Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Exp Orthop ; 6(1): 19, 2019 May 03.
Article in English | MEDLINE | ID: mdl-31053993

ABSTRACT

BACKGROUND: An early detection of Osteoarthritis is urgently needed and still not possible until today. The aim of the study was to assess whether molecular biomarkers of cartilage turnover are associated with longitudinal change in knee cartilage thickness during a 2 year period in individuals with increased risk of developing knee osteoarthritis. A secondary aim was to assess whether prior knee injury or subjective patient-reported outcomes at baseline (BL) were associated with articular cartilage changes. Nineteen volleyball players (mean age 46.5 ± 4.9 years, 47% male) with a 30-year history of regular high impact training were recruited. The serum biomarkers Cpropeptide of type II procollagen (CPII), cartilage oligomeric matrix protein (COMP), collagenase generated carboxy-terminal neoepitope of type II collagen (sC2C), cartilage intermediate layer protein 2 (CILP-2), and the urine biomarkers C-telopeptide of type II collagen (CTX-II) and collagenase-generated peptide(s) of type II collagen (C2C-HUSA) were assessed at BL and at 2 year follow up (FU). Femorotibial cartilage thinning, thickening and absolute thickness change between BL and FU was evaluated from magnetic resonance imaging. Subjective clinical status at BL was evaluated by the International Knee Documentation Committee Subjective Knee Form and the Short-Form 36 Physical Component Score. RESULTS: CILP-2 was significantly higher at FU and linearly associated with the absolute cartilage thickness change during the experimental period. Prior injury was a predictor of increased absolute cartilage thickness change. CONCLUSION: Measuring the change in the cartilage biomarker CILP-2 might be a valid and sensitive method to detect early development of knee osteoarthritis as CILP-2 appears to be related to cartilage thickness loss in certain individuals with increased risk of developing knee osteoarthritis. Prior knee injury may be predictive of increased articular cartilage thickness change.

2.
Am J Pathol ; 187(12): 2674-2685, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28919112

ABSTRACT

Muscle dysfunction is the most important modifiable mediating factor in primary osteoarthritis (OA) because properly contracting muscles are a key absorber of forces acting on a joint. However, the pathological features of disuse muscle atrophy in OA patients have been rarely studied. Vastus medialis muscles of 14 female patients with OA (age range, 69 to 86 years), largely immobile for 1 or more years, were obtained during arthroplastic surgery and analyzed histologically. These were compared with female patients without arthritis, two with patellar fracture and two with patellar subluxation. Areas occupied by myofibers and adipose tissue were quantified. Large numbers of myofibers were lost in the vastus medialis of OA patients. The loss of myofibers was a possible cause of the reduction in muscle strength of the operated on knee. These changes were significantly correlated with an increase in intramuscular ectopic adipose tissue, and not observed in knees of nonarthritic patients. Resident platelet-derived growth factor receptor α-positive mesenchymal progenitor cells contributed to ectopic adipogenesis in vastus medialis muscles of OA patients. The present study suggests that significant loss of myofibers and ectopic adipogenesis in vastus medialis muscles are common pathological features of advanced knee OA patients with long-term loss of mobility. These changes may be related to the loss of joint function in patients with knee OA.


Subject(s)
Adipose Tissue , Choristoma/pathology , Muscular Disorders, Atrophic/pathology , Osteoarthritis/complications , Quadriceps Muscle/pathology , Adipogenesis/physiology , Aged , Aged, 80 and over , Female , Humans , Muscular Disorders, Atrophic/etiology
3.
J Exp Orthop ; 4(1): 7, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28229430

ABSTRACT

BACKGROUND: This study aimed the feasibility to assess longitudinal changes in biomarkers of cartilage turnover and to determine their relationship with patient-rated outcomes over 2 years in volleyball athletes. METHODS: Thirty-seven athletes were studied: 18 adolescents (age 15.9 ± 0.64 years) in a 2-year intensive volleyball training program and 19 adult recreational volleyball players (age 46.5 ± 4.9 years). Blood and serum samples were taken at baseline (BL) and 2-year follow-up (FU). Subjects completed the International Knee Documentation Committee (IKDC) Subjective Knee Form and the Short-Form 36 (SF-36) at BL. RESULTS: Thirteen adolescents (72%) had open growth plates at BL (BL open adolescents), the rest had closed growth plates at BL (BL closed adolescents), and all but one adolescent had closed growth plates at FU as assessed by MRI. BL open and closed adolescents had greater levels of the cartilage degradation-based biomarkers 45 mer collagenase peptide of type II collagen (C2C-HUSA) and C-telopeptide of type II collagen (CTX-II) than adults. BL open adolescents showed decreases in C2CHUSA, collagen synthesis marker C-propeptide of type II procollagen (CPII), and CTXII, and adults showed increases in cartilage intermediate layer protein 2 (CILP-2) and C2C-HUSA. In adolescents, IKDC scores were correlated with CPII changes. In adults, SF-36 Physical Component Scores were correlated with cartilage oligomeric matrix protein (COMP) changes. CONCLUSION: Significant differences in biomarker levels over time show the feasibility to assess their changes. Greater levels of C2C-HUSA and CTX-II in adolescents than in adults may reflect increased cartilage turnover in response to higher joint loading. CPII and COMP may be more reflective of subjective patient outcomes. These biomarkers may thus be useful in assessing mechanical loading-induced cartilage changes, their associated symptoms, and Osteoarthritis risk in athletes.

4.
Int J Nanomedicine ; 11: 5883-5896, 2016.
Article in English | MEDLINE | ID: mdl-27877036

ABSTRACT

Combined individually tailored methods for diagnosis and therapy (theragnostics) could be beneficial in destructive diseases, such as rheumatoid arthritis. Nanoparticles are promising candidates for theragnostics due to their excellent biocompatibility. Nanoparticle modifications, such as improved surface coating, are in development to meet various requirements, although safety concerns mean that modified nanoparticles require further review before their use in medical applications is permitted. We have previously demonstrated that iron oxide nanoparticles with amino-polyvinyl alcohol (a-PVA) adsorbed on their surfaces have the unwanted effect of increasing human immune cell cytokine secretion. We hypothesized that this immune response was caused by free-floating PVA. The aim of the present study was to prevent unwanted immune reactions by further surface modification of the a-PVA nanoparticles. After cross-linking of PVA to nanoparticles to produce PVA-grafted nanoparticles, and reduction of their zeta potential, the effects on cell viability and cytokine secretion were analyzed. PVA-grafted nanoparticles still stimulated elevated cytokine secretion from human immune cells; however, this was inhibited after reduction of the zeta potential. In conclusion, covalent cross-linking of PVA to nanoparticles and adjustment of the surface charge rendered them nontoxic to immune cells, nonimmunogenic, and potentially suitable for use as theragnostic agents.


Subject(s)
Blood Cells/drug effects , Cell Survival/drug effects , Ferric Compounds/chemistry , Magnetite Nanoparticles/administration & dosage , Polyvinyl Alcohol/chemistry , Adsorption , Blood Cells/metabolism , Cytokines/metabolism , Flow Cytometry , Humans , Magnetite Nanoparticles/chemistry
5.
J Rheumatol ; 43(10): 1864-1870, 2016 10.
Article in English | MEDLINE | ID: mdl-27481905

ABSTRACT

OBJECTIVE: To evaluate the association of a sandwich assay for cartilage collagenase-mediated degradation, the C2C human urine sandwich assay (IB-C2C-HUSA), with early and late knee cartilage pathology and with progression of cartilage damage. METHODS: A population-based cohort with knee pain, age 40-79 years, was evaluated at baseline (n = 253) and after mean 3.3 years (n = 161). We evaluated the IB-C2C-HUSA and a related competitive inhibition assay (C2C). The C2C assay was applied to serum (sC2C) and urine (uC2C). Based on knee radiographs and magnetic resonance imaging (MRI), 3 subgroups [no cartilage pathology, preradiographic cartilage pathology, and radiographic osteoarthritis (ROA)] were evaluated cross-sectionally for association with biomarker levels. Longitudinally, we evaluated whether baseline assays predict subsequent progression of cartilage degeneration, defined by MRI cartilage loss. RESULTS: Cross-sectionally, statistically significant differences were seen in the 3 subgroups for IB-C2C-HUSA (p < 0.001), with the highest levels seen in ROA, and for sC2C (p = 0.023), while no differences were seen for uC2C (p = 0.501). Baseline IB-C2C-HUSA levels were higher in progressors vs nonprogressors (p = 0.003). In logistic regression analysis, only baseline IB-C2C-HUSA was associated with an increased risk of progression of cartilage damage (OR 1.78, 95% CI 1.03-3.09). CONCLUSION: The IB-C2C-HUSA degradation assay detects the generation of a pathology-related cartilage collagen peptide(s) that increase(s) with onset of degeneration of knee articular cartilage. The baseline values are associated with progression of cartilage degeneration over 3 subsequent years. This assay may have value in clinical OA trials. Further, it points to collagenase activity as a therapeutic target for controlling degeneration of articular cartilage.


Subject(s)
Cartilage, Articular/pathology , Collagen Type II/urine , Collagenases/urine , Osteoarthritis, Knee/pathology , Adult , Aged , Biomarkers/urine , Cartilage, Articular/diagnostic imaging , Disease Progression , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/urine , Tomography, X-Ray Computed
6.
Int J Rheumatol ; 2016: 6432867, 2016.
Article in English | MEDLINE | ID: mdl-28042296

ABSTRACT

This study reports the effects of the iron chelator deferoxamine (DFO) on collagen cleavage, inflammation, and chondrocyte hypertrophy in relation to energy metabolism-related gene expression in osteoarthritic (OA) articular cartilage. Full-depth explants of human OA knee articular cartilage from arthroplasty were cultured with exogenous DFO (1-50 µM). Type II collagen cleavage and phospho-adenosine monophosphate-activated protein kinase (pAMPK) concentrations were measured using ELISAs. Gene expression studies employed real-time PCR and included AMPK analyses in PBMCs. In OA explants collagen cleavage was frequently downregulated by 10-50 µM DFO. PCR analysis of 7 OA patient cartilages revealed that 10 µM DFO suppressed expression of MMP-1, MMP-13, IL-1ß, and TNFα and a marker of chondrocyte hypertrophy, COL10A1. No changes were observed in the expression of glycolysis-related genes. In contrast, expressions of genes associated with the mitochondrial Krebs cycle (TCA), AMPK, HIF1α, and COL2A1 were upregulated. AMPK gene expression was reduced in OA cartilage and increased in PBMCs from the same patients compared to healthy controls. Our studies demonstrate that DFO is capable of suppressing excessive collagenase-mediated type II collagen cleavage in OA cartilage and reversing phenotypic changes. The concomitant upregulation of proanabolic TCA-related gene expressions points to a potential for availability of energy generating substrates required for matrix repair by end-stage OA chondrocytes. This might normally be prevented by high whole-body energy requirements indicated by elevated AMPK expression in PBMCs of OA patients.

7.
Int J Nanomedicine ; 10: 3429-45, 2015.
Article in English | MEDLINE | ID: mdl-26056442

ABSTRACT

Nanotechnology provides new opportunities in human medicine, mainly for diagnostic and therapeutic purposes. The autoimmune disease rheumatoid arthritis (RA) is often diagnosed after irreversible joint structural damage has occurred. There is an urgent need for a very early diagnosis of RA, which can be achieved by more sensitive imaging methods. Superparamagnetic iron oxide nanoparticles (SPION) are already used in medicine and therefore represent a promising tool for early diagnosis of RA. The focus of our work was to investigate any potentially negative effects resulting from the interactions of newly developed amino-functionalized amino-polyvinyl alcohol coated (a-PVA) SPION (a-PVA-SPION), that are used for imaging, with human immune cells. We analyzed the influence of a-PVA-SPION with regard to cell survival and cell activation in human whole blood in general, and in human monocytes and macrophages representative of professional phagocytes, using flow cytometry, multiplex suspension array, and transmission electron microscopy. We found no effect of a-PVA-SPION on the viability of human immune cells, but cytokine secretion was affected. We further demonstrated that the percentage of viable macrophages increased on exposure to a-PVA-SPION. This effect was even stronger when a-PVA-SPION were added very early in the differentiation process. Additionally, transmission electron microscopy analysis revealed that both monocytes and macrophages are able to endocytose a-PVA-SPION. Our findings demonstrate an interaction between human immune cells and a-PVA-SPION which needs to be taken into account when considering the use of a-PVA-SPION in human medicine.


Subject(s)
Arthritis, Rheumatoid/blood , Magnetite Nanoparticles/chemistry , Polyvinyl Alcohol/chemistry , Cell Survival/drug effects , Cytokines/blood , Endocytosis/drug effects , Humans , Macrophages/drug effects , Magnetite Nanoparticles/adverse effects , Microscopy, Electron, Transmission , Monocytes/drug effects , Toxicity Tests/methods
9.
Arthritis ; 2013: 461486, 2013.
Article in English | MEDLINE | ID: mdl-23864948

ABSTRACT

The gene expression of mTOR, autophagy-related ULK1, caspase 3, CDK-inhibitor p21, and TNF α was measured in the peripheral blood of osteoarthritic (OA) patients at different stages of the disease aiming to establish a gene expression profile that might indicate the activity of the disease and joint destruction. Whole blood of 65 OA outpatients, 27 end-stage OA patients, 27 healthy volunteers, and knee articular cartilages of 28 end-stage OA patients and 26 healthy subjects were examined. OA outpatients were subjected to clinical testing, ultrasonography, and radiographic and WOMAC scoring. Protein levels of p70-S6K, p21, and caspase 3 were quantified by ELISA. Gene expression was measured using real-time RT-PCR. Upregulation of mTOR gene expression was observed in PBMCs of 42 OA outpatients ("High mTOR expression subset") and in PBMCs and articular cartilages of all end-stage OA patients. A positive correlation between mTOR gene expression in PBMCs and cartilage was observed in the end-stage OA patients. 23 OA outpatients in the "Low mTOR expression subset" exhibited significantly lower mTOR gene expression in PBMCs compared to healthy controls. These "Low mTOR" subset subjects experienced significantly more pain upon walking, and standing and increased total joint stiffness versus "High mTOR" subset, while the latter more often exhibited synovitis. The protein concentrations of p70-S6K, p21, and caspase 3 in PBMCs were significantly lower in the "Low" subset versus "High" subset and end-stage subjects. Increases in the expression of mTOR in PBMCs of OA patients are related to disease activity, being associated with synovitis more than with pain.

10.
Arthritis Res Ther ; 14(3): R113, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22584047

ABSTRACT

INTRODUCTION: The intra-helical cleavage of type II collagen by proteases, including collagenases and cathepsin K, is increased with aging and osteoarthritis (OA) in cartilage as determined by immunochemical assays. The distinct sites of collagen cleavage generated by collagenases and cathepsin K in healthy and OA human femoral condylar cartilages were identified and compared. METHODS: Fixed frozen cartilage sections were examined immunohistochemically, using antibodies that react with the collagenase-generated cleavage neoepitopes, C2C and C1,2C, and the primary cleavage neoepitope (C2K) generated in type II collagen by the action of cathepsin K and possibly by other proteases, but not by any collagenases studied to date. RESULTS: In most cases, the staining patterns for collagen cleavage were similar for all three epitopes: weak to moderate mainly pericellular staining in non-OA cartilage from younger individuals and stronger, more widespread staining in aging and OA cartilages that often extended from the superficial to the mid/deep zone of the tissue. In very degenerate OA specimens, with significant disruption of the articular surface, staining was distributed throughout most of the cartilage matrix. CONCLUSIONS: Cleavage of collagen by proteases usually arises pericellularly around chondrocytes at and near the articular surface, subsequently becoming more intense and extending progressively deeper into the cartilage with aging and OA. The close correspondence between the distributions of these products suggests that both collagenases and cathepsin K, and other proteases that may generate this distinct cathepsin K cleavage site, are usually active in the same sites in the degradation of type II collagen.


Subject(s)
Aging , Cartilage, Articular/metabolism , Cathepsin K/metabolism , Collagen Type II/metabolism , Collagenases/metabolism , Osteoarthritis/metabolism , Adult , Aged , Cadaver , Female , Humans , Immunohistochemistry , Male , Middle Aged , Young Adult
11.
HSS J ; 8(1): 4-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-23372516
12.
Arthritis Rheum ; 64(4): 1035-45, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22076945

ABSTRACT

OBJECTIVE: To investigate the relationship between acute-phase serum amyloid A (A-SAA) and joint destruction in inflammatory arthritis. METHODS: Serum A-SAA and C-reactive protein (CRP) levels, the erythrocyte sedimentation rate (ESR), and levels of matrix metalloproteinase 1 (MMP-1), MMP-2, MMP-3, MMP-9, MMP-13, tissue inhibitor of metalloproteinases 1 (TIMP-1), vascular endothelial growth factor (VEGF), and type I and type II collagen-generated biomarkers C2C and C1,2C were measured at 0-3 months in patients with inflammatory arthritis commencing anti-tumor necrosis factor α (anti-TNFα) therapy and were correlated with 1-year radiographic progression. The effects of A-SAA on MMP/TIMP expression on RA fibroblast-like synoviocytes (FLS), primary human chondrocytes, and RA/psoriatic arthritis synovial explant cultures were assessed using real-time polymerase chain reaction, enzyme-linked immunosorbent assay, antibody protein arrays, and gelatin zymography. RESULTS: Serum A-SAA levels were significantly (P < 0.05) correlated with MMP-3, the MMP-3:TIMP-1 ratio, C1,2C, C2C, and VEGF. The baseline A-SAA level but not the ESR or the CRP level correlated with the 28-joint swollen joint count and was independently associated with 1-year radiographic progression (P = 0.038). A-SAA increased MMP-1, MMP-3, MMP-13, and MMP/TIMP expression in RA FLS and synovial explants (P < 0.05). In chondrocytes, A-SAA induced MMP-1, MMP-3, and MMP-13 messenger RNA and protein expression (all P < 0.01), resulting in a significant shift in MMP:TIMP ratios (P < 0.05). Gelatin zymography revealed that A-SAA induced MMP-2 and MMP-9 activity. Blockade of the A-SAA receptor SR-B1 (A-SAA receptor scavenger receptor-class B type 1) inhibited MMP-3, MMP-2, and MMP-9 expression in synovial explant cultures ex vivo. Importantly, we demonstrated that A-SAA has the ability to induce TNFα expression in RA synovial explant cultures (P < 0.05). CONCLUSION: A-SAA may be involved in joint destruction though MMP induction and collagen cleavage in vivo. The ability of A-SAA to regulate TNFα suggests that A-SAA signaling pathways may provide new therapeutic strategies for the treatment of inflammatory arthritis.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/metabolism , Disease Progression , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Serum Amyloid A Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adolescent , Adult , Aged , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/drug therapy , Collagen Type II/metabolism , Female , Follow-Up Studies , Foot Joints/diagnostic imaging , Hand Joints/diagnostic imaging , Humans , Male , Matrix Metalloproteinases/metabolism , Middle Aged , Radiography , Synovial Fluid/metabolism , Synovial Membrane/diagnostic imaging , Synovial Membrane/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Vascular Endothelial Growth Factor A/metabolism
13.
Environ Res ; 111(8): 1208-14, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21839992

ABSTRACT

PURPOSE: To examine associations between biomarkers of joint tissue metabolism and whole blood lead (Pb), separately for men and women in an African American and Caucasian population, which may reflect an underlying pathology. METHODS: Participants in the Johnston County Osteoarthritis Project Metals Exposure Sub-Study (329 men and 342 women) underwent assessment of whole blood Pb and biochemical biomarkers of joint tissue metabolism. Urinary cross-linked N telopeptide of type I collagen (uNTX-I) and C-telopeptide fragments of type II collagen (uCTX-II), serum cleavage neoepitope of type II collagen (C2C), serum type II procollagen synthesis C-propeptide (CPII), and serum hyaluronic acid (HA) were measured using commercially available kits; the ratio of [C2C:CPII] was calculated. Serum cartilage oligomeric matrix protein (COMP) was measured by an in-house assay. Multiple linear regression models were used to examine associations between continuous blood Pb and biomarker outcomes, adjusted for age, race, current smoking status, and body mass index. Results are reported as estimated change in biomarker level for a 5-unit change in Pb level. RESULTS: The median Pb level among men and women was 2.2 and 1.9µg/dL, respectively. Correlations were noted between Pb levels and the biomarkers uNTX-I, uCTX-II, and COMP in women, and between Pb and uCTX-II, COMP, CPII, and the ratio [C2C:CPII] in men. In adjusted models among women, a 5-unit increase in blood Pb level was associated with a 28% increase in uCTX-II and a 45% increase in uNTX-I levels (uCTX-II: 1.28 [95% CI: 1.04-1.58], uNTX-I: 1.45 [95% CI:1.21-1.74]). Among men, levels of Pb and COMP showed a borderline positive association (8% increase in COMP for a 5-unit change in Pb: 1.08 [95% CI: 1.00-1.18]); no other associations were significant after adjustment. CONCLUSIONS: Based upon known biomarker origins, the novel associations between blood Pb and biomarkers appear to be primarily reflective of relationships to bone and calcified cartilage turnover among women and cartilage metabolism among men, suggesting a potential gender-specific effect of Pb on joint tissue metabolism that may be relevant to osteoarthritis.


Subject(s)
Biomarkers/metabolism , Black People , Cartilage, Articular/metabolism , Lead/blood , Osteoarthritis/metabolism , White People , Female , Humans , Male , Middle Aged
14.
Arthritis Res Ther ; 13(2): 110, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21542886

ABSTRACT

Molecular or biochemical biomarkers of joint metabolism offer promise in helping us understand joint pathology, its detection and treatment. But they have often been studied alone and in only one body fluid. Although the synovial joint is usually the focus of most arthritis pathology, it is often difficult, for a variety of reasons, to obtain synovial fluid that should best reflect changes in biomarkers related to pathology. It is therefore very important to see whether analyses of more readily obtainable sera and urine also reflect changes in synovial fluid. Catterall and colleagues, in a paper in Arthritis Research & Therapy that examines very early biomarker changes following joint injury, provide us with some insights into these important questions. As the study was very small and examined very early changes following joint injury, prior to onset of any recognisable pathology, we look forward to future larger biomarker studies of this kind in patients with clinically defined arthritic changes to which we can relate biomarker data.


Subject(s)
Biomarkers/analysis , Collagen/analysis , Knee Injuries/metabolism , Proteoglycans/analysis , Female , Humans , Male
15.
Methods Mol Biol ; 622: 349-66, 2010.
Article in English | MEDLINE | ID: mdl-20135293

ABSTRACT

This chapter describes the production and characterization of antibodies raised against neoepitopes in collagenase-cleaved collagen. It also details the development, validation, and use of immunoassays using such antibodies to measure specifically collagenase-mediated cleavage.


Subject(s)
Collagen Type II/metabolism , Collagen Type I/metabolism , Collagenases/metabolism , Immunoassay/methods , Amino Acid Sequence , Animals , Antibodies/immunology , Antibody Specificity/immunology , Biomarkers/metabolism , Collagen Type II/chemistry , Enzyme-Linked Immunosorbent Assay , Molecular Sequence Data , Reference Standards
16.
Arthritis Rheum ; 60(5): 1372-80, 2009 May.
Article in English | MEDLINE | ID: mdl-19404937

ABSTRACT

OBJECTIVE: To evaluate 10 biomarkers in magnetic resonance imaging (MRI)-determined, pre-radiographically defined osteoarthritis (pre-ROA) and radiographically defined OA (ROA) in a population-based cohort of subjects with symptomatic knee pain. METHODS: Two hundred one white subjects with knee pain, ages 40-79 years, were classified into OA subgroups according to MRI-based cartilage (MRC) scores (range 0-4) and Kellgren/Lawrence (K/L) grades of radiographic severity (range 0-4): no OA (MRC score 0, K/L grade<2), pre-ROA (MRC score>or=1, K/L grade<2), or ROA (MRC score>or=1, K/L grade>or=2). Urine and serum samples were assessed for levels of the following biomarkers: urinary biomarkers C-telopeptide of type II collagen (uCTX-II), type II and types I and II collagen cleavage neoepitopes (uC2C and uC1,2C, respectively), and N-telopeptide of type I collagen, and serum biomarkers sC1,2C, sC2C, C-propeptide of type II procollagen (sCPII), chondroitin sulfate 846 epitope, cartilage oligomeric matrix protein, and hyaluronic acid. Multicategory logistic regression was performed to evaluate the association of OA subgroup with individual biomarker levels and biomarker ratios, adjusted for age, sex, and body mass index. RESULTS: The risk of ROA versus no OA increased with increasing levels of uCTX-II (odds ratio [OR] 3.12, 95% confidence interval [95% CI] 1.35-7.21), uC2C (OR 2.13, 95% CI 1.04-4.37), and uC1,2C (OR 2.07, 95% CI 1.06-4.04), and was reduced in association with high levels of sCPII (OR 0.53, 95% CI 0.30-0.94). The risk of pre-ROA versus no OA increased with increasing levels of uC2C (OR 2.06, 95% CI 1.05-4.01) and uC1,2C (OR 2.06, 95% CI 1.12-3.77). The ratios of type II collagen degradation markers to collagen synthesis markers were better than individual biomarkers at differentiating the OA subgroups, e.g., the ratio of [uCTX-II][uC1,2C] to sCPII was associated with a risk of ROA versus no OA of 3.47 (95% CI 1.34-9.03) and a risk of pre-ROA versus no OA of 2.56 (95% CI 1.03-6.40). CONCLUSION: Different cartilage degradation markers are associated with pre-ROA than are associated with ROA, indicating that their use as diagnostic markers depends on the stage of OA. Biomarker ratios contrasting cartilage degradation with cartilage synthesis are better able to differentiate OA stages compared with levels of the individual markers.


Subject(s)
Biomarkers/analysis , Osteoarthritis, Knee/diagnosis , Adult , Aged , Calcium-Binding Proteins/blood , Cartilage Oligomeric Matrix Protein , Cartilage, Articular/diagnostic imaging , Chondroitin Sulfates/blood , Collagen Type I/urine , Collagen Type II/blood , Extracellular Matrix Proteins/blood , Female , Glycoproteins/blood , Humans , Hyaluronic Acid/blood , Magnetic Resonance Imaging , Male , Matrilin Proteins , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Peptides/urine , Radiography
17.
Am J Pathol ; 173(1): 161-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18511517

ABSTRACT

Cathepsin K is a cysteine protease of the papain family that cleaves triple-helical type II collagen, the major structural component of the extracellular matrix of articular cartilage. In osteoarthritis (OA), the anabolic/catabolic balance of articular cartilage is disrupted with the excessive cleavage of collagen II by collagenases or matrix metalloproteinases. A polyclonal antibody against a C-terminal neoepitope (C2K) generated in triple-helical type II collagen by the proteolytic action of cathepsin K was prepared and used to develop an enzyme-linked immunosorbent assay to study the generation of this epitope and the effects of its presence in normal adult and osteoarthritic femoral condylar articular cartilage. The generation of the C2K epitope in explant culture and the effect of a specific cathepsin K inhibitor were studied. The neoepitope, which is not generated by the collagenase matrix metalloproteinase-13, increased with age in articular cartilage and was significantly elevated in osteoarthritic cartilage compared with adult nonarthritic cartilage. Moreover, in explants from three of eight OA patients, the generation of the neoepitope in culture was significantly reduced by a specific, nontoxic inhibitor of cathepsin K. These data suggest that cathepsin K is involved in the cleavage of type II collagen in human articular cartilage in certain OA patients and that it may play a role in both OA pathophysiology and the aging process.


Subject(s)
Cartilage, Articular/metabolism , Cathepsins/metabolism , Collagen Type II/metabolism , Osteoarthritis, Knee/metabolism , Adolescent , Adult , Aged , Aging/physiology , Base Sequence , Biphenyl Compounds/pharmacology , Cartilage, Articular/physiopathology , Cathepsin K , Cathepsins/drug effects , Collagen Type II/drug effects , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Middle Aged , Osteoarthritis, Knee/physiopathology , Peptides/drug effects , Peptides/metabolism
18.
Menopause ; 15(1): 133-7, 2008.
Article in English | MEDLINE | ID: mdl-18257147

ABSTRACT

OBJECTIVE: It is unclear whether there are changes in cartilage turnover after menopause, although these are well documented in bone. The aim of this study was to explore the possibility that menopause might change cartilage turnover as well as bone turnover. We also examined age and gender to estimate the independent influences of these parameters together with menopause on cartilage and bone turnover. DESIGN: Serum samples were collected from 140 healthy volunteers, 69 men (mean age +/- SD, 42.8 +/- 13.8 y) and 71 women (44.4 +/- 10.5 y) with self-reported pre- or postmenopausal status who had no swelling or pain in their spine and joints. Body mass index was also recorded. The serum concentration of a biomarker of cartilage type II collagen (C2C) degradation and concentrations of serum bone alkaline phosphatase and urine cross-linked type I collagen N-telopeptide, both accepted biomarkers of bone turnover, were measured together. Analyses of covariance were performed to test the mean differences of biomarkers by gender and by menopause status with age adjustment. RESULTS: In men there were no significant correlations between age and the biomarkers. In women C2C concentration decreased with age. Cross-linked type I collagen N-telopeptide and bone alkaline phosphatase levels were both increased after menopause, whereas C2C showed no detectable changes. C2C was not significantly related to body mass index. CONCLUSIONS: This study suggests that there are fundamental differences in the degradation of uncalcified C2C and bone type I collagen degradation and bone assembly in response to menopause and aging.


Subject(s)
Aging/blood , Bone Density , Bone Remodeling , Cartilage, Articular/metabolism , Collagen Type II/blood , Collagen Type I/blood , Osteoporosis/blood , Adult , Alkaline Phosphatase/blood , Biomarkers/blood , Body Mass Index , Female , Humans , Male , Menopause/metabolism , Middle Aged , Peptides/blood , Reference Values
19.
Arthritis Rheum ; 56(9): 2919-28, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17763421

ABSTRACT

OBJECTIVE: To investigate whether short-term changes in serum biomarkers of type II collagen degradation (C2C) and types I and II collagen degradation (C1,2C), as well as the biomarker for the synthesis of type II procollagen (CPII) can predict radiographic progression at 1 year following initiation of biologic therapy in patients with inflammatory arthritis. METHODS: Serum levels of biomarkers were measured at baseline and at 1, 3, 6, 9, and 12 months after initiation of biologic therapy. A composite score reflecting changes from baseline in all 3 biomarkers (DeltaCOL) was calculated. Associations with clinical responses according to the 28-joint count Disease Activity Score and with radiographic progression according to the modified Sharp/van der Heijde score (SHS) were assessed. RESULTS: The 1-year increase in the SHS correlated with the 1-month change in C2C results (r = 0.311, P = 0.028) and the DeltaCOL score (r = 0.342, P = 0.015). Radiographic progression was predicted by increases in serum C2C at 1 month (P = 0.031). The DeltaCOL score was significantly associated with 1-year radiographic progression after 1 (P = 0.022), 3 (P = 0.015), 6 (P = 0.048), and 9 (P = 0.019) months of therapy. Clinical remission was predicted by 1-month decreases in serum levels of C2C (P = 0.008) and C1,2C (P = 0.036). By regression analysis, 1-month changes in C2C, C1,2C, and CPII levels were independently associated with, and correctly predicted radiographic outcome in, 88% of the patients. CONCLUSION: Short-term changes in serum levels of collagen biomarkers following initiation of biologic therapy may better predict long-term clinical and radiographic outcomes. These collagen biomarkers may therefore be valuable new early indicators of short-term biologic treatment efficacy in clinical trials and in individual patients with inflammatory erosive arthritis.


Subject(s)
Arthritis, Psoriatic/blood , Arthritis, Psoriatic/diagnostic imaging , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/diagnostic imaging , Biological Therapy , Collagen Type II/blood , Adolescent , Adult , Aged , Arthritis, Psoriatic/therapy , Arthritis, Rheumatoid/therapy , Biomarkers/blood , Disease Progression , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Radiography , Time Factors
20.
Arthritis Res Ther ; 9(4): R75, 2007.
Article in English | MEDLINE | ID: mdl-17683641

ABSTRACT

Suppression of type II collagen (COL2A1) cleavage by transforming growth factor (TGF)-beta2 in cultured human osteoarthritic cartilage has been shown to be associated with decreased expression of collagenases, cytokines, genes associated with chondrocyte hypertrophy, and upregulation of prostaglandin (PG)E2 production. This results in a normalization of chondrocyte phenotypic expression. Here we tested the hypothesis that PGE2 is associated with the suppressive effects of TGF-beta2 in osteoarthritic (OA) cartilage and is itself capable of downregulating collagen cleavage and hypertrophy in human OA articular cartilage. Full-depth explants of human OA knee articular cartilage from arthroplasty were cultured with a wide range of concentrations of exogenous PGE2 (1 pg/ml to 10 ng/ml). COL2A1 cleavage was measured by ELISA. Proteoglycan content was determined by a colorimetric assay. Gene expression studies were performed with real-time PCR. In explants from patients with OA, collagenase-mediated COL2A1 cleavage was frequently downregulated at 10 pg/ml (in the range 1 pg/ml to 10 ng/ml) by PGE2 as well as by 5 ng/ml TGF-beta2. In control OA cultures (no additions) there was an inverse relationship between PGE2 concentration (range 0 to 70 pg/ml) and collagen cleavage. None of these concentrations of added PGE2 inhibited the degradation of proteoglycan (aggrecan). Real-time PCR analysis of articular cartilage from five patients with OA revealed that PGE2 at 10 pg/ml suppressed the expression of matrix metalloproteinase (MMP)-13 and to a smaller extent MMP-1, as well as the proinflammatory cytokines IL-1beta and TNF-alpha and type X collagen (COL10A1), the last of these being a marker of chondrocyte hypertrophy. These studies show that PGE2 at concentrations much lower than those generated in inflammation is often chondroprotective in that it is frequently capable of selectively suppressing the excessive collagenase-mediated COL2A1 cleavage found in OA cartilage. The results also show that chondrocyte hypertrophy in OA articular cartilage is functionally linked to this increased cleavage and is often suppressed by these low concentrations of added PGE2. Together these initial observations reveal the importance of very low concentrations of PGE2 in maintaining a more normal chondrocyte phenotype.


Subject(s)
Cartilage, Articular/drug effects , Collagen Type X/genetics , Collagenases/genetics , Dinoprostone/administration & dosage , Gene Expression/drug effects , Transforming Growth Factor beta2/genetics , Aged , Aged, 80 and over , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Enlargement/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type II/genetics , Collagen Type II/metabolism , Collagen Type X/metabolism , Collagenases/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Female , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Middle Aged , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/surgery , Proteoglycans/metabolism , Transforming Growth Factor beta2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...