Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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