Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Osteoarthritis Cartilage ; 28(10): 1373-1384, 2020 10.
Article in English | MEDLINE | ID: mdl-32659345

ABSTRACT

OBJECTIVE: TGFß is a key player in cartilage homeostasis and OA pathology. However, few data are available on the role of TGFß signalling in the different OA phenotypes. Here, we analysed the TGFß pathway by transcriptomic analysis in six mouse models of OA. METHOD: We have brought together seven expert laboratories in OA pathophysiology and, used inter-laboratories standard operating procedures and quality controls to increase experimental reproducibility and decrease bias. As none of the available OA models covers the complexity and heterogeneity of the human disease, we used six different murine models of knee OA: from post-traumatic/mechanical models (meniscectomy (MNX), MNX and hypergravity (HG-MNX), MNX and high fat diet (HF-MNX), MNX and seipin knock-out (SP-MNX)) to aging-related OA and inflammatory OA (collagenase-induced OA (CIOA)). Four controls (MNX-sham, young, SP-sham, CIOA-sham) were added. OsteoArthritis Research Society International (OARSI)-based scoring of femoral condyles and ribonucleic acid (RNA) extraction from tibial plateau samples were done by single operators as well as the transcriptomic analysis of the TGFß family pathway by Custom TaqMan® Array Microfluidic Cards. RESULTS: The transcriptomic analysis revealed specific gene signatures in each of the six models; however, no gene was deregulated in all six OA models. Of interest, we found that the combinatorial Gdf5-Cd36-Ltbp4 signature might discriminate distinct subgroups of OA: Cd36 upregulation is a hallmark of MNX-related OA while Gdf5 and Ltbp4 upregulation is related to MNX-induced OA and CIOA. CONCLUSION: These findings stress the OA animal model heterogeneity and the need of caution when extrapolating results from one model to another.


Subject(s)
CD36 Antigens/genetics , Disease Models, Animal , Growth Differentiation Factor 5/genetics , Latent TGF-beta Binding Proteins/genetics , Mice , Osteoarthritis/genetics , Transforming Growth Factor beta/genetics , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/physiopathology , Collagenases , Diet, High-Fat , GTP-Binding Protein gamma Subunits/genetics , Gene Expression Profiling , Hypergravity , Meniscectomy , Metabolic Syndrome , Mice, Knockout , Obesity , Osteoarthritis/metabolism , Osteoarthritis/physiopathology , Transcriptome , Transforming Growth Factor beta/metabolism
2.
Aliment Pharmacol Ther ; 45(8): 1058-1072, 2017 04.
Article in English | MEDLINE | ID: mdl-28247573

ABSTRACT

BACKGROUND: Crohn's disease (CD) and rheumatoid arthritis are chronic, progressive and disabling conditions that frequently lead to structural tissue damage. Based on strategies originally developed for rheumatoid arthritis, the treatment goal for CD has recently moved from exclusively controlling symptoms to both clinical remission and complete mucosal healing (deep remission), with the final aim of preventing bowel damage and disability. AIM: To review the similarities and differences in treatment goals between CD and rheumatoid arthritis. METHODS: This review examined manuscripts from 1982 to 2016 that discussed and/or proposed therapeutic goals with their supportive evidence in CD and rheumatoid arthritis. RESULTS: Proposed therapeutic strategies to improve outcomes in both rheumatoid arthritis and CD include: (i) evaluation of musculoskeletal or organ damage and disability, (ii) tight control, (iii) treat-to-target, (iv) early intervention and (v) disease modification. In contrast to rheumatoid arthritis, there is a paucity of disease-modification trials in CD. CONCLUSIONS: Novel therapeutic strategies in CD based on tight control of objective signs of inflammation are expected to change disease course and patients' lives by halting progression or, ideally, preventing the occurrence of bowel damage. Most of these strategies require validation in prospective studies, whereas several disease-modification trials have addressed these issues in rheumatoid arthritis over the last decade. The recent approval of new drugs in CD such as vedolizumab and ustekinumab should facilitate initiation of disease-modification trials in CD in the near future.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Crohn Disease/drug therapy , Patient Care Planning/trends , Antibodies, Monoclonal, Humanized/therapeutic use , Disease Progression , Goals , Humans , Prospective Studies , Ustekinumab/therapeutic use
3.
Osteoarthritis Cartilage ; 24(11): 1961-1969, 2016 11.
Article in English | MEDLINE | ID: mdl-27307356

ABSTRACT

OBJECTIVE: Fibroblast Growth Factor 23 (FGF23) may represent an attractive candidate that could participate to the osteoarthritic (OA)-induced phenotype switch of chondrocytes. To address this hypothesis, we investigated the expression of FGF23, its receptors (FGFRs) and co-receptor (Klotho) in human cartilage and studied the effects of rhFGF23 on OA chondrocytes. METHOD: Gene expression or protein levels were analysed by RT-PCR and immunohistochemistry. Collagenase 3 (MMP13) activity was measured by a fluorescent assay. MAPK signalling pathways were investigated by phosphoprotein array, immunoblotting and the use of selective inhibitors. RNA silencing was performed to confirm the respective contribution of FGFR1 and Klotho. RESULTS: We showed that the expression of FGF23, FGFR1 and Klotho was up-regulated at both mRNA and protein levels in OA chondrocytes when compared to healthy ones. These overexpressions were markedly elevated in the damaged regions of OA cartilage. When stimulated with rhFGF23, OA chondrocytes displayed an extended expression of FGF23 and of markers of hypertrophy such as MMP13, COL10A1, and VEGF. We demonstrated that FGF23 auto-stimulation was both FGFR1-and Klotho-dependent, whereas the expression of markers of hypertrophy was mainly dependent on FGFR1 alone. Finally, we showed that FGF23-induced MMP13 expression was strongly regulated by the MEK/ERK cascade and to a lesser extent, by the PI-3K/AKT pathway. CONCLUSION: These results demonstrate that FGF23 sustains differentiation of OA chondrocytes in a Klotho-independent manner.


Subject(s)
Chondrocytes , Cartilage, Articular , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Humans , Matrix Metalloproteinase 13 , Osteoarthritis , Phosphatidylinositol 3-Kinases
4.
Bone ; 75: 111-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25667190

ABSTRACT

PURPOSE: Clinical and in vitro studies suggest that subchondral bone sclerosis due to abnormal osteoblasts is involved in the progression of osteoarthritis (OA). Human osteoblasts isolated from sclerotic subchondral OA bone tissue show an altered phenotype, a decreased canonical Wnt/ß-catenin pathway, and a reduced mineralization in vitro as well as in vivo. These alterations were linked with an abnormal response to BMP-2. OA osteoblasts release factors such as the hepatocyte growth factor (HGF) that contribute to cartilage loss whereas chondrocytes do not express HGF. HGF can stimulate BMP-2 expression in human osteoblasts, however, the role of HGF and its effect in OA osteoblasts remains unknown. Here we investigated whether elevated endogenous HGF levels in OA osteoblasts are responsible for their altered response to BMP-2. METHODS: We prepared primary human subchondral osteoblasts using the sclerotic medial portion of the tibial plateaus of OA patients undergoing total knee arthroplasty, or from tibial plateaus of normal individuals obtained at autopsy. The expression of HGF was evaluated by qRT-PCR and the protein production by western blot analysis. HGF expression was reduced with siRNA technique whereas its activity was inhibited using the selective inhibitor PHA665752. Alkaline phosphatase activity (ALPase) and osteocalcin release were measured by substrate hydrolysis and EIA respectively. Canonical Wnt/ß-catenin signaling (cWnt) was evaluated both by target gene expression using the TOPflash TCF/lef luciferase reporter assay and western blot analysis of ß-catenin levels in response to Wnt3a stimulation. Mineralization in response to BMP-2 was evaluated by alizarin red staining. RESULTS: The expression of HGF was increased in OA osteoblasts compared to normal osteoblasts and was maintained during their in vitro differentiation. OA osteoblasts released more HGF than normal osteoblasts as assessed by western blot analysis. HGF stimulated the expression of TGF-ß1. BMP-2 dose-dependently (1 to 100 ng/ml) stimulated both ALPase and osteocalcin in normal osteoblasts whereas, it inhibited them in OA osteoblasts. HGF-siRNA treatments reversed this response in OA osteoblasts and restored the BMP-2 response. cWnt is reduced in OA osteoblasts compared to normal, and HGF-siRNA treatments increased cWnt in OA osteoblasts almost to normal. Smad1/5/8 phosphorylation in response to BMP-2, which is reduced in OA osteoblasts, was corrected when these cells were treated with PHA665752. The BMP-2-dependent mineralization of OA osteoblasts, which is also reduced compared to normal, was only partially restored by PHA665752 treatment whereas 28 days treatment with HGF reduced the mineralization of normal osteoblasts. CONCLUSION: OA osteoblasts expressed more HGF than normal osteoblasts. Increased endogenous HGF production in OA osteoblasts stimulated the expression of TGF-ß1 and reduced their response to BMP-2. Inhibiting HGF expression or HGF signaling restored the response to BMP-2 and Smad1/5/8 signaling. In addition, decreased HGF signaling partly corrects the abnormal mineralization of OA osteoblasts while increased HGF prevents the normal mineralization of normal osteoblasts. In summary, we hypothesize that sustained elevated HGF levels in OA osteoblasts drive their abnormal phenotype and is implicated in OA pathophysiology.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Calcification, Physiologic/physiology , Hepatocyte Growth Factor/metabolism , Osteoarthritis, Knee/metabolism , Osteoblasts/metabolism , Aged , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Osteoarthritis, Knee/pathology , Polymerase Chain Reaction , Signal Transduction/physiology
5.
Free Radic Biol Med ; 76: 114-26, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25106704

ABSTRACT

The inhibitory effect of 15-deoxy-Δ(12,14)-prostaglandin J2 (15d-PGJ2) on proinflammatory gene expression has been extensively documented and frequently ascribed to its ability to prevent NF-κB pathway activation. We and others have previously demonstrated that it was frequently independent of the peroxisome proliferator activated receptor (PPAR)γ activation. Here, we provide evidence that induction of intracellular heat shock protein (HSP)70 by oxidative stress is an additional regulatory loop supporting the anti-inflammatory effect of 15d-PGJ2 in chondrocytes. Using real-time quantitative PCR and Western blotting, we showed that 15d-PGJ2 stimulated HSP70, but not HSP27 expression while increasing oxidative stress as measured by spectrofluorimetry and confocal spectral imaging. Using N-acetylcysteine (NAC) as an antioxidant, we demonstrated further that oxidative stress was thoroughly responsible for the increased expression of HSP70. Finally, using an HSP70 antisense strategy, we showed that the inhibitory effect of 15d-PGJ2 on IL-1-induced activation of the NF-κB pathway, COX-2 and mPGES-1 expression, and PGE2 synthesis was partly supported by HSP70. These data provide a new anti-inflammatory mechanism to support the PPARγ-independent effect of 15d-PGJ2 in chondrocyte and suggest a possible feedback regulatory loop between oxidative stress and inflammation via intracellular HSP70 up-regulation. This cross talk is consistent with 15d-PGJ2 as a putative negative regulator of the inflammatory reaction.


Subject(s)
Chondrocytes/pathology , Gene Expression Regulation/drug effects , HSP70 Heat-Shock Proteins/metabolism , Oxidative Stress/drug effects , Prostaglandin D2/analogs & derivatives , Prostaglandins/metabolism , Signal Transduction/drug effects , Animals , Blotting, Western , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Female , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/genetics , Interleukin-1/pharmacology , NF-kappa B/genetics , NF-kappa B/metabolism , Oligonucleotides, Antisense/pharmacology , PPAR gamma/genetics , PPAR gamma/metabolism , Prostaglandin D2/pharmacology , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
6.
Biomed Mater Eng ; 24(1 Suppl): 3-16, 2014.
Article in English | MEDLINE | ID: mdl-24928913

ABSTRACT

Osteoarthritis (OA) is a whole-joint disease characterized by cartilage degradation and mineralization associated with chondrocyte phenotype changes, subchondral bone sclerosis and mild synovial inflammation. The extracellular levels of inorganic phosphate (ePi) and pyrophosphate (ePPi) are major regulators of the mineralization process but also play a role in the maintenance of the differentiated chondrocyte phenotype. Four membrane proteins control the balance between ePi and ePPi: the transporters ANK (exporting PPi outside the cells) and PiT-1 (importing ePi into the cells), and the enzymes PC-1 (generating ePPi from nucleotides) and Tissue Non-specific Alkaline Phosphatase (TNAP, hydrolyzing ePPi into ePi). In the present work, we investigated the ability of specific microRNAs (miR) to modulate activity and level of the mRNA coding for the regulatory proteins of the ePi/ePPi balance in chondrocytes. The 4 following microRNAs, let7e, miR-9, miR-188 and miR-219, were selected by bioinformatics for their ability to putatively target the mRNA 3'UTRs of these regulators. In IL-1ß-stimulated human chondrocytes, chosen as a model of differentiated phenotype loss implicating the PPi transporter ANK, miR-9, miR-188 and let7e levels increased. However, luciferase reporter assays and transient miR overexpression in the ATDC5 chondrogenic cell line only support that miR-9 was a negative post-transcriptional regulator of PC-1, Pit-1 and TNAP mRNAs. This suggests that miR-9 could contribute to the acquirement of an altered chondrocyte phenotype by disrupting the ePi/ePPi balance.


Subject(s)
Chondrocytes/drug effects , Diphosphates/metabolism , MicroRNAs/genetics , Phosphates/metabolism , Algorithms , Amino Acid Sequence , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Chondrocytes/metabolism , Gene Expression Regulation , Gene Targeting/methods , HeLa Cells , Humans , Membrane Proteins/chemistry , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Osteoarthritis/therapy , Phenotype
7.
Osteoarthritis Cartilage ; 22(3): 519-26, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24462671

ABSTRACT

OBJECTIVE: Conflicting findings raise questions about the role of adiponectin in osteoarthritis (OA). The current study aimed to investigate in OA patients the association between the production of adiponectin and the grade of cartilage destruction, and to provide functional evidence for a potential role of adiponectin in OA. DESIGN: The expression of adiponectin was examined by immunohistochemistry in cartilage obtained from healthy individuals (n = 2; ages 56 and 41 years; 1 male and 1 female) and OA patients (n = 11; ages 64-79 years; 2 male and 9 female). The association between its production in chondrocytes and the grade of cartilage destruction was established on full-depth cartilage biopsies. The functional activity of adiponectin in OA cartilage was determined from the relation between the expression of adiponectin, its receptor, cartilage-specific components and factors involved in matrix degradation, and from the chondrocyte response to the full-length or the globular form of adiponectin. RESULTS: Adiponectin was not detected in healthy cartilage. Conversely, the adipokine was up-regulated in damaged tissue, but no strong association with the grade of cartilage destruction was found. We showed a positive correlation between adiponectin and mPGES or MMP-13 while AdipoR1 was related to the expression of type 2 collagen, aggrecan and Sox9. The full-length form of adiponectin but not the globular isoform, stimulated the production of PGE2 and MMP-13 activity in cultured human chondrocytes. CONCLUSIONS: The elevated level of adiponectin found in chondrocytes from OA patients might contribute to matrix remodelling during OA, the full-length isoform being the single active form.


Subject(s)
Adiponectin/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osteoarthritis, Knee/metabolism , Adult , Aged , Dinoprostone/metabolism , Female , Humans , Male , Matrix Metalloproteinase 13/metabolism , Middle Aged , Receptors, Adiponectin/metabolism
8.
Biorheology ; 45(3-4): 415-32, 2008.
Article in English | MEDLINE | ID: mdl-18836242

ABSTRACT

In inflammatory conditions, chondrocytes produce large amounts of matrix metalloproteases (MMP) and nitric oxide (NO) thought to contribute to joint degradation. We tested the ability of all-trans retinoic acid (ATRA, a retinoic acid receptor (RAR) agonist) to modulate these inflammatory genes in chondrocytes from humans or rats, chosen as representative of animal models of arthritis. All RAR subtypes and RXR-alpha or -beta were expressed at the mRNA level in both species, although IL-1beta (10 ng/ml) inhibited RAR subtypes more markedly in rat than in human cells. ATRA (300 or 1000 nM) inhibited IL-1-induced expression of iNOS and nitrites level in both species, although the NO pathway was induced maximally in rat cells. ATRA displayed controversial effects on MMPs between rat and human chondrocytes, especially for MMP-9 expression. The effects of ATRA were irrelevant to the nuclear translocation of AP-1. The present data underlines that retinoids have a species-dependent impact on IL-1-induced responses in chondrocytes, suggesting that extrapolation of their pharmacological properties from animal cells has a poor relevance to clinical situation.


Subject(s)
Chondrocytes/metabolism , Interleukin-1beta/metabolism , Matrix Metalloproteinases/metabolism , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Transcription Factor AP-1/drug effects , Tretinoin/metabolism , Animals , Arthritis, Rheumatoid , Cell Culture Techniques , Chondrocytes/drug effects , Gene Expression , Humans , Interleukin-1beta/pharmacology , Matrix Metalloproteinases/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Receptors, Retinoic Acid/drug effects , Retinoid X Receptors/drug effects , Species Specificity , Transcription Factor AP-1/metabolism , Tretinoin/pharmacology
9.
Biorheology ; 45(3-4): 439-55, 2008.
Article in English | MEDLINE | ID: mdl-18836244

ABSTRACT

The present work aimed to take advantage of the screening capacity of protein arrays to search for additional targets of rhein in interleukin (IL)-1-stimulated chondrocytes. Primary cultures of chondrocytes from osteoarthritic (OA) patients were stimulated for 24 and 48 h with 1 ng/ml of IL-1alpha, in the presence or absence of 10(-5) M of rhein. Culture supernatants were analyzed with arrays membranes consisting of 120 antibodies directed against cytokines, chemokines, and angiogenic or growth factors and were controlled for 8 proteins by specific immuno-enzymatic assays (ELISA). Protein arrays showed that several CC or CXC chemokines, the growth factor GM-CSF, the cytokines IL-6, IL-7 and IL-10 (but unexpectedly not IL-1beta or TNFalpha) and the adhesion molecule ICAM-1 were induced maximally by IL-1alpha. In IL-1-stimulated chondrocytes, rhein reduced slightly the production of MCP-1 and increased those of IL-1Ra, of the cytokine receptors sgp130, IL-6R, sTNFR I and R II, but also of some chemokines or ICAM-1. Specific ELISAs confirmed the effect of rhein on MCP-1, IL-1Ra, sgp130, IL-6R and sTNFR II but was discrepant for GROalpha and were always more sensitive than protein arrays to detect IL-1 effects such as IL-1Ra and TNFalpha release. The present data show that rhein modulated some IL-1-induced responses contributing possibly to its chondroprotective (IL-1Ra, MCP-1) or cytokine modifying (sTNFR II, sgp130) properties, but that protein arrays were poorly sensitive to check for IL-1- and/or rhein-induced changes.


Subject(s)
Anthraquinones/pharmacology , Antibodies/analysis , Chemokines/drug effects , Chondrocytes/drug effects , Cytokines/drug effects , Interleukin-1/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Chemokines/metabolism , Chondrocytes/immunology , Chondrocytes/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Humans , Inflammation/drug therapy , Nitrites/metabolism , Osteoarthritis/metabolism , Protein Array Analysis/methods
10.
Osteoarthritis Cartilage ; 15(5): 493-505, 2007 May.
Article in English | MEDLINE | ID: mdl-17140817

ABSTRACT

OBJECTIVE: To investigate the potency of selective agonists of peroxisome proliferators-activated receptors' (PPAR) isotypes (alpha, beta/delta or gamma) to modulate the stimulating effect of transforming growth factor-beta1 (TGF-beta1) on proteoglycans' (PGs) synthesis in chondrocytes. METHOD: Rat chondrocytes embedded in alginate beads and cultured under low serum conditions were exposed to TGF-beta1 (10 ng/ml), alone or in combination with the following agonists: Wy14643 for PPARalpha, GW501516 for PPARbeta/delta, rosiglitazone (ROSI) for PPARgamma, in the presence or absence of PPAR antagonists (GW6471 for PPARalpha, GW9662 for PPARgamma). PGs' synthesis was evaluated by radiolabelled sulphate incorporation and glycosaminoglycans' (GAGs) content by Alcian blue staining of beads and colorimetric 1.9 dimethyl-methylene blue assay after beads' solubilization. Phosphorylation of Extracellular Signal-related Kinase1/2 (ERK1/2), Smad2/3 and p38-MAPK was assessed by Western Blot and production of prostaglandin E2 (PGE2) by Enzyme immuno-assay (EIA). Levels of mRNA for PPAR target genes [acyl-CoA oxidase (ACO) for PPARalpha; mitochondrial carnitin palmitoyl transferase-1 (CPT-1) for PPARbeta/delta and adiponectin for PPARgamma], aggrecan, TGF-beta1 and genes controlling GAGs' side chains' synthesis were quantified by real time polymerase chain reaction and normalized over RP29 housekeeping gene. RESULTS: ACO was selectively up-regulated by 100 microM of Wy14643, CPT-1 by 100 nM of GW501516 and adiponectin by 10 microM of ROSI without cell toxicity. TGF-beta1 increased PGs' synthesis by four-fold, GAGs' content and deposition by 3.5-fold and six-fold, respectively, while inducing aggrecan expression around 10-fold without modifying mRNA levels of GAGs' controlling enzymes. PPAR agonists inhibited the stimulating effect of TGF-beta1 by 24-44% on PGs' synthesis and over 75% on aggrecan, GAGs' content and deposition with the following rank order of potency: ROSI>GW501516> or =Wy14643. TGF-beta1-induced phosphorylation of Smad2/3 and ERK1/2 was reduced by ROSI over GW501516 but not by Wy14643 whereas stimulated PGE2 production was inhibited by Wy14643 over GW501516 but not by ROSI. The effect of PPAR agonists on PPAR target genes and TGF-beta1-induced aggrecan expression was reversed selectively by PPAR antagonists. CONCLUSION: In chondrocytes' beads, PPAR agonists reduced the stimulating effect of TGF-beta1 on PGs by inhibiting TGF-beta1-induced aggrecan expression in an isotype-selective manner. Thus, PPAR agonists could be deleterious in situation of cartilage repair although being protective in situation of cartilage degradation.


Subject(s)
Peroxisome Proliferator-Activated Receptors/pharmacology , Proteoglycans/biosynthesis , Transforming Growth Factor beta/agonists , Animals , Chondrocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycosaminoglycans/biosynthesis , Rats , Smad Proteins/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Osteoarthritis Cartilage ; 12(4): 284-95, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15023380

ABSTRACT

OBJECTIVES: To characterize the molecular events underlying cartilage injury in the early phase of mono-iodoacetate-induced osteoarthritis (OA) in rats. METHODS: Experimental osteoarthritis was induced by intra-articular injection of 0.03mg mono-iodoacetate (MIA) in Wistar rats. Animals were killed 2, 5, 10, 15 and 20 days post-injection. Synovial tissue and standardized biopsies from different areas of knee cartilage were examined. Proteoglycan synthesis ((35)S incorporation) and gelatinase activities (zymography), semi-quantitative RT-PCR and immunohistochemistry for IL1beta, iNOS, COX2 and PPARgamma, were performed on these samples. RESULTS: Changes in proteoglycan synthesis and gelatinase activities were time and site-dependent. Proteoglycan synthesis inhibition was maximal by day 2 while the highest gelatinase activities were observed at day 5. Central part of patella and posterior plateaus and condyles, i.e. the weight-bearing cartilage areas, were the most affected. IL1beta and iNOS transcripts were induced early in cartilage at time of maximal proteoglycan synthesis inhibition, especially in weight-bearing areas. COX-2 was slightly up-regulated whereas PPARgamma gene expression remained unchanged. Gene expression profile in synovium paralleled that of cartilage, except for PPARgamma which was up-regulated at day 15 and 20. Immunostaining for IL1beta and iNOS showed that proteins were located in diseased cartilage areas at early stage of the experimental OA (day 2). At later time-points (day 20), IL1beta and iNOS were expressed in perilesional areas whereas immunostaining became below control level for COX-2 and PPARgamma. CONCLUSIONS: Time-dependent degradation of cartilage after injection of low dose of MIA (0.03mg) into rat knee joint can be related to early loss of proteoglycan anabolism, increased gelatinase activities and expression of IL1beta and downstream inflammatory genes. Increased susceptibility to MIA in weight-bearing areas of cartilage further indicate that MIA-induced experimental OA is a relevant model to study not only metabolical but also biomechanical aspects of human OA.


Subject(s)
Cartilage, Articular/metabolism , Gene Expression/genetics , Osteoarthritis/genetics , Animals , Cyclooxygenase 2 , Enzyme Inhibitors , Gelatinases/metabolism , Immunohistochemistry/methods , Interleukin-1/analysis , Iodoacetic Acid , Isoenzymes/analysis , Male , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Osteoarthritis/metabolism , Prostaglandin-Endoperoxide Synthases/analysis , Proteoglycans/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Transcription Factors/analysis
12.
Bull Acad Natl Med ; 185(2): 417-24; discussion 424-6, 2001.
Article in French | MEDLINE | ID: mdl-11474594

ABSTRACT

Familial articular chondrocalcinosis is a chronic articular disease characterized by acute intermittent attacks of arthritis, presence of calcium pyrophosphate dihydrate crystal in synovial fluid, cartilage and periarticular soft tissue and by x rays calcium deposition in articular cartilage. A family originating from Alsace, with an autosomal dominant transmission has been studied. As in English and Argentinean families, a linkage to the short arm of chromosome 5p has been found. These results suggest that a defective gene at this location may be related to the chondrocalcinosis in these families.


Subject(s)
Chondrocalcinosis/genetics , Adult , Chondrocalcinosis/diagnosis , Chondrocalcinosis/physiopathology , France , Humans
13.
Clin Pharmacokinet ; 39(5): 369-82, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11108435

ABSTRACT

OBJECTIVE: To investigate the distribution of meloxicam in the human knee joint and to compare it with the inhibition of cyclo-oxygenase (COX) activity in synovial cells. DESIGN: Prospective pharmacokinetic study and in vitro laboratory investigation. PATIENTS AND PARTICIPANTS: 42 male and female patients aged 26 to 85 years hospitalised for rheumatic disease and requiring a diagnostic and/or therapeutic knee puncture. METHODS: After a single oral dose of meloxicam 15mg, synovial fluid and blood samples were collected once per patient at various intervals after administration. Meloxicam concentrations were determined by a validated high performance liquid chromatography assay, protein binding by equilibrium dialysis, and pharmacokinetic parameters were calculated by noncompartmental analysis from the mean drug concentration-time profiles. The inhibitory effect of meloxicam on COX activity was investigated separately in unstimulated or interleukin-1beta-stimulated human synovial cells from osteoarthritic patients. RESULTS: Meloxicam was found in synovial fluid at the earliest sampling time (1 hour). Peak concentrations were reached approximately 6 hours postdose in both plasma (842 microg/L) and synovial fluid (320 microg/L). A plateau was observed after the distribution phase (6 hours), corresponding to a constant ratio of drug concentration between synovial fluid and plasma of about 0.47. This ratio was higher in patients with acute inflammation (0.58) than in those with no inflammation (0.38). Meloxicam was extensively bound to protein, mainly to serum albumin. The area under the drug concentration-time curve (AUC) in plasma was more than 2.5 times that in synovial fluid. The AUC for free meloxicam was similar in plasma and synovial fluid. The 50% inhibitory concentrations (IC50) for basal and stimulated COX activity in human synovial cells were 33.7 nmol/L (11.8 microg/L) and 2.0 nmol/L (0.70 microg/L), respectively. The free concentration of meloxicam in synovial fluid was higher than the IC50 for stimulated COX activity from 6 to 36 hours postdose. CONCLUSION: On the basis of free synovial concentrations and the IC50 for stimulated COX activity, meloxicam is expected to have a long duration of action. Inhibition of COX activity is expected to be more marked in inflamed synovium compared with non-inflamed synovium.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Cyclooxygenase Inhibitors/pharmacokinetics , Knee Joint/metabolism , Thiazines/pharmacokinetics , Thiazoles/pharmacokinetics , Administration, Oral , Adult , Aged , Aged, 80 and over , Area Under Curve , Diffusion , Female , Humans , Male , Meloxicam , Middle Aged , Prospective Studies , Protein Binding , Synovial Fluid/metabolism , Thiazines/administration & dosage , Thiazines/pharmacology , Thiazoles/administration & dosage , Thiazoles/pharmacology
14.
Rev Med Interne ; 21(11): 978-88, 2000 Nov.
Article in French | MEDLINE | ID: mdl-11109595

ABSTRACT

INTRODUCTION: Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit the production of primary prostanoids by blocking the access of arachidonic acid to the active site of the cyclooxygenases (COXs). Because the prostanoids produced by COX-1 appear to play a physiological role (protection of the gastric mucosa, platelet aggregation, vascular homeostasis, maintenance of renal sodium-water balance) while those produced by COX-2 seem mainly to intervene in the inflammatory response and in certain processes associated with cell proliferation, the hypothesis has been put forward that the NSAIDs that are selective COX-2 inhibitors should theoretically be capable of maintaining NSAID therapeutic properties but also have fewer adverse side effects due to the maintenance of prostaglandin production at normal physiological levels. CURRENT KNOWLEDGE AND KEY POINTS: The hypothesis of COX isoenzyme selectivity has led to a proposed classification for COX inhibitors: 1) COX-1 selective inhibitors (low-dosage aspirin); 2) COX non-selective inhibitors (the majority of classified NSAIDs, which when administered over the long term, e.g., in cases of rheumatoid arthritis, cause duodenal ulcers in 20% of cases and gastric hemorrhage in 1-4% of cases/year); 3) COX-2 preferential inhibitors (meloxicam and nimesulide, which have fewer gastric side effects than standard NSAIDs, but which are not risk-free at high doses); 4) COX-2 selective inhibitors (celecoxib and rofecoxib). Preliminary clinical studies have shown that COX-2 selective inhibitors are as efficient as standard NSAIDs and have fewer adverse digestive side effects, thereby confirming the interest of this proposed classification. In the UK, the aforementioned studies have led to the commercialization of rofecoxib for the treatment of pain and osteoarthritis, while celecoxib has been introduced in medical practice in the USA and other countries for the treatment of rheumatoid arthritis and osteoarthritis. FUTURE PROSPECTS AND PROJECTS: Various epidemiological and laboratory studies have indicated that NSAIDs may be able to reduce the risk of cancer (colorectal cancer in particular) and Alzheimer's disease due to their inhibitory activity on COXs, especially COX-2. The therapeutic contribution of COX-2 specific inhibitors has to be more fully evaluated, particularly as these agents could delay the healing of duodenal ulcers and interfere with several COX-2-induced physiological functions. It is therefore suggested that until further information becomes available, this new class of NSAIDs should be used with caution in certain patient populations.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/classification , Cyclooxygenase Inhibitors/classification , Prostaglandin-Endoperoxide Synthases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/adverse effects , Cyclooxygenase Inhibitors/therapeutic use , Duodenal Ulcer/chemically induced , Gastrointestinal Hemorrhage/chemically induced , Humans , Isoenzymes/metabolism , Membrane Proteins
15.
Biorheology ; 37(1-2): 165-76, 2000.
Article in English | MEDLINE | ID: mdl-10912189

ABSTRACT

The potentialities of a new non-invasive optical scanning microscopy technique were evaluated through 3D analysis of chondrocyte-matrix interactions. Five different 2D or 3D culture systems were used: (1) MonoLayer (ML) of human chondrosarcoma cell line; (2) rat or human chondrocytes encapsulated in Alginate Bead (AB); (3) human chondrocytes encapsulated in Alginate Sponge (AS); (4) Rat Femoral Head Cap (RFHC); (5) slices of knee human Osteoarthritic Cartilage (HOAC). Chondrocytes ML, AB, RFHC were incubated for 24 h in vitro in the presence of recombinant human interleukin1-beta (rhIL1-beta) and the effects on cytoskeleton organisation (F-actin filament), Focal Adhesion Kinase (FAK) expression (tyrosine kinase), collagenase B expression (metalloprotease) were studied. Furthermore, the production of intracellular IL1-beta by LPS- or rhIL1-beta-stimulated chondrocytes was shown to be partly suppressed by rhein (active metabolite of diacerhein) in all culture systems. This high resolution light microscopy gave complementary information that could be important for a better understanding of the interaction of chondrocytes with the extracellular matrix in a variety of culture devices.


Subject(s)
Chondrocytes/metabolism , Extracellular Matrix/metabolism , Integrins/biosynthesis , Microscopy, Confocal , Actins/metabolism , Animals , Cell Culture Techniques , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Culture Techniques , Cytoskeleton/metabolism , Extracellular Matrix/drug effects , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Interleukin-1/biosynthesis , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 9/metabolism , Microspheres , Osteoarthritis/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Recombinant Proteins/pharmacology , Transforming Growth Factor alpha/pharmacology
16.
Presse Med ; 29(5): 267-73, 2000 Feb 12.
Article in French | MEDLINE | ID: mdl-10701410

ABSTRACT

POSSIBLE INFLAMMATORY MECHANISMS: Alzheimer's disease (AD) is a degenerative disease of the brain including possibly inflammatory mechanisms, as illustrated by the presence of activated microglial cells in the periphery of senile plaques and neurofibrillary tangles and the subsequent release of proinflammatory mediators with neurotoxic potency. RATIONALE FOR NSAID USE: Although not firmly demonstrated, the rationale for the prescription of non steroidal anti-inflammatory drugs (NSAIDS) as neuroprotective agents in AD lies on epidemiological data having shown a reduced risk of developing AD in patients on long-term therapy with NSAIDs (non selective cyclo-oxygenase [COX] inhibitors). RATIONALE FOR THE USE OF SELECTIVE COX-2 INHIBITORS: The rationale for the prescription of selective COX-2 inhibitors as neuroprotective drugs in AD lies on: Epidemiological data having shown a reduced risk of developing AD in patients treated with anti-inflammatory doses of classical NSAIDs (inhibition of COX-1 and COX-2) but not with antithrombotic doses of aspirin (selective inhibition of COX-1), Cellular experiments, Demonstration of a better gastro-intestinal (GI) safety profile with selective COX-2 inhibitors than with classical NSAIDs in short-term studies, allowing a possible long-term use in AD. BEFORE PRESCRIBING: COX-2 may have an ambivalent functionality in the brain since the basal production of prostaglandins through COX-2 may participate in neuronal homeostasis whereas the expression of COX-2 is associated with brain development. Classical NSAIDs are ineffective in reducing the formation of senile plaque and neurofibrillary tangles in AD, which is consistent with an ability to reduce inflammation associated with activation of microglia but illustrates their failure to suppress the degenerative process. Prophylactic use of selective COX-2 NSAIDs can be considered on the basis of their good GI safety after 6 months of marketing in United States but need to be confirmed for a longer time. CURRENT TRIALS: Clinical studies focusing on both the prevention and the slowing down of early AD are under way with two recently launched selective COX-2 inhibitors, celecoxib and rofecoxib.


Subject(s)
Alzheimer Disease/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Enzyme Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use , Adult , Age Factors , Aged , Alzheimer Disease/physiopathology , Alzheimer Disease/prevention & control , Animals , Brain/physiopathology , Celecoxib , Child , Female , Humans , Lactones/administration & dosage , Lactones/therapeutic use , Male , Mice , Mice, Transgenic , Oxidative Stress , Pregnancy , Prostaglandins/physiology , Pyrazoles , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use , Sulfones , Time Factors
18.
Arthritis Rheum ; 42(10): 2094-102, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10524680

ABSTRACT

OBJECTIVE: To evaluate the effect of nitric oxide synthase (NOS) inhibitors on proteoglycan synthesis following intraarticular administration of interleukin-1beta (IL-1beta) in rats. METHODS: Recombinant human IL-1beta and NOS inhibitors with different selectivity for inducible NOS (N-monomethyl-L-arginine [L-NMA], N-iminoethyl-L-ornithine [L-NIO], and S-methylisothiourea [SMT]) were simultaneously administered in rats by a single intraarticular injection in each knee. L-NMA was also infused for 72 hours using an Alzet mini osmotic pump implanted into the peritoneal cavity 24 hours before IL-1beta challenge. NO production was determined as nitrate and nitrite, either in synovial fluid or ex vivo in supernatants of synovium and patellae. Proteoglycan synthesis was measured by ex vivo incorporation of 35SO4(2-) into patellar cartilage. RESULTS: IL-1beta induced a time-dependent increase in NO production in synovial fluid. Synovium and patellae released large amounts of nitrate and nitrite under ex vivo conditions, indicating that both tissues are effective sources of NO within the joint. This production of NO was accompanied by a delayed inhibition of proteoglycan synthesis. The intraarticular administration of L-NMA and L-NIO reduced NO release in synovial fluid and resulted in a partial recovery of proteoglycan synthesis. Under our experimental conditions, SMT failed to reduce NO synthesis and to restore proteoglycan synthesis. The protection of cartilage was improved by the systemic and sustained delivery of L-NMA. However, the complete inhibition of NO production in synovial fluid was not sufficient to fully restore cartilage anabolism. CONCLUSION: Our findings show that in rats: 1) NO may be an early mediator of the effect of IL-1beta on cartilage, 2) NO inhibition may have therapeutic relevance, although it is not sufficient to fully reverse the deleterious effects of IL-1beta, 3) among NOS inhibitors tested, only amino acid derivatives are effective, 4) protection can be achieved by local administration of NOS inhibitors, and 5) systemic and sustained delivery of the NOS inhibitor with the highest efficacy after intraarticular injection provides the most benefit.


Subject(s)
Cartilage, Articular/metabolism , Enzyme Inhibitors/pharmacology , Interleukin-1/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/metabolism , Ornithine/analogs & derivatives , Animals , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Drug Antagonism , Humans , Male , Nitric Oxide Synthase/antagonists & inhibitors , Ornithine/pharmacology , Rats , Rats, Wistar
19.
Osteoarthritis Cartilage ; 7(4): 382-5, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10419774

ABSTRACT

Objective To compare the ability of L-arginine (L-arg) analog nitric oxide synthase (NOS) inhibitors and isothioureas to restore the interleukin-1 (IL-1) induced inhibition of proteoglycan (PG) synthesis in rat.Methods Chondrocytes beads and patellae were challenged with IL-1betain vitro and monitored for NO production and proteoglycan synthesis. Rats injected with IL-1beta in knee joints were monitored for NO(2)( - )+NO(3)( - )levels in joint tissues and ex-vivo(35)S sulfate incorporation in patellae. NOS inhibitors were either added to culture medium or injected concomittantly to IL-1beta. Results Ability of NOS inhibitors to reduce NO(2)( - )levels decreased from chondrocytes beads to patellae. Partial restoration of PG synthesis was restricted to L-arg analogs in patellae. After IL-1 injection, NO was produced from patella and synovium. L-arg analogs restored partly PG synthesis when decreasing significantly NO(2)( - )+NO(3)( - )levels in synovial fluid. Isothioureas were ineffective. Conclusions NO accounts importantly for IL-1 induced inhibition of cartilage anabolism in rat. L-arg analog NOS inhibitors are more effective than isothioureas in restoring PG synthesis and have chondroprotective potency when administered locally in diseased joint.


Subject(s)
Cartilage/metabolism , Interleukin-1/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Proteoglycans/biosynthesis , Animals , Chondrocytes/metabolism , Male , Nitric Oxide/metabolism , Nitrogen Oxides/metabolism , Rats , Rats, Wistar , Synovial Fluid/metabolism
20.
Genomics ; 57(3): 352-64, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10329001

ABSTRACT

A cloning of hepatic cDNAs associated with the early phase of an acute, systemic inflammation was carried out by differential screening of arrayed cDNA clones from rat livers obtained at 4-8 h postchallenge with Freund's complete adjuvant. End sequencing of 174 selected clones provided three cDNA groups that coded for: (i) 23 known acute-phase proteins, (ii) 31 known proteins whose change in hepatic synthesis during an acute phase was so far unsuspected, and (iii) 36 novel proteins whose cDNAs were completely sequenced. For 16 proteins in the third group the hepatic mRNA could be detected and quantitated by Northern blot hybridization in Freund's adjuvant-challenged animals, and an extrahepatic expression in healthy animals was further investigated. Matching the open reading frames of the 36 novel proteins with general and specialized data libraries indicated the potential relationships of 16 of these proteins with known protein families/superfamilies and/or the presence of functional domains previously described in other proteins. Overall, our search for novel inflammation-associated proteins selected mostly known or as yet undescribed proteins with an intracellular or membrane location, which extends our knowledge of the proteins involved in the intracellular metabolism of hepatic cells during a systemic, acute-phase response. Finally, some of the cDNAs above allowed us to successfully identify hepatic mRNAs that are differentially expressed in acute vs chronic (polyarthritis) inflammatory conditions in rat.


Subject(s)
Acute-Phase Proteins/genetics , Inflammation/genetics , Liver/metabolism , Acute-Phase Proteins/immunology , Animals , Base Sequence , Blotting, Northern , Cloning, Molecular , DNA Probes , DNA, Complementary , Gene Expression , Genetic Markers , Inflammation/metabolism , Intracellular Fluid , Liver/immunology , Male , Molecular Sequence Data , Nucleic Acid Hybridization , RNA, Messenger , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...