Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Rheumatology (Oxford) ; 58(6): 1065-1074, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30649473

ABSTRACT

OBJECTIVES: Previously, we have shown the involvement of Wnt-activated protein Wnt-1-induced signaling protein 1 (WISP1) in the development of OA in mice. Here, we aimed to characterize the relation between WISP1 expression and human OA and its regulatory epigenetic determinants. METHODS: Preserved and lesioned articular cartilage from end-stage OA patients and non-OA-diagnosed individuals was collected. WISP1 expression was determined using immunohistochemistry and damage was classified using Mankin scoring. RNA expression and DNA methylation were assessed in silico from genome-wide datasets (microarray analysis and RNA sequencing, and 450 k-methylationarrays, respectively). Effects of WISP1 were tested in pellet cultures of primary human chondrocytes. RESULTS: WISP1 expression in cartilage of OA patients was increased compared with non-OA-diagnosed controls and, within OA patients, WISP1 was even higher in lesioned compared with preserved regions, with expression strongly correlating with Mankin score. In early symptomatic OA patients with disease progression, higher synovial WISP1 expression was observed as compared with non-progressors. Notably, increased WISP1 expression was inversely correlated with methylation levels of a positional CpG-dinucleotide (cg10191240), with lesioned areas showing strong hypomethylation for this CpG as compared with preserved cartilage. Additionally, we observed that methylation levels were allele-dependent for an intronic single-nucleotide polymorphism nearby cg10191240. Finally, addition of recombinant WISP1 to pellets of primary chondrocytes strongly inhibited deposition of extracellular matrix as reflected by decreased pellet circumference, proteoglycan content and decreased expression of matrix components. CONCLUSION: Increased WISP1 expression is found in lesioned human articular cartilage, and appears epigenetically regulated via DNA methylation. In vitro assays suggest that increased WISP1 is detrimental for cartilage integrity.


Subject(s)
CCN Intercellular Signaling Proteins/metabolism , Cartilage, Articular/metabolism , Osteoarthritis, Knee/metabolism , Proto-Oncogene Proteins/metabolism , Chondrocytes/metabolism , DNA Methylation , Epigenesis, Genetic , Humans , Knee Joint/metabolism
2.
Rheumatology (Oxford) ; 58(3): 536-546, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30508140

ABSTRACT

OBJECTIVE: To investigate the role of AXL, a member of the anti-inflammatory TYRO3, AXL MER (TAM) receptor family, in arthritis. METHODS: KRN serum transfer arthritis was induced in Axl-/- and wild-type mice. Knee and ankle joints were scored macro- and microscopically. Synovial gene and protein expression of Axl was determined in naïve and TGF-ß1-overexpressing joints. AXL expression was determined in M1-like or M2-like macrophages and RA synovium. Human macrophages, fibroblasts and synovial micromasses were stimulated with TGF-ß1 or the AXL inhibitor R428. RESULTS: Ankle joints of Axl-/- mice showed exacerbated arthritis pathology, whereas no effect of Axl gene deletion was observed on gonarthritis pathology. To explain this spatial difference, we examined the synovium of naïve mice. In contrast to the knee, the ankle synovial cells prominently expressed AXL. Moreover, the M2-like macrophage phenotype was the dominant cell type in the naïve ankle joint. Human M2-like macrophages expressed higher levels of AXL and blocking AXL increased their inflammatory response. In the murine ankle synovium, gene expression of Tgfb1 was increased and Tgb1 correlated with Axl. Moreover, TGFB1 and AXL expression also correlated in human RA synovium. In human macrophages and synovial micromasses, TGF-ß1 enhanced AXL expression. Moreover, TGF-ß1 overexpression in naïve murine knee joints induced synovial AXL expression. CONCLUSION: Differences in synovial AXL expression are in accordance with the observation that AXL dampens arthritis in ankle, but not in knee joints. We provide evidence that the local differences in AXL expression could be due to TGF-ß1, and suggest similar pathways operate in RA synovium.


Subject(s)
Arthritis, Experimental/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Synovial Membrane/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Ankle Joint/metabolism , Arthritis, Experimental/genetics , Fibroblasts/metabolism , Humans , Knee Joint/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Axl Receptor Tyrosine Kinase
3.
Rheumatology (Oxford) ; 57(4): 737-747, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29361119

ABSTRACT

Objectives: RA is a chronic autoimmune disease leading to progressive destruction of cartilage and bone. RA patients show elevated IL-22 levels and the amount of IL-22-producing Th cells positively correlates with the extent of erosive disease, suggesting a role for this cytokine in RA pathogenesis. The purpose of this study was to determine the feasibility of SPECT/CT imaging with 111In-labelled anti-fibroblast activation protein antibody (28H1) to monitor the therapeutic effect of neutralizing IL-22 in experimental arthritis. Methods: Mice (six mice/group) with CIA received anti-IL-22 or isotype control antibodies. To monitor therapeutic effects after treatment, SPECT/CT images were acquired 24 h after injection of 111In-28H1. Imaging results were compared with macroscopic, histologic and radiographic arthritis scores. Results: Neutralizing IL-22 before CIA onset effectively prevented arthritis development, reaching a disease incidence of only 50%, vs 100% in the control group. SPECT imaging showed significantly lower joint tracer uptake in mice treated early with anti-IL-22 antibodies compared with the control-treated group. Reduction of disease activity in those mice was confirmed by macroscopic, histological and radiographic pathology scores. However, when treatment was initiated in a later phase of CIA, progression of joint pathology could not be prevented. Conclusion: These findings suggest that IL-22 plays an important role in CIA development, and neutralizing this cytokine seems an attractive new strategy in RA treatment. Most importantly, SPECT/CT imaging with 111In-28H1 can be used to specifically monitor therapy responses, and is potentially more sensitive in disease monitoring than the gold standard method of macroscopic arthritis scoring.


Subject(s)
Arthritis/diagnostic imaging , Cartilage, Articular/diagnostic imaging , Gelatinases/genetics , Gene Expression Regulation , Interleukins/genetics , Membrane Proteins/genetics , RNA, Messenger/genetics , Serine Endopeptidases/genetics , Single Photon Emission Computed Tomography Computed Tomography/methods , Animals , Arthritis/drug therapy , Arthritis/genetics , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Collagen/toxicity , Disease Models, Animal , Endopeptidases , Gelatinases/biosynthesis , Immunohistochemistry , Interleukins/biosynthesis , Male , Membrane Proteins/biosynthesis , Mice , Mice, Inbred DBA , Real-Time Polymerase Chain Reaction , Serine Endopeptidases/biosynthesis , Synovial Membrane/metabolism , Synovial Membrane/pathology , Interleukin-22
4.
J Cell Physiol ; 232(1): 225-33, 2017 01.
Article in English | MEDLINE | ID: mdl-27138291

ABSTRACT

The general consensus is that milk promotes bone growth and density because is a source of calcium and contains components that enhance intestinal calcium uptake or directly affect bone metabolism. In this study, we investigated the effect of bovine-derived milk 100,000 g pellet (P100), which contains nanoparticles (<220 nm) including extracellular vesicles, on osteoclast differentiation and bone resorption. Bone marrow-derived osteoclast precursor cells were differentiated into osteoclasts by M-CSF and RANKL (control) and in the presence of milk P100. Milk P100 treatment until day 4 increased the number of TRAP-positive mononuclear cells and small (≤5 nuclei) osteoclasts. The number of large (≥6 nuclei) osteoclasts remained the same. These alterations were associated with increased expression of TRAP, NFATc1, and c-Fos. Cells seeded in a calcium-phosphate coated plate or bone slices showed reduced resorption area when exposed to milk P100 during the differentiation phase and even after osteoclast formation. Interestingly, milk P100 treatment enhanced Cathepsin K expression but reduced Carbonic Anhydrase 2 gene expression. Moreover, intracellular acid production was also decreased by milk P100 treatment. Oral delivery of milk P100 to female DBA1/J mice for 7 weeks did not alter bone area; however, increased osteoclast number and area in tibia without changes in serum RANKL and CTX-I levels. We showed for the first time the effect of milk P100 on osteoclast differentiation both in vitro and in vivo and found that milk P100 increased the formation of small osteoclasts but this does not lead to more bone resorption probably due to reduced acid secretion. J. Cell. Physiol. 232: 225-233, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Bone Resorption/drug therapy , Cell Differentiation/drug effects , Milk/metabolism , Nanoparticles/administration & dosage , Osteoclasts/metabolism , Animals , Bone Resorption/metabolism , Calcium Phosphates/pharmacology , Cell Differentiation/physiology , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/drug effects
5.
N Engl J Med ; 370(5): 433-43, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24350901

ABSTRACT

BACKGROUND: Plasmacytoid dendritic cells have been implicated in the pathogenesis of systemic sclerosis through mechanisms beyond the previously suggested production of type I interferon. METHODS: We isolated plasmacytoid dendritic cells from healthy persons and from patients with systemic sclerosis who had distinct clinical phenotypes. We then performed proteome-wide analysis and validated these observations in five large cohorts of patients with systemic sclerosis. Next, we compared the results with those in patients with systemic lupus erythematosus, ankylosing spondylitis, and hepatic fibrosis. We correlated plasma levels of CXCL4 protein with features of systemic sclerosis and studied the direct effects of CXCL4 in vitro and in vivo. RESULTS: Proteome-wide analysis and validation showed that CXCL4 is the predominant protein secreted by plasmacytoid dendritic cells in systemic sclerosis, both in circulation and in skin. The mean (±SD) level of CXCL4 in patients with systemic sclerosis was 25,624±2652 pg per milliliter, which was significantly higher than the level in controls (92.5±77.9 pg per milliliter) and than the level in patients with systemic lupus erythematosus (1346±1011 pg per milliliter), ankylosing spondylitis (1368±1162 pg per milliliter), or liver fibrosis (1668±1263 pg per milliliter). CXCL4 levels correlated with skin and lung fibrosis and with pulmonary arterial hypertension. Among chemokines, only CXCL4 predicted the risk and progression of systemic sclerosis. In vitro, CXCL4 down-regulated expression of transcription factor FLI1, induced markers of endothelial-cell activation, and potentiated responses of toll-like receptors. In vivo, CXCL4 induced the influx of inflammatory cells and skin transcriptome changes, as in systemic sclerosis. CONCLUSIONS: Levels of CXCL4 were elevated in patients with systemic sclerosis and correlated with the presence and progression of complications, such as lung fibrosis and pulmonary arterial hypertension. (Funded by the Dutch Arthritis Association and others.).


Subject(s)
Dendritic Cells/metabolism , Platelet Factor 4/blood , Scleroderma, Systemic/blood , Adult , Animals , Biomarkers/blood , Cytokines/metabolism , Familial Primary Pulmonary Hypertension , Female , Humans , Hypertension, Pulmonary/blood , Male , Mice , Mice, Inbred C57BL , Middle Aged , Platelet Factor 4/metabolism , Proteome , Pulmonary Fibrosis/blood , RNA, Messenger/metabolism , Scleroderma, Systemic/etiology , Skin/pathology
6.
Ann Rheum Dis ; 75(8): 1567-73, 2016 08.
Article in English | MEDLINE | ID: mdl-26371289

ABSTRACT

BACKGROUND: Interferon (IFN) signature has been reported in definite systemic sclerosis (SSc) but it has not been characterised in early SSc (EaSSc). We aim at characterising IFN type I signature in SSc before overt skin fibrosis develops. METHODS: The expression of 11 IFN type I inducible genes was tested in whole-blood samples from 30 healthy controls (HCs), 12 subjects with primary Raynaud's phenomenon (RP), 19 patients with EaSSc, 7 patients with definite SSc without cutaneous fibrosis, 21 limited cutaneous SSc and 10 diffuse cutaneous SSc subjects. The correlation between IFN activity in monocytes, B cell activating factor (BAFF) mRNA expression and type III procollagen N-terminal propeptide (PIIINP) serum levels was tested. RESULTS: In all the SSc groups, higher IFN scores were observed compared with HC. An IFN score ≥7.09 discriminated HCs from patients with SSc (sensitivity=0.7, specificity=0.88, area under receiving operating characteristic (AUROC)=0.82); the prevalence of an elevated IFN score was: HC=3.3%; RP=33.3%, EaSSc=78.9%, definite SSc=100%, limited cutaneous SSc=42.9%, diffuse cutaneous SSc=70.0%. In monocytes an IFN score ≥4.12 distinguished HCs from patients with fibrotic SSc (sensitivity=0.62, specificity=0.85, AUROC=0.76). Compared with IFN-negative subjects, IFN-positive subjects had higher monocyte BAFF mRNA levels (19.7±5.2 vs 15.20±4.0, p=2.1×10(-5)) and serum PIIINP levels (median=6.0 (IQR 5.4-8.9) vs median=3.9 (IQR 3.3-4.7), p=0.0004). CONCLUSIONS: An IFN type I signature is observed in patients with SSc from the earliest phases of the disease, even before overt skin fibrosis. The presence of IFN type I signature in monocytes is correlated with BAFF mRNA expression and serum PIIINP levels, supporting a contribution in the pathogenesis and progression of SSc.


Subject(s)
B-Cell Activating Factor/biosynthesis , Interferon Type I/genetics , Scleroderma, Systemic/genetics , Adult , Aged , B-Cell Activating Factor/genetics , Case-Control Studies , Female , Fibrosis , Gene Expression Regulation , Humans , Interferon Type I/biosynthesis , Male , Middle Aged , Monocytes/metabolism , Peptide Fragments/biosynthesis , Peptide Fragments/blood , Procollagen/biosynthesis , Procollagen/blood , RNA, Messenger/genetics , Scleroderma, Systemic/metabolism , Skin/pathology , Transcriptome
7.
Am J Pathol ; 185(7): 1970-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25976248

ABSTRACT

Proteins from the Wnt signaling pathway are very important for joint development. Curiously, osteoarthritis (OA) is thought to be a recapitulation of developmental processes. Various members of the Wnt signaling pathway are overexpressed in the synovium during experimental OA. Here, we investigated the potency of specific Wnt proteins, when expressed in the synovium, to induce OA pathology. We overexpressed Wnt5a, Wnt8a, Wnt16, and WISP1 in the synovium using adenoviral vectors. We determined whether overexpression resulted in OA pathology by histology, and we measured whether Wnt signaling led to increased protease activity in the joint. Synovial overexpression of Wnt8a and Wnt16 led to canonical Wnt signaling in the cartilage, whereas overexpression of Wnt5a did not. Canonical Wnt signaling increased protease activity and induced cartilage damage shortly after overexpression. Specific blocking of the canonical Wnt signaling pathway with Dickkopf-1 reduced the Wnt-signaling-induced cartilage damage. By contrast, the noncanonical signaling Wnt5a did not cause cartilage lesions. Overexpression of WISP1, a downstream protein of canonical Wnt signaling, resulted in increased cartilage damage. In conclusion, our data show that canonical Wnts and WISP1, which we found overexpressed in the synovium during experimental OA, may conduce to OA pathology.


Subject(s)
CCN Intercellular Signaling Proteins/metabolism , Cartilage, Articular/pathology , Osteoarthritis/pathology , Peptide Hydrolases/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Arthritis, Experimental , Cartilage, Articular/metabolism , Cell Line , Humans , Knee/pathology , Male , Mice , Mice, Inbred C57BL , Osteoarthritis/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology , Wnt Proteins/genetics
8.
Rheumatology (Oxford) ; 55(1): 16-24, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26231344

ABSTRACT

There is increasing evidence that low-density lipoprotein (LDL) cholesterol plays a role in the pathology of OA. Specifically, oxidized LDL (oxLDL), which has been shown to play an essential role during development of atherosclerosis, could be involved in processes such as synovial inflammation, cartilage destruction and bone deformations. OxLDL can activate synovial cells such as macrophages, endothelial cells and synovial fibroblasts, resulting in release of growth factors, MMP and pro-inflammatory cytokines. In this review article, we discuss the role of LDL and oxLDL in OA joint pathology and share our viewpoint of possible mechanisms by which these proteins could influence the development and progression of OA. The proposed theory could provide insight into the aetiopathology of OA and give rise to new potential treatments.


Subject(s)
Cartilage, Articular/metabolism , Cholesterol, LDL/metabolism , Lipoproteins, LDL/metabolism , Osteoarthritis/metabolism , Synovial Membrane/metabolism , Cartilage, Articular/pathology , Cytokines/metabolism , Humans , Osteoarthritis/pathology , Oxidation-Reduction
9.
Rheumatology (Oxford) ; 55(9): 1536-47, 2016 09.
Article in English | MEDLINE | ID: mdl-26667213

ABSTRACT

The Wnt signalling pathway is gaining increasing attention in the field of joint pathologies, attributable to its role in the development and homeostasis of the tissues found in the joint, including bone and cartilage. Imbalance in this pathway has been implicated in the development and progression of OA, and interference with the pathway might therefore depict an effective treatment strategy. Though offering multiple opportunities, it is yet to be decided which starting point will bring forth the most promising results. The complexity of the pathway and its interaction with other pathways (such as the TGF-ß signalling pathway, which also has a central role in the maintenance of joint homeostasis) means that acting directly on proteins in this signalling cascade entails a high risk of undesired side effects. Therefore, interference with Wnt-induced proteins, such as WISP1, might be an overall more effective and safer therapeutic approach to inhibit the pathological events that take place during OA.


Subject(s)
CCN Intercellular Signaling Proteins/physiology , Osteoarthritis/etiology , Proto-Oncogene Proteins/physiology , Transforming Growth Factor beta/physiology , Wnt Proteins/physiology , Wnt Signaling Pathway/physiology , Cartilage Diseases/etiology , Cartilage Diseases/physiopathology , Cell Communication/physiology , Cell Nucleus/physiology , Chondrocytes/physiology , Cytoplasm/physiology , Homeostasis/physiology , Humans , Osteoarthritis/physiopathology , Receptor Cross-Talk/physiology
10.
Rheumatology (Oxford) ; 55(11): 2066-2073, 2016 11.
Article in English | MEDLINE | ID: mdl-27411481

ABSTRACT

OBJECTIVE: SSc is a disease characterized by inflammation and fibrosis. Heme Oxygenase-1 (HO-1) is a haem-degrading enzyme that mediates resolution of inflammation and is induced upon mediators abundantly present in SSc. We aimed to assess whether HO-1 expression/function is disturbed in SSc patients and could therefore be contributing to the ongoing inflammation. METHODS: In total, 92 SSc patients and 48 healthy controls were included. By measuring total bilirubin in plasma in vivo, HO-activity was assessed. HO-1 expression levels were determined with western blot in monocytes before and after induction of HO-1 with cobalt protoporphyrin (CoPP) with or without CXCL4. Monocyte-derived dendritic cells (DCs) were stimulated with several Toll-like receptor (TLR) ligands with or without pre-stimulation with CoPP for 24 h. Cytokine levels were measured in the supernatants using the Luminex Bead Array. RESULTS: SSc patients have lower plasma levels of bilirubin, suggestive of an aberrant HO-1 function. We demonstrated low HO-1 expression in immune cells from SSc patients, whereas induction with CoPP was able to restore HO-1 levels in DCs from SSc patients, almost normalizing the increased TLR response observed in SSc. Co-exposure to CXCL4 completely abrogated CoPP-induced HO-1 expression, suggesting that the high CXCL4 levels present in SSc patients block the normal induction of HO-1 and its function. CONCLUSION: We demonstrate that HO activity in SSc patients is decreased and show its functional consequences. Since CXCL4 blocks the induction of HO-1 expression, neutralization of CXCL4 in SSc patients could have clinical benefits by diminishing overactivation of immune cells and other anti-inflammatory effects of HO-1.


Subject(s)
Heme Oxygenase-1/deficiency , Platelet Factor 4/physiology , Scleroderma, Systemic/enzymology , Toll-Like Receptors/physiology , Adult , Bilirubin/metabolism , Carbon Monoxide/metabolism , Case-Control Studies , Cytokines/metabolism , Dendritic Cells/metabolism , Female , Fibroblasts/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Male
11.
Blood ; 124(17): 2713-24, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25214442

ABSTRACT

During inflammation, neutrophils are rapidly mobilized from the bone marrow storage pool into peripheral blood (PB) to enter lesional sites, where most rapidly undergo apoptosis. Monocytes constitute a second wave of inflammatory immigrates, giving rise to long-lived macrophages and dendritic cell subsets. According to descriptive immunophenotypic and cell culture studies, neutrophils may directly "transdifferentiate" into monocytes/macrophages. We provide mechanistic data in human and murine models supporting the existence of this cellular pathway. First, the inflammatory signal-induced MKK6-p38MAPK cascade activates a monocyte differentiation program in human granulocyte colony-stimulating factor-dependent neutrophils. Second, adoptively transferred neutrophils isolated from G-CSF-pretreated mice rapidly acquired monocyte characteristics in response to inflammatory signals in vivo. Consistently, inflammatory signals led to the recruitment of osteoclast progenitor cell potential from ex vivo-isolated G-CSF-mobilized human blood neutrophils. Monocytic cell differentiation potential was retained in left-shifted band-stage neutrophils but lost in neutrophils from steady-state PB. MKK6-p38MAPK signaling in HL60 model cells led to diminishment of the transcription factor C/EBPα, which enabled the induction of a monocytic cell differentiation program. Gene profiling confirmed lineage conversion from band-stage neutrophils to monocytic cells. Therefore, inflammatory signals relayed by the MKK6-p38MAPK cascade induce monocytic cell differentiation from band-stage neutrophils.


Subject(s)
Cell Differentiation/immunology , Inflammation/immunology , MAP Kinase Kinase 6/immunology , Monocytes/immunology , Neutrophils/immunology , Animals , Blotting, Western , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/immunology , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/immunology , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HL-60 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-1beta/pharmacology , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Neutrophils/metabolism , Oligonucleotide Array Sequence Analysis , Osteoblasts/immunology , Osteoblasts/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Transcriptome/drug effects , Transcriptome/immunology , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Immunol ; 192(9): 4103-11, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24683190

ABSTRACT

Increasing epidemiologic evidence supports a link between periodontitis and rheumatoid arthritis. The actual involvement of periodontitis in the pathogenesis of rheumatoid arthritis and the underlying mechanisms remain, however, poorly understood. We investigated the influence of concomitant periodontitis on clinical and histopathologic characteristics of T cell-mediated experimental arthritis and evaluated modulation of type II collagen (CII)-reactive Th cell phenotype as a potential mechanism. Repeated oral inoculations of periodontal pathogens Porphyromonas gingivalis and Prevotella nigrescens induced periodontitis in mice, as evidenced by alveolar bone resorption. Interestingly, concurrent periodontitis induced by both bacteria significantly aggravated the severity of collagen-induced arthritis. Exacerbation of arthritis was characterized by increased arthritic bone erosion, whereas cartilage damage remained unaffected. Both P. gingivalis and P. nigrescens skewed the CII-specific T cell response in lymph nodes draining arthritic joints toward the Th17 phenotype without affecting Th1. Importantly, the levels of IL-17 induced by periodontal pathogens in CII-specific T cells directly correlated with the intensity of arthritic bone erosion, suggesting relevance in pathology. Furthermore, IL-17 production was significantly correlated with periodontal disease-induced IL-6 in lymph node cell cultures. The effects of the two bacteria diverged in that P. nigrescens, in contrast to P. gingivalis, suppressed the joint-protective type 2 cytokines, including IL-4. Further in vitro studies showed that the Th17 induction strongly depended on TLR2 expression on APCs and was highly promoted by IL-1. Our data provide evidence of the involvement of periodontitis in the pathogenesis of T cell-driven arthritis through induction of Ag-specific Th17 response.


Subject(s)
Arthritis, Experimental/complications , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Periodontal Diseases/complications , Periodontal Diseases/immunology , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/pathology , Interleukin-1/immunology , Mice , Mice, Inbred BALB C , Periodontal Diseases/microbiology , Th17 Cells/immunology , Toll-Like Receptor 2/immunology
13.
Ann Rheum Dis ; 74(11): 2084-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25028707

ABSTRACT

OBJECTIVES: Rheumatoid arthritis is a chronic destructive autoimmune disease, but the course is unpredictable in individual patients. An attractive treatment would provide a disease-regulated therapy that offers personalised drug delivery. Therefore, we expressed the anti-inflammatory interleukin-10 (IL-10) gene under the control of inflammation-dependent promoters in a mouse model of arthritis. METHODS: Proximal promoters of S100a8, Cxcl1, Mmp13, Saa3, IL-1b and Tsg6 were selected by whole-genome expression analysis of inflamed synovial tissues from arthritic mice. Mice were injected intraarticularly in knee joints with lentiviral vectors expressing a luciferase reporter or the therapeutic protein IL-10 under control of the Saa3 or Mmp13 promoter. After 4 days, arthritis was induced by intraarticular injection of streptococcal cell walls (SCW). At different time points after arthritis induction, in vivo bioluminescent imaging was performed and knee joints were dissected for histological and RNA analysis. RESULTS: The disease-regulated promoter-luciferase reporter constructs showed different activation profiles during the course of the disease. The Saa3 and Mmp13 promoters were significantly induced at day 1 or day 4 after arthritis induction respectively and selected for further research. Overexpression of IL-10 using these two disease-inducible promoters resulted in less synovitis and markedly diminished cartilage proteoglycan depletion and in upregulation of IL-1Ra and SOCS3 gene expression. CONCLUSIONS: Our study shows that promoters of genes that are expressed locally during arthritis can be candidates for disease-regulated overexpression of biologics into arthritic joints, as shown for IL-10 in SCW arthritis. The disease-inducible approach might be promising for future tailor-made local gene therapy in arthritis.


Subject(s)
Arthritis, Experimental/therapy , Cartilage, Articular/metabolism , Genetic Therapy , Interleukin-10 , Proteoglycans/metabolism , RNA, Messenger/metabolism , Synovial Membrane/immunology , Synovitis/therapy , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid , Cell Wall/immunology , Gene Expression , Interleukin 1 Receptor Antagonist Protein/genetics , Male , Matrix Metalloproteinase 13/genetics , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Serum Amyloid A Protein/genetics , Streptococcus/immunology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Synovial Membrane/pathology , Synovitis/immunology , Synovitis/pathology
14.
Mol Imaging ; 13: 1-10, 2014.
Article in English | MEDLINE | ID: mdl-24881106

ABSTRACT

Rheumatoid arthritis (RA) and osteoarthritis (OA) are serologically and clinically distinctive, but at the local level, both diseases have many molecular pathways in common. In vivo molecular imaging can unravel the local pathologic processes involved in both diseases. In this study, we investigated matrix metalloproteinase (MMP) and cathepsin activity during cartilage destruction, in an RA and an OA mouse model, using biophotonic imaging of substrate-based probes. Mice with collagen-induced arthritis (CIA) or destabilization of the medial meniscus (DMM) were imaged using near-infrared fluorescent probes, activated by several cathepsins or MMPs. Fluorescence signal intensity was compared to synovial gene expression, histology, and cartilage staining of a neoepitope of aggrecan cleaved by MMPs with the amino acids DIPEN. Increased cathepsin and MMP activity was seen during CIA, whereas the DMM model only showed increased MMP activity. DIPEN expression was seen only during CIA. A possible explanation can be differences in gene expressions; MMP3 and -13, known to produce DIPEN neoepitopes, were upregulated in the CIA model, whereas MMP12, known to be involved in elastin degradation and chemokine inhibition, was upregulated in the DMM model. Thus, molecular imaging showed no cathepsin activity at the time of cartilage damage in the DMM model, whereas both cathepsins and MMPs are active in the CIA model during disease progression.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cathepsins/analysis , Matrix Metalloproteinases/analysis , Molecular Imaging/methods , Osteoarthritis/metabolism , Animals , Arthritis, Experimental/metabolism , Cathepsins/metabolism , Cell Death , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen Type II/adverse effects , Collagen Type II/immunology , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Stifle/chemistry , Stifle/metabolism , Stifle/pathology
15.
Cell Tissue Res ; 355(1): 163-71, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24192939

ABSTRACT

Lysyl hydroxylase 2b (LH2b) is known to increase pyridinoline cross-links, making collagen less susceptible to enzymatic degradation. Previously, we observed a relationship between LH2b and osteoarthritis-related fibrosis in murine knee joint. For this study, we investigate if transforming growth factor-beta (TGF-ß) and connective tissue growth factor (CTGF) regulate procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) (gene encoding LH2b) and LH2b expression differently in osteoarthritic human synovial fibroblasts (hSF). Furthermore, we investigate via which TGF-ß route (Smad2/3P or Smad1/5/8P) LH2b is regulated, to explore options to inhibit LH2b during fibrosis. To answer these questions, fibroblasts were isolated from knee joints of osteoarthritis patients. The hSF were stimulated with TGF-ß with or without a kinase inhibitor of ALK4/5/7 (SB-505124) or ALK1/2/3/6 (dorsomorphin). TGF-ß, CTGF, constitutively active (ca)ALK1 and caALK5 were adenovirally overexpressed in hSF. The gene expression levels of PLOD1/2/3, CTGF and COL1A1 were analyzed with Q-PCR. LH2 protein levels were determined with western blot. As expected, TGF-ß induced PLOD2/LH2 expression in hSF, whereas CTGF did not. PLOD1 and PLOD3 were not affected by either TGF-ß or CTGF. SB-505124 prevented the induction of TGF-ß-induced PLOD2, CTGF and COL1A1. Surprisingly, dorsomorphin completely blocked the induction of CTGF and COL1A1, whereas TGF-ß-induced PLOD2 was only slightly reduced. Overexpression of caALK5 in osteoarthritic hSF significantly induced PLOD2/LH2 expression, whereas caALK1 had no effect. We showed, in osteoarthritic hSF, that TGF-ß induced PLOD2/LH2 via ALK5 Smad2/3P. This elevation of LH2b in osteoarthritic hSF makes LH2b an interesting target to interfere with osteoarthritis-related persistent fibrosis.


Subject(s)
Connective Tissue Growth Factor/metabolism , Fibroblasts/metabolism , Osteoarthritis/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Cells, Cultured , Fibroblasts/pathology , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Gene Expression Regulation , Humans , Osteoarthritis/genetics , Osteoarthritis/pathology , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad1 Protein/metabolism , Smad2 Protein/metabolism
16.
Arthritis Rheum ; 65(9): 2310-22, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23784913

ABSTRACT

OBJECTIVE: Wnt signaling plays a pivotal role in skeletal development and in the control of cartilage and bone turnover. We have recently shown that the secreted Wnt antagonist Wnt inhibitory factor 1 (WIF-1) is mainly expressed in the upper layers of epiphyseal and articular cartilage and, to a lesser extent, in bone. Nevertheless, WIF-1(-/-) mice develop normally. In light of these findings, we undertook this study to analyze the role of WIF-1 in arthritis. METHODS: Expression analyses for WIF-1 were performed by real-time reverse transcription-polymerase chain reaction (RT-PCR). WIF-1(-/-) and tumor necrosis factor (TNF)-transgenic mice were crossbred, and the progression of arthritis in TNF-transgenic WIF-1(-/-) mice and littermate controls was evaluated. Structural joint damage was analyzed by histologic staining, histomorphometry, and micro-computed tomography. Wnt/ß-catenin signaling was investigated by real-time RT-PCR and immunofluorescence on primary chondrocytes. RESULTS: WIF-1 expression was repressed by TNFα in chondrocytes and osteoblasts and down-regulated in experimental arthritis and in articular cartilage from patients with rheumatoid arthritis. WIF-1 deficiency partially protected TNF-transgenic mice against bone erosion and loss of trabecular bone, probably as a result of less osteoclast activity. In contrast, arthritis-related cartilage damage was aggravated by WIF-1 deficiency, while overexpression of WIF-1 attenuated cartilage degradation in TNF-transgenic mice. In chondrocytes, TNFα stimulated canonical Wnt signaling, which could be blocked by WIF-1, indicating a direct effect of TNFα and WIF-1 on Wnt signaling in this system. CONCLUSION: These data suggest that WIF-1 may take part in the fine-tuning of cartilage and bone turnover, promoting the balance of cartilage versus bone anabolism.


Subject(s)
Arthritis, Experimental/metabolism , Bone and Bones/metabolism , Cartilage/metabolism , Extracellular Matrix Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Signal Transducing , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Bone and Bones/pathology , Cartilage/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Progression , Down-Regulation , Extracellular Matrix Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Tumor Necrosis Factor-alpha/genetics
17.
Arthritis Rheum ; 65(10): 2583-93, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23860661

ABSTRACT

OBJECTIVE: Previous studies have demonstrated a protective role of Toll-like receptor 2 (TLR-2) and a proinflammatory function of TLR-4 during chronic T cell-driven arthritis. The involvement of TLRs in T cell-independent arthritic processes, however, remains unclear. This study was undertaken to determine the functional significance of TLR-2 and TLR-4 in T cell-independent immune complex-driven arthritis. METHODS: Serum-transfer arthritis was induced in wild-type and TLR-deficient mice by intraperitoneal injections of arthritogenic K/BxN mouse serum. Arthritis was assessed macroscopically and by histologic analysis. The influence of TLR-2 on macrophage cytokine profile, Fcγ receptor (FcγR) expression, and response to immune complexes was determined. RESULTS: While TLR-4, unexpectedly, did not play any significant role, TLR-2 deficiency accelerated the onset and markedly increased the severity of acute immune complex-driven arthritis in mice. TLR-2 deficiency resulted in a substantial increase in joint inflammation, bone erosion, and cartilage pathology, indicating a protective function of TLR-2 in passive FcγR-driven disease. Ex vivo study of the macrophage inflammatory phenotype revealed increased production of tumor necrosis factor α (TNFα) and interleukin-6 (IL-6) despite similar levels of IL-10, along with a significant increase in FcγR-specific response, in TLR-2-/- mouse macrophages early in the disease. Although distinct FcγR messenger RNA expression was not affected, cell surface protein expression of the inhibitory FcγRIIB in TLR-2-/- naive primary macrophages was specifically diminished, resulting in a higher proinflammatory response. Accordingly, TLR-2 stimulation specifically up-regulated FcγRIIB, but not the activating FcγR, on macrophages. CONCLUSION: TLR-2 regulates acute immune complex-driven arthritis by controlling macrophage FcγR response. Our findings indicate that the protective role of TLR-2 is extended beyond its previously described role in promoting Treg cells during T cell-mediated arthritis.


Subject(s)
Antigen-Antibody Complex/physiology , Arthritis, Experimental/physiopathology , Receptors, IgG/physiology , Signal Transduction/physiology , Toll-Like Receptor 2/physiology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cells, Cultured , Disease Models, Animal , Female , In Vitro Techniques , Interleukin-6/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Serum/immunology , Severity of Illness Index , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology , Tumor Necrosis Factor-alpha/metabolism
18.
J Immunol ; 189(7): 3767-76, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22942425

ABSTRACT

Salivary glands in patients with Sjögren's syndrome (SS) develop ectopic lymphoid structures (ELS) characterized by B/T cell compartmentalization, the formation of high endothelial venules, follicular dendritic cell networks, functional B cell activation with expression of activation-induced cytidine deaminase, as well as local differentiation of autoreactive plasma cells. The mechanisms that trigger ELS formation, autoimmunity, and exocrine dysfunction in SS are largely unknown. In this article, we present a novel model of inducible ectopic lymphoid tissue formation, breach of humoral self-tolerance, and salivary hypofunction after delivery of a replication-deficient adenovirus-5 in submandibular glands of C57BL/6 mice through retrograde excretory duct cannulation. In this model, inflammation rapidly and consistently evolves from diffuse infiltration toward the development of SS-like periductal lymphoid aggregates within 2 wk from AdV delivery. These infiltrates progressively acquire ELS features and support functional GL7(+)/activation-induced cytidine deaminase(+) germinal centers. Formation of ELS is preceded by ectopic expression of lymphoid chemokines CXCL13, CCL19, and lymphotoxin-ß, and is associated with development of anti-nuclear Abs in up to 75% of mice. Finally, reduction in salivary flow was observed over 3 wk post-AdV infection, consistent with exocrine gland dysfunction as a consequence of the inflammatory response. This novel model has the potential to unravel the cellular and molecular mechanisms that regulate ELS formation and their role in exocrine dysfunction and autoimmunity in SS.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Exocrine Glands/physiopathology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Sialadenitis/pathology , Animals , Autoimmune Diseases/physiopathology , Disease Models, Animal , Exocrine Glands/immunology , Exocrine Glands/pathology , Lymphoid Tissue/physiopathology , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Sialadenitis/immunology , Sialadenitis/physiopathology
19.
Proc Natl Acad Sci U S A ; 108(12): 4962-7, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21383200

ABSTRACT

Different splice variants of the proinflammatory cytokine IL-32 are found in various tissues; their putative differences in biological function remain unknown. In the present study, we report that IL-32γ is the most active isoform of the cytokine. Splicing to one less active IL-32ß appears to be a salvage mechanism to reduce inflammation. Adenoviral overexpression of IL-32γ (AdIL-32γ) resulted in exclusion of the IL-32γ-specific exon in vitro as well as in vivo, primarily leading to expression of IL-32ß mRNA and protein. Splicing of the IL-32γ-specific exon was prevented by single-nucleotide mutation, which blocked recognition of the splice site by the spliceosome. Overexpression of splice-resistant IL-32γ in THP1 cells or rheumatoid arthritis (RA) synovial fibroblasts resulted in a greater induction of proinflammatory cytokines such as IL-1ß, compared with IL-32ß. Intraarticular introduction of IL-32γ in mice resulted in joint inflammation and induction of several mediators associated with joint destruction. In RA synovial fibroblasts, overexpression of primarily IL-32ß showed minimal secretion and reduced cytokine production. In contrast, overexpression of splice-resistant IL-32γ in RA synovial fibroblasts exhibited marked secretion of IL-32γ. In RA, we observed increased IL-32γ expression compared with osteoarthritis synovial tissue. Furthermore, expression of TNFα and IL-6 correlated significantly with IL-32γ expression in RA, whereas this was not observed for IL-32ß. These data reveal that naturally occurring IL-32γ can be spliced into IL-32ß, which is a less potent proinflammatory mediator. Splicing of IL-32γ into IL-32ß is a safety switch in controlling the effects of IL-32γ and thereby reduces chronic inflammation.


Subject(s)
Alternative Splicing/immunology , Arthritis, Rheumatoid/immunology , Fibroblasts/immunology , Interleukins/immunology , Synovial Membrane/immunology , 3T3 Cells , Adenoviridae , Alternative Splicing/genetics , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , HeLa Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukins/genetics , Interleukins/metabolism , Mice , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , RNA Splice Sites/genetics , Synovial Membrane/metabolism , Synovial Membrane/pathology
20.
J Biol Chem ; 287(8): 5733-43, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22203669

ABSTRACT

IL-32 can be expressed in several isoforms. The amino acid sequences of the major IL-32 isoforms were used to predict the secondary and tertiary protein structure by I-TASSER software. The secondary protein structure revealed coils and α-helixes, but no ß sheets. Furthermore, IL-32 contains an RGD motif, which potentially activates procaspase-3 intracellular and or binds to integrins. Mutation of the RGD motif did not result in inhibition of the IL-32ß- or IL-32γ-induced cytotoxicity mediated through caspase-3. Although IL-32α interacted with the extracellular part of αVß3 and αVß6 integrins, only the αVß3 binding was inhibited by small RGD peptides. Additionally, IL-32ß was able to bind to αVß3 integrins, whereas this binding was not inhibited by small RGD peptides. In addition to the IL-32/integrin interactions, we observed that IL-32 is also able to interact with intracellular proteins that are involved in integrin and focal adhesion signaling. Modeling of IL-32 revealed a distinct α-helix protein resembling the focal adhesion targeting region of focal adhesion kinase (FAK). Inhibition of FAK resulted in modulation of the IL-32ß- or IL-32γ-induced cytotoxicity. Interestingly, IL-32α binds to paxillin without the RGD motif being involved. Finally, FAK inhibited IL-32α/paxillin binding, whereas FAK also could interact with IL-32α, demonstrating that IL-32 is a member of the focal adhesion protein complex. This study demonstrates for the first time that IL-32 binds to the extracellular domain of integrins and to intracellular proteins like paxillin and FAK, suggesting a dual role for IL-32 in integrin signaling.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Focal Adhesions/metabolism , Interleukins/chemistry , Interleukins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Antigens, Neoplasm/metabolism , Caspase 3/metabolism , Cell Death/drug effects , Computational Biology , Enzyme Activation/drug effects , Focal Adhesions/drug effects , HEK293 Cells , Humans , Integrin alphaVbeta3/metabolism , Integrins/metabolism , Interleukins/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Oligopeptides/pharmacology , Paxillin/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Signal Transduction/drug effects , Software
SELECTION OF CITATIONS
SEARCH DETAIL