Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Article in English | MEDLINE | ID: mdl-38871517

ABSTRACT

Osteoarthritis (OA) is the most common musculoskeletal disease, without any curative treatment. Obesity being the main modifiable risk factor for OA, much attention focused on the role of adipose tissues (AT). In addition to the involvement of visceral and subcutaneous AT via systemic ways, many arguments also highlight the involvement of local AT, present in joint tissues. Local AT include intra-articular AT (IAAT), which border the synovium, and bone marrow AT (BMAT) localized within marrow cavities in the bones. This review describes the known features and involvement of IAAT and BMAT in joint homeostasis and OA. Recent findings evidence that alteration in magnetic resonance imaging signal intensity of infrapatellar fat pad can be predictive of the development and progression of knee OA. IAAT and synovium are partners of the same functional unit; IAAT playing an early and pivotal role in synovial inflammation and fibrosis and OA pain. BMAT, whose functions have only recently begun to be studied, is in close functional interaction with its microenvironment. The volume and molecular profile of BMAT change according to the pathophysiological context, enabling fine regulation of haematopoiesis and bone metabolism. Although its role in OA has not yet been studied, the localization of BMAT, its functions and the importance of the bone remodelling processes that occur in OA argue in favour of a role for BMAT in OA.

2.
Nanoscale ; 14(24): 8691-8708, 2022 Jun 23.
Article in English | MEDLINE | ID: mdl-35673929

ABSTRACT

The extracellular matrix (ECM) of articular cartilage is a three-dimensional network mainly constituted of entangled collagen fibrils and interfibrillar aggrecan aggregates. During the development of osteoarthritis (OA), the most common musculoskeletal disorder, the ECM is subjected to a combination of chemical and structural changes that play a pivotal role in the initiation and the progress of the disease. While the molecular mechanisms involved in the pathological remodelling of the ECM are considered as decisive, they remain, however, not completely elucidated. Herein, we report a relevant way for unravelling the role and nature of OA progress on human cartilage tissues, in terms of chemical composition and morphological and mechanical properties at the level of supramolecular assemblies constituting the cartilage ECM. For this purpose, we used X-ray photoelectron spectroscopy (XPS), and developed an innovative methodological approach that provides the molecular composition of the ECM. Moreover, we used atomic force microscopy (AFM) to probe the tissues at the level of individual collagen fibrils, both imaging and force spectroscopy modes being explored to this end. Taken together, these nanoscale characterization studies reveal the existence of two stages in the OA progress. At the early stage, a marked increase in the aggrecan and collagen content is observed, reflecting the homeostatic chondrocyte activity that tends to repair the cartilage ECM. At the late stage, we observe a failed attempt to stabilize and/or restore the tissue, yielding significant degradation of the supramolecular assemblies. This suggests an imbalance in the chondrocyte activity that turns in favor of catabolic events. Chemical changes are also accompanied by ECM structural changes and stiffening. Interestingly, we showed the possibility to mimic the imbalanced activities of chondrocytes by applying enzymatic digestions of healthy cartilage, through the combined action of hyaluronidase and collagenase. This yields damage strictly analogous to that observed at high OA severity. These findings bring mechanistic insights leading to a better understanding of the mechanism by which OA is initiated and progresses in the cartilage ECM. They offer guidelines for the development of curative treatments, such as targeting the homeostatic balance of chondrocyte metabolism through the control of enzymatic reactions involved in catabolic processes.


Subject(s)
Cartilage, Articular , Osteoarthritis , Aggrecans/metabolism , Cartilage, Articular/pathology , Chondrocytes , Collagen/metabolism , Extracellular Matrix/metabolism , Humans , Osteoarthritis/pathology
3.
Int J Mol Sci ; 23(4)2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35216203

ABSTRACT

Osteoarthritis (OA) is a whole joint disease characterized by an important remodeling of the osteochondral junction. It includes cartilage mineralization due to chondrocyte hypertrophic differentiation and bone sclerosis. Here, we investigated whether gremlin-1 (Grem-1) and its BMP partners could be involved in the remodeling events of the osteochondral junction in OA. We found that Grem-1, BMP-2, and BMP-4 immunostaining was detected in chondrocytes from the deep layer of cartilage and in subchondral bone of knee OA patients, and was positively correlated with cartilage damage. ELISA assays showed that bone released more Grem-1 and BMP-4 than cartilage, which released more BMP-2. In vitro experiments evidenced that compression stimulated the expression and the release of Grem-1 and BMP-4 by osteoblasts. Grem-1 was also overexpressed during the prehypertrophic to hypertrophic differentiation of murine articular chondrocytes. Recombinant Grem-1 stimulated Mmp-3 and Mmp-13 expression in murine chondrocytes and osteoblasts, whereas recombinant BMP-4 stimulated the expression of genes associated with angiogenesis (Angptl4 and osteoclastogenesis (Rankl and Ccl2). In conclusion, Grem-1 and BMP-4, whose expression at the osteochondral junction increased with OA progression, may favor the pathological remodeling of the osteochondral junction by inducing a catabolic and tissue remodeling program in hypertrophic chondrocytes and osteoblasts.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Chondrocytes/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Osteoarthritis, Knee/metabolism , Osteoblasts/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Cartilage, Articular/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Chondrogenesis/physiology , Humans , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Inbred C57BL , Osteogenesis/physiology
4.
Int J Mol Sci ; 23(4)2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35216412

ABSTRACT

Classical treatments of shoulder instability are associated with recurrence. To determine whether the modification of the capsule properties may be an alternative procedure, the effect of crosslinking treatment on the structure and mechanical properties of diseased human shoulder capsules was investigated. Joint capsules harvested from patients during shoulder surgery (n = 5) were treated or not with UV and/or riboflavin (0.1%, 1.0% and 2.5%). The structure and the mechanical properties of the capsules were determined by atomic force microscopy. The effect of treatments on cell death was investigated. Collagen fibrils were well-aligned and adjacent to each other with a D-periodicity of 66.9 ± 3.2 nm and a diameter of 71.8 ± 15.4 nm in control untreated capsules. No effect of treatments was observed on the organization of the collagen fibrils nor on their intrinsic characteristics, including D-periodicity or their mean diameter. The treatments also did not induce cell death. In contrast, UV + 2.5% riboflavin induced capsule stiffness, as revealed by the increased Young's modulus values (p < 0.0001 for each patient). Our results showed that the crosslinking procedure changed the biomechanics of diseased capsules, while keeping their structural organisation unchanged at the single fibril level. The UV/riboflavin crosslinking procedure may be a promising way to preserve the functions of collagen-based tissues and tune their elasticity for clinically relevant treatments.


Subject(s)
Collagen/chemistry , Collagen/pharmacology , Cross-Linking Reagents/pharmacology , Shoulder Joint/drug effects , Shoulder/physiology , Biomechanical Phenomena/drug effects , Cross-Linking Reagents/chemistry , Elastic Modulus/drug effects , Elasticity/drug effects , Extracellular Matrix/drug effects , Humans , Joint Instability , Microscopy, Atomic Force/methods , Riboflavin/chemistry , Riboflavin/pharmacology , Ultraviolet Rays
5.
Nat Rev Rheumatol ; 17(9): 533-549, 2021 09.
Article in English | MEDLINE | ID: mdl-34316066

ABSTRACT

Osteoarthritis (OA) is a whole-joint disease characterized by subchondral bone perfusion abnormalities and neovascular invasion into the synovium and articular cartilage. In addition to local vascular disturbance, mounting evidence suggests a pivotal role for systemic vascular pathology in the aetiology of OA. This Review outlines the current understanding of the close relationship between high blood pressure (hypertension) and OA at the crossroads of epidemiology and molecular biology. As one of the most common comorbidities in patients with OA, hypertension can disrupt joint homeostasis both biophysically and biochemically. High blood pressure can increase intraosseous pressure and cause hypoxia, which in turn triggers subchondral bone and osteochondral junction remodelling. Furthermore, systemic activation of the renin-angiotensin and endothelin systems can affect the Wnt-ß-catenin signalling pathway locally to govern joint disease. The intimate relationship between hypertension and OA indicates that endothelium-targeted strategies, including re-purposed FDA-approved antihypertensive drugs, could be useful in the treatment of OA.


Subject(s)
Hypertension/complications , Osteoarthritis/complications , Animals , Bone and Bones/blood supply , Humans , Hypertension/etiology , Hypertension/metabolism , Joints/blood supply , Joints/metabolism , Joints/pathology , Models, Biological , Osteoarthritis/etiology , Osteoarthritis/metabolism , Synovial Membrane/blood supply
6.
Curr Opin Rheumatol ; 33(1): 84-93, 2021 01.
Article in English | MEDLINE | ID: mdl-33186248

ABSTRACT

PURPOSE OF REVIEW: Epidemiologic studies reveal that the link between obesity and osteoarthritis cannot be uniquely explained by overweight-associated mechanical overload. For this reason, much attention focuses on the endocrine activity of adipose tissues. In addition to the systemic role of visceral and subcutaneous adipose tissues, many arguments highlight the involvement of local adipose tissues in osteoarthritis. RECENT FINDINGS: Alteration in MRI signal intensity of the infrapatellar fat pad may predict both accelerated knee osteoarthritis and joint replacement. In this context, recent studies show that mesenchymal stromal cells could play a pivotal role in the pathological remodelling of intra-articular adipose tissues (IAATs) in osteoarthritis. In parallel, recent findings underline bone marrow adipose tissue as a major player in the control of the bone microenvironment, suggesting its possible role in osteoarthritis. SUMMARY: The recent description of adipose tissues of various phenotypes within an osteoarthritic joint allows us to evoke their direct involvement in the initiation and progression of the osteoarthritic process. We can expect in the near future the discovery of novel molecules targeting these tissues.


Subject(s)
Adipose Tissue/pathology , Osteoarthritis/pathology , Adipokines/metabolism , Adipose Tissue/diagnostic imaging , Bone and Bones/pathology , Disease Progression , Female , Humans , Knee Joint/pathology , Magnetic Resonance Imaging/methods , Male , Mesenchymal Stem Cells/pathology , Obesity/pathology , Osteoarthritis/physiopathology , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/physiopathology , Subcutaneous Fat/pathology
7.
Arthritis Res Ther ; 21(1): 252, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31775901

ABSTRACT

BACKGROUND: Intra-articular adipose tissues (IAATs) are involved in osteoarthritis (OA) pathophysiology. We hypothesize that mesenchymal cells residing in IAATs may account for the specific inflammatory and metabolic patterns in OA patients. METHODS: Adipocyte precursors (preadipocytes and dedifferentiated fat cells (DFATc)) from IAATs (infrapatellar and suprapatellar fat pads) and autologous subcutaneous adipose tissues (SCATs) were isolated from knee OA patients. The ability of these precursors to differentiate into adipocytes was assessed by oil red O staining after 14 days of culture in adipogenic medium. The gene expression of adipocyte-related transcription factors (C/EBP-α and PPAR-γ) and development-related factors (EN1 and SFRP2) were analyzed. The inflammatory pattern was assessed by RT-qPCR and ELISA (interleukin 6 (IL-6), IL-8, Cox2, and prostaglandin E2 (PGE2)) after a 24-h stimulation by IL-1ß (1 ng/mL) and by conditioned medium from OA synovium. RESULTS: IAAT preadipocytes displayed a significantly higher ability to differentiate into adipocytes and expressed significantly more C/EBP-α mRNA than SCAT preadipocytes. IAAT preadipocytes expressed significantly less EN-1 and SFRP2 mRNA than SCAT preadipocytes. Unstimulated IAAT preadipocytes displayed a less inflammatory pattern (IL-6, IL-8, and Cox2/PGE2) than SCAT preadipocytes. In contrast, the response of IAAT preadipocytes to an inflammatory stimulus (IL-1ß and conditioned media of OA synovium) was exacerbated compared to that of SCAT preadipocytes. Similar results were obtained with DFATc. CONCLUSION: IAAT adipocyte precursors from OA patients have a specific phenotype, which may account for the unique phenotype of OA IAATs. The exacerbated response of IAAT preadipocytes to inflammatory stimulation may contribute to OA pathophysiology.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Cartilage, Articular/metabolism , Osteoarthritis, Knee/metabolism , Adipocytes/cytology , Adipogenesis/genetics , Adipose Tissue/cytology , Aged , Aged, 80 and over , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Profiling/methods , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Phenotype , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism , Synovial Membrane/cytology , Synovial Membrane/metabolism
8.
Arthritis Res Ther ; 21(1): 18, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635030

ABSTRACT

BACKGROUND: Accumulation of advanced glycation end-products (AGEs) is involved in age-related osteoarthritis (OA). Glyoxalase (Glo)-1 is the main enzyme involved in the removal of AGE precursors, especially carboxymethyl-lysine (CML). We aimed to investigate the expression of several AGEs and Glo-1 in human OA cartilage and to study chondrocytic Glo-1 regulation by inflammation, mediated by interleukin (IL)-1ß. METHODS: Ex vivo, we quantified AGEs (pentosidine, CML, methylglyoxal-hydroimidazolone-1) in knee cartilage from 30 OA patients. Explants were also incubated with and without IL-1ß, and we assessed Glo-1 protein expression and enzymatic activity. In vitro, primary cultured murine chondrocytes were stimulated with increasing concentrations of IL-1ß to assess Glo-1 enzymatic activity and expression. To investigate the role of oxidative stress in the IL-1ß effect, cells were also treated with inhibitors of mitochondrial oxidative stress or nitric oxide synthase. RESULTS: Ex vivo, only the human cartilage CML content was correlated with patient age (r = 0.78, p = 0.0031). No statistically significant correlation was found between Glo-1 protein expression and enzymatic activity in human cartilage and patient age. We observed that cartilage explant stimulation with IL-1ß decreased Glo-1 protein expression and enzymatic activity. In vitro, we observed a dose-dependent decrease in Glo-1 mRNA, protein quantity, and enzymatic activity in response to IL-1ß in murine chondrocytes. Inhibitors of oxidative stress blunted this downregulation. CONCLUSION: Glo-1 is impaired by inflammation mediated by IL-1ß in chondrocytes through oxidative stress pathways and may explain age-dependent accumulation of the AGE CML in OA cartilage.


Subject(s)
Aging/metabolism , Glycation End Products, Advanced/metabolism , Inflammation Mediators/metabolism , Lactoylglutathione Lyase/biosynthesis , Osteoarthritis/metabolism , Age Factors , Aged , Aged, 80 and over , Aging/pathology , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Osteoarthritis/pathology
9.
J Biol Chem ; 292(35): 14505-14515, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28684418

ABSTRACT

Epidemiological findings support the hypothesis that type 2 diabetes mellitus (T2DM) is a risk factor for osteoarthritis (OA). Moreover, OA cartilage from patients with T2DM exhibits a greater response to inflammatory stress, but the molecular mechanism is unclear. To investigate whether the antioxidant defense system participates in this response, we examined here the expression of nuclear factor-erythroid 2-related factor (Nrf-2), a master antioxidant transcription factor, and of heme oxygenase-1 (HO-1), one of its main target genes, in OA cartilage from T2DM and non-T2DM patients as well as in murine chondrocytes exposed to high glucose (HG). Ex vivo experiments indicated that Nrf-2 and HO-1 expression is reduced in T2DM versus non-T2DM OA cartilage (0.57-fold Nrf-2 and 0.34-fold HO-1), and prostaglandin E2 (PGE2) release was increased in samples with low HO-1 expression. HG-exposed, IL-1ß-stimulated chondrocytes had lower Nrf-2 levels in vitro, particularly in the nuclear fraction, than chondrocytes exposed to normal glucose (NG). Accordingly, HO-1 levels were also decreased (0.49-fold) in these cells. The HO-1 inducer cobalt protoporphyrin IX more efficiently attenuated PGE2 and IL-6 release in HG+IL-1ß-treated cells than in NG+IL-1ß-treated cells. Greater reductions in HO-1 expression and increase in PGE2/IL-6 production were observed in HG+IL-1ß-stimulated chondrocytes from Nrf-2-/- mice than in chondrocytes from wild-type mice. We conclude that the Nrf-2/HO-1 axis is a critical pathway in the hyperglucidic-mediated dysregulation of chondrocytes. Impairments in this antioxidant system may explain the greater inflammatory responsiveness of OA cartilage from T2DM patients and may inform treatments of such patients.


Subject(s)
Chondrocytes/metabolism , Diabetes Mellitus, Type 2/complications , Gene Expression Regulation , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Osteoarthritis/metabolism , Oxidative Stress , Aged , Animals , Animals, Newborn , Cartilage, Articular/cytology , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/immunology , Chondrocytes/pathology , Female , Heme Oxygenase-1/genetics , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , NF-E2-Related Factor 2/genetics , Osteoarthritis/complications , Osteoarthritis/immunology , Osteoarthritis/pathology , Signal Transduction , Specific Pathogen-Free Organisms
10.
Ann Rheum Dis ; 76(6): 1142-1148, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28298375

ABSTRACT

OBJECTIVES: Compared with subcutaneous adipose tissue (SCAT), infrapatellar fat pad (IFP), the main knee intra-articular adipose tissue (IAAT), has an inflammatory phenotype in patients with osteoarthritis (OA). We phenotyped suprapatellar fat pad (SPFP) and hip acetabular fat pad (AFP), two other IAATs, to determinate the unique signature of IAATs compared with SCAT. METHODS: IFP, SPFP, AFP and autologous SCAT were obtained from patients with OA during total knee (n=38) or hip replacement (n=5). Fibrosis and adipocyte area were analysed by histology and vascularisation, leucocyte and mast cell infiltration were analysed by immunohistochemistry for von Willebrand factor, leucocytes and tryptase, respectively. Secretion of interleukin (IL)-6, IL-8 and prostaglandin E2 (PGE2) was assessed by ELISA. The mRNA expression of adipocyte-associated genes (ATGL, LPL, PPAR-γ, FABP4 and CD36) and developmental genes (SFRP2, HoxC9 and EN1) was determined. The inflammatory response of isolated fibroblast-like synoviocytes (FLS) to autologous IFP and SPFP conditioned media was examined. RESULTS: Fibrosis, vascularisation and leucocyte and mast cell infiltration were greater in IAATs than SCAT, and levels of IL-6, IL-8 and PGE2 were greater in all IAATs than SCAT. IFP and SPFP induced a similar inflammatory response to FLS. Adipocyte area was smaller in IAATs than SCAT. Adipocyte-associated and developmental genes showed a similar gene expression pattern in all IAATs, different from SCAT. CONCLUSIONS: IFP but also SPFP and AFP (gathered under the term 'IAAT') may play a deleterious role in OA by affecting joint homeostasis because of their inflammatory phenotype and their close interaction with synovium in the same functional unit.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/pathology , Hip Joint , Knee Joint , Osteoarthritis, Hip/metabolism , Osteoarthritis, Knee/metabolism , RNA, Messenger/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adolescent , Adult , Aged , CD36 Antigens/genetics , Culture Media, Conditioned/pharmacology , Dinoprostone/metabolism , Fatty Acid-Binding Proteins/genetics , Female , Gene Expression , Homeodomain Proteins/genetics , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipase/genetics , Lipoprotein Lipase/genetics , Male , Membrane Proteins/genetics , Middle Aged , Osteoarthritis, Hip/genetics , Osteoarthritis, Knee/genetics , PPAR gamma/genetics , Phenotype , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology , Synoviocytes/drug effects , Young Adult
11.
Sci Rep ; 6: 33527, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27640363

ABSTRACT

Polyphenols exert a large range of beneficial effects in the prevention of age-related diseases. We sought to determine whether an extract of olive and grape seed standardized according to hydroxytyrosol (HT) and procyanidins (PCy) content, exerts preventive anti-osteoathritic effects. To this aim, we evaluated whether the HT/PCy mix could (i) have in vitro anti-inflammatory and chondroprotective actions, (ii) exert anti-osteoarthritis effects in two post-traumatic animal models and (iii) retain its bioactivity after oral administration. Anti-inflammatory and chondroprotective actions of HT/PCy were tested on primary cultured rabbit chondrocytes stimulated by interleukin-1 beta (IL-1ß). The results showed that HT/PCy exerts anti-inflammatory and chondroprotective actions in vitro. The preventive effect of HT/PCy association was assessed in two animal models of post-traumatic OA in mice and rabbits. Diet supplementation with HT/PCy significantly decreased the severity of post-traumatic osteoarthritis in two complementary mice and rabbit models. The bioavailability and bioactivity was evaluated following gavage with HT/PCy in rabbits. Regular metabolites from HT/PCy extract were found in sera from rabbits following oral intake. Finally, sera from rabbits force-fed with HT/PCy conserved anti-IL-1ß effect, suggesting the bioactivity of this extract. To conclude, HT/PCy extract may be of clinical significance for the preventive treatment of osteoarthritis.


Subject(s)
Grape Seed Extract/administration & dosage , Grape Seed Extract/therapeutic use , Interleukin-1beta/metabolism , Olea/chemistry , Osteoarthritis/drug therapy , Osteoarthritis/prevention & control , Wounds and Injuries/complications , Administration, Oral , Animals , Anterior Cruciate Ligament/drug effects , Anterior Cruciate Ligament/surgery , Biflavonoids/pharmacology , Biflavonoids/therapeutic use , Catechin/pharmacology , Catechin/therapeutic use , Cyclooxygenase 2/metabolism , Diet , Dinoprostone/metabolism , Disease Models, Animal , Female , Grape Seed Extract/pharmacology , Male , Mass Spectrometry , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Metabolome , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis/blood , Osteoarthritis/etiology , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/therapeutic use , Proanthocyanidins/pharmacology , Proanthocyanidins/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits
12.
Arthritis Res Ther ; 16(4): R137, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24984954

ABSTRACT

INTRODUCTION: Our objective was to investigate whether a lack of frizzled-related protein B (FrzB), an extracellular antagonist of the Wnt signaling pathways, could enhance cartilage degradation by facilitating the expression, release and activation of matrix metalloproteinases (MMPs) by chondrocytes in response to tissue-damaging stimuli. METHODS: Cartilage explants from FrzB-/- and wild-type mice were challenged by excessive dynamic compression (0.5 Hz and 1 MPa for 6 hours). Load-induced glycosaminoglycan (GAG) release and MMP enzymatic activity were assessed. Interleukin-1ß (IL-1ß) (10, 100 and 1000 pg/mL for 24 hours) was used to stimulate primary cultures of articular chondrocytes from FrzB-/- and wild-type mice. The expression and release of MMP-3 and -13 were determined by RT-PCR, western blot and ELISA. The accumulation of ß-catenin was assessed by RT-PCR and western blot. RESULTS: Cartilage degradation, as revealed by a significant increase in GAG release (2.8-fold, P = 0.014) and MMP activity (4.5-fold, P = 0.014) by explants, was induced by an excessive load. Load-induced MMP activity appeared to be enhanced in FrzB-/- cartilage explants compared to wild-type (P = 0.17). IL-1ß dose-dependently induced Mmp-13 and -3 gene expression and protein release by cultured chondrocytes. IL-1ß-mediated increase in MMP-13 and -3 was slightly enhanced in FrzB-/- chondrocytes compared to wild-type (P = 0.05 and P = 0.10 at gene level, P = 0.17 and P = 0.10 at protein level, respectively). Analysis of Ctnn1b and Lef1 gene expression and ß-catenin accumulation at protein level suggests that the enhanced catabolic response of FrzB-/- chondrocytes to IL-1ß and load may be associated with an over-stimulation of the canonical Wnt/ß-catenin pathway. CONCLUSIONS: Our results suggest that FrzB may have a protective role on cartilage degradation and MMP induction in mouse chondrocytes by attenuating deleterious effects of the activation of the canonical Wnt/ß-catenin pathway.


Subject(s)
Chondrocytes/metabolism , Chondrocytes/pathology , Glycoproteins/metabolism , Matrix Metalloproteinases/metabolism , Animals , Cartilage, Articular/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Osteoarthritis/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Wnt Signaling Pathway/physiology
13.
Arthritis Rheumatol ; 66(8): 2165-74, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24719336

ABSTRACT

OBJECTIVE: The infrapatellar fat pad (IFP) of the knee joint has an inflammatory phenotype in osteoarthritis (OA). Its close proximity to the synovial membrane suggests that the IFP could be involved in the induction of OA synovitis. This study was undertaken to investigate the response of fibroblast-like synoviocytes (FLS) to autologous IFP and subcutaneous adipose tissue (SCAT) from patients with severe knee OA. METHODS: Samples of IFP, SCAT, and autologous synovial membrane tissue close to the IFP were harvested during surgery from 28 patients with end-stage knee OA. FLS from 14 patients were stimulated with autologous IFP- or SCAT-conditioned medium, and levels of messenger RNA (mRNA) expression and protein release of interleukin-6 (IL-6), IL-8, secretory phospholipase A2 (sPLA2 ), cytosolic PLA2 , cyclooxygenase 2 (COX-2), microsomal prostaglandin E synthase, prostaglandin E2 (PGE2 ), and matrix metalloproteinases (MMPs) 1, 3, 9, and 13 were evaluated. Both IFP- and SCAT-conditioned medium were evaluated by enzyme-linked immunosorbent assay for secretion of IL-6, soluble IL-6 receptor (sIL-6R), IL-8, tumor necrosis factor α (TNFα), PGE2 , IL-1ß, and interferon-γ. In addition, OA FLS were treated with PGE2 receptor antagonists to evaluate the contribution of IFP-derived PGE2 to the inflammatory response of FLS to the IFP. RESULTS: Stimulation of OA FLS with IFP-conditioned medium induced the mRNA expression and protein release of IL-6, IL-8, sPLA2 , COX-2, PGE2 , and MMPs 1, 3, 9, and 13. The extent of stimulation was consistently stronger with IFP-conditioned medium than with SCAT-conditioned medium. Moreover, secretion of IL-6, sIL-6R, IL-8, TNFα, and PGE2 was greater in IFP-conditioned medium than in SCAT-conditioned medium, especially PGE2 , whose secretion was 75-fold stronger in IFP-conditioned medium (P < 0.0001). PGE2 receptor antagonists dose-dependently inhibited the release of IL-6, IL-8, and PGE2 by IFP-stimulated FLS. CONCLUSION: This study showed that the IFP has a potential role in the induction of synovial inflammation in patients with severe knee OA. Furthermore, secretion of PGE2 by the IFP may be involved in the OA inflammatory process.


Subject(s)
Adipose Tissue/immunology , Inflammation/genetics , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/immunology , Synovial Membrane/cytology , Adipose Tissue/metabolism , Aged , Aged, 80 and over , Dinoprostone/immunology , Female , Humans , Male , Middle Aged , Patella , Phenotype
14.
Arthritis Res Ther ; 16(1): R38, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24479481

ABSTRACT

INTRODUCTION: Visfatin is an adipokine that may be involved in intertissular joint communication in osteoarthritis (OA). With a homodimeric conformation, it exerts nicotinamide phosphoribosyltransferase (Nampt) enzymatic activity, essential for nicotinamide adenine dinucleotide biosynthesis. We examined the tissular origin and conformation of visfatin/Nampt in human OA joints and investigated the role of visfatin/Nampt in chondrocytes and osteoblasts by studying Nampt enzymatic activity. METHODS: Synovium, cartilage and subchondral bone from human OA joints were used for protein extraction or incubated for 24 hours in serum-free media (conditioned media), and synovial fluid was obtained from OA patients. Visfatin/Nampt expression in tissular extracts and conditioned media was evaluated by western blot and enzyme-linked immunosorbent assay (ELISA), respectively. Nampt activity was assessed in OA synovium by colorimetric assay. Primary cultures of murine chondrocytes and osteoblasts were stimulated with visfatin/Nampt and pretreated or not with APO866, a pharmacologic inhibitor of Nampt activity. The effect on cytokines, chemokines, growth factors and hypertrophic markers expression was examined by quantitative reverse transcriptase polymerase chain reaction and/or ELISA. RESULTS: In tissular explants, conditioned media and synovial fluid, visfatin/Nampt was found as a homodimer, corresponding to the enzymatically active conformation. All human OA joint tissues released visfatin/Nampt (synovium: 628 ± 106 ng/g tissue; subchondral bone: 195 ± 26 ng/g tissue; cartilage: 152 ± 46 ng/g tissue), with significantly higher level for synovium (P <0.0005). Nampt activity was identified ex vivo in synovium. In vitro, visfatin/Nampt significantly induced the expression of interleukin 6, keratinocyte chemoattractant and monocyte chemoattractant protein 1 in chondrocytes and osteoblasts. APO866 decreased the mRNA and protein levels of these pro-inflammatory cytokines in the two cell types (up to 94% and 63% inhibition, respectively). Levels of growth factors (vascular endothelial growth factor, transforming growth factor ß) and hypertrophic genes were unchanged with treatment. CONCLUSION: Visfatin/Nampt is released by all human OA tissues in a dimeric enzymatically active conformation and mostly by the synovium, which displays Nampt activity. The Nampt activity of visfatin is involved in chondrocyte and osteoblast activation, so targeting this enzymatic activity to disrupt joint tissue interactions may be novel in OA therapy.


Subject(s)
Nicotinamide Phosphoribosyltransferase/metabolism , Osteoarthritis/metabolism , Animals , Blotting, Western , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/metabolism , Mice , Osteoblasts/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
15.
Arthritis Res Ther ; 16(1): R16, 2014 Jan 20.
Article in English | MEDLINE | ID: mdl-24438745

ABSTRACT

INTRODUCTION: Nerve growth factor (NGF) level is increased in osteoarthritis (OA) joints and is involved in pain associated with OA. Stimuli responsible for NGF stimulation in chondrocytes are unknown. We investigated whether mechanical stress and proinflammatory cytokines may influence NGF synthesis by chondrocytes. METHODS: Primary cultures of human OA chondrocytes, newborn mouse articular chondrocytes or cartilage explants were stimulated by increasing amounts of IL-1ß, prostaglandin E2 (PGE2), visfatin/nicotinamide phosphoribosyltransferase (NAMPT) or by cyclic mechanical compression (0.5 Hz, 1 MPa). Before stimulation, chondrocytes were pretreated with indomethacin, Apo866, a specific inhibitor of NAMPT enzymatic activity, or transfected by siRNA targeting visfatin/NAMPT. mRNA NGF levels were assessed by real-time quantitative PCR and NGF released into media was determined by ELISA. RESULTS: Unstimulated human and mouse articular chondrocytes expressed low levels of NGF (19.2 ± 8.7 pg/mL, 13.5 ± 1.0 pg/mL and 4.4 ± 0.8 pg/mL/mg tissue for human and mouse articular chondrocytes and costal explants, respectively). Mechanical stress induced NGF release in conditioned media. When stimulated by IL-1ß or visfatin/NAMPT, a proinflammatory adipokine produced by chondocytes in response to IL-1ß, a dose-dependent increase in NGF mRNA expression and NGF release in both human and mouse chondrocyte conditioned media was observed. Visfatin/NAMPT is also an intracellular enzyme acting as the rate-limiting enzyme of the generation of NAD. The expression of NGF induced by visfatin/NAMPT was inhibited by Apo866, whereas IL-1ß-mediated NGF expression was not modified by siRNA targeting visfatin/NAMPT. Interestingly, PGE2, which is produced by chondrocytes in response to IL-1ß and visfatin/NAMPT, did not stimulate NGF production. Consistently, indomethacin, a cyclooxygenase inhibitor, did not counteract IL-1ß-induced NGF production. CONCLUSIONS: These results show that mechanical stress, IL-1ß and extracellular visfatin/NAMPT, all stimulated the expression and release of NGF by chondrocytes and thus suggest that the overexpression of visfatin/NAMPT and IL-1ß in the OA joint and the increased mechanical loading of cartilage may mediate OA pain via the stimulation of NGF expression and release by chondrocytes.


Subject(s)
Chondrocytes/metabolism , Inflammation/metabolism , Nerve Growth Factor/biosynthesis , Osteoarthritis/metabolism , Animals , Cartilage, Articular/metabolism , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Osteoarthritis/complications , Pain/etiology , Pain/metabolism , Physical Stimulation , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Transfection
16.
Curr Rheumatol Rep ; 15(11): 375, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24072604

ABSTRACT

Osteoarthritis (OA) is a whole joint disease, in which thinning and disappearance of cartilage is a critical determinant in OA progression. The rupture of cartilage homeostasis whatever its cause (aging, genetic predisposition, trauma or metabolic disorder) induces profound phenotypic modifications of chondrocytes, which then promote the synthesis of a subset of factors that induce cartilage damage and target other joint tissues. Interestingly, among these factors are numerous components of the inflammatory pathways. Chondrocytes produce cytokines, chemokines, alarmins, prostanoids, and adipokines and express numerous cell surface receptors for cytokines and chemokines, as well as Toll-like receptors. These receptors activate intracellular signaling pathways involved in inflammatory and stress responses of chondrocytes in OA joints. This review focuses on mechanisms responsible for the maintenance of cartilage homeostasis and highlights the role of inflammatory processes in OA progression.


Subject(s)
Cartilage, Articular/physiopathology , Homeostasis/physiology , Inflammation/complications , Osteoarthritis/physiopathology , Adipokines/physiology , Cartilage, Articular/pathology , Chemokines/physiology , Humans , Inflammation/physiopathology , Mechanotransduction, Cellular/physiology , Osteoarthritis/etiology , Osteoarthritis/pathology , Signal Transduction/physiology , Toll-Like Receptors/physiology
17.
Front Biosci (Elite Ed) ; 5(3): 911-21, 2013 06 01.
Article in English | MEDLINE | ID: mdl-23747906

ABSTRACT

Aging tendons or chronic tendinopathy are frequent conditions responsible for handicap in middle-aged and aging populations. Current therapies fail to relieve handicap. Medical treatment can sometimes be efficient, but surgical procedures often fail to restore tendon function. Cell therapy with platelets, based on tendon histological modifications and the capacity of such tissue to respond to growth factors, is an ever-expanding field of clinical research. In the current review, we compare the histological properties of normal tendons, aging tendons and chronic tendinopathy. We explain the natural healing process of such tendons and the rationale for using, or not, autologous growth factors. We review current clinical studies exploring the effect of concentrated autologous growth factor injection in chronic lesions and attempt to explain why, to date, all clinical studies have demonstrated no effect of such therapies.


Subject(s)
Aging/physiology , Intercellular Signaling Peptides and Proteins/therapeutic use , Tendinopathy/drug therapy , Animals , Chronic Disease , Humans , Tendinopathy/physiopathology
18.
Arthritis Rheum ; 65(7): 1831-42, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23552998

ABSTRACT

OBJECTIVE: Mechanical stress plays an important role in cartilage degradation and subchondral bone remodeling in osteoarthritis (OA). The remodeling of the subchondral bone could initiate cartilage loss in OA through the interplay of bone and cartilage. The aim of this study was to identify soluble mediators released by loaded osteoblasts/osteocytes that could induce the release of catabolic factors by chondrocytes. METHODS: Murine osteoblasts/osteocytes were subjected to cyclic compression, and then conditioned medium from either compressed (CCM) or uncompressed (UCM) cells was used to stimulate mouse chondrocytes. Chondrocyte expression of matrix metalloproteinase 3 (MMP-3), MMP-13, type II collagen, and aggrecan was assessed by reverse transcription-polymerase chain reaction, Western blotting, and enzyme-linked immunosorbent assay. Soluble mediators released by compressed osteoblasts/osteocytes were identified using iTRAQ (isobaric tags for relative and absolute quantification), a differential secretome analysis. Subchondral bone and cartilage samples were isolated from OA patients, and culture medium conditioned with OA subchondral bone or cartilage was used to stimulate human chondrocytes. RESULTS: Stimulation of mouse chondrocytes with CCM strongly induced the messenger RNA (mRNA) expression and protein release of MMP-3 and MMP-13 and inhibited the mRNA expression of type II collagen and aggrecan. Differential secretome analysis revealed that 10 proteins were up-regulated in compressed osteoblasts/osteocytes. Among them, soluble 14-3-3∊ (s14-3-3∊) dose-dependently induced the release of catabolic factors by chondrocytes, mimicking the effects of cell compression. Addition of a 14-3-3∊ blocking antibody greatly attenuated the CCM-mediated induction of MMP-3 and MMP-13 expression. Furthermore, in human OA subchondral bone, s14-3-3∊ was strongly released, and in cultures of human OA chondrocytes, s14-3-3∊ stimulated MMP-3 expression. CONCLUSION: The results of this study identify s14-3-3∊ as a novel soluble mediator critical in the communication between subchondral bone and cartilage in OA. Thus, s14-3-3∊ may be a potential target for future therapeutic or prognostic applications in OA.


Subject(s)
14-3-3 Proteins/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osteoarthritis/metabolism , Osteoblasts/metabolism , Osteocytes/metabolism , Aggrecans/metabolism , Animals , Bone Remodeling/physiology , Cells, Cultured , Collagen Type II/metabolism , Humans , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3 , Mice , Stress, Mechanical
19.
Curr Opin Rheumatol ; 25(1): 114-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23090672

ABSTRACT

PURPOSE OF REVIEW: Obesity is one of the main risk factors of the incidence and prevalence of knee osteoarthritis. Recent epidemiological data showing an increased risk of hand osteoarthritis in obese patients opened the door to a role of systemic inflammatory mediators, adipokines, released by adipose tissue. RECENT FINDINGS: Recent experimental studies confirm the critical roles of adipokines in the pathophysiologic features of osteoarthritis, with an emphasis on a new member, chemerin. Animal models of diet-induced obesity show that overload cannot completely explain the aggravation of spontaneous or posttraumatic knee osteoarthritis. We now have data suggesting that some adipokines may be surrogate biomarkers for severity of osteoarthritis. SUMMARY: Preclinical studies targeting adipokines are now expected to provide new hope for patients with osteoarthritis, especially those with metabolic syndrome.


Subject(s)
Inflammation/complications , Obesity/complications , Osteoarthritis/etiology , Adipokines/blood , Adipokines/physiology , Animals , Arthritis, Experimental/etiology , Biomarkers/blood , Humans , Mice , Osteoarthritis/diagnosis , Osteoarthritis, Knee/etiology , Rabbits
20.
Arthritis Rheum ; 64(12): 3972-81, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22933232

ABSTRACT

OBJECTIVE: The main feature of osteoarthritis (OA) is degradation and loss of articular cartilage. Interleukin-1ß (IL-1ß) is thought to have a prominent role in shifting the metabolic balance toward degradation. IL-1ß is first synthesized as an inactive precursor that is cleaved to the secreted active form mainly in the "inflammasome," a complex of initiators (including NLRP3), adaptor molecule ASC, and caspase 1. The aim of this study was to clarify the roles of IL-1ß and the inflammasome in cartilage breakdown. METHODS: We assessed IL-1ß release by cartilage explants from 18 patients with OA. We also evaluated the lipopolysaccharide (LPS)-, IL-1α-, and tumor necrosis factor α (TNFα)-induced activity of matrix metalloproteinase 3 (MMP-3), MMP-9, and MMP-13 in NLRP3-knockout mice and wild-type mice and the inhibition of caspase 1 with Z-YVAD-FMK and the blockade of IL-1ß with IL-1 receptor antagonist (IL-1Ra). Cartilage explants from NLRP3-knockout mice and IL-1R type I (IL-1RI)-knockout mice were subjected to excessive dynamic compression (0.5 Hz, 1 MPa) to trigger degradation, followed by assessment of load-induced glycosaminoglycan (GAG) release and MMP enzymatic activity. RESULTS: Despite the expression of NLRP3, ASC, and caspase 1, OA cartilage was not able to produce active IL-1ß. LPS, IL-1α, and TNFα dose-dependently increased MMP-3, MMP-9, and MMP-13 activity in cultured chondrocytes and in NLRP3(-/-) chondrocytes, and this effect was not changed by inhibiting caspase 1 or IL-1ß. The load-induced increase in GAG release and MMP activity was not affected by knockout of NLRP3 or IL-1RI in cartilage explants. CONCLUSION: OA cartilage may be degraded independently of any inflammasome activity, which may explain, at least in part, the lack of effect of IL-1ß inhibitors observed in previous trials.


Subject(s)
Carrier Proteins/physiology , Cartilage, Articular/physiopathology , Inflammasomes/physiology , Osteoarthritis, Knee/physiopathology , Stress, Mechanical , Animals , Carrier Proteins/genetics , Caspase 1/metabolism , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Humans , Interleukin-1beta/physiology , Lipopolysaccharides/pharmacology , Matrix Metalloproteinases/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Osteoarthritis, Knee/metabolism , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...