Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 273
1.
Front Immunol ; 12: 778480, 2021.
Article En | MEDLINE | ID: mdl-34887865

Objectives: This study aims to investigate if addition of fibroblast-stromal cell markers to a classification of synovial pathotypes improves their predictive value on clinical outcomes in rheumatoid arthritis (RA). Methods: Active RA patients with a knee needle synovial biopsy at baseline and finished 1-year follow-up were recruited from a real-world prospective cohort. Positive staining for CD20, CD38, CD3, CD68, CD31, and CD90 were scored semiquantitatively (0-4). The primary outcome was radiographic progression defined as a minimum increase of 0.5 units of the modified total Sharp score from baseline to 1 year. Results: Among 150 recruited RA patients, 123 (82%) had qualified synovial tissue. Higher scores of CD20+ B cells, sublining CD68+ macrophages, CD31+ endothelial cells, and CD90+ fibroblasts were associated with less decrease in disease activity and greater increase in radiographic progression. A new fibroblast-based classification of synovial pathotypes giving more priority to myeloid and stromal cells classified samples as myeloid-stromal (57.7%, 71/123), lymphoid (31.7%, 39/123), and paucicellular pathotypes (10.6%, 13/123). RA patients with myeloid-stromal pathotype showed the highest rate of radiographic progression (43.7% vs. 23.1% vs. 7.7%, p = 0.011), together with the lowest rate of Boolean remission at 3, 6, and 12 months. Baseline synovial myeloid-stromal pathotype independently predicted radiographic progression at 1 year (adjusted OR: 3.199, 95% confidence interval (95% CI): 1.278, 8.010). Similar results were obtained in a subgroup analysis of treatment-naive RA. Conclusions: This novel fibroblast-based myeloid-stromal pathotype could predict radiographic progression at 1 year in active RA patients which may contribute to the shift of therapeutic decision in RA.


Antigens, CD/analysis , Arthritis, Rheumatoid/immunology , Fibroblasts/immunology , Immunohistochemistry , Knee Joint/immunology , Stromal Cells/immunology , Synovial Membrane/immunology , Adult , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Biomarkers/analysis , Biopsy, Needle , Disease Progression , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Knee Joint/diagnostic imaging , Knee Joint/drug effects , Knee Joint/pathology , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Remission Induction , Stromal Cells/drug effects , Stromal Cells/pathology , Synovial Membrane/diagnostic imaging , Synovial Membrane/drug effects , Synovial Membrane/pathology , Time Factors , Treatment Outcome
2.
Front Immunol ; 12: 651049, 2021.
Article En | MEDLINE | ID: mdl-34276648

Objective: Autoantibodies are detected in most patients with rheumatoid arthritis (RA) and can be of the IgM, IgG or IgA subclass. Correlations between IgA autoantibodies and more severe disease activity have been previously reported, but the functional role of IgA autoantibodies in the pathogenesis of RA is ill understood. In this study, we explored the effect of IgA immune complexes on osteoclast mediated bone resorption. Methods: Anti-citrullinated peptide antibody (ACPA) and anti-carbamylated protein (anti-CarP) antibody levels of the IgA and IgG isotype and rheumatoid factor (RF) IgA were determined in synovial fluid (SF) of RA patients. Monocytes, neutrophils, and osteoclasts were stimulated with precipitated immune complexes from SF of RA patients or IgA- and IgG-coated beads. Activation was determined by neutrophil extracellular trap (NET) release, cytokine secretion, and bone resorption. Results: NET formation by neutrophils was enhanced by SF immune complexes compared to immune complexes from healthy or RA serum. Monocytes stimulated with isolated SF immune complexes released IL-6 and IL-8, which correlated with the levels of ACPA IgA levels in SF. Osteoclasts cultured in the presence of supernatant of IgA-activated monocytes resorbed significantly more bone compared to osteoclasts that were cultured in supernatant of IgG-activated monocytes (p=0.0233). Osteoclasts expressed the Fc receptor for IgA (FcαRI; CD89) and Fc gamma receptors. IgA-activated osteoclasts however produced significantly increased levels of IL-6 (p<0.0001) and IL-8 (p=0.0007) compared to IgG-activated osteoclasts. Both IL-6 (p=0.03) and IL-8 (p=0.0054) significantly enhanced bone resorption by osteoclasts. Conclusion: IgA autoantibodies induce release of IL-6 and IL-8 by immune cells as well as osteoclasts, which enhances bone resorption by osteoclasts. We anticipate that this will result in more severe disease activity in RA patients. Targeting IgA-FcαRI interactions therefore represents a promising novel therapeutic strategy for RA patients with IgA autoantibodies.


Arthritis, Rheumatoid/immunology , Autoantibodies/immunology , Bone Resorption/immunology , Immunoglobulin A/immunology , Osteoclasts/immunology , Animals , Antigen-Antibody Complex/immunology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology , Bone Resorption/blood , Bone Resorption/pathology , Cattle , Extracellular Traps/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Knee Joint/immunology , Knee Joint/pathology , Osteoclasts/metabolism , Synovial Fluid/immunology
3.
Int J Mol Sci ; 22(5)2021 Mar 08.
Article En | MEDLINE | ID: mdl-33800401

Osteoarthritis (OA) is a progressive degenerative disease that manifests as pain and inflammation and often results in total joint replacement. There is significant interest in understanding how intra-articular injections made from autologous blood or bone marrow could alleviate symptoms and potentially intervene in the progression of the disease. There is in vitro an in vivo evidence that suggests that these therapies, including platelet-rich plasma (PRP), autologous anti-inflammatories (AAIs), and concentrated bone marrow aspirate (cBMA), can interrupt cartilage matrix degradation driven by pro-inflammatory cytokines. This review analyzes the evidence for and against inclusion of white blood cells, the potential role of platelets, and the less studied potential role of blood plasma when combining these components to create an autologous point-of-care therapy to treat OA. There has been significant focus on the differences between the various autologous therapies. However, evidence suggests that there may be more in common between groups and perhaps we should be thinking of these therapies on a spectrum of the same technology, each providing significant levels of anti-inflammatory cytokines that can be antagonists against the inflammatory cytokines driving OA symptoms and progression. While clinical data have demonstrated symptom alleviation, more studies will need to be conducted to determine whether these preclinical disease-modifying findings translate into clinical practice.


Anti-Inflammatory Agents/therapeutic use , Osteoarthritis, Knee/therapy , Platelet-Rich Plasma , Point-of-Care Systems , Humans , Injections, Intra-Articular , Knee Joint/immunology , Knee Joint/pathology , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/pathology
4.
J Orthop Res ; 39(4): 821-830, 2021 04.
Article En | MEDLINE | ID: mdl-33107629

γ-Tilmanocept (99m Tc-tilmanocept) is a receptor-directed, radiolabeled tracer that is FDA-approved for guiding sentinel lymph node biopsy. Tilmanocept binds the C-type lectin mannose receptor (MR, CD206) on macrophages. In this study, nonradioactive, fluorescently-labeled Cy3-tilmanocept was used to detect CD206+ mononuclear cells in the cartilage of mice with antibody-induced arthritis and in the synovial fluid and tissue of human subjects with rheumatoid arthritis (RA) for comparison with osteoarthritis (OA), and healthy volunteer (HV) controls. Murine arthritis was induced by injection of monoclonal anti-cartilage antibody followed by injection of Escherichia coli lipopolysaccharide. Post-arthritis development (7-11 days), the mice were injected intravenously with Cy3-tilmanocept followed by in vivo and ex vivo epifluorescence imaging. Two-photon imaging, immunofluorescence, and immunohistochemistry were used to identify articular and synovial macrophages (CD206, F4/80, and Cy3-tilmanocept binding) in murine tissues. Cy3-tilmanocept epifluorescence was present in arthritic knees and elbows of murine tissues; no radiographic changes were noted in the skeletons. However, inflammatory arthritic changes were apparent by histopathology and immunohistochemistry (F4/80), immunofluorescence (CD206) and Cy3-tilmanocept binding. In human RA synovial fluid, Cy3-tilmanocept staining correlated with CD206+ /CD16+ cells; negligible labeling was observed in OA samples. Cy3-tilmanocept colocalized with CD206 and staining was significantly higher in RA synovial tissue compared to OA or HV. Our results demonstrate that imaging with Cy3-tilmanocept can detect in vivo inflammatory, CD206+ macrophages in an early arthritis animal model and in human RA patients. These data establish a novel tool for preclinical research of early arthritis and have implications for early RA detection and monitoring of therapeutic efficacy in humans.


Arthritis, Rheumatoid/diagnostic imaging , Joints/diagnostic imaging , Macrophages/immunology , Synovial Membrane/diagnostic imaging , Animals , Antibodies, Monoclonal , Arthritis, Rheumatoid/immunology , Carbocyanines/pharmacology , Dextrans/chemistry , Escherichia coli/metabolism , Healthy Volunteers , Humans , Inflammation , Joints/immunology , Knee Joint/diagnostic imaging , Knee Joint/immunology , Lectins, C-Type/chemistry , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/chemistry , Male , Mannans/chemistry , Mannose Receptor , Mannose-Binding Lectins/chemistry , Mice , Mice, Inbred DBA , Microscopy, Fluorescence , Osteoarthritis/diagnostic imaging , Osteoarthritis/immunology , Photons , Receptors, Cell Surface/chemistry , Synovial Membrane/immunology , Technetium/chemistry , Technetium Tc 99m Pentetate/analogs & derivatives , Technetium Tc 99m Pentetate/chemistry
5.
Int J Surg Pathol ; 29(2): 216-222, 2021 Apr.
Article En | MEDLINE | ID: mdl-32631118

Inflammatory myofibroblastic tumor (IMT) is a lesion of intermediate biological potential with local recurrences and rare metastases found in multiple anatomical locations. We present a case of a pure intraarticular IMT of the knee, a location that has not been previously documented, with genetic confirmation of ALK-CARS fusion detected with next-generation sequencing. A 20-year-old healthy male was admitted to the orthopedic oncology department due to several months of pain and restriction in movement of his left knee. On magnetic resonance imaging, multiple intraarticular nodular lesions were seen. The patient underwent 2 synovectomies within the course of 1 year. The initial biopsy was interpreted as nodular fasciitis. The second biopsy revealed exuberant tissue displaying compact fascicles of spindle cells intermixed with myxoid areas in a background of inflammatory cells, highly suggestive for IMT. Due to the unusual intraarticular location, equivocal ALK immunostaining and the differential diagnosis with nodular fasciitis, we performed targeted next-generation sequencing using Archer FusionPlex Sarcoma panel, which can identify multiple fusions in a single assay. An ALK-CARS fusion was found, supporting the diagnosis of IMT. This report emphasizes the added value of broad molecular analysis in cases with unusual clinical presentation, equivocal immunohistochemistry, and a wide differential diagnosis.


Knee Joint/pathology , Oncogene Proteins, Fusion/genetics , Soft Tissue Neoplasms/diagnosis , Synovial Membrane/pathology , Amino Acyl-tRNA Synthetases/genetics , Anaplastic Lymphoma Kinase/genetics , Biopsy , Cytoreduction Surgical Procedures , Diagnosis, Differential , Fasciitis/diagnosis , High-Throughput Nucleotide Sequencing , Humans , Knee Joint/diagnostic imaging , Knee Joint/immunology , Knee Joint/surgery , Magnetic Resonance Imaging , Male , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/immunology , Soft Tissue Neoplasms/surgery , Synovectomy , Synovial Membrane/diagnostic imaging , Synovial Membrane/immunology , Young Adult
6.
Biosci Rep ; 40(9)2020 09 30.
Article En | MEDLINE | ID: mdl-32840301

OBJECTIVES: Rheumatoid arthritis (RA) is the most common inflammatory arthritis in the world, but its underlying mechanism is still unclear. The present study aims to screen and verify the potential biomarkers of RA. METHODS: We searched the Gene Expression Omnibus (GEO) database for synovial expression profiling from different RA microarray studies to perform a systematic analysis. Functional annotation of differentially expressed genes (DEGs) was conducted, including GO enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The protein-protein interaction (PPI) networks of the DEGs were constructed based on data from the STRING database. The expression levels of the hub genes in normal membranes and RA synovium were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot system. RESULTS: A total of 444 differential expression genes were identified, including 172 up-regulated and 272 down-regulated genes in RA synovium compared with normal controls. The top ten hub genes; protein tyrosine phosphatase receptor type C (PTPRC), LCK proto-oncogene (LCK), cell division cycle 20 (CDC20), Jun proto-oncogene (JUN), cyclin-dependent kinase 1 (CDK1), kinesin family member 11 (KIF11), epidermal growth factor receptor (epidermal growth factor receptor (EGFR), vascular endothelial growth factor A (VEGFA), mitotic arrest deficient 2 like 1 (MAD2L1), and signal transducer and activator of transcription 1 (STAT1) were identified from the PPI network, and the expression level of VEGFA and EGFR was significantly increased in RA membranes (P<0.05). CONCLUSION: Our results indicate that the hub genes VEGFA and EGFR may have essential effects during the development of RA and can be used as potential biomarkers of RA.


Arthritis, Rheumatoid/diagnosis , Synovial Membrane/pathology , Synovitis/diagnosis , Vascular Endothelial Growth Factor A/analysis , Adult , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/surgery , Arthroplasty, Replacement, Knee , Biomarkers/analysis , Biomarkers/metabolism , Case-Control Studies , Computational Biology , Datasets as Topic , ErbB Receptors/analysis , ErbB Receptors/metabolism , Female , Gene Expression Profiling , Humans , Knee Joint/immunology , Knee Joint/pathology , Knee Joint/surgery , Male , Middle Aged , Proto-Oncogene Mas , Synovial Membrane/immunology , Synovitis/immunology , Synovitis/pathology , Synovitis/surgery , Tissue Array Analysis , Vascular Endothelial Growth Factor A/metabolism
7.
Int Immunopharmacol ; 85: 106598, 2020 Aug.
Article En | MEDLINE | ID: mdl-32442901

Previous studies in a rat model of Sephadex induced lung inflammation showed that 4-Thiouridine (4SU), a thiol substituted nucleoside, was very effective in reducing edema, leukocyte influx and TNF levels in bronchoalvelolar lavage fluid. However, little is known about the factors and mechanisms underlying these effects. In the present study, we have used two separate mouse models of chronic inflammation, a model of dextran sulphate sodium (DSS) induced colitis and a model of antigen induced arthritis, to evaluate the anti-inflammatory effect of 4-thiouridine. We have analyzed a broad spectrum of inflammatory mediators in order to delineate the mechanisms behind a potential anti-inflammatory effect of 4SU. Colitis was induced in C57BL/6 mice by administration of 3.5% DSS in drinking water for 5 days and the potential anti-colitic effect of 4SU was assessed by monitoring the disease activity index (DAI), measurement of colon length and histopathological analysis of colon tissue. We analyzed tissue myeloperoxidase (MPO) activity, serum pro-inflammatory cytokines (IL-1ß, IL-6 and TNF), mRNA and protein expression of pro-inflammatory cytokines, COX-2, and NF-κB activity in colitis tissue. Intracolonic administration of 4SU (5 mg/kg & 10 mg/kg.) significantly inhibited MPO activity and reduced the levels of pro-inflammatory cytokines (IL-1ß, IL-6 and TNF) as well as COX-2. Further, NF-κB activation was also blocked by attenuating the phosphorylation of IkB kinase (IKK α/ß) in DSS-induced colitis tissues. Arthritis was induced by intra-articular injection of mBSA in the knee of NMRI mice pre-immunized with mBSA and 4SU was administered locally by direct injection into the knee joint. The antiarthritic potential of 4SU was calculated by histopathological scores and histochemical analysis of joint tissue. Further, immunohistochemistry was used to study inflammatory cell infiltration and expression of cytokines and adhesion molecules in the synovium. Local administration of 50-100 mg/kg 4SU at the time of arthritis onset clearly prevented development of joint inflammation and efficiently inhibited synovial expression of CD18, local cytokine production and recruitment of leukocytes to the synovium. Taken together, our data clearly demonstrates a potent anti-inflammatory effect of 4SU in two experimental models. In conclusion 4SU could be a new promising candidate for therapeutic modulation of chronic inflammatory diseases like ulcerative colitis and arthritis.


Anti-Inflammatory Agents/therapeutic use , Arthritis/drug therapy , Colitis, Ulcerative/drug therapy , Thiouridine/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Antigens , Arthritis/immunology , Arthritis/pathology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Cyclooxygenase 2/immunology , Cytokines/blood , Cytokines/genetics , Cytokines/immunology , Dextran Sulfate , Disease Models, Animal , Female , Knee Joint/drug effects , Knee Joint/immunology , Knee Joint/pathology , Male , Mice, Inbred C57BL , NF-kappa B/immunology , Serum Albumin, Bovine , Thiouridine/pharmacology
8.
Biomed Res Int ; 2020: 9647072, 2020.
Article En | MEDLINE | ID: mdl-32258161

BACKGROUND: Immunological mechanisms play a vital role in the pathogenesis of knee osteoarthritis (KOA). Moreover, the immune phenotype is a relevant prognostic factor in various immune-related diseases. In this study, we used CIBERSORT for deconvolution of global gene expression data to define the immune cell landscape of different structures of knee in osteoarthritis. Methods and Findings. By applying CIBERSORT, we assessed the relative proportions of immune cells in 76 samples of knee cartilage, 146 samples of knee synovial tissue, 40 samples of meniscus, and 50 samples of knee subchondral bone. Enumeration and activation status of 22 immune cell subtypes were provided by the obtained immune cell profiles. In synovial tissues, the differences in proportions of plasma cells, M1 macrophages, M2 macrophages, activated dendritic cells, resting mast cells, and eosinophils between normal tissues and osteoarthritic tissues were statistically significant (P < 0.05). The area under the curve was relatively large in resting mast cells, dendritic cells, and M2 macrophages in receiver operating characteristic analyses. In subchondral bones, the differences in proportions of resting master cells and neutrophils between normal tissues and osteoarthritic tissues were statistically significant (P < 0.05). In subchondral bones, the proportions of immune cells, from the principle component analyses, displayed distinct group-bias clustering. Resting mast cells and T cell CD8 were the major component of first component. Moreover, we revealed the potential interaction between immune cells. There was almost no infiltration of immune cells in the meniscus and cartilage of the knee joint. CONCLUSIONS: The immune cell composition in KOA differed substantially from that of healthy joint tissue, while it also differed in different anatomical structures of the knee. Meanwhile, activated mast cells were mainly associated with high immune cell infiltration in OA. Furthermore, we speculate M2 macrophages in synovium and mast cells in subchondral bone may play an important role in the pathogenesis of OA.


Cartilage, Articular/immunology , Gene Expression Regulation/immunology , Knee Joint/immunology , Osteoarthritis, Knee/immunology , Cartilage, Articular/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Eosinophils/immunology , Eosinophils/pathology , Female , Humans , Knee Joint/pathology , Macrophages/immunology , Macrophages/pathology , Male , Mast Cells/immunology , Mast Cells/pathology , Osteoarthritis, Knee/pathology , Plasma Cells/immunology , Plasma Cells/pathology
9.
Eur J Pharmacol ; 879: 173054, 2020 Jul 15.
Article En | MEDLINE | ID: mdl-32145326

We have previously shown that endogenous adenosine 5'-triphosphate (ATP), via P2X3 and P2X2/3 receptors, plays an essential role in carrageenan-induced articular hyperalgesia model in rats' knee joint. In the present study, we used the rat knee joint incapacitation test, Enzyme-Linked Immunosorbent Assay (ELISA), and myeloperoxidase enzyme activity assay, to test the hypothesis that the activation of P2X3 and P2X2/3 receptors by their agonist induces articular hyperalgesia mediated by the inflammatory mediators bradykinin, prostaglandin, sympathomimetic amines, pro-inflammatory cytokines and by neutrophil migration. We also tested the hypothesis that the activation of P2X3 and P2X2/3 receptors contributes to the articular hyperalgesia induced by the inflammatory mediators belonging to carrageenan inflammatory cascade. The non-selective P2X3 and P2X2/3 receptors agonist αß-meATP induced a dose-dependent articular hyperalgesia, which was significantly reduced by the selective antagonists for P2X3 and P2X2/3 receptors (A-317491), bradykinin B1- (DALBK) or B2-receptors (bradyzide), ß1-(atenolol) or ß2-adrenoceptors (ICI-118,551), by the pre-treatment with cyclooxygenase inhibitor (indomethacin) or with the nonspecific selectin inhibitor (Fucoidan). αß-meATP induced the release of pro-inflammatory cytokines TNFα, IL-1ß, IL-6, and CINC-1, as well as the neutrophil migration. Moreover, the co-administration of A-317491 significantly reduced the articular hyperalgesia induced by bradykinin, prostaglandin E2 (PGE2), and dopamine. These findings suggest that peripheral P2X3 and P2X2/3 receptors activation induces articular hyperalgesia by an indirect sensitization of the primary afferent nociceptor of rats' knee joint through the release of inflammatory mediators. Further, they also indicate that the activation of these purinergic receptors by endogenous ATP mediates the bradykinin-, PGE2-, and dopamine-induced articular hyperalgesia.


Hyperalgesia/metabolism , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X3/metabolism , Adenosine Triphosphate/analogs & derivatives , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Bradykinin , Cytokines/immunology , Dinoprostone , Dopamine , Hyperalgesia/chemically induced , Hyperalgesia/immunology , Knee Joint/immunology , Knee Joint/metabolism , Male , Neutrophils/drug effects , Phenols/pharmacology , Phenols/therapeutic use , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Purinergic P2X Receptor Agonists , Purinergic P2X Receptor Antagonists/pharmacology , Purinergic P2X Receptor Antagonists/therapeutic use , Rats, Wistar
10.
Zhonghua Nei Ke Za Zhi ; 58(12): 911-914, 2019 Dec 01.
Article Zh | MEDLINE | ID: mdl-31775456

The purpose of this study was to explore the role and mechanism of transient receptor potential M(2) (TRPM(2)) in antigen-induced arthritis (AIA) mice. Twelve C57BL/6 mice and 12 TRPM(2) knockout mice were divided into 4 groups, includingwild type control group, wild type AIA group, TRPM(2) knockout control group and TRPM(2) knockout AIA group, with 6 mice in each group. Methylated bovine serum albumin (mBSA) was used to establish AIA mouse model. The degree of joint swelling and inflammatory cell infiltration were recorded, as well as synovial hyperplasia of the knee joints. Real-time quantitative polymerase chain reaction (PCR) was used to detect the expression of interleukin (IL)-6, IL-8, chemokine ligand 6 (CXCL-6) and tumor necrosis factor alpha (TNFα) mRNA in synovial cells of knee joints. The results showed that compared with the wild-type AIA group, the TRPM(2) knockout AIA group had more significant synovial proliferation and inflammatory cell infiltration in the synovial tissue.The neutrophil and macrophage counts rather than monocytes in the knee joints of TRPM(2) knockout AIA group were higher than those in wild-type AIA mice. The expression of IL-6, IL-8 and CXCL-6 mRNA were significantly increased in the knock out mice. In summary, TRPM(2) may inhibit inflammatory cytokines such as IL-6 and IL-8 in knee joints of AIA mice by reducing the infiltration of neutrophils and macrophages, the refore alleviates the manifestations of knee arthritis.


Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Chemokine CXCL6/immunology , Interleukin-6/immunology , Interleukin-8/immunology , Serum Albumin, Bovine/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antigens/immunology , Arthritis, Rheumatoid/genetics , Chemokine CXCL6/genetics , Interleukin-6/genetics , Interleukin-8/genetics , Knee Joint/immunology , Knee Joint/pathology , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Synovial Membrane , Tumor Necrosis Factor-alpha/genetics
11.
Int Immunopharmacol ; 77: 105900, 2019 Dec.
Article En | MEDLINE | ID: mdl-31644963

Via promoting synovitis, pannus growth and cartilage/bone destruction, fibroblast-like synovial cells (FLSs) play a significant role in the pathogenesis of rheumatoid arthritis (RA). In our study, rats were induced with complete freund's adjuvant (CFA) to be animal models for studying the RA pathogenesis. Microtubule-associated Serine/Threonine-protein kinase 3 (MAST3) has been documented to play a critical role in regulating the immune response of IBD (Inflammatory bowel disease) and involved in the process of cytoskeleton organization, intracellular signal transduction and peptidyl-serine phosphorylation, but its role in the progression of RA remains unknown and is warranted for investigation. So, we tried our best to investigate the mechanism and signaling pathway of MAST3 in RA progression. In the synovial tissue and FLSs of AA rats, we have found that MAST3 was significantly up-regulated than normal. Furthermore, MAST3 overexpression could promote proliferation and inflammatory response of FLSs. In the aspect of mechanism, we discovered that the expression of MAST3 might involve in NF-κB signaling pathway in RA. On the whole, our results suggested that MAST3 might promote the proliferation and inflammation of FLSs by regulating NF-κB signaling pathway.


Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Microtubule-Associated Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Synoviocytes/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cell Proliferation , Cells, Cultured , Female , Inflammation/genetics , Inflammation/immunology , Knee Joint/immunology , Knee Joint/pathology , Microtubule-Associated Proteins/genetics , NF-kappa B/immunology , Protein Serine-Threonine Kinases/genetics , Rats, Sprague-Dawley
12.
Bull Hosp Jt Dis (2013) ; 77(3): 183-188, 2019 Sep.
Article En | MEDLINE | ID: mdl-31487483

PURPOSE: This study attempts to establish whether local adiposity of the knee at the level of the joint line is associated with alterations in synovial fluid biomarker concentrations in patients undergoing ACL reconstruction with or without an associated meniscectomy. METHODS: Patients undergoing ACL reconstruction were prospectively enrolled at the time of surgery from July 2011 to January 2015. Synovial fluid samples were collected just prior to incision and the concentrations of 10 biomarkers of interest were determined using a multiplex magnetic bead immunoassay. Knee adiposity was assessed via measures of leg fat area using magnetic resonance axial T2 images at the level of the joint line. Measurement was determined by subtracting the sum of the joint area, consisting of bony and muscle areas, from the total leg area with six different ratios assessed. Groups were evaluated by injury type (isolated ACL, ACL + meniscal injury, and total cohort). The correlation between synovial fluid biomarker levels and leg fat area ratios was evaluated using Spearman's correlation. RESULTS: There were 22 females and 26 males, with a mean age of 33.8 years (± 10.5) and a mean BMI of 25.3 (± 4.0). In the setting of isolated ACL injury, there was a statistically significant correlation between leg fat ratios and interleukin- 6, vascular endothelial growth factor, and interleukin-1 receptor antagonist. In patients with concomitant meniscal tears, there was an inverse correlation between leg fat ratios and monocyte chemoattractant protein-1. CONCLUSION: The leg fat to total leg volume ratio and leg fat to joint space volume ratio were the most consistent measures for alterations in post-injury synovial fluid biomarker concentrations. Analysis of synovial fluid at the time of ACL reconstruction demonstrated significant correlations between specific leg-fat area ratios and synovial fluid biomarker concentrations. Local adiposity around the knee joint appears to modulate the biochemical environment of the joint and can clinically help guide prognostic discussions with the patient.


Adiposity , Anterior Cruciate Ligament Injuries/surgery , Anterior Cruciate Ligament Reconstruction , Chemokine CCL2/metabolism , Knee Joint , Synovial Fluid , Adult , Anterior Cruciate Ligament Reconstruction/adverse effects , Anterior Cruciate Ligament Reconstruction/methods , Biomarkers/metabolism , Body Mass Index , Female , Humans , Interleukins/metabolism , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/pathology , Male , Meniscectomy/methods , Prognosis , Synovial Fluid/immunology , Synovial Fluid/metabolism , Vascular Endothelial Growth Factor A/metabolism
13.
Bull Hosp Jt Dis (2013) ; 77(3): 189-193, 2019 Sep.
Article En | MEDLINE | ID: mdl-31487484

BACKGROUND: Synovial fluid biomarkers can highlight the molecular milieu associated with knee pathology and have been shown to be significantly different in patients with anterior cruciate ligament (ACL) injuries compared to uninjured controls. The purpose of the current study was to establish how synovial fluid biomarker concentrations change in patients undergoing ACL reconstruction between the immediate preoperative period to the acute postoperative period. METHODS: Patients were prospectively enrolled at the time of surgery from September 2016 to March 2017. Patients who had an operative knee synovial fluid sample obtained at the time of ACL reconstruction and provided a synovial fluid sample at their first postoperative appointment were included. The concentrations of 10 biomarkers were determined using a multiplex magnetic bead immunoassay. Biomarker concentrations before and after surgery were compared using a paired sample t-test. RESULTS: Eight patients with mean age of 33.4 years who underwent isolated ACL reconstruction using a bonepatellar tendon-bone autograft were included. The mean time between surgery and postoperative office visit was 10.4 days. There was a statistically significant increase in the concentrations of interleukin-6 (IL-6, p = 0.014), monocyte chemoattractant protein-1 (MCP-1, p = 0.024), human matrix metalloproteinase 3 (MMP-3, p = 0.00002), macrophage inflammatory protein-1 beta (MIP-1ß, p = 0.006), human interleukin-1 receptor antagonist (IL-1Ra, p = 0.017), and vascular endothelial growth factor (VEGF, p = 0.023) between the time of surgery and the first postoperative visit and a decrease in the concentration of tissue inhibitor of metalloproteinase-2 (p = 0.050). CONCLUSION: The molecular profile of the synovial fluid changes in the early postoperative period following arthroscopic ACL reconstruction. The concentration of proinflammatory markers (such as IL-6, MCP-1, MMP-3, and MIP-1ß) and growth factors including VEGF increases. The concentration of the anti-inflammatory marker tissue inhibitor of metalloproteinase-2 (TIMP-2) appears to decrease postoperatively.


Anterior Cruciate Ligament Injuries/surgery , Biomarkers/analysis , Synovial Fluid , Adult , Anterior Cruciate Ligament Reconstruction/adverse effects , Anterior Cruciate Ligament Reconstruction/methods , Arthroscopy/methods , Chemokine CCL2/analysis , Chemokine CCL4/analysis , Female , Humans , Interleukin 1 Receptor Antagonist Protein/analysis , Interleukin-6/analysis , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/surgery , Male , Matrix Metalloproteinase 3/analysis , Perioperative Period , Synovial Fluid/immunology , Synovial Fluid/metabolism , Vascular Endothelial Growth Factor A/analysis
14.
Pharmacol Res ; 147: 104351, 2019 09.
Article En | MEDLINE | ID: mdl-31315067

Gout is a paradigm of acute, self-limiting inflammation caused by the deposition of monosodium urate (MSU) crystals within intra-and/or peri-articular areas, leading to excruciating pain, joint swelling and stiffness. The infiltration of leukocytes drives the inflammatory response and remains an attractive target for therapeutic intervention. In this context, emerging evidence supports the view that systemic differentiation of Th17 cells and their in situ infiltration as one of the potential mechanisms by which these cells, and their main product IL-17, causes damage to target tissues. To test if IL-17 was having a detrimental role in gouty onset and progression we targeted this cytokine, using a neutralizing antibody strategy, in an experimental model of gout. Joint inflammation was induced in CD-1 mice by the intra-articular (i.a.) administration of MSU crystals (200 µg/20 µl). Animals from IL-17Ab-treated groups received 1, 3 and 10 µg (i.a.) in 20 µl of neutralizing antibody after MSU crystals administration. Thereafter, joints were scored macroscopically, and knee joint oedema determined with a caliper. Histological analysis, myeloperoxidase assay and western blots analysis for COX-2/mPGEs-1/IL-17R pathway were conducted at 18 h (peak of inflammation) to evaluate leukocytes infiltration and activation, followed by the analysis, in situ, of pro/anti-inflammatory cytokines and chemokines. Flow cytometry was also used to evaluate the modulation of infiltrated inflammatory monocytes and systemic Th17 and Treg profile. Treatment with IL-17Ab revealed a dose-dependent reduction of joint inflammation scores with maximal inhibition at 10 µg. The neutralizing antibody was also able to significantly reduce leukocytes infiltration and MPO activity as well the expression of JE, IL-1α, IL-1ß, IL-16, IL-17, C5a, BLC and, with a less extent IP-10, Rantes, KC, TIMP-1, SDF-1 and metalloproteinases in inflamed tissues. Biochemical analysis also revealed that IL-17Ab treatment modulated COX-2/mPGEs-1 pathway (and related PGE2 production) without interfering with IL-17R expression. Furthermore, flow cytometry analysis highlighted a selective modulation of infiltrating inflammatory monocytes (B220-/GR1hi-F480hi/CD115+) and circulating Th17, but not Treg, cells after IL-17Ab treatment. Collectively the results of this study report for the first time, that i.a. injection of MSU crystals stimulates in vivo production of Th17 cells and Th17-related inflammatory cyto-chemokines. In addition, we have demonstrated that the administration of a neutralizing antibody against IL-17 attenuates joint symptoms, swelling and leukocytes infiltration to the inflamed tissue, possibly providing a new strategy for the treatment of gouty inflammation and/or arthritis.


Antibodies, Neutralizing/immunology , Gout/immunology , Interleukin-17/immunology , Uric Acid , Animals , Edema/immunology , Edema/pathology , Gout/pathology , Inflammation/immunology , Inflammation/pathology , Injections, Intra-Articular , Knee Joint/immunology , Knee Joint/pathology , Male , Mice
15.
Drug Dev Res ; 80(3): 353-359, 2019 05.
Article En | MEDLINE | ID: mdl-30663793

Preclinical Research & Development Curcumin has been shown to possess a series of beneficial effects, such as antiinflammatory, antioxidant, analgesic, and promoting healing. However, the effect and relative mechanism of curcumin on knee osteoarthritis (OA) have not been elucidated. The aim of this study is to explore the protective effect of curcumin on monosodium iodoacetate (MIA)-induced OA. Forty-eight rats were randomized into four experimental groups: control group, OA group, OA + PBS group, and OA + curcumin group, respectively. A single intraarticular injection of MIA was applied to establish the rat model of knee OA. Hematoxylin-eosin staining was used to evaluate histological changes of knee joint. The paw withdrawal threshold was collected and the expression of synovial fluid cytokine levels was measured by ELISA. The protein expression of TRL-4, MyD88, p-IκBα, NF-κB, TNF-α, IL-1ß, and IL6 was measured by western blot. Treating with curcumin can significantly reduce joint diameter and Mankin's score, and increase the paw withdrawal threshold. The expression of synovial fluid inflammatory biomarkers, IL-6, IL-1ß, and TNF-α in the OA + curcumin group were lower than that in OA and OA + PBS group. The protein expression of the TLR4 receptor was increased in the OA, OA + PBS, and OA + curcumin group compared to the control group. However, curcumin treatment can significantly decrease the expression of MyD88, p-IκBα, NF-κB, TNF-α, IL-1ß, and IL6 in OA + curcumin group. These findings may indicate that curcumin could block TLR4/NF-κB signal pathway, and reduce inflammation level to prevent knee wound in OA rats. Curcumin may be a feasible kind of medicament in the treatment of knee OA.


Anti-Inflammatory Agents/therapeutic use , Curcumin/therapeutic use , Osteoarthritis, Knee/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Curcumin/pharmacology , Cytokines/immunology , Female , Iodoacetic Acid , Knee Joint/drug effects , Knee Joint/immunology , Knee Joint/pathology , Myeloid Differentiation Factor 88/immunology , NF-kappa B/immunology , Osteoarthritis, Knee/chemically induced , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Synovial Fluid/drug effects , Synovial Fluid/immunology , Toll-Like Receptor 4/immunology
16.
J Knee Surg ; 32(10): 1015-1019, 2019 Oct.
Article En | MEDLINE | ID: mdl-30396205

Tissues from a periprosthetic joint infection (PJI) of the knee contain a heavy neutrophil polymorph (NP) infiltrate (> 5 NPs per high-powered field [HPF] by Musculoskeletal Infection Society [MSIS] criteria). PJI of the knee can be treated by a two-stage procedure and our aim was to determine whether the MSIS histological criteria for PJI diagnosis are valid in a second-stage revision knee arthroplasty. Periprosthetic tissues from 45 second-stage revision knee cases were analyzed histologically by hematoxylin-eosin and chloroacetate esterase (CAE) staining for the identification of NPs. The number of NPs was determined semiquantitatively and results correlated with the microbiological and clinical findings. In 9 of the 45 cases, an organism was cultured in two or more samples, meeting MSIS microbiological criteria for a definite diagnosis of PJI; histologically, seven of these cases contained > 5 per NPs per HPF on average, with the remaining two cases containing 1 NP and 2 NPs per HPF. In noninfected second-stage revisions, NPs were not seen in 30 cases with 6 cases showing less than 1 NP per HPF on average. The sensitivity, specificity, accuracy, and positive and negative predictive values of MSIS histological criteria (> 5 NPs per HPF) to diagnose PJI were 78%, 100%, 96%, 100%, and 95%, respectively. MSIS histological criteria for the diagnosis of PJI are valid for most but not all infected second-stage revision knee arthroplasties. Correlation of histology with clinical, microbiology and other laboratory findings is required to establish a diagnosis of PJI in second-stage revision knee arthroplasties.


Arthritis, Infectious/pathology , Arthroplasty, Replacement, Knee/methods , Knee Joint/pathology , Neutrophil Infiltration/immunology , Prosthesis-Related Infections/pathology , Adult , Aged , Aged, 80 and over , Arthritis, Infectious/etiology , Arthritis, Infectious/immunology , Arthritis, Infectious/microbiology , Arthroplasty, Replacement, Knee/adverse effects , Female , Humans , Knee Joint/immunology , Knee Joint/microbiology , Knee Joint/surgery , Leukocyte Count/methods , Male , Middle Aged , Prosthesis-Related Infections/etiology , Prosthesis-Related Infections/immunology , Prosthesis-Related Infections/microbiology , Reoperation
17.
Clin Exp Immunol ; 195(3): 395-406, 2019 03.
Article En | MEDLINE | ID: mdl-30368774

Thorough understanding of the complex pathophysiology of osteoarthritis (OA) is necessary in order to open new avenues for treatment. The aim of this study was to characterize the CD4+ T cell population and evaluate their activation and polarization status in OA joints. Fifty-five patients with end-stage knee OA (Kellgren-Lawrence grades III-IV) who underwent surgery for total knee arthroplasty (TKA) were enrolled into this study. Matched samples of synovial membrane (SM), synovial fluid (SF) and peripheral blood (PB) were analysed for CD3+ CD4+ CD8- T cell subsets [T helper type 1 (Th1), Th2, Th17, regulatory T cells] and activation status (CD25, CD69, CD45RO, CD45RA, CD62L) by flow cytometry. Subset-specific cytokines were analysed by cytometric bead array (CBA). SM and SF samples showed a distinct infiltration pattern of CD4+ T cells. In comparison to PB, a higher amount of joint-derived T cells was polarized into CD3+ CD4+ CD8- T cell subsets, with the most significant increase for proinflammatory Th1 cells in SF. CBA analysis revealed significantly increased immunomodulating cytokines [interferon (IFN)-γ, interleukin (IL)-2 and IL-10] in SF compared to PB. Whereas in PB only a small proportion of CD4+ T cells were activated, the majority of joint-derived CD4+ T cells can be characterized as activated effector memory cells (CD69+ CD45RO+ CD62L- ). End-stage OA knees are characterized by an increased CD4+ T cell polarization towards activated Th1 cells and cytokine secretion compared to PB. This local inflammation may contribute to disease aggravation and eventually perpetuate the disease process.


Knee Joint/immunology , Osteoarthritis, Knee/immunology , Synovial Fluid/immunology , Synovial Membrane/immunology , Th1 Cells/physiology , Adult , Aged , Aged, 80 and over , Cell Polarity , Cytokines/analysis , Female , Humans , Lymphocyte Activation , Male , Middle Aged
18.
Theranostics ; 8(20): 5519-5528, 2018.
Article En | MEDLINE | ID: mdl-30555561

Rationale: Recent studies confirmed that osteoarthritis (OA) is associated with systemic inflammation. Adipose-derived stromal cells (ASCs) could become the most promising cell-based therapy in OA, based not only on their differentiation capacities and trophic and paracrine effects on the existing cartilage, but also on their immunomodulatory properties. Here, we wanted to determine the biological effect of autologous ASC intra-articular (IA) injection. Method: To this aim, we monitored the profile of immune cells in fresh peripheral blood after IA injection of autologous ASCs in the knee of 18 patients with severe OA (ADIPOA phase I study). Specifically, we used 8-color flow cytometry antibody panels to characterize the frequencies of innate and adaptive immune cell subsets (monocytes, dendritic cells, regulatory T cells and B cells) in blood samples at baseline (before injection) and one week, one month and three months after ASC injection. Results: We found that the percentage of CD4+CD25highCD127lowFOXP3+ regulatory T cells was significantly increased at 1 month after ASC injection, and this effect persisted for at least 3 months. Moreover, CD24highCD38high transitional B cells also were increased, whereas the percentage of classical CD14+ monocytes was decreased, at 3 months after ASC injection. These results suggest a global switch toward regulatory immune cells following IA injection of ASCs, underscoring the safety of ASC-based therapy. We did not find any correlation between the scores for the Visual Analogic Scale for pain, the Western Ontario and McMaster Universities Osteoarthritis Index (pain subscale and total score) at baseline and the immune cell profile changes, but this could be due to the small number of analyzed patients. Conclusion: ASCs may drive an immediate local response by releasing paracrine factors and cytokines, and our results suggest that ASCs could also initiate a cascade resulting in a long-lasting systemic immune modulation.


Adipose Tissue/cytology , Mesenchymal Stem Cells/cytology , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/therapy , Stromal Cells/cytology , Flow Cytometry , Humans , Knee Joint/cytology , Knee Joint/immunology , Prospective Studies
19.
J Proteome Res ; 17(11): 3780-3790, 2018 11 02.
Article En | MEDLINE | ID: mdl-30229649

Despite osteoarthritis (OA) and rheumatoid arthritis (RA) being typically age-related, their underlying etiologies are markedly different. We used 1H nuclear magnetic resonance (NMR) spectroscopy to identify differences in metabolite profiles in low volumes of OA and RA synovial fluid (SF). SF was aspirated from knee joints of 10 OA and 14 RA patients. 100 µL SF was analyzed using a 700 MHz Avance IIIHD Bruker NMR spectrometer with a TCI cryoprobe. Spectra were analyzed by Chenomx, Bruker TopSpin and AMIX software. Statistical analysis was undertaken using Metaboanalyst. 50 metabolites were annotated, including amino acids, saccharides, nucleotides and soluble lipids. Discriminant analysis identified group separation between OA and RA cohorts, with 32 metabolites significantly different between OA and RA SF (false discovery rate (FDR) < 0.05). Metabolites of glycolysis and the tricarboxylic acid cycle were lower in RA compared to OA; these results concur with higher levels of inflammation, synovial proliferation and hypoxia found in RA compared to OA. Elevated taurine in OA may indicate increased subchondral bone sclerosis. We demonstrate that quantifiable differences in metabolite abundance can be measured in low volumes of SF by 1H NMR spectroscopy, which may be clinically useful to aid diagnosis and improve understanding of disease pathogenesis.


Arthritis, Rheumatoid/metabolism , Magnetic Resonance Spectroscopy/methods , Metabolome , Metabolomics/methods , Osteoarthritis/metabolism , Synovial Fluid/chemistry , Aged , Amino Acids/chemistry , Amino Acids/classification , Amino Acids/isolation & purification , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Citric Acid Cycle/immunology , Cohort Studies , Female , Glycolysis/immunology , Humans , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/pathology , Lipids/chemistry , Lipids/classification , Lipids/isolation & purification , Male , Metabolomics/instrumentation , Middle Aged , Nucleotides/chemistry , Nucleotides/classification , Nucleotides/isolation & purification , Oligosaccharides/chemistry , Oligosaccharides/classification , Oligosaccharides/isolation & purification , Osteoarthritis/immunology , Osteoarthritis/pathology , Synovial Fluid/metabolism
20.
Arthritis Res Ther ; 20(1): 80, 2018 05 02.
Article En | MEDLINE | ID: mdl-29720243

BACKGROUND: Osteoclast-mediated bone erosion is a central feature of rheumatoid arthritis (RA). Immune complexes, present in a large percentage of patients, bind to Fcγ receptors (FcγRs), thereby modulating the activity of immune cells. In this study, we investigated the contribution of FcγRs, and FcγRIV in particular, during antigen-induced arthritis (AIA). METHODS: AIA was induced in knee joints of wild-type (WT), FcγRI,II,III-/-, and FcγRI,II,III,IV-/- mice. Bone destruction, numbers of tartrate-resistant acid phosphatase-positive (TRAP+) osteoclasts, and inflammation were evaluated using histology; expression of the macrophage marker F4/80, neutrophil marker NIMPR14, and alarmin S100A8 was evaluated using immunohistochemistry. The percentage of osteoclast precursors in the bone marrow was determined using flow cytometry. In vitro osteoclastogenesis was evaluated with TRAP staining, and gene expression was assessed using real-time PCR. RESULTS: FcγRI,II,III,IV-/- mice showed decreased bone erosion compared with WT mice during AIA, whereas both the humoral and cellular immune responses against methylated bovine serum albumin were not impaired in FcγRI,II,III,IV-/- mice. The percentage of osteoclast precursors in the bone marrow of arthritic mice and their ability to differentiate into osteoclasts in vitro were comparable between FcγRI,II,III,IV-/- and WT mice. In line with these observations, numbers of TRAP+ osteoclasts on the bone surface during AIA were comparable between the two groups. Inflammation, a process that strongly activates osteoclast activity, was reduced in FcγRI,II,III,IV-/- mice, and of note, mainly decreased numbers of neutrophils were present in the joint. In contrast to FcγRI,II,III,IV-/- mice, AIA induction in knee joints of FcγRI,II,III-/- mice resulted in increased bone erosion, inflammation, and numbers of neutrophils, suggesting a crucial role for FcγRIV in the joint pathology by the recruitment of neutrophils. Finally, significant correlations were found between bone erosion and the number of neutrophils present in the joint as well as between bone erosion and the number of S100A8-positive cells, with S100A8 being an alarmin strongly produced by neutrophils that stimulates osteoclast resorbing activity. CONCLUSIONS: FcγRs play a crucial role in the development of bone erosion during AIA by inducing inflammation. In particular, FcγRIV mediates bone erosion in AIA by inducing the influx of S100A8/A9-producing neutrophils into the arthritic joint.


Arthritis, Experimental/immunology , Bone and Bones/immunology , Calgranulin A/immunology , Calgranulin B/immunology , Neutrophils/immunology , Receptors, IgG/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Bone and Bones/metabolism , Bone and Bones/pathology , Calgranulin A/metabolism , Calgranulin B/metabolism , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/pathology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Osteoclasts/immunology , Osteoclasts/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Tartrate-Resistant Acid Phosphatase/immunology , Tartrate-Resistant Acid Phosphatase/metabolism
...