necrosis/immunology
15.
Ann Rheum Dis ; 71(2): 281-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22072017

ABSTRACT

OBJECTIVES: To define the cell type (myeloid vs other cells) specific effect of interleukin 1 (IL-1) receptor antagonist (IL-1Ra) deficiency on the acute inflammatory phase of arthritis. METHODS: Arthritis was induced by K/BxN serum transfer in wild-type (WT), IL-1Ra-deficient (IL-1Ra(-/-)) and conditional knockout mice. In the latter, IL-1Ra production was specifically targeted in myeloid cells (IL-1Ra(ΔM)) or in both hepatocytes and myeloid cells (IL-1Ra(ΔH+M)). Arthritis severity was clinically evaluated and ankle sections were scored for synovial inflammation and cartilage erosion. Quantitative RT-PCR, western blot and immunohistochemical analyses measured expression, localisation and cellular sources of the different IL-1Ra isoforms in arthritic joints. RESULTS: Total and myeloid cell-specific IL-1Ra deficiency was associated with increased arthritis severity, although disease incidence was similar to that of WT mice. Increased clinical scores were associated with exacerbated synovial inflammation. All IL-1Ra isoforms, except for intracellular (ic)IL-1Ra2, were expressed in arthritic joints of WT mice. In contrast, production of secreted (s)IL-1Ra and icIL-1Ra3 isoforms was markedly decreased in arthritic joints of both IL-1Ra(ΔM) and IL-1Ra(ΔH+M) mice. Immunohistochemical and western blot analyses suggested that the icIL-1Ra1 isoform is produced primarily by synovial fibroblasts. CONCLUSION: Myeloid cell-derived IL-1Ra, including both sIL-1Ra and icIL-1Ra3 isoforms, controls articular inflammation during the acute phase of K/BxN serum transfer-induced arthritis.


Subject(s)
Arthritis, Experimental/metabolism , Interleukin 1 Receptor Antagonist Protein/physiology , Myeloid Cells/metabolism , Acute Disease , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Disease Progression , Female , Interleukin 1 Receptor Antagonist Protein/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms/deficiency , Protein Isoforms/physiology , Serum , Severity of Illness Index , Synovitis/metabolism , Synovitis/pathology
16.
Arthritis Rheum ; 63(11): 3354-63, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21739421

ABSTRACT

OBJECTIVE: To determine the regulation of class II major histocompatibility complex (MHC) expression in fibroblast-like synoviocytes (FLS) in order to investigate their role as nonprofessional antigen-presenting cells in collagen-induced arthritis (CIA). METHODS: Expression of class II MHC, class II MHC transactivator (CIITA), and Ciita isoforms PI, PIII, and PIV was examined by real-time quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry in human synovial tissues, arthritic mouse joints, and human and murine FLS. CIA was induced in mice in which isoform PIV of Ciita was knocked out (PIV(-/-) ), in PIV(-/-) mice transgenic for CIITA in the thymus (K14 CIITA), and in their control littermates. RESULTS: HLA-DRA, total CIITA, and CIITA PIII messenger RNA levels were significantly increased in synovial tissue samples from patients with rheumatoid arthritis compared with the levels in tissue from patients with osteoarthritis. Human FLS expressed surface class II MHC via CIITA PIII and PIV, while class II MHC expression in murine FLS was entirely mediated by PIV. Mice with a targeted deletion of CIITA PIV lack CD4+ T cells and were protected against CIA. The expression of CIITA was restored in the thymus of PIV(-/-) K14 CIITA-transgenic mice, which had a normal CD4+ T cell repertoire and normal surface levels of class II MHC on professional antigen-presenting cells, but did not induce class II MHC on FLS. Synovial inflammation and immune responses against type II collagen were similar in PIV(-/-) K14 CIITA-transgenic mice and control mice with CIA, but bone erosion was significantly reduced in the absence of PIV. CONCLUSION: Overexpression of class II MHC is tightly correlated with CIITA expression in arthritic synovium and in FLS. Selective targeting of Ciita PIV in peripheral tissues abrogates class II MHC expression by murine FLS but does not protect against inflammation and autoimmune responses in CIA.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Autoimmunity/genetics , Histocompatibility Antigens Class II/metabolism , Inflammation/immunology , Nuclear Proteins/metabolism , T-Lymphocytes/metabolism , Trans-Activators/metabolism , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Cartilage, Articular/immunology , Cartilage, Articular/metabolism , Hip Joint/immunology , Hip Joint/metabolism , Histocompatibility Antigens Class II/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Knee Joint/immunology , Knee Joint/metabolism , Mice , Mice, Knockout , Nuclear Proteins/genetics , Promoter Regions, Genetic , Synovial Membrane/immunology , Synovial Membrane/metabolism , T-Lymphocytes/immunology , Trans-Activators/genetics
17.
Nephrol Dial Transplant ; 26(4): 1413-6, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21220755

ABSTRACT

BACKGROUND: Toll-like receptors (TLR) recognize a variety of ligands, including pathogen-associated molecular patterns and link innate and adaptive immunity. Individual receptors can be up-regulated during infection and inflammation. We examined the expression of selected TLRs at the protein level in various types of renal disease. METHODS: Frozen sections of renal biopsies were stained with monoclonal antibodies to TLR-2, -4 and -9. RESULTS: Up-regulation of the three TLRs studied was seen, although the extent was modest. TLR-2- and -4-positive cells belonged to the population of infiltrating inflammatory cells; only in the case of TLR-9 were intrinsic glomerular cells positive in polyoma virus infection and haemolytic uraemic syndrome (HUS). CONCLUSIONS: Evidence for the involvement of the three TLRs tested in a variety of human renal diseases was found. These findings add to our understanding of the role of the innate immune system in kidney disease.


Subject(s)
Biomarkers/metabolism , Kidney Diseases/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Humans , Kidney/metabolism , Kidney Diseases/pathology , Prognosis
18.
Arthritis Rheum ; 62(3): 765-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20187132

ABSTRACT

OBJECTIVE: RNA interference (RNAi) is a powerful tool for sequence-specific gene silencing, and interest in its application in human diseases is growing. Given the success of recent strategies for administering gene therapy in rheumatoid arthritis using recombinant vectors such as adeno-associated virus type 5 (rAAV5) for optimized intraarticular gene transfer, we undertook the present study to determine the feasibility of using rAAV5-mediated RNAi-based therapy in arthritis. METHODS: We developed rAAV5 vectors expressing short hairpin small interfering RNA (shRNA) against tumor necrosis factor alpha (TNFalpha) under H1 promoter, and carrying the enhanced green fluorescent protein (eGFP) reporter gene under cytomegalovirus promoter (rAAV5-shTNF). TNFalpha gene silencing was validated in vitro with mouse macrophages. Mice with collagen-induced arthritis were injected in the ankle and knee joints, at disease onset, with either rAAV5-shTNF or control rAAV5-eGFP vectors (5 x 10(9) particles). Arthritis severity was assessed clinically and histologically, and immunologic response was examined. Local and systemic transgene expression was monitored using quantitative reverse transcriptase-polymerase chain reaction, immunohistochemical analysis, and enzyme-linked immunosorbent assay. RESULTS: After a single injection of rAAV5-shTNF into inflamed joints, local TNFalpha gene silencing provided rapid and long-term suppression of arthritis progression and reduced joint damage compared with that observed in control groups. Treatment with rAAV5-shTNF was associated with decreased proliferation and interferon-gamma production by antigen-stimulated T cells from draining lymph nodes, and the potency of this treatment was similar to that observed with other treatment strategies targeting TNFalpha at the protein level, either locally or systemically. CONCLUSION: Our data present the first proof-of-concept for the application of rAAV5-mediated RNAi-based gene therapy for local blockade of inflammation in experimental arthritis.


Subject(s)
Arthritis, Experimental/therapy , RNA Interference/physiology , RNA, Small Interfering/administration & dosage , Tumor Necrosis Factor-alpha/genetics , Animals , Dependovirus , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Gene Silencing/physiology , Genetic Vectors , Green Fluorescent Proteins/genetics , Immunohistochemistry , Injections, Intra-Articular , Mice , Tumor Necrosis Factor-alpha/metabolism
19.
Arthritis Rheum ; 62(2): 452-62, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20112392

ABSTRACT

OBJECTIVE: The balance between interleukin-1 (IL-1) and its specific inhibitor, the IL-1 receptor antagonist (IL-1Ra), plays a major role in the development of arthritis. The purpose of this study was to investigate the role of IL-1Ra produced specifically by myeloid cells in the control of collagen-induced arthritis (CIA) by using myeloid cell-specific IL-1Ra-deficient mice (IL-1Ra(DeltaM)). METHODS: IL-1Ra(DeltaM) mice were generated by using the loxP/Cre recombinase system. CIA was induced in IL-1Ra(DeltaM) mice and littermate control mice by a single immunization with bovine type II collagen (CII) in Freund's complete adjuvant. Arthritis severity was assessed by clinical and histologic scoring. Draining lymph node (DLN) cell responses were examined ex vivo, and ankle extracts were used in the quantification of cytokines and chemokines. RESULTS: Clinical and histopathologic evaluations revealed an early disease onset and a severe form of CIA in IL-1Ra(DeltaM) mice. This was characterized by increased production of interferon-gamma (IFNgamma) and IL-17 by CII-stimulated DLN cells. We also observed that the CII-specific CD4+ T cell response shifted in vivo, from a dominant Th1 response early in the course of the arthritis to the presence of both Th1 and Th17 cytokines later in the disease course. Interestingly, IL-1Ra levels were higher in the arthritic joints of IL-1Ra(DeltaM) mice as compared with the controls, indicating that nonmyeloid cells strongly contribute to the local production of IL-1Ra. However, this enhanced IL-1Ra production was not sufficient to limit joint inflammation and tissue damage. CONCLUSION: Our results suggest that myeloid cell-derived IL-1Ra plays a critical role in the control of the development and the severity of CIA by modulating Th1 and Th17 responses in lymphoid organs.


Subject(s)
Arthritis, Experimental/immunology , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-17/metabolism , Myeloid Cells/immunology , Th1 Cells/immunology , Animals , Arthritis, Experimental/pathology , Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Female , Integrases/genetics , Interferon-gamma/metabolism , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myeloid Cells/pathology , Pregnancy , Severity of Illness Index , Th1 Cells/pathology
20.
Arthritis Res Ther ; 11(5): R144, 2009.
Article in English | MEDLINE | ID: mdl-19788740

ABSTRACT

INTRODUCTION: A proliferation-inducing ligand (APRIL) from the TNF family, owing to its role in the generation and survival of plasma cells (PCs), is currently targeted for rheumatoid arthritis (RA) treatment. However, little is known about APRIL expression in RA lesions, hampering our understanding of the way APRIL may modulate this autoimmune disease. METHODS: We performed immunological staining of human normal, non-RA and RA synovial tissues with a pair of antibodies specifically recognizing APRIL-producing cells and secreted APRIL. RESULTS: We detected significant amounts of secreted APRIL in normal synovium mostly concentrated around blood vessels and at the lining layer, but no cells producing APRIL. Meanwhile, we observed that blood neutrophils constitutively secrete APRIL, indicating that blood APRIL may diffuse into the synovium via its fenestrated vessels. Synovium from non-RA and RA patients retained similarly secreted APRIL, but in this case APRIL-producing cells, including neutrophils and macrophages, were present in the tissue. Notably, PCs--when present in RA synovium--accumulated in areas of APRIL retention, spreading from blood vessels towards the lining layer. CONCLUSIONS: PCs accumulate in synovial zones rich in secreted APRIL, consistent with a pro-survival role of APRIL for PCs in RA. The concentration of APRIL by normal synovium indicates that this tissue may constitute a proper environment for PCs even before RA onset.


Subject(s)
Arthritis, Rheumatoid/metabolism , Plasma Cells/metabolism , Synovial Membrane/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Adult , Aged , Aged, 80 and over , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Middle Aged , Neutrophils/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...