Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Med ; 20(10): 1165-73, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25216639

ABSTRACT

Neurons have an important role in retinal vascular development. Here we show that the G protein-coupled receptor (GPCR) coagulation factor II receptor-like 1 (F2rl1, previously known as Par2) is abundant in retinal ganglion cells and is associated with new blood vessel formation during retinal development and in ischemic retinopathy. After stimulation, F2rl1 in retinal ganglion cells translocates from the plasma membrane to the cell nucleus using a microtubule-dependent shuttle that requires sorting nexin 11 (Snx11). At the nucleus, F2rl1 facilitates recruitment of the transcription factor Sp1 to trigger Vegfa expression and, in turn, neovascularization. In contrast, classical plasma membrane activation of F2rl1 leads to the expression of distinct genes, including Ang1, that are involved in vessel maturation. Mutant versions of F2rl1 that prevent nuclear relocalization but not plasma membrane activation interfere with Vegfa but not Ang1 expression. Complementary angiogenic factors are therefore regulated by the subcellular localization of a receptor (F2rl1) that governs angiogenesis. These findings may have implications for the selectivity of drug actions based on the subcellular distribution of their targets.


Subject(s)
Neovascularization, Physiologic , Neurons/metabolism , Receptor, PAR-2/metabolism , Active Transport, Cell Nucleus , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubules/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neovascularization, Pathologic , Neovascularization, Physiologic/genetics , Promoter Regions, Genetic , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Retinal Ganglion Cells/metabolism , Retinal Vessels/growth & development , Retinal Vessels/metabolism , Sorting Nexins/metabolism , Sp1 Transcription Factor/metabolism , Subcellular Fractions/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
J Rheumatol ; 38(5): 911-20, 2011 May.
Article in English | MEDLINE | ID: mdl-21285164

ABSTRACT

OBJECTIVE: Evidence indicates that proteinase-activated receptor (PAR)-2 participates in the degradative processes of human osteoarthritis (OA). We evaluated the in vivo effect of PAR-2 on articular lesions in a PAR-2-knockout (KO) mouse model of OA. METHODS: OA was surgically induced by destabilization of the medial meniscus of the right knee in C57Bl/6 wild-type (WT) and PAR-2 KO mice. Knee swelling was measured throughout the duration of the study (8 weeks postsurgery) and histologic evaluation of cartilage was done to assess structure, cellularity, matrix staining, and remodeling in the deep zone. Morphometric analysis of subchondral bone was also performed. RESULTS: Data showed significant knee swelling in the operated WT mice immediately following surgery, which increased with time (8 weeks post-surgery). Knee swelling was significantly lower (p ≤ 0.0001) in PAR-2 KO mice than in WT mice at both 4 and 8 weeks postsurgery. Cartilage damage was found in both operated WT and PAR-2 KO mice; however, lesions were significantly less severe (global score; p ≤ 0.05) in the PAR-2 KO mice at 4 weeks postsurgery. Operated WT mice showed reduced subchondral bone surface and trabecular thickness with significance reached at 4 weeks (p ≤ 0.03 and p ≤ 0.05, respectively), while PAR-2 KO mice demonstrated a gradual increase in subchondral bone surface with significance reached at 8 weeks (p ≤ 0.007). CONCLUSION: We demonstrated the in vivo implication of PAR-2 in the development of experimental OA, thus confirming its involvement in OA joint structural changes and reinforcing the therapeutic potential of a PAR-2 antagonist for treatment of OA.


Subject(s)
Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , Knee Joint/metabolism , Osteoarthritis, Knee/metabolism , Receptor, PAR-2/physiology , Animals , Cartilage, Articular/physiopathology , Female , Gene Targeting , Inflammation/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis, Knee/drug therapy
3.
J Rheumatol ; 38(1): 118-28, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20952474

ABSTRACT

OBJECTIVE: to investigate over a 1-year period in dogs that underwent extracapsular stabilization surgery (ECS) following anterior cruciate ligament (ACL) transection: whether reconstructive surgery could prevent osteoarthritis (OA) progression and whether treatment with the bisphosphonate tiludronic acid (TA) could improve the chronic evolution of OA structural changes. METHODS: ACL transection was performed on dogs on Day 0 and ECS on Day 28. Dogs were randomly divided into 2 groups: 15 received placebo and 16 were treated with TA (2 mg/kg subcutaneous injection) on Days 14, 28, 56, and 84. Magnetic resonance images were acquired on Days -10, 26, 91, 210, and 357, and cartilage volume was quantified. At sacrifice (Day 364), cartilage from femoral condyles and tibial plateaus was macroscopically and histologically evaluated. Expression levels of MMP-1, -3, -13, ADAMTS-4, -5, BMP-2, FGF-2, IGF-1, TGF-ß1, collagen type II, and aggrecan were determined using real-time RT-PCR. RESULTS: the loss of cartilage volume observed after ACL transection stabilized following ECS. Thereafter, a gradual gain occurred, with the cartilage volume loss on the tibial plateaus reduced at Day 91 (p < 0.02) and Day 210 (p < 0.001) in the TA-treated dogs. At sacrifice, TA-treated dogs presented a reduction in the severity of macroscopic (p = 0.03 for plateaus) and histologic (p = 0.07 for plateaus) cartilage lesions, had a better preserved collagen network, and showed decreased MMP-13 (p = 0.04), MMP-1 and MMP-3 levels. CONCLUSION: our findings indicate that in dogs with ACL transection, ECS greatly prevents development of cartilage volume loss. Treatment with TA provided an additional benefit of reducing the development of OA lesions.


Subject(s)
Anterior Cruciate Ligament/surgery , Bone Density Conservation Agents/therapeutic use , Cartilage, Articular/pathology , Diphosphonates/therapeutic use , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Animals , Anterior Cruciate Ligament/pathology , Dogs , Humans , Magnetic Resonance Imaging/methods
4.
Ann Rheum Dis ; 69(6): 1179-84, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19546097

ABSTRACT

OBJECTIVE: The aims of this study were to evaluate the effect of oral treatment with a whole plant extract of Brachystemma calycinum D don (BCD) on the development of osteoarthritic lesions and symptoms in the experimental dog anterior cruciate ligament (ACL) transection model and to document its mechanism of action. METHODS: Osteoarthritis was induced by sectioning the ACL of the right knee in crossbred dogs. There were two experimental groups (n=6-7 dogs/group): placebo and BCD extract (200 mg/kg per day) given orally for 8 weeks. Macroscopic and histopathological evaluation of cartilage lesions and immunohistochemical analysis of cartilage to assess levels of inducible nitric oxide synthase (iNOS), matrix metalloprotease 13 (MMP-13) and protease activated receptor 2 (PAR-2) were done. A gait analysis of dogs was performed. RESULTS: Treatment with BCD reduced the severity (depth) (p=0.04) and histopathological score (p<0.02) of osteoarthritis cartilage lesions. BCD treatment also significantly reduced the osteoarthritis chondrocyte level of key inflammatory and catabolic factors (iNOS, p=0.009 and MMP-13, p=0.003) as well as the level of PAR-2 (p=0.03). Dogs treated with BCD showed a significant improvement in peak vertical force measured at 8 weeks (p<0.05). CONCLUSIONS: Treatment with BCD extract exerts a positive effect on the prevention of cartilage lesions induced by joint instability, and improves joint function. This effect was associated with the inhibition of major catabolic and inflammatory mediators. This study is the first to demonstrate that a therapeutic intervention that can inhibit PAR-2 is associated with a disease-modifying osteoarthritis effect.


Subject(s)
Arthritis, Experimental/prevention & control , Caryophyllaceae , Osteoarthritis/prevention & control , Phytotherapy/methods , Receptor, PAR-2/antagonists & inhibitors , Administration, Oral , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Dogs , Drug Evaluation, Preclinical/methods , Gait , Osteoarthritis/metabolism , Osteoarthritis/pathology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Receptor, PAR-2/metabolism
5.
Rheumatology (Oxford) ; 48(12): 1482-90, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19762475

ABSTRACT

OBJECTIVES: Earlier studies suggest the involvement of osteoprotegerin (OPG), RANK and RANK ligand (RANKL) in OA subchondral bone metabolism; however, few studies have looked at their functional consequences on chondrocytes. We compared the expression/production of OPG, RANK and RANKL on human normal and OA chondrocytes, and evaluated, on OA chondrocytes, their modulation by some catabolic factors. Furthermore, the role of OPG and RANKL on the production of catabolic/anabolic factors was assessed. METHODS: Expression was determined using real-time PCR, production of RANK and RANKL by flow cytometry and that of OPG by ELISA. Modulation of these factors was determined upon treatment with IL-1beta, TNF-alpha and PGE(2). The functional consequences were examined following treatment with soluble RANKL or OPG-Fc (OPG without the heparin-binding domain). RESULTS: OPG, RANK and RANKL were expressed and produced by human chondrocytes. Membranous RANK was produced only by an OA chondrocyte subpopulation (29%) localized throughout the cartilage. The OPG/RANKL ratio was significantly (P = 0.05) reduced on the OA chondrocytes, whereas the RANK/RANKL ratio was significantly (P < 0.03) increased. OPG and membranous RANKL levels were significantly enhanced by IL-1beta, TNF-alpha and PGE(2), whereas membranous RANK was significantly increased only with IL-1beta. Administration of soluble RANKL had no effect on the OA chondrocytes. However, addition of OPG-Fc significantly stimulated MMP-13 (P = 0.05) and protease-activated receptor-2 (PAR-2) (P < 0.04) production. CONCLUSIONS: Our findings showed that human chondrocytes express and produce OPG, RANK and RANKL. OA chondrocyte treatment with catabolic factors pointed towards an increased biological effect of OPG. Interestingly, OPG appears to be involved in OA progression by increasing two catabolic factors involved in cartilage pathophysiology.


Subject(s)
Chondrocytes/physiology , Osteoarthritis, Knee/metabolism , Osteoprotegerin/biosynthesis , Receptor Activator of Nuclear Factor-kappa B/biosynthesis , Adult , Aged , Cells, Cultured , Chondrocytes/drug effects , Dinoprostone/pharmacology , Humans , Interleukin-1beta/pharmacology , Middle Aged , Osteoprotegerin/physiology , Polymerase Chain Reaction/methods , RANK Ligand/biosynthesis , RANK Ligand/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
6.
Arthritis Res Ther ; 11(4): R119, 2009.
Article in English | MEDLINE | ID: mdl-19664212

ABSTRACT

INTRODUCTION: Members of the ephrin system, the ephrin receptor erythropoietin-producing hepatocellular B4 (EphB4) and its specific ligand, ephrin B2, appear to be involved in the bone remodelling process. We recently showed that their interaction inhibits the resorptive activity of human osteoarthritic (OA) subchondral bone osteoblasts. Hence, we further investigated the possible implication of these ephrin members on the catabolic/anabolic activities of human OA chondrocytes. METHODS: EphB4 receptor and ephrin B2 levels were determined by quantitative PCR and immunohistochemistry, and the effects of ephrin B2 on the expression/production of factors involved in the OA process. RESULTS: EphB4 receptors and ephrin B2 ligands are expressed and produced by human normal and OA chondrocytes. Ephrin B2 protein was found at similar levels in both cartilage types, whereas EphB4 receptor expression (P < 0.0001) and production (P < 0.01) levels were significantly increased in OA chondrocytes/cartilage. Ephrin B2 treatment significantly inhibited the interleukin (IL)-1beta, IL-6, matrix metalloproteinase-1 (MMP-1), MMP-9, MMP-13, and proteinase-activated receptor-2 (PAR-2) gene expression levels, whereas MMP-2 was unaffected, and significantly increased collagen type II, a cartilage specific macromolecule. It also inhibited the IL-1beta stimulated protein production of IL-6, MMP-1 and MMP-13. CONCLUSIONS: Our study is the first to provide data on the presence and role of ephrin B2/EphB4 receptors in human chondrocytes/cartilage. Data showed that ephrin B2 treatment positively impacts the abnormal metabolism of OA cartilage by inhibiting important catabolic factors involved in this disease at the same time as increasing anabolic activity.


Subject(s)
Chondrocytes/metabolism , Ephrin-B2/metabolism , Osteoarthritis/metabolism , Receptor, EphB4/metabolism , Aged , Cartilage, Articular/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression , Humans , Immunohistochemistry , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 13/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
7.
Arthritis Res Ther ; 11(2): R41, 2009.
Article in English | MEDLINE | ID: mdl-19291317

ABSTRACT

INTRODUCTION: The aims of this study were, first, to investigate the in vivo effects of treatment with avocado/soybean unsaponifiables on the development of osteoarthritic structural changes in the anterior cruciate ligament dog model and, second, to explore their mode of action. METHODS: Osteoarthritis was induced by anterior cruciate ligament transection of the right knee in crossbred dogs. There were two treatment groups (n = 8 dogs/group), in which the animals received either placebo or avocado/soybean unsaponifiables (10 mg/kg per day), which were given orally for the entire duration of the study (8 weeks). We conducted macroscopic and histomorphological analyses of cartilage and subchondral bone of the femoral condyles and/or tibial plateaus. We also conducted immunohistochemical analyses in cartilage for the following antigens: inducible nitric oxide synthase, matrix metalloproteinase (MMP)-1, MMP-13, a disintegrin and metalloproteinase domain with thrombospondin motifs (ADAMTS)4 and ADAMTS5. RESULTS: The size of macroscopic lesions on the tibial plateaus was decreased (P = 0.04) in dogs treated with the avocado/soybean unsaponifiables. Histologically, in these animals the severity of cartilage lesions on both tibial plateaus and femoral condyles, and the cellular infiltration in synovium were significantly decreased (P = 0.0002 and P = 0.04, respectively). Treatment with avocado/soybean unsaponifiables also reduced loss of subchondral bone volume (P < 0.05) and calcified cartilage thickness (P = 0.01) compared with placebo. Immunohistochemical analysis of cartilage revealed that avocado/soybean unsaponifiables significantly reduced the level of inducible nitric oxide synthase (P < 0.05) and MMP-13 (P = 0.01) in cartilage. CONCLUSIONS: This study demonstrates that treatment with avocado/soybean unsaponifiables can reduce the development of early osteoarthritic cartilage and subchondral bone lesions in the anterior cruciate ligament dog model of osteoarthritis. This effect appears to be mediated through the inhibition of inducible nitric oxide synthase and MMP-13, which are key mediators of the structural changes that take place in osteoarthritis.


Subject(s)
Glycine max/chemistry , Matrix Metalloproteinase 13/drug effects , Nitric Oxide Synthase/drug effects , Osteoarthritis/drug therapy , Persea/chemistry , Phytosterols/therapeutic use , Plant Extracts/therapeutic use , Vitamin E/therapeutic use , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Bone and Bones/drug effects , Bone and Bones/pathology , Cartilage/drug effects , Cartilage/pathology , Dogs , Drug Combinations , Immunohistochemistry , Matrix Metalloproteinase 13/metabolism , Nitric Oxide Synthase/metabolism , Osteoarthritis/pathology
8.
Bone ; 44(6): 1143-50, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19264156

ABSTRACT

INTRODUCTION: In osteoarthritis (OA), the subchondral bone undergoes a remodelling process involving several factors synthesized by osteoblasts. In this study, we investigated the expression, production, modulation, and role of PAR-2 in human OA subchondral bone osteoblasts. MATERIALS AND METHODS: PAR-2 expression and production were determined by real-time PCR and flow cytometry, respectively. PAR-2 modulation was investigated in OA subchondral bone osteoblasts treated with IL-1 beta (100 pg/ml), TNF-alpha (5 ng/ml), TGF-beta1 (10 ng/ml), PGE(2) (500 nM), IL-6 (10 ng/ml) and IL-17 (10 ng/ml). Membranous RANKL protein was assessed by flow cytometry, and OPG, MMP-1, MMP-9, MMP-13, IL-6 and intracellular signalling pathways by specific ELISAs. Bone resorptive activity was measured by using a co-culture model of human PBMC and OA subchondral bone osteoblasts. RESULTS: PAR-2 expression and production (p<0.05) were markedly increased when human OA subchondral bone osteoblasts were compared to normal. On OA osteoblasts, PAR-2 production was significantly increased by IL-1 beta, TNF-alpha and PGE(2). Activation of PAR-2 with a specific agonist, SLIGKV-NH(2), induced a significant up-regulation of MMP-1, MMP-9, IL-6, and membranous RANKL, but had no effect on MMP-13 or OPG production. Interestingly, bone resorptive activity was also significantly enhanced following PAR-2 activation. The PAR-2 effect was mediated by activation of the MAP kinases Erk1/2 and JNK. CONCLUSION: This study is the first to demonstrate that PAR-2 activation plays a role in OA subchondral bone resorption via an up-regulation of major bone remodelling factors. These results shed new light on the potential of PAR-2 as a therapeutic target in OA.


Subject(s)
Bone Resorption/metabolism , Osteoarthritis/metabolism , Osteoblasts/metabolism , Receptor, PAR-2/metabolism , Aged , Aged, 80 and over , Bone Resorption/pathology , Cells, Cultured , Dinoprostone/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interleukin-17/pharmacology , Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/metabolism , Middle Aged , Osteoarthritis/pathology , Osteoblasts/drug effects , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor, PAR-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
9.
Arthritis Rheum ; 58(12): 3820-30, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19035475

ABSTRACT

OBJECTIVE: Abnormal subchondral bone metabolism is involved in osteoarthritis (OA). It has been suggested that ephrin B2 and its specific receptor EphB4 participate in bone homeostasis. We previously reported that human OA subchondral bone osteoblasts could be classified into 2 subpopulations: low (L), having proresorption properties, and high (H), having proformation properties. The purpose of this study was to investigate the importance of the ephrin system in OA subchondral bone osteoblasts. METHODS: The presence of the EphB4 receptor was determined by immunohistochemistry, and its expression level, modulation upon treatment, and consequences of activation by ephrin B2 were determined by quantitative polymerase chain reaction. The effects of ephrin B2 activation of the EphB4 receptor on bone resorption activity were also determined. EphB4 receptor activation signaling pathways were investigated by specific enzyme-linked immunosorbent assay. RESULTS: EphB4 receptors were present in subchondral bone osteoblasts and osteocytes. Compared with normal and H-OA osteoblasts, EphB4 receptor expression levels were significantly increased in L-OA osteoblasts, with no difference between normal and H-OA osteoblasts. EphB4 receptor levels in L-OA osteoblasts were significantly up-regulated by prostaglandin E2 (PGE2) and interleukin-17 (IL-17). Ephrin B2, PGE2, and IL-17 significantly inhibited bone resorption activity in these cells. EphB4 activation by ephrin B2 significantly inhibited the expression of IL-1beta, IL-6, matrix metalloproteinase 1 (MMP-1), MMP-9, MMP-13, and RANKL, but not MMP-2 and osteoprotegerin. EphB4 receptor activation significantly inhibited the phosphatidylinositol 3-kinase/Akt pathway. CONCLUSION: This study is the first to provide evidence that EphB4 receptor activation by ephrin B2 in OA subchondral bone could affect abnormal metabolism in this tissue by inhibiting resorption factors and their activities. Ephrin B2 could be targeted as a specific therapeutic approach in the development of a disease-modifying OA drug.


Subject(s)
Ephrin-B2/metabolism , Osteoarthritis, Knee/physiopathology , Osteoblasts/physiology , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Aged , Aged, 80 and over , Cells, Cultured , Gene Expression/physiology , Humans , Ligands , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoblasts/cytology , Osteocytes/cytology , Osteocytes/physiology , Signal Transduction/physiology
10.
Arthritis Res Ther ; 10(3): R71, 2008.
Article in English | MEDLINE | ID: mdl-18578867

ABSTRACT

INTRODUCTION: Subchondral bone alterations represent an essential component of osteoarthritis (OA). Modifying the abnormal subchondral bone metabolism may be indicated to treat OA. We investigated the effect of diacerein and rhein on the changes occurring in subchondral bone during OA. To this end, we determined the drugs' effects on metalloprotease-13 (MMP-13) synthesis on subchondral bone and on the osteoblast signalling pathways. In osteoclasts, we studied MMP-13 and cathepsin K production as well as cell differentiation, proliferation, and survival. METHODS: The effect of diacerein/rhein on the production of subchondral bone MMP-13 was determined by enzyme-linked immunosorbent assay. Signalling pathways were evaluated on osteoblasts by Western blot. Osteoclast experiments were performed using cells from the pre-osteoclastic murine cell line Raw 264.7. Osteoclast MMP-13 and cathepsin K activities were determined by specific bioassays and differentiation of these cells quantified by tartrate-resistant acid phosphatase staining. RESULTS: Diacerein and rhein reduced, in a dose-dependent manner, the interleukin-1-beta (IL-1beta)-induced MMP-13 production in OA subchondral bone. This effect occurred through the inhibition of ERK1/2 (extracellular signal-regulated kinase-1/2) and p38. In osteoclasts, they significantly reduced the activity of MMP-13 and cathepsin K. Moreover, these drugs effectively blocked the IL-1beta effect on the osteoclast differentiation process and the survival of mature osteoclasts. CONCLUSION: Altogether, these data suggest that diacerein/rhein could impact the abnormal subchondral bone metabolism in OA by reducing the synthesis of resorptive factors and osteoclast formation.


Subject(s)
Anthraquinones/therapeutic use , Bone Remodeling/physiology , Cartilage, Articular/enzymology , Cell Differentiation/physiology , Enzyme Inhibitors/pharmacology , Osteoarthritis/enzymology , Osteoclasts/enzymology , Aged , Aged, 80 and over , Animals , Anthraquinones/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bone Remodeling/drug effects , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Osteoarthritis/pathology , Osteoarthritis/prevention & control , Osteoclasts/cytology , Osteoclasts/drug effects
11.
Best Pract Res Clin Rheumatol ; 22(2): 351-84, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18455690

ABSTRACT

The preservation of articular cartilage depends on keeping the cartilage architecture intact. Cartilage strength and function depend on both the properties of the tissue and on their structural parameters. The main structural macromolecules are collagen and proteoglycans (aggrecan). During life, cartilage matrix turnover is mediated by a multitude of complex autocrine and paracrine anabolic and catabolic factors. These act on the chondrocytes and can lead to repair, remodeling or catabolic processes like those that occur in osteoarthritis. Osteoarthritis is characterized by degradation and loss of articular cartilage, subchondral bone remodeling, and, at the clinical stage of the disease, inflammation of the synovial membrane. The alterations in osteoarthritic cartilage are numerous and involve morphologic and metabolic changes in chondrocytes, as well as biochemical and structural alterations in the extracellular matrix macromolecules.


Subject(s)
Cartilage/metabolism , Osteoarthritis/metabolism , Bone Remodeling/physiology , Cartilage/anatomy & histology , Cartilage/chemistry , Humans , Joints/metabolism , Joints/pathology , Joints/physiopathology , Osteoarthritis/pathology , Osteoarthritis/physiopathology , Synovial Membrane/physiopathology
12.
Arthritis Res Ther ; 9(6): R121, 2007.
Article in English | MEDLINE | ID: mdl-18031579

ABSTRACT

Proteinase-activated receptors (PARs) belong to a family of G protein-coupled receptors. PARs are activated by a serine-dependent cleavage generating a tethered activating ligand. PAR-2 was shown to be involved in inflammatory pathways. We investigated the in situ levels and modulation of PAR-2 in human normal and osteoarthritis (OA) cartilage/chondrocytes. Furthermore, we evaluated the role of PAR-2 on the synthesis of the major catabolic factors in OA cartilage, including metalloproteinase (MMP)-1 and MMP-13 and the inflammatory mediator cyclooxygenase 2 (COX-2), as well as the PAR-2-activated signalling pathways in OA chondrocytes. PAR-2 expression was determined using real-time reverse transcription-polymerase chain reaction and protein levels by immunohistochemistry in normal and OA cartilage. Protein modulation was investigated in OA cartilage explants treated with a specific PAR-2-activating peptide (PAR-2-AP), SLIGKV-NH2 (1 to 400 microM), interleukin 1 beta (IL-1beta) (100 pg/mL), tumor necrosis factor-alpha (TNF-alpha) (5 ng/mL), transforming growth factor-beta-1 (TGF-beta1) (10 ng/mL), or the signalling pathway inhibitors of p38 (SB202190), MEK1/2 (mitogen-activated protein kinase kinase) (PD98059), and nuclear factor-kappa B (NF-kappaB) (SN50), and PAR-2 levels were determined by immunohistochemistry. Signalling pathways were analyzed on OA chondrocytes by Western blot using specific phospho-antibodies against extracellular signal-regulated kinase 1/2 (Erk1/2), p38, JNK (c-jun N-terminal kinase), and NF-kappaB in the presence or absence of the PAR-2-AP and/or IL-1beta. PAR-2-induced MMP and COX-2 levels in cartilage were determined by immunohistochemistry. PAR-2 is produced by human chondrocytes and is significantly upregulated in OA compared with normal chondrocytes (p < 0.04 and p < 0.03, respectively). The receptor levels were significantly upregulated by IL-1beta (p < 0.006) and TNF-alpha (p < 0.002) as well as by the PAR-2-AP at 10, 100, and 400 microM (p < 0.02) and were downregulated by the inhibition of p38. After 48 hours of incubation, PAR-2 activation significantly induced MMP-1 and COX-2 starting at 10 microM (both p < 0.005) and MMP-13 at 100 microM (p < 0.02) as well as the phosphorylation of Erk1/2 and p38 within 5 minutes of incubation (p < 0.03). Though not statistically significant, IL-1beta produced an additional effect on the activation of Erk1/2 and p38. This study documents, for the first time, functional consequences of PAR-2 activation in human OA cartilage, identifies p38 as the major signalling pathway regulating its synthesis, and demonstrates that specific PAR-2 activation induces Erk1/2 and p38 in OA chondrocytes. These results suggest PAR-2 as a potential new therapeutic target for the treatment of OA.


Subject(s)
Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Receptor, PAR-2/metabolism , Adult , Aged , Aged, 80 and over , Base Sequence , Cartilage, Articular/pathology , Case-Control Studies , Chondrocytes/metabolism , DNA Primers/genetics , Humans , Inflammation Mediators/metabolism , Middle Aged , Osteoarthritis/genetics , Osteoarthritis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, PAR-2/genetics , Signal Transduction , Tissue Culture Techniques , Up-Regulation
13.
Arthritis Rheum ; 56(7): 2288-98, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17599749

ABSTRACT

OBJECTIVE: Emerging evidence indicates that peroxisome proliferator-activated receptor gamma (PPARgamma) may have protective effects in osteoarthritis (OA). The aim of this study was to evaluate the in vivo effect of a PPARgamma agonist, pioglitazone, on the development of lesions in a canine model of OA, and to explore the influence of pioglitazone on the major signaling and metabolic pathways involved in OA pathophysiologic changes. METHODS: OA was surgically induced in dogs by sectioning of the anterior cruciate ligament. The dogs were then randomly divided into 3 treatment groups in which they were administered either placebo, 15 mg/day pioglitazone, or 30 mg/day pioglitazone orally for 8 weeks. Following treatment, the severity of cartilage lesions was scored. Cartilage specimens were processed for histologic and immunohistochemical evaluations; specific antibodies were used to study the levels of matrix metalloproteinase 1 (MMP-1), ADAMTS-5, and inducible nitric oxide synthase (iNOS), as well as phosphorylated MAPKs ERK-1/2, p38, JNK, and NF-kappaB p65. RESULTS: Pioglitazone reduced the development of cartilage lesions in a dose-dependent manner, with the highest dosage producing a statistically significant change (P < 0.05). This decrease in lesions correlated with lower cartilage histologic scores. In addition, pioglitazone significantly reduced the synthesis of the key OA mediators MMP-1, ADAMTS-5, and iNOS and, at the same time, inhibited the activation of the signaling pathways for MAPKs ERK-1/2, p38, and NF-kappaB. CONCLUSION: These results indicate the efficacy of pioglitazone in reducing cartilage lesions in vivo. The results also provide new and interesting insights into a therapeutic intervention for OA in which PPARgamma activation can inhibit major signaling pathways of inflammation and reduce the synthesis of cartilage catabolic factors responsible for articular cartilage degradation.


Subject(s)
Cartilage/pathology , Osteoarthritis/pathology , PPAR gamma/agonists , Thiazolidinediones/therapeutic use , Animals , Cartilage/drug effects , Disease Models, Animal , Dogs , Femur , Hypoglycemic Agents/therapeutic use , Osteoarthritis/drug therapy , Pioglitazone , Tibia
14.
Arthritis Res Ther ; 9(1): R20, 2007.
Article in English | MEDLINE | ID: mdl-17326835

ABSTRACT

In this study we examine the extracellular role of galectin-3 (gal-3) in joint tissues. Following intra-articular injection of gal-3 or vehicle in knee joints of mice, histological evaluation of articular cartilage and subchondral bone was performed. Further studies were then performed using human osteoarthritic (OA) chondrocytes and subchondral bone osteoblasts, in which the effect of gal-3 (0 to 10 microg/ml) was analyzed. Osteoblasts were incubated in the presence of vitamin D3 (50 nM), which is an inducer of osteocalcin, encoded by an osteoblast terminal differentiation gene. Genes of interest mainly expressed in either chondrocytes or osteoblasts were analyzed with real-time RT-PCR and enzyme immunoassays. Signalling pathways regulating osteocalcin were analyzed in the presence of gal-3. Intra-articular injection of gal-3 induced knee swelling and lesions in both cartilage and subchondral bone. On human OA chondrocytes, gal-3 at 1 microg/ml stimulated ADAMTS-5 expression in chondrocytes and, at higher concentrations (5 and 10 microg/ml), matrix metalloproteinase-3 expression. Experiments performed with osteoblasts showed a weak but bipolar effect on alkaline phosphatase expression: stimulation at 1 microg/ml or inhibition at 10 microg/ml. In the absence of vitamin D3, type I collagen alpha 1 chain expression was inhibited by 10 microg/ml of gal-3. The vitamin D3 induced osteocalcin was strongly inhibited in a dose-dependent manner in the presence of gal-3, at both the mRNA and protein levels. This inhibition was mainly mediated by phosphatidylinositol-3-kinase. These findings indicate that high levels of extracellular gal-3, which could be encountered locally during the inflammatory process, have deleterious effects in both cartilage and subchondral bone tissues.


Subject(s)
Extracellular Fluid/chemistry , Extracellular Fluid/metabolism , Galectin 3/metabolism , Joints/chemistry , Joints/metabolism , Aged , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Female , Galectin 3/administration & dosage , Galectin 3/physiology , Humans , Inflammation/metabolism , Joints/pathology , Knee Joint/metabolism , Knee Joint/pathology , Male , Mice , Middle Aged , Osteoblasts/metabolism , Osteoblasts/pathology
15.
Arthritis Rheum ; 54(8): 2471-80, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16868967

ABSTRACT

OBJECTIVE: Matrix metalloprotease 13 (MMP-13) plays a major role in osteoarthritic (OA) processes. We previously identified the AG-rich element (AGRE) regulatory site (GAAAAGAAAAAG) in the proximal promoter of this gene. Electrophoretic mobility shift assays (EMSAs) done with nuclear extracts from OA chondrocytes showed the presence of 2 AGRE protein-binding complexes, the formation of which depended on the pathophysiologic state (high or low) of the cells; the low OA (L-OA) chondrocytes have low MMP-13 basal levels and high interleukin-1beta (IL-1beta) inducibility, and the high OA (H-OA) chondrocytes have high MMP-13 basal levels and low IL-1beta inducibility. In this study, we sought to determine the importance of individual AGRE bases in promoter activity and to identify AGRE binding proteins from L-OA and H-OA chondrocyte complexes. METHODS: Promoter activity was determined following transient transfection into human OA chondrocytes. AGRE binding proteins were identified by mass spectroscopy. RESULTS: Individual mutations of the AGRE site differentially modulated promoter activity, indicating that the intact AGRE site is required for optimal MMP-13 expression. Damage-specific DNA binding protein 1 (DDB-1) was identified in the L-OA chondrocyte-binding complex. EMSA experiments performed with the mutation of the left AGRE site (GTGCTGAAAAAG) and nuclear extracts of L-OA chondrocytes reproduced the pattern seen in the H-OA chondrocytes. Mass spectroscopy identified p130cas as one of the proteins in this complex. Supershift experiments showed the presence of p130cas and nuclear matrix transcription factor 4 (NMP-4) in the wild-type AGRE/H-OA chondrocyte complex. CONCLUSION: These data suggest that the binding of p130(cas) and NMP-4 to the AGRE site regulates MMP-13 expression and may trigger the change in human chondrocytes from the L-OA state to the H-OA state.


Subject(s)
Cartilage, Articular/enzymology , Chondrocytes/enzymology , Collagenases/metabolism , DNA-Binding Proteins/metabolism , Osteoarthritis, Knee/metabolism , Promoter Regions, Genetic , Base Sequence , Cartilage, Articular/chemistry , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/pathology , Collagenases/analysis , DNA-Binding Proteins/analysis , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Matrix Metalloproteinase 13 , Molecular Sequence Data , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/surgery , Promoter Regions, Genetic/physiology , Synovial Membrane/chemistry , Synovial Membrane/enzymology , Synovial Membrane/pathology , Trans-Activators/analysis , Trans-Activators/metabolism
16.
J Rheumatol ; 33(6): 1176-83, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16652435

ABSTRACT

OBJECTIVE: We investigated the effectiveness of licofelone, a combined 5-lipoxygenase and cyclooxygenase inhibitor, on structural changes in the anterior cruciate ligament (ACL) experimental dog model of osteoarthritis (OA) under therapeutic conditions. The effect of drug treatment on the expression and activity of metalloproteases in the OA cartilage was also studied. METHODS: The cranial cruciate ligament of the right stifle joint was surgically sectioned in 14 dogs to create OA lesions. Of these dogs, 7 received placebo treatment and served as OA controls, while 7 were treated with licofelone 2.5 mg/kg twice daily for an 8-week period, starting 4 weeks after surgery. At necropsy, macroscopic evaluations were made of the size of osteophytes and the severity of cartilage lesions on femoral condyles and tibial plateaus. Collagenase and other metalloprotease activity levels in cartilage were measured. Levels of gene expression of matrix metalloprotease (MMP-1), MMP-13, cathepsin K, and ADAMTS-5 were quantified by RT-PCR. RESULTS: Licofelone treatment reduced the development of osteophytes and size of cartilage lesions on the femoral condyles and on the tibial plateaus (p < 0.04). Drug treatment also significantly decreased collagenase (p < 0.02) and metalloprotease (p < 0.04) activities, as well as the levels of gene expression of MMP-1 (p < 0.01), MMP-13 (p < 0.05), cathepsin K, and ADAMTS-5 (p = 0.01). CONCLUSION: Under therapeutic conditions licofelone showed the ability to reduce the progression of structural changes in experimental dog OA. This beneficial effect is likely mediated through decrease in the synthesis of a number of catabolic factors, including proteolytic enzymes, involved in cartilage breakdown.


Subject(s)
Acetates/therapeutic use , Enzyme Inhibitors/therapeutic use , Metalloproteases/metabolism , Osteoarthritis, Knee/drug therapy , Pyrroles/therapeutic use , Animals , Anterior Cruciate Ligament/surgery , Cartilage, Articular/drug effects , Cartilage, Articular/enzymology , Cartilage, Articular/pathology , Collagenases/metabolism , Disease Models, Animal , Disease Progression , Dogs , Metalloproteases/genetics , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Stifle/surgery
17.
Arthritis Res Ther ; 7(5): R1091-102, 2005.
Article in English | MEDLINE | ID: mdl-16207326

ABSTRACT

This study sought to evaluate the levels of mRNA expression and protein synthesis of MMP-13, cathepsin K, aggrecanase-1 (ADAMTS-4), aggrecanase-2 (ADAMTS-5) and 5-lipoxygenase (5-LOX) in cartilage in the experimental anterior cruciate ligament (ACL) dog model of osteoarthritis (OA), and to examine the effects of treatment with licofelone, a 5-lipoxygenase (LOX)/cyclooxygenase (COX) inhibitor, on the levels of these catabolic factors. Sectioning of the ACL of the right knee was performed in three experimental groups: group 1 received no active treatment (placebo group); and groups 2 and 3 received therapeutic concentrations of licofelone (2.5 or 5.0 mg/kg/day orally, respectively) for 8 weeks, beginning the day following surgery. A fourth group consisted of untreated dogs that were used as normal controls. Specimens of cartilage were selected from lesional areas of OA femoral condyles and tibial plateaus, and were processed for real-time quantitative PCR and immunohistochemical analyses. The levels of MMP-13, cathepsin K, ADAMTS-4, ADAMTS-5 and 5-LOX were found to be significantly increased in OA cartilage. Licofelone treatment decreased the levels of both mRNA expression and protein synthesis of the factors studied. Of note was the marked reduction in the level of 5-LOX gene expression. The effects of the drug were about the same at both tested dosages. In vivo treatment with therapeutic dosages of licofelone has been found to reduce the degradation of OA cartilage in experimental OA. This, coupled with the results of the present study, indicates that the effects of licofelone are mediated by the inhibition of the major cartilage catabolic pathways involved in the destruction of cartilage matrix macromolecules. Moreover, our findings also indicate the possible auto-regulation of 5-LOX gene expression by licofelone in OA cartilage.


Subject(s)
ADAM Proteins/biosynthesis , Acetates/therapeutic use , Anterior Cruciate Ligament/drug effects , Arachidonate 5-Lipoxygenase/biosynthesis , Cathepsins/biosynthesis , Collagenases/biosynthesis , Cyclooxygenase Inhibitors/therapeutic use , Lipoxygenase Inhibitors/therapeutic use , Osteoarthritis/drug therapy , Procollagen N-Endopeptidase/biosynthesis , Pyrroles/therapeutic use , ADAM Proteins/genetics , ADAMTS4 Protein , Acetates/pharmacology , Animals , Anterior Cruciate Ligament/metabolism , Anterior Cruciate Ligament/pathology , Arachidonate 5-Lipoxygenase/genetics , Cathepsin K , Cathepsins/genetics , Collagenases/genetics , Cyclooxygenase Inhibitors/pharmacology , Dogs , Down-Regulation/drug effects , Drug Evaluation , Enzyme Induction/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Lipoxygenase Inhibitors/pharmacology , Matrix Metalloproteinase 13 , Osteoarthritis/genetics , Osteoarthritis/pathology , Procollagen N-Endopeptidase/genetics , Pyrroles/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
18.
Arthritis Rheum ; 52(2): 488-500, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15693013

ABSTRACT

OBJECTIVE: To examine the in vivo effects of PD-0200347, an alpha(2)delta ligand of voltage-activated Ca(2+) channels and a compound chemically related to pregabalin and gabapentin, on the development of cartilage structural changes in an experimental dog model of osteoarthritis (OA). The effects of PD-0200347 on the major pathways involved in OA cartilage degradation, including matrix metalloproteinases (MMPs) and the inducible form of nitric oxide synthase (iNOS), were also studied. METHODS: OA was surgically induced in dogs by sectioning the anterior cruciate ligament. OA dogs were randomly distributed into 3 groups and treated orally with either 1) placebo, 2) 15 mg/kg/day of PD-0200347, or 3) 90 mg/kg/day of PD-0200347. Dogs were killed 12 weeks after surgery. The severity of the lesions was scored macroscopically and histologically. Cartilage specimens from the femoral condyles and tibial plateaus were processed for RNA extraction and quantitative reverse transcription-polymerase chain reaction (RT-PCR) or immunohistochemistry. Specific probes and antibodies were used to study the messenger RNA and protein levels of iNOS, MMP-1, MMP-3, and MMP-13. RESULTS: No clinical signs of drug toxicity were noted in the treated animals. Treatment with PD-0200347 at both dosages tested (15 and 90 mg/kg/day) reduced the development of cartilage lesions. There was a reduction in the score of lesions, with a statistically significant (P = 0.01) difference when the highest dosage of the drug was administered. The reduction in the score was mainly related to a decrease in the surface size of the lesions. Quantitative RT-PCR showed that PD-0200347 significantly reduced the expression of MMP-13, a key mediator in OA. Immunohistochemical analyses showed that treatment with PD-0200347 significantly reduced the synthesis of all key OA mediators studied. CONCLUSION: This study demonstrated the efficacy of PD-0200347 in reducing the progression of cartilage structural changes in a dog model of OA. It also showed that this effect is linked to the inhibition of the major pathophysiologic mediators responsible for cartilage degradation.


Subject(s)
Amines/administration & dosage , Cartilage, Articular/enzymology , Chondrocytes/enzymology , Cyclohexanecarboxylic Acids/administration & dosage , Metalloproteases/biosynthesis , Metalloproteases/genetics , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Osteoarthritis/prevention & control , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/administration & dosage , Administration, Oral , Animals , Cartilage, Articular/pathology , Disease Models, Animal , Disease Progression , Dogs , Gabapentin , Gene Expression/drug effects , Immunohistochemistry , Metalloproteases/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Osteoarthritis/pathology , Pregabalin , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , gamma-Aminobutyric Acid/pharmacology
19.
Arthritis Rheum ; 50(12): 3925-33, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15593193

ABSTRACT

OBJECTIVE: To explore the modulation of 5-lipoxygenase-activating protein (FLAP) and 5-lipoxygenase (5-LOX) expression in human osteoarthritic (OA) chondrocytes, their relative implications in leukotriene B(4) (LTB(4)) production, the effect of different factors on this system, and the influence of increased LTB(4) production on the synthesis of catabolic factors of cartilage. METHODS: FLAP and 5-LOX expression and LTB(4) production were monitored following treatment with transforming growth factor beta1 (TGFbeta1; 5 ng/ml) and 1,25-dihydroxyvitamin D(3) (1,25[OH](2)D(3); 50 nM) alone or in combination with selective or nonselective cyclooxygenase (COX) inhibitors, naproxen (90 mug/ml), NS-398 (10 muM), or FR122047 (5 muM), or a dual inhibitor of COX/5-LOX activity, licofelone (2.6 muM). LTB(4), prostaglandin E(2) (PGE(2)), and matrix metalloprotease 1 (MMP-1) production were measured by specific enzyme-linked immunosorbent assays, nitric oxide by the Griess reaction, and FLAP and 5-LOX expression by quantitative polymerase chain reaction. RESULTS: Human OA chondrocytes expressed both FLAP and 5-LOX. TGFbeta1 and/or 1,25(OH)(2)D(3) induced a rapid and marked enhancement ( approximately 4-13-fold) in FLAP messenger RNA (mRNA) levels, which was associated with a subsequent and late increase in LTB(4) production and PGE(2) synthesis. Treatment with COX inhibitors in the absence or presence of TGFbeta1 and 1,25(OH)(2)D(3) induced a rapid increase in LTB(4) production; this response was mediated by the sustained and significant (P < 0.01) up-regulation ( approximately 1.5-fold) of 5-LOX mRNA levels. Conversely, treatment with licofelone showed no effect on 5-LOX but significantly reduced FLAP expression levels. Coincubation of licofelone with TGFbeta1 plus 1,25(OH)(2)D(3) did not affect FLAP or 5-LOX levels. In the presence of TGFbeta1 plus 1,25(OH)(2)D(3), naproxen, but not licofelone, induced MMP-1 production and both drugs decreased nitric oxide levels. CONCLUSION: Both the eicosanoids PGE(2) and LTB(4) are important cofactors in regulating FLAP/5-LOX expression; the inhibition of PGE(2) up-regulates 5-LOX while down-regulating FLAP gene expression, and LTB(4) appears to be an up-regulating factor on the 5-LOX gene. Importantly, nonsteroidal antiinflammatory drugs up-regulate the synthesis of LTB(4), supporting the shunt hypothesis from COX to 5-LOX. We also demonstrated that LTB(4) likely contributes to the up-regulation of important catabolic factors involved in the pathophysiology of OA, such as MMP.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Carrier Proteins/metabolism , Cartilage, Articular/enzymology , Chondrocytes/enzymology , Leukotriene B4/biosynthesis , Membrane Proteins/metabolism , Osteoarthritis, Knee/enzymology , Transforming Growth Factor beta/physiology , 5-Lipoxygenase-Activating Proteins , Arachidonate 5-Lipoxygenase/genetics , Calcitriol/pharmacology , Carrier Proteins/genetics , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/pathology , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/metabolism , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Humans , Matrix Metalloproteinase 1/metabolism , Membrane Proteins/genetics , Middle Aged , Nitric Oxide/metabolism , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , RNA, Messenger/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Up-Regulation
20.
Arthritis Rheum ; 50(8): 2521-30, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15334466

ABSTRACT

OBJECTIVE: To compare gene expression in normal and osteoarthritic (OA) human chondrocytes using microarray technology. Of the novel genes identified, we selected follistatin, a bone morphogenetic protein (BMP) antagonist, and investigated its expression/regulation as well as that of 3 other antagonists, gremlin, chordin, and noggin, in normal and OA chondrocytes and synovial fibroblasts. METHODS: Basal and induced gene expression were determined using real-time polymerase chain reaction. Gene regulation was monitored following treatment with inflammatory, antiinflammatory, growth, and developmental factors. Follistatin protein production was measured using a specific enzyme-linked immunosorbent assay, and localization of follistatin and gremlin in cartilage was determined by immunohistochemical analysis. RESULTS: All BMP antagonists except noggin were expressed in chondrocytes and synovial fibroblasts. Follistatin and gremlin were significantly up-regulated in OA chondrocytes but not in OA synovial fibroblasts. Chordin was weakly expressed in normal and OA cells. Production of follistatin protein paralleled the gene expression pattern. Follistatin and gremlin were expressed preferentially by the chondrocytes at the superficial layers of cartilage. Tumor necrosis factor alpha and interferon-gamma significantly stimulated follistatin expression but down-regulated expression of gremlin. Interleukin-1beta (IL-1beta) had no effect on follistatin but reduced gremlin expression. Conversely, BMP-2 and BMP-4 significantly stimulated expression of gremlin but down-regulated that of follistatin. IL-13, dexamethasone, transforming growth factor beta1, basic fibroblast growth factor, platelet-derived growth factor type BB, and endothelial cell growth factor down-regulated the expression of both antagonists. CONCLUSION: This study is the first to show the possible involvement of follistatin and gremlin in OA pathophysiology. The increased activin/BMP-binding activities of these antagonists could affect tissue remodeling. The data suggest that follistatin and gremlin might appear at different stages during the OA process, making them interesting targets for the treatment of this disease.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Chondrocytes/metabolism , Fibroblasts/metabolism , Follistatin/analysis , Gene Expression Regulation/physiology , Gene Expression/physiology , Intercellular Signaling Peptides and Proteins/analysis , Osteoarthritis/physiopathology , Synovial Membrane/cytology , Transforming Growth Factor beta , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins , Cells, Cultured , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Follistatin/physiology , Glycoproteins/analysis , Glycoproteins/physiology , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/physiology , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Osteoarthritis/metabolism , Protein Array Analysis , Proteins/analysis , Proteins/physiology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...