Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Osteoarthritis Cartilage ; 23(11): 1879-89, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26521733

ABSTRACT

OBJECTIVE: Articular cartilage defects commonly result from traumatic injury and predispose to degenerative joint diseases. To test the hypothesis that aberrant healing responses and chronic inflammation lead to osteoarthritis (OA), we examined spatiotemporal changes in joint tissues after cartilage injury in murine knees. Since intra-articular injection of hyaluronan (HA) can attenuate injury-induced osteoarthritis in wild-type (WT) mice, we investigated a role for HA in the response to cartilage injury in mice lacking HA synthase 1 (Has1(-/-)). DESIGN: Femoral groove cartilage of WT and Has1(-/-) mice was debrided to generate a non-bleeding wound. Macroscopic imaging, histology, and gene expression were used to evaluate naïve, sham-operated, and injured joints. RESULTS: Acute responses (1-2 weeks) in injured joints from WT mice included synovial hyperplasia with HA deposition and joint-wide increases in expression of genes associated with inflammation, fibrosis, and extracellular matrix (ECM) production. By 4 weeks, some resurfacing of damaged cartilage occurred, and early cell responses were normalized. Cartilage damage in Has1(-/-) mice also induced early responses; however, at 4 weeks, inflammation and fibrosis genes remained elevated with widespread cartilage degeneration and fibrotic scarring in the synovium and joint capsule. CONCLUSIONS: We conclude that the ineffective repair of injured cartilage in Has1(-/-) joints can be at least partly explained by the markedly enhanced expression of particular genes in pathways linked to ECM turnover, IL-17/IL-6 cytokine signaling, and apoptosis. Notably, Has1 ablation does not alter gross HA content in the ECM, suggesting that HAS1 has a unique function in the metabolism of inflammatory HA matrices.


Subject(s)
Cartilage, Articular/pathology , Gene Expression Regulation , Glucuronosyltransferase/deficiency , Glucuronosyltransferase/genetics , Knee Joint/pathology , Osteoarthritis, Knee/enzymology , RNA/genetics , Animals , Cartilage, Articular/enzymology , Cartilage, Articular/injuries , Chronic Disease , Disease Models, Animal , Fibrosis/enzymology , Fibrosis/pathology , Glucuronosyltransferase/biosynthesis , Hyaluronan Synthases , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Knee Joint/enzymology , Male , Mice , Mice, Inbred C57BL , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Polymerase Chain Reaction
2.
Osteoarthritis Cartilage ; 23(11): 1939-45, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26521740

ABSTRACT

A major objective of this article is to examine the research implications of recently available genome-wide expression profiles of cartilage from human osteoarthritis (OA) joints. We propose that, when viewed in the light of extensive earlier work, this novel data provides a unique opportunity to reorient the design of experimental systems toward clinical relevance. Specifically, in the area of cartilage explant biology, this will require a fresh evaluation of existing paradigms, so as to optimize the choices of tissue source, cytokine/growth factor/nutrient addition, and biomechanical environment for discovery. Within this context, we firstly discuss the literature on the nature and role of potential catabolic mediators in OA pathology, including data from human OA cartilage, animal models of OA, and ex vivo studies. Secondly, due to the number and breadth of studies on IL-1ß in this area, a major focus of the article is a critical analysis of the design and interpretation of cartilage studies where IL-1ß has been used as a model cytokine. Thirdly, the article provides a data-driven perspective (including genome-wide analysis of clinical samples, studies on mutant mice, and clinical trials), which concludes that IL-1ß should be replaced by soluble mediators such as IL-17 or TGF-ß1, which are much more likely to mimic the disease in OA model systems. We also discuss the evidence that changes in early OA can be attributed to the activity of such soluble mediators, whereas late-stage disease results more from a chronic biomechanical effect on the matrix and cells of the remaining cartilage and on other local mediator-secreting cells. Lastly, an updated protocol for in vitro studies with cartilage explants and chondrocytes (including the use of specific gene expression arrays) is provided to motivate more disease-relevant studies on the interplay of cytokines, growth factors, and biomechanics on cellular behavior.


Subject(s)
Cartilage, Articular/metabolism , Gene Expression , Genome, Human , Inflammation Mediators/metabolism , Osteoarthritis , Animals , Biomechanical Phenomena , Cartilage, Articular/physiopathology , Chondrocytes/metabolism , Chondrocytes/pathology , DNA/genetics , Humans , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/physiopathology
3.
Osteoarthritis Cartilage ; 20(8): 887-95, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22609479

ABSTRACT

OBJECTIVE: The current study was undertaken to adapt Equilibrium Partitioning of an Ionic Contrast agent via microcomputed tomography (EPIC-µCT) to mouse articular cartilage (AC), which presents a particular challenge because it is thin (30 µm) and has a small volume (0.2-0.4 mm(3)), meaning there is only approximately 2-4 µg of chondroitin sulfate (CS) glycosaminoglycan per joint surface cartilage. DESIGN: Using 6 µm isotropic voxels and the negatively charged contrast agent ioxaglate (Hexabrix), we optimized contrast agent concentration and incubation time, assessed two methods of tissue preservation (formalin fixation and freezing), examined the effect of ex vivo chondroitinase ABC digestion on X-ray attenuation, assessed accuracy and precision, compared young and skeletally mature cartilage, and determined patterns of degradation in a murine cartilage damage model induced by treadmill running. RESULTS: The optimal concentration of the contrast agent was 15%, formalin fixation was preferred to freezing, and 2 h of incubation was needed to reach contrast agent equilibrium with formalin-fixed specimens. There was good agreement with histologic measurements of cartilage thickness, although µCT over-estimated thickness by 13% (5 µm) in 6-week-old mice. Enzymatic release of 0.8 µg of chondrotin sulfate (about 40% of the total) increased X-ray attenuation by 17%. There was a 15% increase in X-ray attenuation in 14-week-old mice compared to 6-week-old mice (P < 0.001) and this corresponded to 65% decrease in CS content at 14 weeks. The older mice also had reductions of 33% in cartilage thickness and 44% in cartilage volume (P < 0.001). Treadmill running induced a 16% decrease in cartilage thickness (P = 0.012) and a 12% increase in X-ray attenuation (P = 0.006) in 14-week-old mice. CONCLUSION: This technique enables non-destructive visualization and quantification of murine femoral AC in three dimensions with anatomic specificity and should prove to be a useful new tool in studying degeneration of cartilage in mouse models.


Subject(s)
Aging/pathology , Cartilage, Articular/diagnostic imaging , Running/injuries , X-Ray Microtomography/methods , Animals , Contrast Media , Femur/diagnostic imaging , Hindlimb/pathology , Ioxaglic Acid , Male , Mice , Mice, Inbred C57BL
4.
Osteoarthritis Cartilage ; 19(9): 1081-90, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21624477

ABSTRACT

OBJECTIVE: To review the literature on modulation of chondrocyte activities in the osteoarthritic joint, and to discuss these changes in relation to established hard and soft tissue repair paradigms, with an emphasis on transforming growth factor beta (TGFß1)-mediated signaling which can promote either a chondrogenic or fibrogenic phenotype. METHODS: Papers addressing the close relationship between repair in general, and the specific post-injury response of joint tissues are summarized. Different interpretations of the role of TGFß1 in the emergence of an "osteoarthritic" chondrocyte are compared and the phenotypic plasticity of "reparative" progenitor cells is examined. Lastly, emerging data on a central role for A-Disintegrin-And-Metalloproteinase-with-Thrombospondin-like-Sequences-5 (ADAMTS5) activity in modulating TGFß1 signaling through activin receptor-like kinase 1 (ALK1) and activin receptor-like kinase 5 (ALK5) pathways is discussed. RESULTS: The review illustrates how a transition from ALK5-mediated fibrogenic signaling to ALK1-mediated chondrogenic signaling in joint cells represents the critical transition from a non-reparative to a reparative cell phenotype. Data from cell and in vivo studies illustrates the mechanism by which ablation of ADAMTS5 activity allows the transition to reparative chondrogenesis. Multiple large gene expression studies of normal and osteoarthritis (OA) human cartilages (CAs) also support an important role for TGFß1-mediated pro-fibrogenic activities during disease progression. CONCLUSIONS: We conclude that progressive articular CA damage in post-injury OA results primarily from biomechanical, cell biologic and mediator changes that promote a fibroblastic phenotype in joint cells. Since ADAMTS5 and TGFß1 appear to control this process, agents which interfere with their activities may not only enhance endogenous CA repair in vivo, but also improve the properties of tissue-engineered CA for implantation.


Subject(s)
Cartilage, Articular/physiopathology , Osteoarthritis/physiopathology , Transforming Growth Factor beta1/metabolism , ADAM Proteins/metabolism , ADAMTS5 Protein , Activin Receptors/metabolism , Animals , Cartilage, Articular/injuries , Chondrocytes/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Osteoarthritis/etiology , Osteoarthritis/metabolism , Rats , Synovial Fluid/chemistry , Wounds and Injuries/complications
5.
Osteoarthritis Cartilage ; 18(11): 1477-86, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20692354

ABSTRACT

OBJECTIVE: To quantify the structural characteristics and nanomechanical properties of aggrecan produced by adult bone marrow stromal cells (BMSCs) in peptide hydrogel scaffolds and compare to aggrecan from adult articular cartilage. DESIGN: Adult equine BMSCs were encapsulated in 3D-peptide hydrogels and cultured for 21 days with TGF-ß1 to induce chondrogenic differentiation. BMSC-aggrecan was extracted and compared with aggrecan from age-matched adult equine articular cartilage. Single molecules of aggrecan were visualized by atomic force microscopy-based imaging and aggrecan nanomechanical stiffness was quantified by high resolution force microscopy. Population-averaged measures of aggrecan hydrodynamic size, core protein structures and CS sulfation compositions were determined by size-exclusion chromatography, Western analysis, and fluorescence-assisted carbohydrate electrophoresis (FACE). RESULTS: BMSC-aggrecan was primarily full-length while cartilage-aggrecan had many fragments. Single molecule measurements showed that core protein and GAG chains of BMSC-aggrecan were markedly longer than those of cartilage-aggrecan. Comparing full-length aggrecan of both species, BMSC-aggrecan had longer GAG chains, while the core protein trace lengths were similar. FACE analysis detected a ∼ 1:1 ratio of chondroitin-4-sulfate to chondroitin-6-sulfate in BMSC-GAG, a phenotype consistent with aggrecan from skeletally-immature cartilage. The nanomechanical stiffness of BMSC-aggrecan was demonstrably greater than that of cartilage-aggrecan at the same total sGAG (fixed charge) density. CONCLUSIONS: The higher proportion of full-length monomers, longer GAG chains and greater stiffness of the BMSC-aggrecan makes it biomechanically superior to adult cartilage-aggrecan. Aggrecan stiffness was not solely dependent on fixed charge density, but also on GAG molecular ultrastructure. These results support the use of adult BMSCs for cell-based cartilage repair.


Subject(s)
Aggrecans/chemistry , Aggrecans/ultrastructure , Cartilage, Articular/chemistry , Cartilage, Articular/ultrastructure , Tissue Engineering/methods , Aggrecans/biosynthesis , Animals , Biomechanical Phenomena , Blotting, Western , Bone Marrow Cells/cytology , Electrophoresis/methods , Glycosaminoglycans/chemistry , Glycosaminoglycans/ultrastructure , Horses , Microscopy, Atomic Force , Nanotechnology , Stromal Cells/cytology
6.
Matrix Biol ; 29(5): 427-38, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20153827

ABSTRACT

Our objective was to evaluate the age-dependent mechanical phenotype of bone marrow stromal cell- (BMSC-) and chondrocyte-produced cartilage-like neo-tissue and to elucidate the matrix-associated mechanisms which generate this phenotype. Cells from both immature (2-4 month-old foals) and skeletally-mature (2-5 year-old adults) mixed-breed horses were isolated from animal-matched bone marrow and cartilage tissue, encapsulated in self-assembling-peptide hydrogels, and cultured with and without TGF-beta1 supplementation. BMSCs and chondrocytes from both donor ages were encapsulated with high viability. BMSCs from both ages produced neo-tissue with higher mechanical stiffness than that produced by either young or adult chondrocytes. Young, but not adult, chondrocytes proliferated in response to TGF-beta1 while BMSCs from both age groups proliferated with TGF-beta1. Young chondrocytes stimulated by TGF-beta1 accumulated ECM with 10-fold higher sulfated-glycosaminoglycan content than adult chondrocytes and 2-3-fold higher than BMSCs of either age. The opposite trend was observed for hydroxyproline content, with BMSCs accumulating 2-3-fold more than chondrocytes, independent of age. Size-exclusion chromatography of extracted proteoglycans showed that an aggrecan-like peak was the predominant sulfated proteoglycan for all cell types. Direct measurement of aggrecan core protein length and chondroitin sulfate chain length by single molecule atomic force microscopy imaging revealed that, independent of age, BMSCs produced longer core protein and longer chondroitin sulfate chains, and fewer short core protein molecules than chondrocytes, suggesting that the BMSC-produced aggrecan has a phenotype more characteristic of young tissue than chondrocyte-produced aggrecan. Aggrecan ultrastructure, ECM composition, and cellular proliferation combine to suggest a mechanism by which BMSCs produce a superior cartilage-like neo-tissue than either young or adult chondrocytes.


Subject(s)
Aggrecans/biosynthesis , Bone Marrow Cells/physiology , Cartilage/physiology , Chondrocytes/physiology , Extracellular Matrix/physiology , Horses/physiology , Animals , Bone Marrow Cells/cytology , Cartilage/ultrastructure , Cell Survival/physiology , Chondrocytes/cytology , Chromatography, Gel , Extracellular Matrix/ultrastructure , Hydrogels/pharmacology , Hydroxyproline/physiology , Male , Microscopy, Atomic Force , Stress, Mechanical , Tissue Engineering/methods , Transforming Growth Factor beta/pharmacology
7.
Osteoarthritis Cartilage ; 18(3): 297-302, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19912983

ABSTRACT

OBJECTIVE: As part of the National Institutes of Health (NIH)-sponsored Glucosamine/Chondroitin sulfate Arthritis Intervention Trial (GAIT) our objective here was to examine (1) the pharmacokinetics (PK) of glucosamine (GlcN) and chondroitin sulfate (CS) when taken separately or in combination as a single dose in normal individuals (n=29) and (2) the PK of GlcN and CS when taken as a single dose after 3 months daily dosing with GlcN, CS or GlcN+CS, in patients with symptomatic knee pain (n=28). METHODS: The concentration of GlcN in the circulation was determined by established fluorophore-assisted carbohydrate electrophoresis (FACE) methods. The hydrodynamic size and disaccharide composition of CS chains in the circulation and dosage samples was determined by Superose 6 chromatography and FACE. RESULTS: We show that circulating levels of CS in human plasma are about 20 microg/ml. Most significantly, the endogenous concentration and CS disaccharide composition were not detectably altered by ingestion of CS, when the CS was taken alone or in combination with GlcN. On the other hand, the Cmax (single-dose study) and AUC values (multiple-dose study) for ingested GlcN were significantly reduced by combination dosing with CS, relative to GlcN dosing alone. CONCLUSIONS: We conclude that pain relief perceived following ingestion of CS probably does not depend on simultaneous or prior intake of GlcN. Further, such effects on joint pain, if present, probably do not result from ingested CS reaching the joint space but may result from changes in cellular activities in the gut lining or in the liver, where concentrations of ingested CS, or its breakdown products, could be substantially elevated following oral ingestion. Moreover, since combined dosing of GlcN with CS was found to reduce the plasma levels seen with GlcN dosing alone, any improved pain relief by combination dosing cannot be explained by higher circulating concentrations of GlcN.


Subject(s)
Arthralgia/metabolism , Chondroitin Sulfates/pharmacokinetics , Glucosamine/pharmacokinetics , Osteoarthritis/drug therapy , Administration, Oral , Adult , Chondroitin Sulfates/administration & dosage , Clinical Trials as Topic , Dose-Response Relationship, Drug , Drug Combinations , Drug Therapy, Combination , Female , Glucosamine/administration & dosage , Humans , Male , Middle Aged , Pain Measurement , Treatment Outcome , Young Adult
8.
Osteoarthritis Cartilage ; 18(1): 5-11, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19733270

ABSTRACT

OBJECTIVE: Oral glucosamine (GlcN) has been widely studied for its potential therapeutic benefits in alleviating the pain and disability of osteoarthritis (OA). Its popularity has grown despite ongoing controversy regarding its effectiveness vs placebo in clinical trials, and lack of information regarding possible mechanisms of action. Here, we review the state of knowledge concerning the biology of GlcN as it relates to OA, and discuss a framework for future research directions. METHODS: An editorial "narrative" review of peer-reviewed publications is organized into four topics (1) Chemistry and pharmacokinetics of GlcN salts (2) Biological effects of GlcN salts in vitro (3) Therapeutic effects of GlcN salts in animal models of OA and (4) GlcN salts in the treatment of clinical OA. RESULTS: Data reporting potent pleiotropic activities of GlcN in in vitro cell and explant cultures are discussed in the context of the established pharmacokinetic data in humans and animals. The available clinical trial data are discussed to place the patient in the context of controlled research on disease management. CONCLUSIONS: Future research to determine therapeutic mechanisms of GlcN salt preparations will require use of standardized and clinically relevant in vitro assay systems and in vivo animal models for testing, as well as development of new outcome measures for inflammation and pain pathways in human OA.


Subject(s)
Glucosamine/pharmacokinetics , Glucosamine/therapeutic use , Joints/drug effects , Osteoarthritis/drug therapy , Administration, Oral , Animals , Cattle , Dogs , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Horses , Humans , Pain/drug therapy , Rabbits , Rats
9.
Arch Biochem Biophys ; 489(1-2): 118-26, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19607802

ABSTRACT

We studied changes in chondrocyte gene expression, aggrecan degradation, and aggrecanase production and activity in normal and mechanically injured cartilage co-cultured with joint capsule tissue. Chondrocyte expression of 21 genes was measured at 1, 2, 4, 6, 12, and 24h after treatment; clustering analysis enabled identification of co-expression profiles. Aggrecan fragments retained in cartilage and released to medium and loss of cartilage sGAG were quantified. Increased expression of MMP-13 and ADAMTS4 clustered with effects of co-culture, while increased expression of ADAMTS5, MMP-3, TGF-beta, c-fos, c-jun clustered with cartilage injury. ADAMTS5 protein within cartilage (immunohistochemistry) increased following injury and with co-culture. Cartilage sGAG decreased over 16-days, most severely following injury plus co-culture. Cartilage aggrecan was cleaved at aggrecanase sites in the interglobular and C-terminal domains, resulting in loss of the G3 domain, especially after injury plus co-culture. Together, these results support the hypothesis that interactions between injured cartilage and other joint tissues are important in matrix catabolism after joint injury.


Subject(s)
ADAM Proteins/biosynthesis , Cartilage/injuries , Cartilage/metabolism , Chondrocytes/metabolism , Gene Expression Regulation , Joint Capsule/metabolism , Aggrecans/metabolism , Animals , Cartilage/pathology , Cattle , Chondrocytes/pathology , Coculture Techniques , Endopeptidases/metabolism , Joint Capsule/pathology , Matrix Metalloproteinase 13/biosynthesis , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , Time Factors , Transforming Growth Factor beta/biosynthesis
10.
Matrix Biol ; 28(1): 53-61, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19041721

ABSTRACT

The microstructure of keratan sulphate purified from the interglobular domain, the keratan sulphate-rich region and total aggrecan was compared using fluorophore-assisted-carbohydrate-electrophoresis. Keratan sulphate in the interglobular domain was substantially less sulphated than keratan sulphate elsewhere on aggrecan, based on the ratio of unsulphated: monosulphated disaccharides generated by endo-beta-galactosidase digestion, and the ratio of monosulphated: disulphated disaccharides generated by keratanase II digestion. The ratio of unsulphated: monosulphated: disulphated disaccharides was 1:4:5 for keratan sulphate from total aggrecan and the keratan sulphate-rich region, but only 1:0.9:0.8 for the interglobular domain. These results show that keratan sulphate in the interglobular domain of pig aggrecan has a microstructure that is distinct from keratan sulphate in the keratan sulphate-rich region.


Subject(s)
Aggrecans/chemistry , Keratan Sulfate/chemistry , Keratan Sulfate/metabolism , Swine , Aggrecans/isolation & purification , Amino Acid Sequence , Animals , Conserved Sequence , Humans , Molecular Sequence Data , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Protein Structure, Tertiary , Sequence Alignment
11.
Osteoarthritis Cartilage ; 16(11): 1343-55, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18524635

ABSTRACT

OBJECTIVE: We have examined the occurrence of the inflammation-associated inter-alpha-trypsin inhibitor (IalphaI) components, bikunin, heavy chain (HC)1 and HC2 in normal cartilage and osteoarthritis (OA) cartilage and synovial fluids. DESIGN/METHODS: Cartilage extracts from normal donors and late-stage OA patients, and synovial fluids from OA patients were studied by Western blot with multiple antibodies to bikunin, HC1 and HC2. Cell and matrix localization was determined by immunohistochemistry and mRNA by RT-PCR. RESULTS: Bikunin.chondroitin sulfate (CS) and IalphaI were abundant in OA cartilages, but virtually undetectable in normal. In both OA and normal cartilages, HCs were largely present in a novel C-terminally truncated 50-kDa form, with most, if not all of these being attached to CS on a proteoglycan other than bikunin. Synovial fluids from OA patients contained bikunin.CS and full-length (approximately 90 kDa) HCs linked to hyaluronan (HA) as HC.HA (SHAP.HA). Immunohistochemistry showed intracellular and cell-associated staining for bikunin and HCs, consistent with their synthesis by superficial zone chondrocytes. PCR on multiple human normal and OA cartilage samples detected transcripts for HC1 and HC2 but not for bikunin. In OA cartilages, immunostaining was predominantly matrix-associated, being most intense in regions with a pannus-like fibrotic overgrowth. CONCLUSION: The truncated structure of HCs, their attachment to a proteoglycan other than bikunin, PCR data and intracellular staining are all consistent with synthesis of HC1 and HC2 by human articular chondrocytes. The presence of bikunin.CS and IalphaI in OA cartilage, but not in normal, appears to be due to diffusional uptake and retention through fibrillated (but not deeply fissured) cartilage surfaces.


Subject(s)
Alpha-Globulins/biosynthesis , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osteoarthritis/metabolism , Proteoglycans/metabolism , Alpha-Globulins/chemistry , Blotting, Western , Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfates/chemistry , Humans , Hyaluronic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synovial Fluid
12.
Osteoarthritis Cartilage ; 15(11): 1318-25, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17543547

ABSTRACT

OBJECTIVES: (1) To investigate the effects of exercise and osteochondral (OC) injury on synovial fluid (SF) chondroitin sulfate (CS) and hyaluronan (HA) concentration and chain length, (2) to compare SF and cartilage CS data from joints with OC fragmentation, and (3) to compare SF CS and HA profiles with those seen in serum from the same horses. METHODS: Serum and SF were obtained from (1) normal horses after 8 weeks rest, (2) the same horses after 9 months treadmill training, and (3) horses with OC injury from racing. Articular cartilage was also collected from group 3 horses. Concentrations and chain lengths of CS and HA were determined by gel chromatography and fluorophore-assisted carbohydrate electrophoresis. RESULTS: SF CS peak chain length in the OC injury group increased significantly (18.7kDa) when compared to rested horses (11.6kDa), with exercise producing an intermediate chain length (15.6kDa). Cartilage and serum from the OC injury group had the abnormally long CS chains seen in SF from these horses. Total SF HA was significantly lower in the OC injury group compared to the rested group. Both the OC injury group and the exercised group had significant decreases in SF HA chain length compared to the rested group. CONCLUSIONS: Chain length of SF CS was increased by exercise and OC injury. Exercise resulted in a modest increase, whereas OC injury caused a marked increase. In contrast to CS, SF HA chain length was decreased by OC injury, and to a lesser extent by exercise. Chain length analysis of SF CS and HA may provide a useful tool for evaluation of joint health.


Subject(s)
Cartilage, Articular/metabolism , Chondroitin Sulfates/chemistry , Hyaluronic Acid/chemistry , Physical Conditioning, Animal/physiology , Synovial Fluid/physiology , Animals , Cartilage, Articular/injuries , Chondroitin Sulfates/blood , Chondroitin Sulfates/metabolism , Chromatography, Gel , Electrophoresis , Horses , Hyaluronic Acid/metabolism , Synovial Fluid/metabolism
13.
Osteoarthritis Cartilage ; 15(7): 719-34, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17360199

ABSTRACT

OBJECTIVE: Human osteoarthritis (OA) is characterized by aggrecanase-mediated depletion of cartilage aggrecan. We have examined the abundance, location and some biochemical properties of the six known aggrecanases (A disintegrin and metalloproteinase with thrombospondin-like motifs 1 (ADAMTS1) 4, 5, 8, 9 and 15) in normal and OA human cartilages. METHODS: Formalin-fixed, ethylenediamine tetraacetic acid (EDTA)-decalcified sections of full-depth cartilage from human OA tibial plateaus and normal control samples were studied by confocal imaging. Probes included specific antibodies to aggrecanases and two aggrecan epitopes, as well as biotinylated hyaluronan binding protein (HABP) for hyaluronan (HA) visualization. Cartilage extracts were analyzed by Western blot for the individual proteinases and aggrecan fragments. RESULTS: ADAMTS5 was present in association with cells throughout normal cartilage and was markedly increased in OA, particularly in clonal groups in the superficial and transitional zones, where it was predominantly co-localized with HA. Consistent with the confocal analysis, a high molecular weight complex of ADAMTS5 and HA was isolated from human OA cartilage by isotonic salt extraction and chromatography on Superose 6. The complex eluted with an apparent molecular size of about 2x10(6) and contained major ADAMTS5 forms of 150, 60, 40 and 30kDa. The yield of most forms on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) was markedly enhanced by prior digestion of the complex with either Streptomyces hyaluronidase or chondroitinase ABC. CONCLUSION: ADAMTS5 abundance and distribution in human OA cartilages is consistent with a central role for this enzyme in destructive aggrecanolysis. HA-dependent sequestration of ADAMTS5 in the pericellular matrix may be a mechanism for regulating the activity of this proteinase in human OA cartilage.


Subject(s)
ADAM Proteins/metabolism , Aggrecans/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Endopeptidases/metabolism , Hyaluronic Acid/metabolism , Aged , Female , Humans , Male , Microscopy, Confocal/methods , Middle Aged , Osteoarthritis/metabolism
14.
Osteoarthritis Cartilage ; 14(4): 392-402, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16406703

ABSTRACT

OBJECTIVE: Aggrecan degradation by aggrecanases [a disintegrin and metalloproteinase with thrombospondin-like motifs (ADAMTS) 1, 4, 5, 8, 9, 15] is considered to initiate much of the cartilage pathology seen in human arthritis, however, the proteinase responsible and its mode of control is unclear. The present work was done to examine mechanisms of aggrecanase control in a novel murine epiphyseal cell system and to determine whether ADAMTS5 alone is responsible for aggrecanolysis by these cells. METHODS: Epiphyseal cells from 4-day-old mice (wild type, TS-5 (-/-), CD44(-/-), syndecan-1(-/-), membrane type-4 matrix metalloproteinase [MT4MMP(-/-)]) were maintained in non-adherent aggregate cultures and aggrecanolysis studied by biochemical and histochemical methods. Confocal immunolocalization analyses were done with specific probes for ADAMTS5, hyaluronan (HA) and aggrecanase-generated fragments of aggrecan. RESULTS: Aggrecanolysis by these cells was specifically aggrecanase-mediated and it occurred spontaneously without the need for addition of catabolic stimulators. Chondrocytes from ADAMTS5-null mice were aggrecanase-inactive whereas all other mutant cells behaved as wild type in this regard suggesting that ADAMTS5 activity is not controlled by CD44, syndecan-1 or MT4MMP in this system. Immunohistochemical analysis supported the central role for ADAMTS5 in the degradative pathway and indicated that aggrecanolysis occurs primarily in the HA-poor pericellular region in these cultures. CONCLUSION: These findings are consistent with published in vivo studies showing that single-gene ADAMTS5 ablation confers significant protection on cartilage in murine arthritis. We propose that this culture system and the analytical approaches described provide a valuable framework to further delineate the expression, activity and control of ADAMTS-mediated aggrecanolysis in human arthritis.


Subject(s)
Chondrocytes/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Extracellular Matrix Proteins/metabolism , Growth Plate/metabolism , Lectins, C-Type/metabolism , ADAM Proteins , Aggrecans , Animals , Growth Plate/pathology , Hyaluronan Receptors/metabolism , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases, Membrane-Associated , Membrane Glycoproteins/metabolism , Mice , Proteoglycans/metabolism , Syndecan-1 , Syndecans
15.
Osteoarthritis Cartilage ; 13(9): 828-36, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16006153

ABSTRACT

OBJECTIVE: To compare matrix composition and glycosaminoglycan (GAG) fine structure among five scaffolds commonly used for in vitro chondrocyte culture and cartilage tissue engineering. DESIGN: Bovine articular chondrocytes were seeded into agarose, alginate, collagen I, fibrin and polyglycolic acid (PGA) constructs and cultured for 20 or 40 days. In addition to construct DNA and sulfated GAG (sGAG) contents, the delta-disaccharide compositions of the chondroitin/dermatan sulfate GAGs were determined for each scaffold group via fluorophore-assisted carbohydrate electrophoresis (FACE). RESULTS: Significant differences were found in cell proliferation and extracellular matrix accumulation among the five scaffold groups. Significant cell proliferation was observed for all scaffold types but occurred later (20-40 days) in PGA constructs compared to the other groups (0-20 days). By 40 days, agarose constructs had the highest sGAG to DNA ratio, while alginate and collagen I had the lowest levels. Quantitative differences in the Delta-disaccharide composition of the GAGs accumulated in the different scaffolds were also found, with the most striking variations in unsulfated and disulfated delta-disaccharides. Agarose constructs had the highest fraction of disulfated residues and the lowest fraction of unsulfated residues, with a 6-sulfated/4-sulfated disaccharide ratio most similar to that of native articular cartilage. CONCLUSIONS: The similarities and differences among scaffolds in proteoglycan accumulation and GAG composition suggest that the scaffold material directly or indirectly influences chondrocyte proteoglycan metabolism and may have an influence on the quality of tissue engineered cartilage.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Extracellular Matrix/chemistry , Glycosaminoglycans/analysis , Proteoglycans/analysis , Tissue Engineering/methods , Animals , Biocompatible Materials , Cattle , Cell Culture Techniques , Chondrogenesis , Collagen Type II/analysis , DNA/analysis , Disaccharides/analysis , Electrophoresis/methods
16.
Osteoarthritis Cartilage ; 9 Suppl A: S16-22, 2001.
Article in English | MEDLINE | ID: mdl-11680680

ABSTRACT

OBJECTIVE: Quantitation and analyses of the fine structure of glycosaminoglycans are increasingly important for understanding many biological processes, including those most critical for understanding skeletal biology. We have developed a novel procedure, fluorophore-assisted carbohydrate electrophoresis (FACE), for determination of glycosaminoglycan fine structure and estimation of chain length. DESIGN: FACE utilizes enzymes that cleave glycosaminoglycans to create products, usually disaccharides, characteristic of the enzyme specificity. Each cleavage exposes a new reducing terminus that is fluorotagged by reductive amination with 2-aminoacridone. The tagged products are then displayed by electrophoresis, identified by their characteristic migration and chemistry, and quantitated by their molar fluorescence. RESULTS: Each class of glycosaminoglycan and the enzymes specific for each class are discussed. Specific application of the FACE technology is shown for analysis of the glycosaminoglycans on aggrecan isolated from knee cartilage of 5- and 68-year-old patients, and assessment of hyaluronan oligosaccharides. CONCLUSIONS: The FACE technology is a powerful tool for analysis of all four classes of glycosaminoglycans obtained from a wide variety of biologic sources. While the FACE protocols are relative simple, they provide a wealth of information including quantitation in the pmole range, determination of fine structure, and estimation of chain length.


Subject(s)
Cartilage, Articular/chemistry , Electrophoresis/methods , Glycosaminoglycans/analysis , Aged , Child, Preschool , Chondroitin Sulfates/analysis , Dermatan Sulfate/analysis , Heparin/analysis , Heparitin Sulfate/analysis , Humans , Hyaluronic Acid/analysis , Keratan Sulfate/analysis , Sensitivity and Specificity
17.
Glycobiology ; 11(10): 779-90, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11588154

ABSTRACT

Many tissues contain glycoproteins and proteoglycans, which are substituted with N-or O-linked keratan sulfate, a glycosaminoglycan in which the lactosamine (-galbeta1,4glcNAc-) disaccharide backbone is variably modified by sulfation, fucosylation, and sialylation. We report here a rapid, sensitive, and quantitative procedure for obtaining a complete disaccharide compositional analyses for keratan sulfates after FACE separation of products generated by hydrolysis of the glycosaminoglycans with B. fragillis keratanase II and E. freundii endo-beta-galactosidase. Seven digestion end products are separable in a single electrophoretic step using Monosaccharide composition gels. These are: the unsulfated disaccharide, glcNAcbeta1,3gal, the fucosylated trisaccharide, galbeta1,2[fucalpha1,3]glcNAc6S, the mono- and disulfated disaccharides, galbeta1,4glcNAc6S or gal6Sbeta1,4glcNAc6S from the chain interior, and the sialylated mono- and disulfated trisaccharides neuAalpha2,3galbeta1,4glcNAc6S or neuAalpha2,3gal6Sbeta1,4glcNAc6S from the nonreducing terminus. FACE analyses also revealed the presence of a contaminant beta-galactosidase activity in keratanase II enzyme preparations which cleaves the disaccharide, galbeta1,4glcNAc6S to its constituent monosaccharides, gal and glcNAc6S. It was particularly prominent at enzyme concentrations > 2 mU per nmole substrate glcNH(2) or after prolonged digestion times (> 12 h), and was not inhibitable by thiogalactosides or N-acetyl-lactosamine. As these monosaccharide products would not be detectable using the commonly described analytical methods for KS hydrolase products, such as (1)H-NMR and HPLC analyses, our data illustrate that the FACE procedure represents an improved approach for accurate compositional microanalyses of corneal and skeletal keratan sulfates, especially applicable to experimentation involving small amounts (1-2 microg) of this glycosaminoglycan.


Subject(s)
Acetylglucosaminidase/metabolism , Electrophoresis/methods , Glycoside Hydrolases , beta-Galactosidase/metabolism , Acetylglucosaminidase/chemistry , Adolescent , Aged , Animals , Carbohydrate Sequence , Cartilage, Articular/enzymology , Cattle , Child, Preschool , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Cornea/enzymology , Humans , Hydrolysis , Molecular Sequence Data , Muscle, Skeletal/enzymology , Nuclear Magnetic Resonance, Biomolecular , beta-Galactosidase/chemistry
18.
J Biol Chem ; 276(43): 39788-96, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11514545

ABSTRACT

The content and fine structure of keratan and chondroitin/dermatan sulfate in normal human corneas and corneas affected by macular corneal dystrophies (MCD) types I and II were examined by fluorophore-assisted carbohydrate electrophoresis. Normal tissues (n = 11) contained 15 microg of keratan sulfate and 8 microg of chondroitin/dermatan sulfate per mg dry weight. Keratan sulfates consisted of approximately 4% unsulfated, 42% monosulfated, and 54% disulfated disaccharides with number of average chain lengths of approximately 14 disaccharides. Chondroitin/dermatan sulfates were significantly longer, approximately 40 disaccharides per chain, and consisted of approximately 64% unsulfated, 28% 4-sulfated, and 8% 6-sulfated disaccharides. The fine structural parameters were altered in all diseased tissues. Keratan sulfate chain size was reduced to 3-4 disaccharides; chain sulfation was absent in MCD type I corneas and cartilages, and sulfation of both GlcNAc and Gal was significantly reduced in MCD type II. Chondroitin/dermatan sulfate chain sizes were also decreased in all diseased corneas to approximately 15 disaccharides, and the contents of 4- and 6-sulfated disaccharides were proportionally increased. Tissue concentrations (nanomole of chains per mg dry weight) of all glycosaminoglycan types were affected in the disease types. Keratan sulfate chain concentrations were reduced by approximately 24 and approximately 75% in type I corneas and cartilages, respectively, and by approximately 50% in type II corneas. Conversely, chondroitin/dermatan sulfate chain concentrations were increased by 60-70% in types I and II corneas. Such changes imply a modified tissue content of individual proteoglycans and/or an altered efficiency of chain substitution on the core proteins. Together with the finding that hyaluronan, not normally present in healthy adult corneas, was also detected in both disease subtypes, the data support the conclusion that a wide range of keratocyte-specific proteoglycan and glycosaminoglycan remodeling processes are activated during degeneration of the stromal matrix in the macular corneal dystrophies.


Subject(s)
Cornea/chemistry , Corneal Dystrophies, Hereditary/pathology , Glycosaminoglycans/chemistry , Acetylglucosamine/analysis , Adult , Aged , Carbohydrate Sequence , Cartilage/chemistry , Chondroitin Sulfates/chemistry , Dermatan Sulfate/chemistry , Fucose/analysis , Glycopeptides/chemistry , Humans , Keratan Sulfate/chemistry , Middle Aged , Molecular Sequence Data , Oligosaccharides/chemistry
20.
J Orthop Res ; 19(2): 269-77, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11347701

ABSTRACT

The isolated mouse tail tendon fascicle, a functional and homogenous volume of tendon extracellular matrix, was utilized as an experimental system to examine the structure function relationships in tendon. Our previous work using this model system demonstrated relationships between mean collagen fibril diameter and fascicle mechanical properties in isolated tail tendon fascicles from three different groups of mice (3-week and 8-week control and 8-week Mov13 transgenic) K.A. Derwin, L.J. Soslowsky, J. Biomech. Eng. 121 (1999) 598-604. These groups of mice were chosen to obtain tendon tissues with varying collagen fibril structure and/or biochemistry, such that relationships with material properties could be investigated. To further investigate the molecular details of matrix composition and organization underlying tendon function, we report now on the preparation, characterization, and quantitation of fascicle PGs (proteoglycans) from these three groups. The chondroitin sulfate/dermatan sulfate (CS/DS)-substituted PGs, biglycan and decorin, which are the abundant proteoglycans of whole tendons, were also shown to be the predominant PGs in isolated fascicles. Furthermore, similar to the postnatal maturation changes in matrix composition previously reported for whole tendons, isolated fascicles from 8-week mice had lower CS/DS PG contents (both decorin and biglycan) and a higher collagen content than 3-week mice. In addition, CS/DS chains substituted on PGs from 8-week fascicles were shorter (based on a number average) and richer in disulfated disaccharide residues than chains from 3-week mice. Fascicles from 8-week Mov13 transgenic mice were found to contain similar amounts of total collagen and total CS/DS PG as age-matched controls, and CS/DS chain lengths and sulfation also appeared normal. However, both decorin and biglycan in Mov13 tissue migrated slightly faster on sodium dodecyl sulfate polyacrylamide gel electorphoresis (SDS-PAGE) than the corresponding species from 8-week control, and biglycan from the 8-week Mov 13 fascicles appeared to migrate as a more polydisperse band, suggesting the presence of a unique PG population in the transgenic tissue. These observations, together with our biomechanical data [Derwin and Soslowsky, 1999] suggest that compensatory pathways of extracellular matrix assembly and maturation may exist, and that tissue mechanical properties may not be simply determined by the contents of individual matrix components or collagen fibril size.


Subject(s)
Glycosaminoglycans/metabolism , Glycosaminoglycans/ultrastructure , Proteoglycans/metabolism , Proteoglycans/ultrastructure , Tendons/metabolism , Tendons/ultrastructure , Animals , Collagen/metabolism , Collagen Type I, alpha 1 Chain , Male , Mice , Mice, Inbred Strains , Tail
SELECTION OF CITATIONS
SEARCH DETAIL
...