Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Biomaterials ; 247: 119998, 2020 07.
Article in English | MEDLINE | ID: mdl-32251928

ABSTRACT

Many skeletal tissue regenerative strategies centre around the multifunctional properties of bone marrow derived stromal cells (BMSC) or mesenchymal stem/stromal cells (MSC)/bone marrow derived skeletal stem cells (SSC). Specific identification of these particular stem cells has been inconclusive. However, enriching these heterogeneous bone marrow cell populations with characterised skeletal progenitor markers has been a contributing factor in successful skeletal bone regeneration and repair strategies. In the current studies we have isolated, characterised and enriched ovine bone marrow mesenchymal stromal cells (oBMSCs) using a specific antibody, Stro-4, examined their multipotential differentiation capacity and, in translational studies combined Stro-4+ oBMSCs with a bovine extracellular matrix (bECM) hydrogel and a biocompatible melt electro-written medical-grade polycaprolactone scaffold, and tested their bone regenerative capacity in a small in vivo, highly vascularised, chick chorioallantoic membrane (CAM) model and a preclinical, critical-sized ovine segmental tibial defect model. Proliferation rates and CFU-F formation were similar between unselected and Stro-4+ oBMSCs. Col1A1, Col2A1, mSOX-9, PPARG gene expression were upregulated in respective osteogenic, chondrogenic and adipogenic culture conditions compared to basal conditions with no significant difference between Stro-4+ and unselected oBMSCs. In contrast, proteoglycan expression, alkaline phosphatase activity and adipogenesis were significantly upregulated in the Stro-4+ cells. Furthermore, with extended cultures, the oBMSCs had a predisposition to maintain a strong chondrogenic phenotype. In the CAM model Stro-4+ oBMSCs/bECM hydrogel was able to induce bone formation at a femur fracture site compared to bECM hydrogel and control blank defect alone. Translational studies in a critical-sized ovine tibial defect showed autograft samples contained significantly more bone, (4250.63 mm3, SD = 1485.57) than blank (1045.29 mm3, SD = 219.68) ECM-hydrogel (1152.58 mm3, SD = 191.95) and Stro-4+/ECM-hydrogel (1127.95 mm3, SD = 166.44) groups. Stro-4+ oBMSCs demonstrated a potential to aid bone repair in vitro and in a small in vivo bone defect model using select scaffolds. However, critically, translation to a large related preclinical model demonstrated the complexities of bringing small scale reported stem-cell material therapies to a clinically relevant model and thus facilitate progression to the clinic.


Subject(s)
Mesenchymal Stem Cells , Animals , Bone Marrow , Bone Marrow Cells , Cattle , Cell Differentiation , Cells, Cultured , Extracellular Matrix , Hydrogels , Osteogenesis , Polyesters , Sheep
2.
Osteoarthritis Cartilage ; 24(11): 1951-1960, 2016 11.
Article in English | MEDLINE | ID: mdl-27307355

ABSTRACT

OBJECTIVE: To examine the methylation profile of the nuclear factor (NF)-κB enhancer region at -5.8 kb of inducible nitric oxide synthase (iNOS) and the subsequent role in the induction of osteoarthritis (OA) via cell cycle regulation. METHODS: Percentage methylation was determined by pyrosequencing, gene expression by qRT-PCR and cell proliferation was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Transient transfections were induced to determine the effect of the NF-κB enhancer region on cell proliferation and the influence of DNA methylation. RESULTS: In vitro de-methylation with 5-aza-dC showed decreased levels of DNA methylation at CpG sites localised at -5.8 kb, which correlated with higher levels of iNOS expression. In vitro methylation of the NF-κB enhancer region at -5.8 kb increased the percentage of cells at G0/G1 cell cycle phase. Loss of methylation within this region correlated with, enhanced proliferation and increased number of cells at G2/M phase. OA chondrocytes demonstrated up-regulation of the G0/G1 cell cycle progression markers Cyclin D1 and CDK6 in contrast to control cells. We demonstrate the loss of methylation that occurs at specific CpG sites localised at the -5.8 kb NF-κB enhancer region of the iNOS gene in OA chondrocytes permits the binding of this transcription factor activating the expression of iNOS. This results in subsequent altered cell cycle regulation, altered proliferative phenotype and transmission of the pathogenic phenotype to daughter cells. CONCLUSIONS: This study indicates that inhibition of cell cycle progression by iNOS enhancer hypermethylation is capable of reducing pro-inflammatory responses via down-regulation of NF-κB with important therapeutic implications in OA.


Subject(s)
Osteoarthritis , Cell Cycle , Chondrocytes , Demethylation , Enhancer Elements, Genetic , Humans , NF-kappa B
3.
Osteoarthritis Cartilage ; 23(11): 1946-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26521741

ABSTRACT

OBJECTIVE: To determine whether altered IL8 methylation status is associated with increased expression of IL8 in human osteoarthritic (OA) chondrocytes. METHODS: IL8 expression levels and the percentage CpG methylation in human chondrocytes were quantified by qRT-PCR and pyrosequencing in OA patients and in non-OA osteoporotic controls. The effect of CpG methylation on IL8 promoter activity was determined using a CpG-free vector; co-transfections with expression vectors encoding nuclear factor-kappa B (NF-κB), AP-1 and C/EBP were subsequently undertaken to analyse for IL8 promoter activity in response to changes in methylation status. RESULTS: IL8 expression in OA patients was 37-fold higher than in osteoporotic controls. Three CpG sites in the IL8 promoter were significantly demethylated in OA patients. Multiple regression analysis revealed that the degree of methylation of the CpG site located at -116-bp was the strongest predictor of IL8 expression. In vitro DNA methylation was noted to decrease IL8 promoter basal activity. Furthermore, NF-κB, AP-1 and C/EBP strongly enhanced IL8 promoter activity whilst DNA methylation inhibited the effects of these three transcription factors. CONCLUSIONS: The present study demonstrates the key role of DNA methylation status on the expression of IL8 in human chondrocytes. We demonstrate a quantitative relationship between percentage methylation and gene expression within clinical samples. These studies provide direct evidence linking the activation of IL8, DNA demethylation and the induction of the OA process with important therapeutic implications therein for patients with this debilitating disease.


Subject(s)
Chemokines/genetics , Chondrocytes/metabolism , DNA/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation , Interleukin-8/genetics , Osteoarthritis/genetics , Adult , Aged , Cells, Cultured , Chemokines/biosynthesis , Chondrocytes/pathology , DNA Methylation , Female , Humans , Interleukin-8/biosynthesis , Male , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction
4.
Biochem Biophys Res Commun ; 405(3): 362-7, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21219853

ABSTRACT

OBJECTIVE: Idiopathic osteoarthritis is the most common form of osteoarthritis (OA) world-wide and remains the leading cause of disability and the associated socio-economic burden in an increasing aging population. Traditionally, OA has been viewed as a degenerative joint disease characterized by progressive destruction of the articular cartilage and changes in the subchondral bone culminating in joint failure. However, the etiology of OA is multifactorial involving genetic, mechanical and environmental factors. Treatment modalities include analgesia, joint injection with steroids or hyaluronic acid, oral supplements including glucosamine and chondroitin sulfate, as well as physiotherapy. Thus, there is significant interest in the discovery of disease modifying agents. One such agent, glucosamine (GlcN) is commonly prescribed even though the therapeutic efficacy and mechanism of action remain controversial. Inflammatory cytokines, including IL-1ß, and proteinases such as MMP-13 have been implicated in the pathogenesis and progression of OA together with an associated CpG demethylation in their promoters. We have investigated the potential of GlcN to modulate NF-kB activity and cytokine-induced abnormal gene expression in articular chondrocytes and, critically, whether this is associated with an epigenetic process. METHOD: Human chondrocytes were isolated from the articular cartilage of femoral heads, obtained with ethical permission, following fractured neck of femur surgery. Chondrocytes were cultured for 5 weeks in six separate groups; (i) control culture, (ii) cultured with a mixture of 2.5 ng/ml IL-1ß and 2.5 ng/ml oncostatin M (OSM), (iii) cultured with 2mM N-acetyl GlcN (Sigma-Aldrich), (iv) cultured with a mixture of 2.5 ng/ml IL-1ß, 2.5 ng/ml OSM and 2mM GlcN, (v) cultured with 1.0 µM BAY 11-7082 (BAY; NF-kB inhibitor: Calbiochem, Darmstadt, Germany) and, (vi) cultured with a mixture of 2.5 ng/ml IL-1ß, 2.5 ng/ml OSM and 1.0 µM BAY. The levels of IL1B and MMP13 mRNA were examined using qRT-PCR. The percentage DNA methylation in the CpG sites of the IL1ß and MMP13 proximal promoter were quantified by pyrosequencing. RESULT: IL1ß expression was enhanced over 580-fold in articular chondrocytes treated with IL-1ß and OSM. GlcN dramatically ameliorated the cytokine-induced expression by 4-fold. BAY alone increased IL1ß expression by 3-fold. In the presence of BAY, IL-1ß induced IL1B mRNA levels were decreased by 6-fold. The observed average percentage methylation of the -256 CpG site in the IL1ß promoter was 65% in control cultures and decreased to 36% in the presence of IL-1ß/OSM. GlcN and BAY alone had a negligible effect on the methylation status of the IL1B promoter. The cytokine-induced loss of methylation status in the IL1B promoter was ameliorated by both GlcN and BAY to 44% and 53%, respectively. IL-1ß/OSM treatment increased MMP13 mRNA levels independently of either GlcN or BAY and no change in the methylation status of the MMP13 promoter was observed. CONCLUSION: We demonstrate for the first time that GlcN and BAY can prevent cytokine-induced demethylation of a specific CpG site in the IL1ß promoter and this was associated with decreased expression of IL1ß. These studies provide a potential mechanism of action for OA disease modifying agents via NF-kB and, critically, demonstrate the need for further studies to elucidate the role that NF-kB may play in DNA demethylation in human chondrocytes.


Subject(s)
Chondrocytes/drug effects , Epigenesis, Genetic/drug effects , Glucosamine/pharmacology , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Osteoarthritis/metabolism , Sulfones/pharmacology , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/drug effects , Down-Regulation , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/genetics , Promoter Regions, Genetic/drug effects
5.
Ann Rheum Dis ; 64(3): 388-95, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15708893

ABSTRACT

OBJECTIVE: To investigate the effect of nitric oxide (NO) on mitochondrial activity and its relation with the apoptosis of human articular chondrocytes. MATERIALS AND METHODS: Mitochondrial function was evaluated by analysing respiratory chain enzyme complexes, citrate synthase (CS) activities, and mitochondrial membrane potential (Delta psi m). The activities of the mitochondrial respiratory chain (MRC) complexes (complex I: NADH CoQ(1) reductase, complex II: succinate dehydrogenase, complex III: ubiquinol cytochrome c reductase, complex IV: cytochrome c oxidase) and CS were measured in human articular chondrocytes isolated from normal cartilage. The Delta psi m was measured by 5,5',6,6'-tetracholoro-1,1',3,3'-tetraethylbenzimidazole carbocyanide iodide (JC-1) using flow cytometry. Apoptosis was analysed by flow cytometry. The mRNA expression of caspases was analysed by ribonuclease protection analysis and the detection of protein synthesis by western blotting. Sodium nitroprusside (SNP) was used as an NO compound donor. RESULTS: SNP at concentrations higher than 0.5 mmol/l for 24 hours induced cellular changes characteristic of apoptosis. SNP elicited mRNA expression of caspase-3 and caspase-7 and down regulated bcl-2 synthesis in a dose and time dependent manner. Furthermore, 0.5 mM SNP induced depolarisation of the mitochondrial membrane at 5, 12, and 24 hours. Analysis of the MRC showed that at 5 hours, 0.5 mM SNP reduced the activity of complex IV by 33%. The individual inhibition of mitochondrial complex IV with azide modified the Delta psi m and induced apoptosis. CONCLUSIONS: This study suggests that the effect of NO on chondrocyte survival is mediated by its effect on complex IV of the MRC.


Subject(s)
Cartilage, Articular/drug effects , Chondrocytes/drug effects , Mitochondria/physiology , Nitric Oxide/pharmacology , Adult , Aged , Apoptosis/drug effects , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cell Respiration/drug effects , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/metabolism , Dose-Response Relationship, Drug , Electron Transport/drug effects , Humans , Membrane Potentials/drug effects , Microscopy, Fluorescence , Middle Aged , Mitochondria/drug effects , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology
6.
Clin Exp Rheumatol ; 22(3): 307-12, 2004.
Article in English | MEDLINE | ID: mdl-15144124

ABSTRACT

OBJECTIVES: The intra-articular injection of hyaluronan (HA) was originally used in the treatment of osteoarthritis (OA) to increase the viscosity of synovial fluid. However, some findings suggest that the activity of HA cannot be solely explained by its biomechanical properties. The aim of this study was to analyze the in vitro biological effects of HA on human OA chondrocytes and the impact of its molecular weight (MW) on those effects. METHODS: Cells were isolated from cartilage obtained during joint replacement surgery in OA patients. The chondrocytes were cultured for 24 hours to detect prostaglandin E2 (PGE2) and for 48 hours to measure nitric oxide (NO), after which they were pre-incubated with HA and stimulated with interleukin-1 (IL-1) at 5 ng/ml. Two commercial HA preparations with different MWs were used: Hyalgan (500-730 kDa, HA, Bioibérica S.A.) and Synvisc (hylan of 6,000 kDa, Biomatrix Inc). NO was detected by the Greiss reaction and PGE2 was quantified by a commercial EIA in the supernatant. Apoptosis was induced by an NO donor (sodium nitroprusside, SNP) and the effect of HA on apoptosis was quantified by flow cytometry. RESULTS: Neither HA preparation studied had any effect on the basal production of NO or PGE2. However, the 500-730 kDa HA at 200 microg/ml reduced the synthesis of both IL-1-induced NO and PGE2 by 70% and 45% respectively. Furthermore both HA preparations at 200 microg/ml decreased the apoptosis induced by SNP, 500-730 kDa to 40% and 6,000 kDa to 36%. CONCLUSION: HA may induce biological effects in addition to acting as a viscoelastic substance. This study suggests that HA preparations are different due to differences in biological activity resulting from MW.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Hyaluronic Acid/pharmacology , Osteoarthritis , Adjuvants, Immunologic/antagonists & inhibitors , Adjuvants, Immunologic/chemistry , Apoptosis/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Drug Antagonism , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/chemistry , Interleukin-1/pharmacology , Molecular Weight , Nitric Oxide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...