Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Osteoarthritis Cartilage ; 25(12): 2108-2118, 2017 12.
Article in English | MEDLINE | ID: mdl-28919430

ABSTRACT

OBJECTIVE: Animal models recapitulating post-traumatic osteoarthritis (OA) suggest that subchondral bone (SCB) properties and remodeling may play major roles in disease initiation and progression. Thus, we investigated the role of SCB properties and its effects on load-induced OA progression by applying a tibial loading model on two distinct mouse strains treated with alendronate (ALN). DESIGN: Cyclic compression was applied to the left tibia of 26-week-old male C57Bl/6 (B6, low bone mass) and FVB (high bone mass) mice. Mice were treated with ALN (26 µg/kg/day) or vehicle (VEH) for loading durations of 1, 2, or 6 weeks. Changes in articular cartilage and subchondral and epiphyseal cancellous bone were analyzed using histology and microcomputed tomography. RESULTS: FVB mice exhibited thicker cartilage, a thicker SCB plate, and higher epiphyseal cancellous bone mass and tissue mineral density than B6 mice. Loading induced cartilage pathology, osteophyte formation, and SCB changes; however, lower initial SCB mass and stiffness in B6 mice did not attenuate load-induced OA severity compared to FVB mice. By contrast, FVB mice exhibited less cartilage damage, and slower-growing and less mature osteophytes. In B6 mice, inhibiting bone remodeling via ALN treatment exacerbated cartilage pathology after 6 weeks of loading, while in FVB mice, inhibiting bone remodeling protected limbs from load-induced cartilage loss. CONCLUSIONS: Intrinsically lower SCB properties were not associated with attenuated load-induced cartilage loss. However, inhibiting bone remodeling produced differential patterns of OA pathology in animals with low compared to high SCB properties, indicating that these factors do influence load-induced OA progression.


Subject(s)
Cancellous Bone/diagnostic imaging , Cartilage, Articular/diagnostic imaging , Osteoarthritis, Knee/diagnostic imaging , Tibia/diagnostic imaging , Weight-Bearing , Alendronate/pharmacology , Animals , Bone Density , Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Cancellous Bone/drug effects , Cancellous Bone/pathology , Cartilage, Articular/pathology , Disease Models, Animal , Epiphyses/diagnostic imaging , Epiphyses/pathology , Male , Mice , Mice, Inbred C57BL , Osteoarthritis, Knee/pathology , Osteophyte , Tibia/drug effects , Tibia/pathology , X-Ray Microtomography
2.
Osteoarthritis Cartilage ; 24(6): 1021-8, 2016 06.
Article in English | MEDLINE | ID: mdl-26746148

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) is the most common form of arthritis and a leading cause of disability. OA is characterized by articular chondrocyte deterioration, subchondral bone changes and debilitating pain. One strategy to promote cartilage regeneration and repair is to accelerate proliferation and matrix production of articular chondrocytes. We previously reported that the protein phosphatase Phlpp1 controls chondrocyte differentiation by regulating the activities of anabolic kinases. Here we examined the role of Phlpp1 in OA progression in a murine model. We also assessed PHLPP1 expression and promoter methylation. DESIGN: Knee joints of WT and Phlpp1(-/-) mice were surgically destabilized by transection of the medial meniscal ligament (DMM). Mice were assessed for signs of OA progression via radiographic and histological analyses, and pain assessment for mechanical hypersensitivity using the von Frey assay. Methylation of the PHLPP1 promoter and PHLPP1 expression were evaluated in human articular cartilage and chondrocyte cell lines. RESULTS: Following DMM surgeries, Phlpp1 deficient mice showed fewer signs of OA and cartilage degeneration. Mechanical allodynia associated with DMM surgeries was also attenuated in Phlpp1(-/-) mice. PHLPP1 was highly expressed in human articular cartilage from OA patients, but was undetectable in cartilage specimens from femoral neck fractures (FNFxs). Higher PHLPP1 levels correlated with less PHLPP1 promoter CpG methylation in cartilage from OA patients. Blocking cytosine methylation or treatment with inflammatory mediators enhanced PHLPP1 expression in human chondrocyte cell lines. CONCLUSION: Phlpp1 deficiency protects against OA progression while CpG demethylation and inflammatory cytokines promote PHLPP1 expression.


Subject(s)
Osteoarthritis/etiology , Animals , Cartilage, Articular , Chondrocytes , Demethylation , Disease Models, Animal , Humans , Inflammation , Mice , Nuclear Proteins , Phosphoprotein Phosphatases
3.
Osteoarthritis Cartilage ; 24(5): 892-901, 2016 May.
Article in English | MEDLINE | ID: mdl-26687824

ABSTRACT

OBJECTIVE: To determine the strain-induced signaling pathways involved in regulating the transactivation of the transcription regulator Cbp/p300 Interacting Transactivator with ED-rich tail 2 (CITED2) and downstream targets in chondrocytes. METHODS: Primary human chondrocytes or C28/I2 chondrocytic cells were subjected to various strain regimes. C57BL/6 mice were subjected to treadmill running. Loss-of-function was carried out using siRNA or inhibitors specific for targeted molecules. mRNA levels were assayed by RT-qPCR, and proteins by western blotting, immunofluorescence, and/or immunohistochemical staining. CITED2 promoter activity was assayed in chondrocytes using wild-type or mutant constructs. RESULTS: Cyclic strain at 5%, 1 Hz induced CITED2 expression and suppressed expression of matrix metalloproteinase (MMP)-1 and -13 at the messenger RNA (mRNA) and protein levels in human chondrocytes. Abolishing primary cilia through knockdown of intraflagellar transport protein (IFT88) attenuated CITED2 gene expression and decreased protein levels. Similar effects were observed with inhibitors of extracellular adenosine triphosphate (ATP) or P2 purinergic receptors, or antagonists of Ca(2+) signaling. Knockdown of IFT88 in articular chondrocytes in vivo diminished treadmill induced-CITED2 expression and upregulated MMPs. Knockdown of hypoxia-inducible factor (HIF)1α, specificity protein 1 (Sp1), or deletion of the shear stress response element (SSRE) in the CITED2 promoter limited cyclic strain-induced transactivation of CITED2. However, the strain induced-transactivation of CITED2 was abolished only on knockdown of HIF1α, Sp1, and SSRE or by loss-of-function of IFT88 or extracellular-signal-regulated kinases (ERK)1/2. CONCLUSIONS: CITED2 transactivation is a critical event in signaling generated by strain and transduced by primary cilia, extracellular ATP, P2 purinergic receptors, and Ca(2+) signaling. Strain-induced CITED2 transactivation requires HIF1α, Sp1, and an intact SSRE and leads to the downregulation of MMPs such as MMP-1 and MMP-13.


Subject(s)
Adenosine Triphosphate/physiology , Calcium Signaling/physiology , Chondrocytes/metabolism , Mechanotransduction, Cellular/physiology , Repressor Proteins/biosynthesis , Trans-Activators/biosynthesis , Animals , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/physiology , Cilia/metabolism , Down-Regulation , Humans , MAP Kinase Signaling System/physiology , Male , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 13/genetics , Mice, Inbred C57BL , Physical Exertion/physiology , RNA, Messenger/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Transcriptional Activation/physiology
4.
J Biol Regul Homeost Agents ; 29(4 Suppl): 1-14, 2015.
Article in English | MEDLINE | ID: mdl-26652486

ABSTRACT

The exact mechanisms controlling the development and progression of osteoarthritis have not yet been clarified. Our aim was to investigate new pathomechanisms, with an emphasis on novel molecular targets that might regulate human chondrocytes in osteoarthritis. As a model for studying cell survival and metabolism, C-28/I2 and T/C-28a4 human chondrocytes were grown in complete medium, in dex-tran-coated charcoal treated medium and in serum-free medium. Healthy and osteoarthritic human cartilage samples were obtained from discarded surgical material. Cell survival, PTEN, AKT, Beclin1, AMBRA, AMPK and glucose/triglyceride metabolism were evaluated by immunoblotting and spectro-photometric assays. Starvation and steroids depletion decreased cell survival concomitantly with PTEN elevation, repression of the PI3K/AKT signaling axis and autophagy activation. These experimental conditions promoted the accumulation of glucose, decreased levels of G6PDH and resulted in differen-tial expression of OXPHOS complexes. Furthermore, they induced the expression of AMPK, reduced triglyceride levels and increased lipase activity, which was accompanied by a change in chondrocytes toward a fibroblast-like morphology. In osteoarthritic human cartilage, increased PTEN, AMPK and autophagy reflected the chondrocyte responses observed during starvation and steroids depletion. In conclusion, we defined the metabolic phenotype of human chondrocytes, in which both starvation and steroids depletion induce the activation of PTEN, AMPK and autophagy signaling, concomitant with metabolic reprogramming. Our data may aid in the development of novel in vitro models for the discovery and design of drugs or nutraceuticals capable of ameliorating the course of osteoarthritis.

5.
Osteoarthritis Cartilage ; 22(4): 586-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24530659

ABSTRACT

OBJECTIVE: The development of chondrocytic cell lines has enabled the investigation of the role of cellular phenotype and mechanisms in articular cartilage biology and physiopathology of several rheumatic diseases. Among them, the T/C-28a2 cell line has become a common tool in cartilage research. Recent results from our group have revealed that primary human chondrocytes in tissue and in monolayer culture contain high levels of connexin 43 (Cx43) and are able to directly communicate through gap junction (GJ) channels. These results challenge the existing thesis of cartilage physiology, that chondrocytes do not have the capacity to physically communicate with each other. Established cell lines offer the advantage of convenience and uniformity; however, the establishment process may cause a disruption of GJ. This study was performed to investigate if T/C-28a2 cells contain Cx43 protein and form functional channels. METHODS: Cx43 was characterized by RT-qPCR, Western blotting, and immunohistochemistry (IHC). Electrophysiology experiments, Lucifer Yellow (LY) uptake, electroporation in situ and scrape loading assay were performed to test the functionality of GJs. RESULTS: T/C-28a2 cells express Cx43. Electrophysiology experiments and LY uptake confirmed the capacity of these cells to communicate through GJ channels, although these cells contain significant levels of active c-Src kinase, presumably due to their immortalization with the Simian Virus 40 large T antigen. The results were validated using primary chondrocytes (PC). CONCLUSIONS: These results reveal that the T/C-28a2 line may provide a useful in vitro model for the study of Cx43 function and cell communication to understand the physiology of chondrocytes and cartilage.


Subject(s)
Cartilage, Articular/metabolism , Cell Communication/physiology , Chondrocytes/metabolism , Connexin 43/metabolism , Gap Junctions/metabolism , Blotting, Western , CSK Tyrosine-Protein Kinase , Cartilage, Articular/cytology , Cell Communication/genetics , Cell Line , Chondrocytes/cytology , Connexin 43/genetics , Gap Junctions/genetics , Humans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Staining and Labeling/methods , src-Family Kinases/metabolism
6.
Osteoarthritis Cartilage ; 21(11): 1781-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23948978

ABSTRACT

OBJECTIVES: To clarify whether there is oxidative stress in Kashin-Beck disease (KBD) and if cartilage damage from reactive oxygen species (ROS) and oxidative stress mediate the chondral necrosis in articular cartilage of KBD. METHODS: We recruited 64 KBD patients, 46 healthy children from severely affected KBD regions, 81 healthy children from a non-severely affected KBD endemic regions, and 91 healthy control children from a non-KBD region. Ten patients with KBD from the non-severely affected KBD regions were included in the experiment. The 2,3-DAN fluorescence technique was used to test selenium in the hair and blood. The biochemical techniques used to test the indicators of oxidative stress included thiobarbituric acid reactive substances (TBARS) levels, and antioxidant enzyme activities in serum samples. Histochemical staining was used to detect proteoglycans in cartilage sections. The 4-hydroxy-2-nonenal (4-HNE) and 8-hydroxydeoxyguanisine (8-OHdG) were localized by immunohistochemistry. RESULTS: The levels of TBARS in serum were significantly increased in KBD children. The levels of antioxidants in serum were significantly higher in both KBD and normal children from KBD regions than in the normal children from non-KBD regions. The percentage of chondrocytes staining for 4-HNE and 8-OHdG in KBD patients was significantly higher than in controls. Staining for 4-HNE and 8-OHdG in KBD patients was prominent in all zones of articular cartilage, especially in the necrotic chondrocytes of the deep zone. CONCLUSION: KBD is an oxidative stress-related disease, and the oxidative stress in cartilage contributes to the pathology of cartilage damage in KBD.


Subject(s)
Antioxidants/metabolism , Kashin-Beck Disease/physiopathology , Oxidative Stress/physiology , 8-Hydroxy-2'-Deoxyguanosine , Aldehydes/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Case-Control Studies , Child , Child, Preschool , Chondrocytes/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Female , Hair/chemistry , Humans , Kashin-Beck Disease/metabolism , Lipid Peroxidation/physiology , Male , Proteoglycans/metabolism , Selenium/analysis , Selenium/blood , Thiobarbiturates/blood
8.
Osteoarthritis Cartilage ; 20(7): 791-5, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22484575

ABSTRACT

OBJECTIVE: The purpose of this case report was to investigate local immune mechanisms present during an acute inflammatory flare initiated by viscosupplementation with hylan G-F 20 in a patient with osteoarthritis (OA) and past meniscectomy. EXPERIMENTAL DESIGN: A patient with a history of bilateral OA and partial left knee meniscectomy, who had received three injections of hylan G-F 20, was diagnosed with an acute flare reaction in the left knee. Her chart was evaluated for clinical, radiological, and laboratory findings and for clinical follow-up. Histopathological synovial examination and real-time polymerase chain reaction (RT-PCR) for genes with major roles in local inflammation and enzyme-linked immunosorbent assays (ELISAs) for markers of complement activation and cytokines were performed. To study the impact of the inflammatory and immune features we compared the case patient with groups of three representative OA and three rheumatoid arthritis (RA) patients. RESULTS: The patient exhibited evidence of highly increased acute phase reactant C-reactive protein (CRP) in the blood. The pathological examination of the synovial membrane identified abundant fibrinous exudate with numerous particles of hyaluronan surrounded by a dense infiltrate of neutrophils and eosinophils. The synovium had moderate hypertrophy and sclerosis as well as an inflammatory infiltrate predominantly composed of T lymphocytes and macrophages with scattered perivascular eosinophils and neutrophils. Immunoperoxidase staining identified numerous deposits of C5b-9 in the fibrinous exudates and the synovial membrane of the patient. Similar findings were observed in the RA patients, whereas deposits were rare in OA synovial samples. In addition, both anaphylatoxin C5a and the terminal complement complex C5b-9 were present at high levels, comparable to those in RA patients. The levels of mRNA for interleukin-1 beta (IL-1ß), IL-6, and the neutrophil marker myeloperoxidase (MPO) were markedly increased compared to those in the RA and OA patients. CONCLUSIONS: This present study is indicative of a pseudo-septic acute inflammatory reaction in response to local accumulation of hylan G-F 20 with the activation of complement and local invasion of pro-inflammatory cells.


Subject(s)
Arthritis/chemically induced , Complement C5a/biosynthesis , Complement Membrane Attack Complex/biosynthesis , Hyaluronic Acid/analogs & derivatives , Viscosupplements/adverse effects , Acute Disease , Arthritis/immunology , Arthritis/pathology , Biocompatible Materials , C-Reactive Protein/metabolism , Drug Administration Schedule , Female , Humans , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/adverse effects , Injections, Intra-Articular , Knee Joint/immunology , Knee Joint/pathology , Middle Aged , Osteoarthritis, Knee/drug therapy , Viscosupplementation/adverse effects , Viscosupplementation/methods , Viscosupplements/administration & dosage
9.
Biochem Pharmacol ; 82(12): 1919-29, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21946086

ABSTRACT

Osteoarthritis is a highly prevalent and disabling disease for which we do not have a cure. The identification of suitable molecular targets is hindered by the lack of standardized, reproducible and convenient screening assays. Following extensive comparisons of a number of chondrocytic cell lines, culture conditions, and readouts, we have optimized an assay utilizing C-28/I2, a chondrocytic cell line cultured in high-density micromasses. Utilizing molecules with known effects on cartilage (e.g. IL-1ß, TGFß1, BMP-2), we have exploited this improved protocol to (i) evoke responses characteristic of primary chondrocytes; (ii) assess the pharmacodynamics of gene over-expression using non-viral expression vectors; (iii) establish the response profiles of known pharmacological treatments; and (iv) investigate their mechanisms of action. These data indicate that we have established a medium-throughput methodology for studying chondrocyte-specific cellular and molecular responses (from gene expression to rapid quantitative measurement of sulfated glycosaminoglycans by Alcian blue staining) that may enable the discovery of novel therapeutics for pharmacological modulation of chondrocyte activation in osteoarthritis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chondrocytes/cytology , Chondrocytes/drug effects , Naproxen/pharmacology , Prednisolone/pharmacology , Alcian Blue , Cartilage, Articular/cytology , Cell Line , Cell Proliferation , Chondrocytes/physiology , Gene Expression Regulation/physiology , Humans , Interleukin-1beta/pharmacology
10.
Osteoarthritis Cartilage ; 18(11): 1518-27, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20833252

ABSTRACT

OBJECTIVE: Transforming growth factor-ß (TGF-ß) plays a critical role in cartilage homeostasis and deregulation of its signalling is implicated in osteoarthritis (OA). TGF-ß isoforms signal through a pair of transmembrane serine/threonine kinases known as the type I and type II TGF-ß receptors. Endoglin is a TGF-ß co-receptor that binds TGF-ß with high affinity in the presence of the type II TGF-ß receptor. We have previously shown that endoglin is expressed in human chondrocytes and that it forms a complex with the TGF-ß signalling receptors. However, the functional significance of endoglin expression in chondrocytes is unknown. Our objective was to determine whether endoglin regulates TGF-ß/Smad signalling and extracellular matrix (ECM) production in human chondrocytes and whether its expression varies with chondrocyte differentiation state. METHOD: Endoglin function was determined by overexpression or antisense morpholino/siRNA knockdown of endoglin in human chondrocytes and measuring TGF-ß-induced Smad phosphorylation, transcriptional activity and ECM production. Alterations in endoglin expression levels were determined during subculture-induced dedifferentiation of human chondrocytes and in normal vs OA cartilage samples. RESULTS: Endoglin enhances TGF-ß1-induced Smad1/5 phosphorylation and inhibits TGF-ß1-induced Smad2 phosphorylation, Smad3-driven transcriptional activity and ECM production in human chondrocytes. In addition, the enhancing effect of endoglin siRNA knockdown on TGF-ß1-induced Smad3-driven transcription is reversed by ALK1 overexpression. Furthermore, endoglin levels are increased in chondrocytes following subculture-induced dedifferentiation and in OA cartilage as compared to normal cartilage. CONCLUSION: Together, our results suggest that endoglin regulates the balance between TGF-ß/ALK1/Smad1/5 and ALK5/Smad2/3 signalling and ECM production in human chondrocytes and that endoglin may represent a marker for chondrocyte phenotype.


Subject(s)
Antigens, CD/metabolism , Antigens, CD/pharmacology , Chondrocytes/metabolism , Extracellular Matrix/metabolism , Receptors, Cell Surface/metabolism , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/pharmacology , Blotting, Western , Cartilage, Articular/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chondrocytes/cytology , Endoglin , Gene Expression Regulation , Humans , Immunohistochemistry , Osteoarthritis/metabolism , Phosphorylation/drug effects
11.
Osteoarthritis Cartilage ; 18(2): 240-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19800998

ABSTRACT

OBJECTIVE: Sialic acids frequently occur at the terminal positions of glycoprotein N-glycans present at chondrocyte surfaces or in the cartilage matrix. Sialic acids are transferred to glycoproteins in either alpha-2,3 or alpha-2,6 linkage by specific sialyltransferases (SiaTs) and can potentially affect cell functions and cell-matrix interactions. The present study aimed to assess the relationship between the expression of the human chondrocyte phenotype and the sialylation of chondrocyte glycoprotein N-glycans. METHODS: The transcription of 5 SiaT was quantified using real-time Reverse transcription polymerase chain reaction (RT-PCR) assays. N-glycan analysis was performed using LC-ESI-MS. Primary human chondrocytes were cultured in monolayer or alginate beads and compared to the chondrocyte cell lines C-28/I2 and SW1353. In addition, effects of interleukin-1beta (IL-1beta) or tumour necrosis factor-alpha (TNF-alpha) on primary cells were assessed. RESULTS: Primary human chondrocytes predominantly express alpha-2,6-specific SiaTs and accordingly, alpha-2,6-linked sialic acid residues in glycoprotein N-glycans. In contrast, the preponderance of alpha-2,3-linked sialyl residues and, correspondingly, reduced levels of alpha-2,6-specific SiaTs are associated with the altered chondrocyte phenotype of C-28/I2 and SW1353 cells. Importantly, a considerable shift towards alpha-2,3-linked sialic acids and alpha-2,3-specific SiaT mRNA levels occurred in primary chondrocytes treated with IL-1beta or tumour necrosis factor-alpha (TNF-alpha). CONCLUSION: The expression of the differentiated chondrocyte phenotype is linked to the ratio of alpha-2,6- to alpha-2,3-linked sialic acids in chondrocyte glycoprotein N-glycans. A shift towards altered sialylation might contribute to impaired cell-matrix interactions in disease conditions.


Subject(s)
Chondrocytes/metabolism , Glycoproteins/chemistry , Sialyltransferases/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chondrocytes/drug effects , Cytokines/pharmacology , Gene Expression , Humans , Interleukin-1beta/pharmacology , Phenotype , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sialyltransferases/chemistry , Sialyltransferases/genetics , Tumor Necrosis Factor-alpha/pharmacology
12.
In Vitro Cell Dev Biol Anim ; 45(7): 351-60, 2009.
Article in English | MEDLINE | ID: mdl-19263178

ABSTRACT

In vitro studies using chondrocyte cell cultures have increased our understanding of cartilage physiology and the altered chondrocytic cell phenotype in joint diseases. Beside the use of primary cells isolated from cartilage specimens of donors, immortalized chondrocyte cell lines such as C-28/I2 and T/C-28a2 have facilitated reproducible and standardized experiments. Although carbohydrate structures appear of significance for cartilage function, the contribution of the chondrocyte glycocalyx to matrix assembly and alterations of the chondrocyte phenotype is poorly understood. Therefore, the present study aimed to evaluate the glycoprofile of primary human chondrocytes as well as of C-28/I2 and T/C-28a2 cells in culture. First, the chondrocytic phenotype of primary and immortalized cells was assessed using real-time reverse transcriptase polymerase chain reaction, immunofluorescence, and glycosaminoglycans staining. Then, a panel of lectins was selected to probe for a range of oligosaccharide sequences determining specific products of the O-glycosylation and N-glycosylation pathways. We found that differences in the molecular phenotype between primary chondrocytes and the immortalized chondrocyte cell models C-28/I2 and T/C-28a2 are reflected in the glycoprofile of the cells. In this regard, the glycocalyx of immortalized chondrocytes was characterized by reduced levels of high-mannose type and sialic acid-capped N-glycans as well as increased fucosylated O-glycosylation products. In summary, the present report emphasizes the glycophenotype as an integral part of the chondrocyte phenotype and points at a significant role of the glycophenotype in chondrocyte differentiation.


Subject(s)
Chondrocytes/metabolism , Lectins/metabolism , Cell Differentiation , Cell Line , Chondrocytes/cytology , Flow Cytometry , Glycocalyx/physiology , Glycosaminoglycans/metabolism , Humans , Models, Biological , Phenotype
13.
Ann Rheum Dis ; 67 Suppl 3: iii75-82, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19022820

ABSTRACT

In osteoarthritis (OA), adult articular chondrocytes undergo phenotypic modulation in response to alterations in the environment owing to mechanical injury and inflammation. These processes not only stimulate the production of enzymes that degrade the cartilage matrix but also inhibit repair. With the use of in vitro and in vivo models, new genes, not known previously to act in cartilage, have been identified and their roles in chondrocyte differentiation during development and in dysregulated chondrocyte function in OA have been examined. These new genes include growth arrest and DNA damage (GADD)45beta and the epithelial-specific ETS (ESE)-1 transcription factor, induced by bone morphogenetic protein (BMP)-2 and inflammatory cytokines, respectively. Both genes are induced by NF-kappaB, suppress COL2A1 and upregulate matrix meatalloproteinase-13 (MMP-13) expression. These genes have also been examined in mouse models of OA, in which discoidin domain receptor 2 is associated with MMP-13-mediated remodelling, in order to understand their roles in physiological cartilage homoeostasis and joint disease.


Subject(s)
Cartilage, Articular/metabolism , Cytokines/physiology , Inflammation Mediators/physiology , Osteoarthritis/metabolism , Antigens, Differentiation/physiology , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinase 13/physiology , Signal Transduction , Transcription, Genetic
14.
Osteoarthritis Cartilage ; 16(11): 1413-20, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18485748

ABSTRACT

OBJECTIVE: As we previously reported, ADAMTS-7 and ADAMTS-12, two members of ADAMTS (a disintegrin and metalloprotease with thrombospondin motifs) family, degrade cartilage oligomeric matrix protein (COMP) in vitro and are significantly induced in the cartilage and synovium of arthritic patients [Liu CJ, Kong W, Ilalov K, Yu S, Xu K, Prazak L, et al. ADAMTS-7: a metalloproteinase that directly binds to and degrades cartilage oligomeric matrix protein. FASEB J 2006;20(7):988-90; Liu CJ, Kong W, Xu K, Luan Y, Ilalov K, Sehgal B, et al. ADAMTS-12 associates with and degrades cartilage oligomeric matrix protein. J Biol Chem 2006;281(23):15800-8]. The purpose of this study was to determine (1) whether cleavage activity of ADAMTS-7 and ADAMTS-12 of COMP are associated with COMP degradation in osteoarthritis (OA); (2) whether alpha-2-macroglobulin (a(2)M) is a novel substrate for ADAMTS-7 and ADAMTS-12; and (3) whether a(2)M inhibits ADAMTS-7 or ADAMTS-12 cleavage of COMP. METHODS: An in vitro digestion assay was used to examine the degradation of COMP by ADAMTS-7 and ADAMTS-12 in the cartilage of OA patients; in cartilage explants incubated with tumor necrosis factor-alpha (TNF-alpha) or interleukin-1-beta (IL-1beta) with or without blocking antibodies; and in human chondrocytes treated with specific small interfering RNA (siRNA) to knockdown ADAMTS-7 or/and ADAMTS-12. Digestion of a(2)M by ADAMTS-7 and ADAMTS-12 in vitro and the inhibition of ADAMTS-7 or ADAMTS-12-mediated digestion of COMP by a(2)M were also analyzed. RESULTS: The molecular mass of the COMP fragments produced by either ADAMTS-7 or ADAMTS-12 were similar to those observed in OA patients. Specific blocking antibodies against ADAMTS-7 and ADAMTS-12 dramatically inhibited TNF-alpha- or IL-1beta-induced COMP degradation in the cultured cartilage explants. The suppression of ADAMTS-7 or ADAMTS-12 expression by siRNA silencing in the human chondrocytes also prevented TNF-alpha- or IL-1beta-induced COMP degradation. Both ADAMTS-7 and ADAMTS-12 were able to cleave a(2)M, giving rise to 180- and 105-kDa cleavage products, respectively. Furthermore, a(2)M inhibited both ADAMTS-7- and ADAMTS-12-mediated COMP degradation in a concentration (or dose)-dependent manner. CONCLUSION: Our observations demonstrate the importance of COMP degradation by ADAMTS-7 and ADAMTS-12 in vivo. Furthermore, a(2)M is a novel substrate for ADAMTS-7 and ADAMTS-12. More significantly, a(2)M represents the first endogenous inhibitor of ADAMTS-7 and ADAMTS-12.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Cartilage, Articular/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , alpha-Macroglobulins/physiology , ADAMTS Proteins , ADAMTS7 Protein , Adult , Blotting, Western , Cartilage Oligomeric Matrix Protein , Humans , Matrilin Proteins , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/metabolism
15.
Osteoarthritis Cartilage ; 16(10): 1263-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18396421

ABSTRACT

OBJECTIVE: The heterogeneity of biosynthesis in human-derived cartilage explants poses a challenge to its use in experiments. The aim of this study was to determine the consistency with which two consecutive measures of biosynthesis could be made in individual human articular cartilage explants using a dual proline radiolabeling protocol. METHODS: Full-thickness cartilage explants were harvested from young bovine or human (total knee replacement) tibial plateaus. Two consecutive measurements of biosynthesis were obtained by measuring (3)H-proline and (14)C-proline incorporation. Each sample's ratio of (14)C-/(3)H-proline incorporation was computed. For comparison to traditional experimental designs, the (14)C-proline incorporation ratio was computed for adjacent cartilage samples. The number of samples needed to observe a change in the proline incorporation ratio of 10, 20, and 50% was determined for both methods. RESULTS: The dual-label ratio was consistent across samples from the same plateau [95% confidence interval (CI): +/-20% (human) and +/-30% (bovine) of median]. Adjacent human sample pairs had much greater variability in their (14)C-proline incorporation (95% CI: +/-50% of median). Adjacent bovine sample pairs had CIs that were similar in magnitude to those for the dual-label approach. In the human plateaus, ratio changes of 10, 20 and 50% could be detected using dramatically fewer samples than the adjacent pair method. For bovine samples, the two methods required a similar number of samples per group. CONCLUSION: The consistency of the dual-label approach may overcome the difficulties in studying the effects of interventions on biosynthesis in human cartilage in vitro.


Subject(s)
Cartilage, Articular/metabolism , Isotope Labeling/methods , Proline/metabolism , Radioisotopes/metabolism , Aged , Animals , Cattle , Cells, Cultured , Female , Humans , Middle Aged , Predictive Value of Tests , Sensitivity and Specificity
16.
Osteoarthritis Cartilage ; 16(10): 1205-12, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18321735

ABSTRACT

OBJECTIVE: To compare the effects of glucosamine (GlcN), curcumin, and diacerein in immortalized human C-28/I2 chondrocytes at the cellular and the gene expression level. This study aimed to provide insights into the proposed beneficial effects of these agents and to assess the applicability of the C-28/I2 cell line as a model for the evaluation of chondroprotective action. METHODS: Interleukin-1beta (IL-1beta)-stimulated C-28/I2 cells were cultured in the presence of GlcN, curcumin, and diacerein prior to the evaluation of parameters such as viability, morphology and proliferation. The impact of GlcN, curcumin, and diacerein on gene expression was determined using quantitative real-time RT-PCR (qPCR). RESULTS: At the transcriptional level, 5 mM GlcN and 50 microM diacerein increased the expression of cartilage-specific genes such as aggrecan (AGC) and collagen type II (COL2), while reducing collagen type I (COL1) mRNA levels. Moreover, the IL-1beta-mediated shift in gene expression pattern was antagonized by GlcN and diacerein. These effects were associated with a significant reduction in cellular proliferation and the development of chondrocyte-specific cell morphology. In contrast, curcumin was not effective at lower concentrations but even damaged the cells at higher amounts. CONCLUSIONS: Both GlcN and diacerein promoted a differentiated chondrocytic phenotype of immortalized human C-28/I2 chondrocytes by altering proliferation, morphology, and COL2/COL1 mRNA ratios. Moreover, both agents antagonized inhibitory effects of IL-1beta by enhancing AGC and COL2 as well as by reducing COL1 mRNA levels.


Subject(s)
Anthraquinones/pharmacology , Chondrocytes/drug effects , Curcumin/pharmacology , Glucosamine/pharmacology , Osteoarthritis/metabolism , Protective Agents/pharmacology , Cell Proliferation/drug effects , Cells, Cultured/metabolism , Gene Expression/drug effects , Glucosamine/genetics , Humans , Interleukin-1beta/genetics , Models, Biological , Osteoarthritis/genetics , Polymerase Chain Reaction
17.
Osteoarthritis Cartilage ; 15(2): 155-62, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16949312

ABSTRACT

OBJECTIVE: Pleiotrophin (PTN) is a secreted heparin-binding peptide expressed in mesodermal and neuroectodermal cells during development, but rarely in adult tissues. In fetal and juvenile, but not in mature cartilage, PTN is abundant. Furthermore, PTN is re-expressed in chondrocytes in early stages of osteoarthritis (OA). Since little is known about the functions of PTN in cartilage, we investigated the occurrence of PTN receptors in human articular cartilage in situ and PTN effects on human primary and immortalized chondrocytes in vitro. METHODS: Receptor expression and regulation was monitored by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry. PTN effects and signal transduction were studied by electrophoretic mobility shift, Boyden chamber cell migration and proliferation assays, effects on gene expression by real time RT-PCR and that on nitric oxide (NO) by the Griess reaction. RESULTS: Of the putative PTN signaling receptors, immortalized and primary chondrocytes (pc) expressed the anaplastic lymphoma kinase (ALK), less the receptor-type protein tyrosine phosphatase zeta/beta (PTPzeta). ALK expression was upregulated upon ligand exposure. PTN stimulation activated the AP-1 (activator protein-1) transcription factor and altered gene expression. Prolonged stimulation induced PTN mRNA expression slightly, reduced vascular endothelial growth factor (VEGF) mRNA as well as NO production. Whereas mRNA expression of matrix metalloproteinases (MMPs) MMP-1 and MMP-13 was reduced, their inhibitors TIMP-1 and TIMP-2 were induced. Furthermore, PTN stimulated chondrocyte migration and proliferation. CONCLUSIONS: These results show that PTN is an autocrine growth factor in cartilage. We suggest that PTN may be involved in the clustering and proliferation of chondrocytes observed in the early stages of OA.


Subject(s)
Carrier Proteins/genetics , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Cytokines/genetics , Growth Substances/genetics , Osteoarthritis/metabolism , Signal Transduction , Animals , Cattle , Humans , Osteoarthritis/genetics
18.
J Control Release ; 117(1): 121-9, 2007 Jan 22.
Article in English | MEDLINE | ID: mdl-17126445

ABSTRACT

The present study was performed to evaluate the applicability of plant lectins as mediators of bioadhesion in cartilage research using human chondrocyte cell lines C-28/I2 and T/C-28a2. The bioadhesive properties of fluorescein-labelled lectins with different carbohydrate specificities were investigated by flow cytometry. Specificity of the lectin-cell interactions was ascertained by competitive inhibition using complementary carbohydrates. As compared to that of other lectins, the interaction between wheat germ agglutinin (WGA) and chondrocytic cells was characterised by remarkable cytoadhesion, adequate binding strength and a high degree of specificity for N-acetyl-glucosamine as contained in hyaluronan chains. We therefore suggest WGA to be a promising candidate for mediating bioadhesion to low-adhesive scaffolds in cartilage tissue engineering. Moreover, the WGA-association rate of C-28/I2 and T/C-28a2 cells was dependent on temperature indicating cellular uptake of membrane-bound WGA. Intracellular enrichment was confirmed by confocal microscopy. Equilibration of intracellular pH gradients with monensin resulted in the reversal of quenching effects indicating accumulation of WGA within acid compartments of chondrocytic cells. Thus, WGA might be internalised into chondrocytes together with hyaluronan via the CD44 receptor-mediated endocytosis pathway and accumulated within lysosomes. This physiological process could represent a feasible pathway to target WGA-functionalised drug delivery devices into chondrocytes.


Subject(s)
Cartilage/physiology , Chondrocytes/metabolism , Drug Delivery Systems , Tissue Engineering , Acetylglucosamine , Binding, Competitive/drug effects , Carbohydrates/chemistry , Cartilage/cytology , Cell Adhesion , Cell Line , Cell Survival , Flow Cytometry , Humans , Hyaluronic Acid/chemistry , Lectins , Microscopy, Confocal , Wheat Germ Agglutinins
20.
J Pathol ; 210(3): 315-24, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16981238

ABSTRACT

Bacterial arthritis is a progressive joint disease which includes rapid destruction of articular cartilage even after clearance of the causal factor. The resulting post-infectious arthropathy is mainly characterized by self-perpetuating joint destruction and extensive angiogenesis in the emerging pannus-like synovial membrane, but the underlying molecular mechanisms of the bacteria-initiated process remain incompletely understood. This study was conducted to elucidate the expression and regulation of angiogenic and cartilage-destructive vascular endothelial growth factor (VEGF) in septic arthritis. For that purpose, aspirates of synovial fluid from patients with pyogenic arthritis were examined for VEGF levels by ELISA. In vitro studies with primary and immortalized chondrocytes were performed to determine whether Gram-positive and Gram-negative bacteria induce VEGF expression, by using real-time RT-PCR, ELISA, and immunohistochemistry. Activation of the transcription factor AP-1 was assessed by EMSA experiments. The necessity of the Toll-like receptor-2 (TLR-2), ERK-1/-2, and AP-1 pathway for infectious VEGF induction in chondrocytes was examined by using specific blocking reagents. ELISA experiments revealed that aspirates of synovial fluid from patients with pyogenic arthritis contain elevated levels of VEGF. The in vitro results confirmed the transcriptional induction of VEGF in chondrocytes after bacterial challenge by real-time RT-PCR, ELISA, and immunohistochemistry. This activation was mediated by a TLR-2-, ERK-1/-2-, and AP-1-dependent pathway. The findings demonstrate the expression of Toll-like receptors on mesenchymal articular chondrocytes and reveal TLR-2-mediated VEGF induction in human chondrocytes after Gram-positive bacterial sensing. Since VEGF is a potent angiogenic and tissue remodelling factor, evidence that Toll-like receptors contribute to destructive arthropathy after microbial joint infection is provided. VEGF may be a therapeutic target in the future for the prevention of post-infectious cartilage degradation in articular joints.


Subject(s)
Arthritis, Infectious/metabolism , Cartilage, Articular/metabolism , Toll-Like Receptor 2/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cadaver , Cells, Cultured , Chondrocytes/metabolism , Culture Media , Humans , Immunohistochemistry/methods , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pseudomonas Infections/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/physiology , Staphylococcal Infections/metabolism , Synovial Fluid/metabolism , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...