Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Orthop Res ; 32(12): 1549-56, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224138

ABSTRACT

Activated Protein C (APC) is an anticoagulant with strong cytoprotective properties that has been shown to promote wound healing. In this study APC was investigated for its potential orthopedic application using a Bone Morphogenetic Protein 2 (rhBMP-2) induced ectopic bone formation model. Local co-administration of 10 µg rhBMP-2 with 10 µg or 25 µg APC increased bone volume at 3 weeks by 32% (N.S.) and 74% (p<0.01) compared to rhBMP-2 alone. This was associated with a significant increase in CD31+ and TRAP+ cells in tissue sections of ectopic bone, consistent with enhanced vascularity and bone turnover. The actions of APC are largely mediated by its receptors endothelial protein C receptor (EPCR) and protease-activated receptors (PARs). Cultured pre-osteoblasts and bone nodule tissue sections were shown to express PAR1/2 and EPCR. When pre-osteoblasts were treated with APC, cell viability and phosphorylation of ERK1/2, Akt, and p38 were increased. Inhibition with PAR1 and sometimes PAR2 antagonists, but not with EPCR blocking antibodies, ameliorated the effects of APC on cell viability and kinase phosphorylation. These data indicate that APC can affect osteoblast viability and signaling, and may have in vivo applications with rhBMP-2 for bone repair.


Subject(s)
Bone and Bones/drug effects , Protein C/pharmacology , Receptor, PAR-1/physiology , Receptor, PAR-2/physiology , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Resorption/prevention & control , Bone and Bones/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteogenesis/drug effects , Protein C/metabolism
2.
Rheumatology (Oxford) ; 53(12): 2270-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24982240

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the effect of endogenous matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9) on the invasive characteristics of RA synovial fibroblasts. METHODS: Synovial fibroblasts isolated from patients with RA or OA were treated with MMP small interfering RNA (siRNA), inhibitors and recombinant proteins or TNF-α, with or without cartilage explants. Cell viability and proliferation were measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide and 5-bromo-2-deoxyuridine (BrdU) proliferation assays, respectively; apoptosis by an in situ cell death detection kit; migration and invasion by CytoSelect invasion assay, scratch migration and collagen gel assays; cartilage degradation by 1,9-dimethylmethylene blue assay; and inflammatory mediators and MMPs by ELISA, western blot and zymography. RESULTS: MMP-2 was expressed by both OA and RA synovial fibroblasts, whereas only RA synovial fibroblasts expressed MMP-9. Suppressing MMP-2 or MMP-9 reduced RA synovial fibroblast proliferation equally. However, MMP-9 siRNA had greater effects compared with MMP-2 siRNA on promoting apoptosis and suppressing RA synovial fibroblast viability, migration and invasion. Suppression/inhibition of MMP-9 also decreased the production of IL-1ß, IL-6, IL-8 and TNF-α, inactivated nuclear factor κB (NF-κB), extracellular signal-regulated kinase (ERK) and c-Jun NH2-terminal kinase (JNK) and suppressed RA synovial fibroblast-mediated cartilage degradation. In contrast, suppression/inhibition of MMP-2 stimulated TNF-α and IL-17 secretion and activated NF-κB, while recombinant MMP-2 (rMMP-2) inactivated NF-κB and suppressed RA synovial fibroblast-mediated cartilage degradation. Results using specific inhibitors and rMMPs provided supportive evidence for the siRNA results. CONCLUSION: Endogenous MMP-2 or MMP-9 contribute to RA synovial fibroblast survival, proliferation, migration and invasion, with MMP-9 having more potent effects. Additionally, MMP-9 stimulates RA synovial fibroblast-mediated inflammation and degradation of cartilage, whereas MMP-2 inhibits these parameters. Overall, our data indicate that MMP-9 derived from RA synovial fibroblasts may directly contribute to joint destruction in RA.


Subject(s)
Arthritis, Rheumatoid/pathology , Cartilage, Articular/metabolism , Fibroblasts/physiology , Matrix Metalloproteinase 9/physiology , Synovial Membrane/pathology , Aged , Apoptosis/physiology , Arthritis, Rheumatoid/enzymology , Cell Movement/physiology , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Cytokines/biosynthesis , Female , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Inflammation Mediators/metabolism , Male , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinase 9/genetics , Middle Aged , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/pathology , RNA Interference , RNA, Small Interfering/genetics , Synovial Membrane/enzymology
3.
Arthritis Res Ther ; 16(1): R44, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24495480

ABSTRACT

INTRODUCTION: Rheumatoid synovial fibroblasts (RASFs) mediate joint inflammation and destruction in rheumatoid arthritis (RA). Endothelial protein C receptor (EPCR) is a specific receptor for the natural anticoagulant activated protein C (APC). It mediates the cytoprotective properties of APC and is expressed in rheumatoid synovial tissue. A recent report shows that group V secretory phospholipase A2 (sPLA2V) prevents APC from binding to EPCR in endothelium and inhibits EPCR/APC function. The aim of this study was to investigate the expression and function of EPCR on RASFs. METHODS: Human synovial fibroblasts (SFs) were isolated from RA or osteoarthritis (OA) synovial tissues and treated with control, EPCR, or sPLA2V small interfering RNA (siRNA); recombinant human APC, tumor necrosis factor-alpha (TNF-α), or sPLA2V. RASF viability and migration/invasion were measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) and collagen gel migration/invasion assays, respectively, and cartilage degradation by 1,9-dimethylmethylene blue (DMMB) assay in the presence of human OA articular cartilage explants. The expression or activation of cytokines, EPCR, cadherin-11, mitogen-activated protein (MAP) kinases, and nuclear factor-kappa-B (NF-κB) or both were detected by enzyme-linked immunosorbent assay, Western blotting, or immunostaining. RESULTS: EPCR was expressed by both OASFs and RASFs but was markedly increased in RASFs. When EPCR was suppressed by siRNA or blocking antibody cell viability, cell invasion and cartilage degradation were reduced by more than 30%. Inflammatory mediators interleukin-1-beta (IL-1ß), cadherin-11, and NF-κB were significantly reduced by EPCR suppression under control or TNF-α-stimulated conditions. The expression or activation (or both) of MAP kinases ERK, p38, and JNK were also markedly decreased in cells transfected with EPCR siRNA. Further analysis revealed that sPLA2V co-localized with EPCR on RASFs. Suppression of sPLA2V reduced cell viability and cartilage degradation and increased APC binding to RASFs. Conversely, recombinant sPLA2V increased cartilage degradation, blocked APC binding to RASFs, and could not rescue the effects induced by EPCR suppression. CONCLUSIONS: Our results demonstrate that EPCR is overexpressed by RASFs and mediates the aggressive behavior of RASFs. This function of EPCR is contrary to its cytoprotective role in other settings and is likely driven by sPLA2V.


Subject(s)
Antigens, CD/metabolism , Arthritis, Rheumatoid/pathology , Fibroblasts/pathology , Group V Phospholipases A2/metabolism , Receptors, Cell Surface/metabolism , Synovial Membrane/metabolism , Aged , Arthritis, Rheumatoid/metabolism , Blotting, Western , Cells, Cultured , Endothelial Protein C Receptor , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , RNA, Small Interfering , Transfection
4.
Mol Med ; 19: 324-31, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24096826

ABSTRACT

Synovial fibroblast proliferation is a hallmark of the invasive pannus in the rheumatoid joint. Activated protein C (APC) is a natural anticoagulant that exerts antiinflammatory and cyto-protective effects in various diseases via endothelial protein C receptor (EPCR) and proteinase-activated receptor (PAR)-mediated pathways. In this study, we investigated the effect and the underlying cellular signaling mechanisms of APC on proliferation of human rheumatoid synovial fibroblasts (RSFs). We found that APC stimulated proliferation of mouse dermal fibroblasts (MDFs) and normal human dermal fibroblasts (HDFs) by up to 60%, but robustly downregulated proliferation of RSFs. APC induced the phosphorylation of extracellular signal-regulated protein kinase (ERK) and enhanced expression of p21 and p27 in a dose-dependent manner in RSFs. The latter effect was inhibited by pre-treatment with the ERK inhibitors PD98059 and U0126 but not by p38 inhibitor SB203580. In addition, APC significantly downregulated tumor necrosis factor (TNF)α-stimulated cell proliferation and activation of p38, c-Jun NH2-terminal kinase (JNK) and Akt in RSFs. These results provide the first evidence that APC selectively inhibits proliferation and the inflammatory signaling pathways of RSFs. Thus, APC may reduce synovial hyperplasia and pannus invasion in rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/pathology , Dermis/cytology , Fibroblasts/physiology , MAP Kinase Signaling System , Protein C/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Aged , Animals , Cell Proliferation , Cells, Cultured , Female , Fibroblasts/cytology , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
5.
Arthritis Rheum ; 64(1): 88-98, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21905006

ABSTRACT

OBJECTIVE: To investigate whether protease-activated receptor 1 (PAR-1) and/or PAR-2 promotes the invasiveness/proliferation of synovial fibroblasts (SFs) and to determine the signaling mechanisms of these pathways. METHODS: SFs were isolated from the synovial tissue of patients with rheumatoid arthritis (RA), patients with osteoarthritis (OA), and PAR-1- or PAR-2-knockout (KO) mice. Expression of PAR-1 and PAR-2 was detected by immunofluorescence and Western blotting. The invasion and proliferation of SFs were measured by invasion assay and MTT assay, respectively. Matrix metalloproteinase 2 (MMP-2) and MMP-9 were detected by zymography, and cytokines were measured by enzyme-linked immunosorbent assay. RESULTS: PAR-1 and PAR-2 were colocalized with SFs in RA and OA synovium and, to a considerably lesser extent, in normal synovium. Inhibition of PAR-2 by small interfering RNA (siRNA) inhibited RASF invasion and proliferation, whereas blocking of PAR-1 by siRNA had the reverse effects. SFs from PAR-2-KO mice exhibited slower rates of proliferation and invasion. SFs from PAR-1-KO mice produced less MMP-2 and, in response to tumor necrosis factor α (TNFα) stimulation, had increased MMP-9 secretion when compared to SFs from wild-type and PAR-2-KO mice. Inhibition of PAR-1, but not PAR-2, stimulated the secretion of interleukin-17 (IL-17) and TNFα by RASFs. Furthermore, PAR-1 and PAR-2 had opposing effects on the activation of ERK, p38, and NF-κB. CONCLUSION: Activation of PAR-1 stimulates MMP-2 secretion, inhibits RASF growth and invasion, and decreases production of IL-17 and TNFα by RASFs, whereas activation of PAR-2 stimulates RASF growth and invasion and increases production of TNFα. Thus, although PAR-1 and PAR-2 are coexpressed by RASFs, PAR-2 alone appears to be responsible for the aggressive properties of RASFs and is likely to contribute to the pathologic progression of RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Fibroblasts/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Synovial Membrane/metabolism , Aged , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cell Movement , Cell Proliferation , Cell Survival , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Formazans/metabolism , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptor, PAR-1/deficiency , Receptor, PAR-1/genetics , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Synovial Membrane/drug effects , Synovial Membrane/pathology , Tetrazolium Salts/metabolism , Transfection , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...