1
|
Roy HS, Murugesan P, Kulkarni C, Arora M, Nagar GK, Guha R, Chattopadhyay N, Ghosh D. On-demand release of a selective MMP-13 blocker from an enzyme-responsive injectable hydrogel protects cartilage from degenerative progression in osteoarthritis. J Mater Chem B 2024; 12:5325-5338. [PMID: 38669084 DOI: 10.1039/d3tb02871b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
In osteoarthritis (OA), the degradation of cartilage is primarily driven by matrix metalloprotease-13 (MMP-13). Hence, the inhibition of MMP-13 has emerged as an attractive target for OA treatment. Among the various approaches that are being explored for MMP-13 regulation, blocking of the enzyme with specific binding molecules appears to be a more promising strategy for preventing cartilage degeneration. To enhance effectiveness and ensure patient compliance, it is preferable for the binding molecule to exhibit sustained activity when administered directly into the joint. Herein, we present an enzyme-responsive hydrogel that was designed to exhibit on-demand, the sustained release of BI-4394, a potent and highly selective MMP-13 blocker. The stable and compatible hydrogel was prepared using triglycerol monostearate. The efficacy of the hydrogel to prevent cartilage damage was assessed in a rat model of OA induced by anterior cruciate ligament transection (ACLT). The results revealed that in comparison to the rats administrated weekly with intra-articular BI-4394, the hydrogel implanted rats had reduced levels of inflammation and bone erosion. In comparison to untreated control, the cartilage in animals administered with BI-4394/hydrogel exhibited significant levels of collagen-2 and aggrecan along with reduced MMP-13. Overall, this study confirmed the potential of BI-4394 delivery using an enzyme-responsive hydrogel as a promising treatment option to treat the early stages of OA by preventing further cartilage degradation.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Preethi Murugesan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Chirag Kulkarni
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Malika Arora
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Geet Kumar Nagar
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Rajdeep Guha
- Division of Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Deepa Ghosh
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| |
Collapse
|
2
|
Bihlet AR, Balchen T, Goteti K, Sonne J, Ladel C, Karsdal MA, Ona V, Moreau F, Waterhouse R, Bay‐Jensen A, Guehring H. Safety, Tolerability, and Pharmacodynamics of the ADAMTS-5 Nanobody M6495: Two Phase 1, Single-Center, Double-Blind, Randomized, Placebo-Controlled Studies in Healthy Subjects and Patients With Osteoarthritis. ACR Open Rheumatol 2024; 6:205-213. [PMID: 38311369 PMCID: PMC11016567 DOI: 10.1002/acr2.11610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVE To assess the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of single and multiple injections of M6495, a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS-5) nanobody, in healthy volunteers and patients with osteoarthritis. METHODS Two randomized, placebo-controlled, double-blind studies were performed. Study 1 enrolled 54 healthy male volunteers who received one subcutaneous (s.c.) injection of M6495 (1-300 mg) or placebo (ratio 2:1), evaluating safety, PK, and PD as changes in the serum aggrecan fragment alanine-arginine-glycine-serine (ARGS). Study 2 enrolled 32 patients with osteoarthritis with Kellgren-Lawrence grades 2 to 4 and pain greater than or equal to 40 on the Western Ontario and McMaster Universities Arthritis Index pain subscale at screening and evaluated the safety, PK, and PD of three doses every two weeks (75-300 mg per dose) or six once-weekly M6495 s.c. doses (300 mg) or placebo (ratio 3:1) over 106 days' follow-up. RESULTS M6495 in single and multiple doses of less than or equal to 300 mg s.c. weekly was well tolerated with no clinically significant changes in any safety parameter. Adverse events more frequently reported in the M6495 groups were mostly mild cases of injection site reactions, myalgia, and nausea, which resolved after treatment cessation. The elimination half-life of single s.c. doses of M6495 ranged from 79 to 267 hours. M6495 administration substantially reduced serum ARGS levels, indicative of target engagement and indicating disease-modifying potential of M6495. CONCLUSION Treatment with M6495 in single and multiple doses up to and including 300 mg s.c. was found to be well tolerated and adequately safe for further clinical evaluation of potential disease-modifying effects.
Collapse
Affiliation(s)
| | | | | | - Jesper Sonne
- Bispebjerg Hospital and Copenhagen UniversityCopenhagenDenmark
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
4
|
Rousseau JC, Chapurlat R, Garnero P. Soluble biological markers in osteoarthritis. Ther Adv Musculoskelet Dis 2021; 13:1759720X211040300. [PMID: 34616494 PMCID: PMC8488516 DOI: 10.1177/1759720x211040300] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
In recent years, markers research has focused on the structural components of cartilage matrix. Specifically, a second generation of degradation markers has been developed against type II collagen neoepitopes generated by specific enzymes. A particular effort has been made to measure the degradation of minor collagens III and X of the cartilage matrix. However, because clinical data, including longitudinal controlled studies, are very scarce, it remains unclear whether they will be useful as an alternative to or in combination with current more established collagen biological markers to assess patients with osteoarthritis (OA). In addition, new approaches using high-throughput technologies allowed to detect new types of markers and improve the knowledge about the metabolic changes linked to OA. The relative advances coming from phenotype research are a first attempt to classify the heterogeneity of OA, and several markers could improve the phenotype characterization. These phenotypes could improve the selection of patients in clinical trials limiting the size of the studies by selecting patients with OA characteristics corresponding to the metabolic pathway targeted by the molecules evaluated. In addition, the inclusion of rapid progressors only in clinical trials would facilitate the demonstration of efficacy of the investigative drug to reduce joint degradation. The combination of selective biochemical markers appears as a promising and cost-effective approach to fulfill this unmet clinical need. Among the various potential roles of biomarkers in OA, their ability to monitor drug efficacy is probably one of the most important, in association with clinical and imaging parameters. Biochemical markers have the unique property to detect changes in joint tissue metabolism within a few weeks.
Collapse
Affiliation(s)
- Jean-Charles Rousseau
- INSERM Unit 1033, Pavillon F, Hôpital E. Herriot, 5 Place d’Arsonval, 69437 Lyon Cedex 03, France
- Biochemical Marker Assay Laboratory for Clinical Research (PMO-Lab), Lyon, France
- INSERM 1033, Lyon, France
| | - Roland Chapurlat
- Biochemical Marker Assay Laboratory for Clinical Research (PMO-Lab), Lyon, France
- INSERM UMR 1033, Lyon, France
- Université de Lyon, Lyon, France
- Hôpital Edouard Herriot, Hospice Civils de Lyon, Lyon, France
| | - Patrick Garnero
- Biochemical Marker Assay Laboratory for Clinical Research (PMO-Lab), Lyon, France
- INSERM UMR 1033, Lyon, France
| |
Collapse
|
5
|
Ferreira A, Duarte T, Marques S, Costa P, Neves S, dos Santos T, Granja P, Porto G. Iron triggers the early stages of cartilage degeneration in vitro: The role of articular chondrocytes. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100145. [DOI: 10.1016/j.ocarto.2021.100145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
|
6
|
Sprifermin (rhFGF18) versus vehicle induces a biphasic process of extracellular matrix remodeling in human knee OA articular cartilage ex vivo. Sci Rep 2020; 10:6011. [PMID: 32265494 PMCID: PMC7138815 DOI: 10.1038/s41598-020-63216-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Sprifermin, recombinant human fibroblast growth factor 18 (rhFGF18), induces cartilage regeneration in knees of patients with osteoarthritis (OA). We hypothesized that a temporal multiphasic process of extracellular matrix (ECM) degradation and formation underlie this effect. We aimed to characterize the temporal ECM remodeling of human knee OA articular cartilage in response to sprifermin treatment. Articular cartilage explants from patients with knee OA (npatients = 14) were cultured for 70 days, with permanent exposure to sprifermin (900, 450, 225 ng/mL), FGF18 (450 ng/mL), insulin-like growth factor-1 (100 ng/mL, positive control) or vehicle (nreplicates/treatment/patient = 2). Metabolic activity (AlamarBlue) and biomarkers of type IIB collagen (PIIBNP) formation (Pro-C2 enzyme-linked immunosorbent assay [ELISA]) and aggrecanase-mediated aggrecan neo-epitope NITEGE (AGNx1 ELISA) were quantified once a week. At end of culture (day 70), gene expression (quantitative reverse transcription polymerase chain reaction) and proteoglycan content (Safranin O/Fast green staining) were quantified. The cartilage had continuously increased metabolic activity, when treated with sprifermin/FGF18 compared to vehicle. During days 7-28 PIIBNP was decreased and NITEGE was increased, and during days 35-70 PIIBNP was increased. At end of culture, the cartilage had sustained proteoglycan content and relative expression of ACAN < COL2A1 < SOX9 < COL1A1, indicating that functional chondrocytes remained in the explants. Sprifermin induces a temporal biphasic cartilage remodeling in human knee OA articular cartilage explants, with early-phase increased aggrecanase activity and late-phase increased type II collagen formation.
Collapse
|
7
|
van Geffen EW, van Caam APM, Schreurs W, van de Loo FA, van Lent PLEM, Koenders MI, Thudium CS, Bay-Jensen AC, Blaney Davidson EN, van der Kraan PM. IL-37 diminishes proteoglycan loss in human OA cartilage: donor-specific link between IL-37 and MMP-3. Osteoarthritis Cartilage 2019; 27:148-157. [PMID: 30201492 DOI: 10.1016/j.joca.2018.08.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 07/23/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE A hallmark of osteoarthritis (OA) is degradation of articular cartilage proteoglycans. In isolated human OA chondrocytes, the anti-inflammatory cytokine Interleukin-37 (IL-37) lowers the expression of the proteolytic MMP and ADAMTS enzymes, which mediate this degradation. Therefore, we investigated if IL-37 protects against proteoglycan loss in freshly obtained human OA explants. MATERIAL AND METHODS Human OA cartilage explants were incubated with IL-37. Release of sulphated proteoglycans (sGAGs) was measured with the dimethylmethylene-blue assay. Production and degradation of newly synthesized proteoglycans was measured using 35S-sulphate. Proteoglycan and proteolytic enzyme expression were analyzed by qPCR and Western Blot. Proteolytic activity was determined by measuring MMP- and ADAMTS-generated aggrecan neo-epitopes with ELISA and by using MMP-3-, MMP-13- or ADAMTS-5-inhibitors. RESULTS Over time, a linear release of sGAGs from OA cartilage was measured. IL-37 reduced this release by 87 μg/ml (24%) 95%CI [21.04-141.4]. IL-37 did not affect 35S-sulphate incorporation or proteoglycan gene expression. In contrast, IL-37 reduced loss of 35S-sulphate labeled GAGs and reduced MMP-3 protein expression, indicating that IL-37 inhibits proteoglycan degradation. Remarkably, we observed two groups of patients; one group in which MMP-3-inhibition lowered sGAG release, and one group in which ADAMTS5-inhibition had this effect. Remarkably, IL-37 was only functional in the group of patients that responded to MMP-3-inhibition. CONCLUSION We identified a relationship between IL-37 and reduced sGAG loss in OA cartilage. Most likely, this effect is mediated by inhibition of MMP-3 expression. These results suggest that IL-37 could be applied as therapy in a subgroup of OA patients, in which cartilage degradation is mediated by MMP-3.
Collapse
Affiliation(s)
- E W van Geffen
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A P M van Caam
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Schreurs
- Department of Orthopaedics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - F A van de Loo
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P L E M van Lent
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M I Koenders
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - C S Thudium
- Department of Rheumatology, Nordic Bioscience, Copenhagen, Denmark
| | - A C Bay-Jensen
- Department of Rheumatology, Nordic Bioscience, Copenhagen, Denmark
| | - E N Blaney Davidson
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P M van der Kraan
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
8
|
Biochemical marker discovery, testing and evaluation for facilitating OA drug discovery and development. Drug Discov Today 2018; 23:349-358. [DOI: 10.1016/j.drudis.2017.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 09/18/2017] [Accepted: 10/06/2017] [Indexed: 01/25/2023]
|
9
|
Reker D, Kjelgaard-Petersen CF, Siebuhr AS, Michaelis M, Gigout A, Karsdal MA, Ladel C, Bay-Jensen AC. Sprifermin (rhFGF18) modulates extracellular matrix turnover in cartilage explants ex vivo. J Transl Med 2017; 15:250. [PMID: 29233174 PMCID: PMC5727954 DOI: 10.1186/s12967-017-1356-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/04/2017] [Indexed: 12/19/2022] Open
Abstract
Background Sprifermin (recombinant human fibroblast growth factor 18) is in clinical development as a potential disease-modifying osteoarthritis drug (DMOAD). In vitro studies have shown that cartilage regenerative properties of sprifermin involve chondrocyte proliferation and extracellular matrix (ECM) production. To gain further insight into the process of sprifermin in the cartilage tissue, this study aimed at investigating the ECM turnover of articular cartilage explants in a longitudinal manner. Methods Bovine full-depth articular cartilage explants were stimulated with sprifermin or placebo at weekly intervals, similar to the dosing regimen used in clinical trials. Pre-culturing with oncostatin M and tumour necrosis factor-α, was also used to induce an inflammatory state before treatment. Metabolic activity was measured using AlamarBlue, and chondrocyte proliferation was visualized by immuno-histochemical detection of proliferating cell nuclear antigen. ECM turnover was quantified by biomarker ELISAs; ProC2 reflecting type II collagen formation, CS846 reflecting aggrecan formation, active MMP9, C2M and AGNx2 reflecting matrix metalloproteinase activity, and AGNx1 reflecting aggrecanase activity. Results Sprifermin was able to reach the chondrocytes through the extracellular matrix, as it increased cell proliferation and metabolic activity of explants. ProC2 and CS846 was dose-dependently increased (P < 0.05) by sprifermin compared to placebo, while C2M and AGNx2 were unaffected, active MMP9 was slightly decreased, and AGNx1 was slightly increased. Over the course of treatment, the temporal order of ECM turnover responses was AGNx1, then ProC2, followed by CS846 and MMP9. Pro-inflammatory activation of the explants diminished the ECM turnover responses otherwise observed under non-inflammatory conditions. Conclusions The data suggest that sprifermin has chondrogenic effects on articular cartilage ex vivo, exerted through a sequential process of ECM turnover; aggrecan degradation seems to occur first, while type II collagen and aggrecan production increased at a later time point. In addition, it was observed that these chondrogenic effects are dependent on the inflammatory status of the cartilage prior to treatment. Electronic supplementary material The online version of this article (10.1186/s12967-017-1356-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ditte Reker
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark. .,Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen Ø, Denmark.
| | - Cecilie F Kjelgaard-Petersen
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark.,Technical University of Denmark, Anker Engelunds Vej 1, 2800, Kgs. Lyngby, Denmark
| | - Anne Sofie Siebuhr
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Martin Michaelis
- Osteoarthritis Research, Merck-KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Anne Gigout
- Osteoarthritis Research, Merck-KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Morten A Karsdal
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Christoph Ladel
- Osteoarthritis Research, Merck-KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Anne C Bay-Jensen
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark.
| |
Collapse
|
10
|
Kar S, Smith DW, Gardiner BS, Li Y, Wang Y, Grodzinsky AJ. Modeling IL-1 induced degradation of articular cartilage. Arch Biochem Biophys 2016; 594:37-53. [PMID: 26874194 DOI: 10.1016/j.abb.2016.02.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
Abstract
In this study, we develop a computational model to simulate the in vitro biochemical degradation of articular cartilage explants sourced from the femoropatellar grooves of bovine calves. Cartilage explants were incubated in culture medium with and without the inflammatory cytokine IL-1α. The spatio-temporal evolution of the cartilage explant's extracellular matrix components is modelled. Key variables in the model include chondrocytes, aggrecan, collagen, aggrecanase, collagenase and IL-1α. The model is first calibrated for aggrecan homeostasis of cartilage in vivo, then for data on (explant) controls, and finally for data on the IL-1α driven proteolysis of aggrecan and collagen over a 4-week period. The model was found to fit the experimental data best when: (i) chondrocytes continue to synthesize aggrecan during the cytokine challenge, (ii) a one to two day delay is introduced between the addition of IL-1α to the culture medium and subsequent aggrecanolysis, (iii) collagen degradation does not commence until the total concentration of aggrecan (i.e. both intact and degraded aggrecan) at any specific location within the explant becomes ≤ 1.5 mg/ml and (iv) degraded aggrecan formed due to the IL-1α induced proteolysis of intact aggrecan protects the collagen network while collagen degrades in a two-step process which, together, significantly modulate the collagen network degradation. Under simulated in vivo conditions, the model predicts increased aggrecan turnover rates in the presence of synovial IL-1α, consistent with experimental observations. Such models may help to infer the course of events in vivo following traumatic joint injury, and may also prove useful in quantitatively evaluating the efficiency of various therapeutic molecules that could be employed to avoid or modify the course of cartilage disease states.
Collapse
Affiliation(s)
- Saptarshi Kar
- School of Computer Science and Software Engineering, University of Western Australia, Crawley, WA, Australia
| | - David W Smith
- School of Computer Science and Software Engineering, University of Western Australia, Crawley, WA, Australia.
| | - Bruce S Gardiner
- School of Computer Science and Software Engineering, University of Western Australia, Crawley, WA, Australia
| | - Yang Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yang Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
11
|
The effect of protease inhibitors on the induction of osteoarthritis-related biomarkers in bovine full-depth cartilage explants. PLoS One 2015; 10:e0122700. [PMID: 25909781 PMCID: PMC4409205 DOI: 10.1371/journal.pone.0122700] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 02/12/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The specific degradation of type II collagen and aggrecan by matrix metalloproteinase (MMP)-9, -13 and ADAMTS-4 and -5 (aggrecanase-1 and -2) in the cartilage matrix is a critical step in pathology of osteoarthritis (OA). The aims of this study were: i) To investigate the relative contribution of ADAMTS-4 and ADAMTS-5 to cartilage degradation upon catabolic stimulation; ii) To investigate the effect of regulating the activities of key enzymes by mean of broad-spectrum inhibitors. METHODS Bovine full-depth cartilage explants stimulated with tumor necrosis factor alpha (TNF-α) and Oncostatin M (OSM) were cultured for 21 days with or without a number of inhibitors targeting different types of proteases. Monoclonal antibodies were raised against the active sites of ADAMTS-4, -5, MMP-9 and -13, and 4 ELISAs were developed and technically validated. In addition, the established AGNxI (ADAMTS-degraded aggrecan), AGNxII (MMP-degraded aggrecan), and CTX-II (MMP-derived type II collagen) were quantified in the explants-conditioned media. RESULTS We found that: i) Active ADAMTS-4, MMP-9, -13 were released in the late stage of TNF-α/ OSM stimulation, whereas no significant active ADAMTS-5 was detected in either extracts or supernatants; ii) Active ADAMTS-4 was primarily responsible for E373-374A bond cleavage in aggrecan in this setting; and iii) The compensatory mechanism could be triggered following the blockage of the enzyme caused by inhibitors. CONCLUSIONS ADAMTS-4 appeared to be the major protease for the generation of 374ARGS aggrecan fragment in the TNF-α/OSM stimulated bovine cartilage explants. This study addresses the need to determine the roles of ADAMTS-4 and ADAMTS-5 in human articular degradation in OA and hence identify the attractive target for slowing down human cartilage breakdown.
Collapse
|
12
|
Siebuhr AS, He Y, Gudmann NS, Gram A, Kjelgaard-Petersen CF, Qvist P, Karsdal MA, Bay-Jensen AC. Biomarkers of cartilage and surrounding joint tissue. Biomark Med 2014; 8:713-31. [DOI: 10.2217/bmm.13.144] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The identification and clinical demonstration of efficacy and safety of osteo- and chondro-protective drugs are met with certain difficulties. During the last few decades, the pharmaceutical industry has, in the field of rheumatology, experienced disappointments associated with the development of disease modification. Today, the vast amount of patients suffering from serious, chronic joint diseases can only be offered treatments aimed at improving symptoms, such as pain and acute inflammation, and are not aimed at protecting the joint tissue. This huge, unmet medical need has been the driver behind the development of improved analytical techniques allowing better and more efficient clinical trial design, implementation and analysis. With this review, we aim to provide a brief and general overview of biochemical markers of joint tissue, with special focus on neoepitopes. Furthermore, we highlight recent studies applying biochemical markers in joint degenerative diseases. These disorders, including osteoarthritis, rheumatoid arthritis and spondyloarthropathies, are the most predominant disorders in Europe and the USA, and have enormous socioeconomical impact.
Collapse
Affiliation(s)
- Anne S Siebuhr
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Yi He
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Natasja S Gudmann
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Aurelie Gram
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | | | - Per Qvist
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Morten A Karsdal
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Anne C Bay-Jensen
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| |
Collapse
|
13
|
An ARGS-aggrecan assay for analysis in blood and synovial fluid. Osteoarthritis Cartilage 2014; 22:242-9. [PMID: 24361794 DOI: 10.1016/j.joca.2013.12.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 11/29/2013] [Accepted: 12/09/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To validate a modified ligand-binding assay for the detection of aggrecanase generated aggrecan fragments with the ARGS neoepitope in synovial fluid (SF) and blood, and to verify the identity of aggrecan fragments found in blood. DESIGN An enzyme-linked immunosorbent assay (ELISA) on the Meso Scale Discovery (MSD) platform for detection of ARGS-aggrecan was validated, using a standard made from recombinant human aggrecan. Matched samples of SF, serum, plasma, and urine were obtained from 36 subjects at different time points after knee injury, and analysed for ARGS-aggrecan content. Aggrecan was purified from serum and plasma pools and analysed by Western blot. RESULTS The limits of quantification for the ARGS-aggrecan assay was between 0.2 and 0.025 pmol ARGS/ml, and the sensitivity of the assay was improved two-fold compared to when using a standard purified from human donors. The ARGS concentrations were highest in SF (mean, range; 3.02, 0.36-30.22 pmol/ml), 20 times lower in the blood samples (0.14, 0.055-0.28 pmol/ml serum and 0.13, 0.053-0.28 pmol/ml plasma), and 80 times lower in urine (0.036, below detection - 0.087 pmol/ml). Serum-ARGS and plasma-ARGS concentrations were similar, and correlated (r(S) = 0.773, P < 0.001). SF concentration correlated with serum concentrations (r(S) = 0.420, P = 0.011). In blood, we identified 129-138 kDa aggrecan fragments containing the ARGS neoepitope. CONCLUSIONS This novel ARGS-aggrecan assay is highly sensitive and suited for analysis of SF and blood samples. Both SF and blood contains ARGS-aggrecan, and ARGS concentrations in SF and serum are correlated.
Collapse
|
14
|
Kalai E, Bahlous A, Charni N, Bouzid K, Sahli H, Laadhar L, Chelly M, Rajhi H, Zouari B, Makni S, Abdelmoula J, Sallemi S, Garnero P. Association of serum levels of aggrecan ARGS, NITEGE fragments and radiologic knee osteoarthritis in Tunisian patients. Joint Bone Spine 2012; 79:610-5. [DOI: 10.1016/j.jbspin.2011.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 12/03/2011] [Indexed: 10/14/2022]
|
15
|
Rousseau JC, Garnero P. Biological markers in osteoarthritis. Bone 2012; 51:265-77. [PMID: 22538364 DOI: 10.1016/j.bone.2012.04.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 03/07/2012] [Accepted: 04/03/2012] [Indexed: 12/01/2022]
Abstract
Osteoarthritis (OA) is considered as a chronic disease with a long "silent" period. The diagnosis is generally based on clinical symptoms and radiographic changes. However X-ray has a poor sensitivity and a relatively large precision error that does not allow an early detection of OA or the monitoring of joint damage progression. The limitations of the tools that are currently available for OA assessment have been the impetus to identify specific biological markers that reflect quantitative and dynamic variations in joint remodeling. Research has focused on the structural components of cartilage matrix, especially type II collagen degradation markers. In spite of a significant increase of some markers in individuals with early stage of OA, the large overlap with control subjects indicates that the current biomarkers used alone have limited diagnostic potential. However, the combination of specific markers seems to improve the prediction of disease progression at the individual level. Several types of treatment have been investigated but the lack of medications with definitively demonstrated chondroprotective activity has limited the assessment of the potential role of biomarkers for monitoring patients' responses to the treatment of OA. In this review, we will use the BIPED classification that appeared in 2006 for OA markers to describe the potential usage of a given marker [5]. This article is part of a Special Issue entitled "Osteoarthritis".
Collapse
Affiliation(s)
- J Ch Rousseau
- INSERM Unit 1033; Pathophysiology, Diagnosis and Treatments of bone diseases, Université de Lyon, Lyon, France.
| | | |
Collapse
|
16
|
Böhm M, Grässel S. Role of proopiomelanocortin-derived peptides and their receptors in the osteoarticular system: from basic to translational research. Endocr Rev 2012; 33:623-51. [PMID: 22736674 PMCID: PMC3410228 DOI: 10.1210/er.2011-1016] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proopiomelanocortin (POMC)-derived peptides such as melanocortins and β-endorphin (β-ED) exert their pleiotropic effects via binding to melanocortin receptors (MCR) and opioid receptors (OR). There is now compelling evidence for the existence of a functional POMC system within the osteoarticular system. Accordingly, distinct cell types of the synovial tissue and bone have been identified to generate POMC-derived peptides like β-ED, ACTH, or α-MSH. MCR subtypes, especially MC1R, MC2R (the ACTH receptor), MC3R, and MC4R, but also the μ-OR and δ-OR, have been detected in various cells of the synovium, cartilage, and bone. The respective ligands of these POMC-derived peptide receptors mediate an increasing number of newly recognized biological effects in the osteoarticular system. These include bone mineralization and longitudinal growth, cell proliferation and differentiation, extracellular matrix synthesis, osteoprotection, and immunomodulation. Importantly, bone formation is also regulated by the central melanocortin system via a complex hormonal interplay with other organs and tissues involved in energy metabolism. Among the POMC-derived peptides examined in cell culture systems from osteoarticular tissue and in animal models of experimentally induced arthritis, α-MSH, ACTH, and MC3R-specific agonists appear to have the most promising antiinflammatory actions. The effects of these melanocortin peptides may be exploited in future for the treatment of patients with inflammatory and degenerative joint diseases.
Collapse
Affiliation(s)
- Markus Böhm
- Laboratory for Neuroendocrinology of the Skin and Interdisciplinary Endocrinology, Department of Dermatology, University of Münster, Von Esmarch-Strasse 58, D-48149 Münster, Germany.
| | | |
Collapse
|
17
|
Chen-An P, Andreassen KV, Henriksen K, Li Y, Karsdal MA, Bay-Jensen AC. The inhibitory effect of salmon calcitonin on tri-iodothyronine induction of early hypertrophy in articular cartilage. PLoS One 2012; 7:e40081. [PMID: 22768225 PMCID: PMC3386925 DOI: 10.1371/journal.pone.0040081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 06/05/2012] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE Salmon calcitonin has chondroprotective effect both in vitro and in vivo, and is therefore being tested as a candidate drug for cartilage degenerative diseases. Recent studies have indicated that different chondrocyte phenotypes may express the calcitonin receptor (CTR) differentially. We tested for the presence of the CTR in chondrocytes from tri-iodothyronin (T3)-induced bovine articular cartilage explants. Moreover, investigated the effects of human and salmon calcitonin on the explants. METHODS Early chondrocyte hypertrophy was induced in bovine articular cartilage explants by stimulation over four days with 20 ng/mL T3. The degree of hypertrophy was investigated by molecular markers of hypertrophy (ALP, IHH, COLX and MMP13), by biochemical markers of cartilage turnover (C2M, P2NP and AGNxII) and histology. The expression of the CTR was detected by qPCR and immunohistochemistry. T3-induced explants were treated with salmon or human calcitonin. Calcitonin down-stream signaling was measured by levels of cAMP, and by the molecular markers. RESULTS Compared with untreated control explants, T3 induction increased expression of the hypertrophic markers (p<0.05), of cartilage turnover (p<0.05), and of CTR (p<0.01). Salmon, but not human, calcitonin induced cAMP release (p<0.001). Salmon calcitonin also inhibited expression of markers of hypertrophy and cartilage turnover (p<0.05). CONCLUSIONS T3 induced early hypertrophy of chondrocytes, which showed an elevated expression of the CTR and was thus a target for salmon calcitonin. Molecular marker levels indicated salmon, but not human, calcitonin protected the cartilage from hypertrophy. These results confirm that salmon calcitonin is able to modulate the CTR and thus have chondroprotective effects.
Collapse
Affiliation(s)
- Pingping Chen-An
- Cartilage Biology and Biomarkers, Nordic Bioscience A/S, Herlev, Denmark
| | | | - Kim Henriksen
- Bone Biology and Pharmacology, Nordic Bioscience A/S, Herlev, Denmark
| | - Yadong Li
- Orthopedic Surgery Unit, Beijing Friendship Hospital, Beijing, People’s Republic of China
| | | | | |
Collapse
|
18
|
Investigation of chondrocyte hypertrophy and cartilage calcification in a full-depth articular cartilage explants model. Rheumatol Int 2012; 33:401-11. [DOI: 10.1007/s00296-012-2368-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 03/11/2012] [Indexed: 11/26/2022]
|
19
|
Madsen SH, Andreassen KV, Christensen ST, Karsdal MA, Sverdrup FM, Bay-Jensen AC, Henriksen K. Glucocorticoids exert context-dependent effects on cells of the joint in vitro. Steroids 2011; 76:1474-82. [PMID: 21855558 DOI: 10.1016/j.steroids.2011.07.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 07/27/2011] [Accepted: 07/28/2011] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Glucocorticoids are known to attenuate bone formation in vivo leading to decreased bone volume and increased risk of fractures, whereas effects on the joint tissue are less characterized. However, glucocorticoids appear to have a reducing effect on inflammation and pain in osteoarthritis. This study aimed at characterizing the effect of glucocorticoids on chondrocytes, osteoclasts, and osteoblasts. EXPERIMENTAL We used four model systems to investigate how glucocorticoids affect the cells of the joint; two intact tissues (femoral head- and cartilage-explants), and two separate cell cultures of osteoblasts (2T3-pre-osteoblasts) and osteoclasts (CD14(+)-monocytes). The model systems were cultured in the presence of two glucocorticoids; prednisolone or dexamethasone. To induce anabolic and catabolic conditions, cultures were activated by insulin-like growth factor I/bone morphogenetic protein 2 and oncostatin M/tumor necrosis factor-α, respectively. Histology and markers of bone- and cartilage-turnover were used to evaluate effects of glucocorticoid treatment. RESULTS Prednisolone treatment decreased collagen type-II degradation in immature cartilage, whereas glucocorticoids did not affect collagen type-II in mature cartilage. Glucocorticoids had an anti-catabolic effect on catabolic-activated cartilage from a bovine stifle joint and murine femoral heads. Glucocorticoids decreased viability of all bone cells, leading to a reduction in osteoclastogenesis and bone resorption; however, bone morphogenetic protein 2-stimulated osteoblasts increased bone formation, as opposed to non-stimulated osteoblasts. CONCLUSIONS Using highly robust in vitro models of bone and cartilage turnover, we suggest that effects of glucocorticoids highly depend on the activation and differential stage of the cell targeted in the joint. Present data indicated that glucocorticoid treatment may be beneficial for articular cartilage, although detrimental effects on bone should be taken into account.
Collapse
Affiliation(s)
- Suzi H Madsen
- Cartilage Biology and Biomarkers, Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Denmark.
| | | | | | | | | | | | | |
Collapse
|
20
|
Pedersen SJ, Sørensen IJ, Lambert RGW, Hermann KGA, Garnero P, Johansen JS, Madsen OR, Hansen A, Hansen MS, Thamsborg G, Andersen LS, Majgaard O, Loft AG, Erlendsson J, Asmussen KH, Jurik AG, Møller J, Hasselquist M, Mikkelsen D, Østergaard M. Radiographic progression is associated with resolution of systemic inflammation in patients with axial spondylarthritis treated with tumor necrosis factor α inhibitors: A study of radiographic progression, inflammation on magnetic resonance imaging, and c. ACTA ACUST UNITED AC 2011; 63:3789-800. [DOI: 10.1002/art.30627] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Abstract
OBJECTIVE: Exposure of articular cartilage to interleukin-1 (IL-1) results in increased synthesis of matrix degrading enzymes. Previously mechanical load applied together with IL-1 stimulation was found to reduce aggrecan cleavage by ADAMTS-4 and 5 and MMP-1, -3, -9, and -13 and reduce proteoglycan loss from the extracellular matrix. To further delineate the inhibition mechanism the gene expression of ADAMTS-4 and 5; MMP-1, -3, -9, and -13; and TIMP-1, -2, and -3 were measured. DESIGN: Mature bovine articular cartilage was stimulated with a 0.5 MPa compressive stress and 10 ng/ml of IL-1α for 3 days and then allowed to recover without stimulation for 1 additional day. The media was assayed for proteoglycan content on a daily basis, while chondrocyte gene expression (mRNA) was measured during stimulation and 1 day of recovery. RESULTS: Mechanical load alone did not change the gene expression for ADAMTS, MMP, or TIMP. IL-1 caused an increase in gene expression for all enzymes after 1 day of stimulation while not affecting the TIMP levels. Load applied together with IL-1 decreased the expression levels of ADAMTS-4 and -5 and MMP-1 and -3 and increased TIMP-3 expression. CONCLUSIONS: A mechanical load appears to modify cartilage degradation by IL-1 at the cellular level by reducing mRNA.
Collapse
Affiliation(s)
- P. A. Torzilli
- Laboratory for Soft Tissue Research, Tissue Engineering, Regeneration and Repair Program, Hospital for Special Surgery, New York, NY, USA,Peter A. Torzilli, PhD, Laboratory for Soft Tissue Research, The Hospital for Special Surgery, 471 East 71st Street, Room 524, New York, NY 10021-4010.
| | - M. Bhargava
- Laboratory for Soft Tissue Research, Tissue Engineering, Regeneration and Repair Program, Hospital for Special Surgery, New York, NY, USA
| | - C. T. Chen
- Laboratory for Soft Tissue Research, Tissue Engineering, Regeneration and Repair Program, Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
22
|
Pedersen SJ, Sørensen IJ, Garnero P, Johansen JS, Madsen OR, Tvede N, Hansen MS, Thamsborg G, Andersen LS, Majgaard O, Loft AG, Erlendsson J, Asmussen K, Jurik AG, Møller J, Hasselquist M, Mikkelsen D, Skjødt T, Lambert R, Hansen A, Østergaard M. ASDAS, BASDAI and different treatment responses and their relation to biomarkers of inflammation, cartilage and bone turnover in patients with axial spondyloarthritis treated with TNFα inhibitors. Ann Rheum Dis 2011; 70:1375-81. [PMID: 21551511 DOI: 10.1136/ard.2010.138883] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To investigate the relation between ankylosing spondylitis disease activity score (ASDAS), Bath ankylosing spondylitis disease activity index (BASDAI) and treatment response and biomarkers of inflammation (C-reactive protein (CRP), interleukin-6 (IL-6), YKL-40), angiogenesis (vascular endothelial growth factor (VEGF)), cartilage (C-terminal crosslinking telopeptide of type II collagen (CTX-II), matrix metalloproteinase-3 (MMP-3), total aggrecan, cartilage oligomeric matrix protein) and bone (C-terminal crosslinking telopeptide of type I collagen, osteocalcin) turnover in 60 patients with axial spondyloarthritis initiating tumour necrosis factor alpha (TNFα) inhibitor therapy. METHODS ASDAS (CRP-based), BASDAI and biomarkers were determined before and seven times during 46 weeks of TNFα inhibitor therapy. RESULTS Very high ASDAS were associated with high levels of inflammatory biomarkers, while high BASDAI were not related to any biomarkers. Mixed modeling demonstrated significant longitudinal associations between ASDAS and IL-6, VEGF, MMP-3 and osteocalcin and between BASDAI and CRP, IL-6 and VEGF. Major improvement in ASDAS was associated with larger percentage decreases in biomarkers of inflammation, angiogenesis, MMP-3 and increases in aggrecan and osteocalcin. BASDAI response was associated with larger decreases in CRP and IL-6. Biomarkers with moderate/high differences in responsiveness for major versus no/clinically important improvement in ASDAS were CRP, IL-6, VEGF, aggrecan and osteocalcin, and VEGF and CTX-II for BASDAI response versus non-response. CONCLUSION Levels and changes of 10 biomarkers in patients with axial spondyloarthritis during anti-TNFα therapy were documented. Construct validity and responsiveness of IL-6, VEGF, MMP-3, total aggrecan and osteocalcin were demonstrated. ASDAS was more associated with these biomarkers than BASDAI, and may better reflect the inflammatory disease processes. ClinicalTrials.gov identifier NCT00133315.
Collapse
|
23
|
Human aggrecanase generated synovial fluid fragment levels are elevated directly after knee injuries due to proteolysis both in the inter globular and chondroitin sulfate domains. Osteoarthritis Cartilage 2011; 19:1047-57. [PMID: 21664283 DOI: 10.1016/j.joca.2011.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 05/08/2011] [Accepted: 05/16/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To examine different aggrecanase generated fragments in synovial fluid (SF) from patients with acute and chronic knee injuries and from knee healthy subjects. METHODS We prepared SF-D1 samples from acute (n=35) and chronic (n=35) knee injury patients and knee healthy subjects (n=10). Aggrecan fragments were analyzed in the SF-D1 samples by quantitative (G1, ARGS, KEEE and G3 antibodies) and non-quantitative (GRGT and AGEG antibodies) Western blot. RESULTS ARGS-SELE, ARGS-chondroitin sulfate (CS)1, GRGT-, GLGS- and AGEG-G3 fragments were the main ARGS and G3 fragments in injured and reference samples. In the acute injury samples the concentrations of these fragments were increased compared to the reference, and the level of the ARGS-SELE remained elevated for at least 2 years after the joint injury. Both SF ARGS fragments and aggrecanase generated G3 fragments had high sensitivity and specificity as biomarkers in distinguishing injured from healthy knee joints, although the ARGS fragments had higher area under the receiver operating characteristic curve (AUC) values for injuries (74-86%) than the G3 fragments (AUC values 63-68%). CONCLUSION Our results suggest that during the acute phase after knee injury there is an increased aggrecanase activity against both the interglobular domain (IGD) and the CS2 cleavage sites of joint cartilage aggrecan. This increase in SF aggrecanolytic fragments is present for several years after the injury. SF ARGS fragments are better biomarkers than the aggrecanase generated G3-fragments in distinguishing injured from healthy knee joints.
Collapse
|
24
|
De Ceuninck F, Sabatini M, Pastoureau P. Recent progress toward biomarker identification in osteoarthritis. Drug Discov Today 2011; 16:443-9. [DOI: 10.1016/j.drudis.2011.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 01/14/2011] [Indexed: 11/30/2022]
|
25
|
Karsdal MA, Woodworth T, Henriksen K, Maksymowych WP, Genant H, Vergnaud P, Christiansen C, Schubert T, Qvist P, Schett G, Platt A, Bay-Jensen AC. Biochemical markers of ongoing joint damage in rheumatoid arthritis--current and future applications, limitations and opportunities. Arthritis Res Ther 2011; 13:215. [PMID: 21539724 PMCID: PMC3132026 DOI: 10.1186/ar3280] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease associated with potentially debilitating joint inflammation, as well as altered skeletal bone metabolism and co-morbid conditions. Early diagnosis and aggressive treatment to control disease activity offers the highest likelihood of preserving function and preventing disability. Joint inflammation is characterized by synovitis, osteitis, and/or peri-articular osteopenia, often accompanied by development of subchondral bone erosions, as well as progressive joint space narrowing. Biochemical markers of joint cartilage and bone degradation may enable timely detection and assessment of ongoing joint damage, and their use in facilitating treatment strategies is under investigation. Early detection of joint damage may be assisted by the characterization of biochemical markers that identify patients whose joint damage is progressing rapidly and who are thus most in need of aggressive treatment, and that, alone or in combination, identify those individuals who are likely to respond best to a potential treatment, both in terms of limiting joint damage and relieving symptoms. The aims of this review are to describe currently available biochemical markers of joint metabolism in relation to the pathobiology of joint damage and systemic bone loss in RA; to assess the limitations of, and need for additional, novel biochemical markers in RA and other rheumatic diseases, and the strategies used for assay development; and to examine the feasibility of advancement of personalized health care using biochemical markers to select therapeutic agents to which a patient is most likely to respond.
Collapse
Affiliation(s)
- Morten A Karsdal
- Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
- Southern University of Denmark (SDU), Campusvej 55DK-5230 Odense M Denmark
| | - Thasia Woodworth
- Leading Edge Clinical Research LLC, 3901 SE St Lucie Blvd unit 20, Stuart, Florida 34997, USA
| | - Kim Henriksen
- Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Walter P Maksymowych
- University of Alberta, 562 Heritage Medical Research Building, Edmonton, AB T6G 2S2, Canada
| | | | | | | | | | - Per Qvist
- Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Georg Schett
- University of Erlangen-Nurnberg Department of Internal Medicine, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Adam Platt
- Roche Products Limited, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW, UK
| | | |
Collapse
|
26
|
Bay-Jensen AC, Liu Q, Byrjalsen I, Li Y, Wang J, Pedersen C, Leeming DJ, Dam EB, Zheng Q, Qvist P, Karsdal MA. Enzyme-linked immunosorbent assay (ELISAs) for metalloproteinase derived type II collagen neoepitope, CIIM--increased serum CIIM in subjects with severe radiographic osteoarthritis. Clin Biochem 2011; 44:423-9. [PMID: 21223960 DOI: 10.1016/j.clinbiochem.2011.01.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Revised: 12/30/2010] [Accepted: 01/01/2011] [Indexed: 12/13/2022]
Abstract
OBJECTIVES In joint degenerative diseases, the collagens are degraded by matrix metalloproteinases and protein fragments are released to serum as potential biomarkers. METHODS A collagen type II specific neoepitope, CIIM, was identified (…RDGAAG(1053)) by mass spectrometry. Two ELISAs against the neoepitope were developed. CIIM was measured in cartilage explants in the presence or absence of protease inhibitors. CIIM was measured in OA synovial fluid (n=51) and serum (n=156). Knee OA was graded by standard Kellgren-Lawrence (KL) score. RESULTS The ELISAs showed good technical performance; CV%, <13%. CIIM release from cartilage explants was blocked by the MMP inhibitor. CIIM was detected in synovial fluid. Furthermore, serum CIIM levels were significantly higher (P<0.05) in those individuals with mild or severe OA than in those with no OA. CONCLUSION We developed a new biomarker for joint degenerative diseases, which we demonstrated was derived from MMP-degraded type II collagen.
Collapse
|
27
|
Ly M, Laremore TN, Linhardt RJ. Proteoglycomics: recent progress and future challenges. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2010; 14:389-99. [PMID: 20450439 DOI: 10.1089/omi.2009.0123] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Proteoglycomics is a systematic study of structure, expression, and function of proteoglycans, a posttranslationally modified subset of a proteome. Although relying on the established technologies of proteomics and glycomics, proteoglycomics research requires unique approaches for elucidating structure-function relationships of both proteoglycan components, glycosaminoglycan chain, and core protein. This review discusses our current understanding of structure and function of proteoglycans, major players in the development, normal physiology, and disease. A brief outline of the proteoglycomic sample preparation and analysis is provided along with examples of several recent proteoglycomic studies. Unique challenges in the characterization of glycosaminoglycan component of proteoglycans are discussed, with emphasis on the many analytical tools used and the types of information they provide.
Collapse
Affiliation(s)
- Mellisa Ly
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, USA
| | | | | |
Collapse
|
28
|
Settle S, Vickery L, Nemirovskiy O, Vidmar T, Bendele A, Messing D, Ruminski P, Schnute M, Sunyer T. Cartilage degradation biomarkers predict efficacy of a novel, highly selective matrix metalloproteinase 13 inhibitor in a dog model of osteoarthritis: confirmation by multivariate analysis that modulation of type II collagen and aggrecan degradation peptides parallels pathologic changes. ACTA ACUST UNITED AC 2010; 62:3006-15. [PMID: 20533541 DOI: 10.1002/art.27596] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To demonstrate that the novel highly selective matrix metalloproteinase 13 (MMP-13) inhibitor PF152 reduces joint lesions in adult dogs with osteoarthritis (OA) and decreases biomarkers of cartilage degradation. METHODS The potency and selectivity of PF152 were evaluated in vitro using 16 MMPs, TACE, and ADAMTS-4 and ADAMTS-5, as well as ex vivo in human cartilage explants. In vivo effects were evaluated at 3 concentrations in mature beagles with partial medial meniscectomy. Gross and histologic changes in the femorotibial joints were evaluated using various measures of cartilage degeneration. Biomarkers of cartilage turnover were examined in serum, urine, or synovial fluid. Results were analyzed individually and in combination using multivariate analysis. RESULTS The potent and selective MMP-13 inhibitor PF152 decreased human cartilage degradation ex vivo in a dose-dependent manner. PF152 treatment of dogs with OA reduced cartilage lesions and decreased biomarkers of type II collagen (type II collagen neoepitope) and aggrecan (peptides ending in ARGN or AGEG) degradation. The dose required for significant inhibition varied with the measure used, but multivariate analysis of 6 gross and histologic measures indicated that all doses differed significantly from vehicle but not from each other. Combined analysis of cartilage degradation markers showed similar results. CONCLUSION This highly selective MMP-13 inhibitor exhibits chondroprotective effects in mature animals. Biomarkers of cartilage degradation, when evaluated in combination, parallel the joint structural changes induced by the MMP-13 inhibitor. These data support the potential therapeutic value of selective MMP-13 inhibitors and the use of a set of appropriate biomarkers to predict efficacy in OA clinical trials.
Collapse
Affiliation(s)
- Steven Settle
- Pfizer Global Research and Development, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pedersen SJ, Hetland ML, Sørensen IJ, Ostergaard M, Nielsen HJ, Johansen JS. Circulating levels of interleukin-6, vascular endothelial growth factor, YKL-40, matrix metalloproteinase-3, and total aggrecan in spondyloarthritis patients during 3 years of treatment with TNFα inhibitors. Clin Rheumatol 2010; 29:1301-9. [PMID: 20640910 DOI: 10.1007/s10067-010-1528-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 06/07/2010] [Accepted: 06/19/2010] [Indexed: 11/24/2022]
Abstract
The objectives of the study were to investigate short and long-term changes and relations to treatment response of plasma interleukin-6 (IL-6), vascular endothelial growth factor (VEGF), YKL-40, matrix metalloproteinase-3 (MMP-3), and total aggrecan in patients with spondyloarthritis (SpA) treated with tumor necrosis factor-alpha (TNFα) inhibitors and to compare with levels in healthy subjects. Biomarkers were measured in an observational cohort of 49 SpA patients (ankylosing spondylitis, n=32, and psoriatic arthritis, n=17) initiating TNFα inhibitor therapy (infliximab, n=38; etanercept, n=8; and adalimumab, n=3) and compared with levels in healthy subjects. Clinical parameters and biomarkers were measured at baseline, weeks 2, 6, and every 6-12 weeks for up to 3 years. Patients with co-morbidities (n=4), missing baseline samples (n=3), and adverse events (n=5) were excluded. Patients with SpA had compared with healthy subjects elevated IL-6 (median 8.5 ng/l (range, 0.98-64) vs. 1.3 (0.33-26)), VEGF (105 ng/l (22-752) vs. 45 (12-351)), YKL-40 (74 μg/l (14-572) vs. 43 (20-184)), and MMP-3 (43 μg/l (9.1-401) vs. 16 (2.5-47), p≤0.001), whereas total aggrecan was lower (662 μg/l (223-2,219) vs. 816 (399-2,190), p≤0.001). Two weeks after first treatment, all biomarker levels changed towards normal levels (p≤0.03) in clinical responders (n=24), and persistent reductions over 3 years were found in IL-6, VEGF, YKL-40, and MMP-3. Only MMP-3 decreased (p≤0.02) in non-responders (n=13). The study demonstrated changes of plasma IL-6, VEGF, YKL-40, MMP-3, and total aggrecan and a potential value for monitoring disease activity and treatment response in SpA patients. Larger prospective studies are required to clarify clinical utility of these biomarkers.
Collapse
Affiliation(s)
- Susanne Juhl Pedersen
- Department of Rheumatology, Copenhagen University Hospital, Herlev and Gentofte, Denmark.
| | | | | | | | | | | |
Collapse
|
30
|
Zhang A, Cao JL, Yang B, Chen JH, Zhang ZT, Li SY, Fu Q, Hugnes CE, Caterson B. Effects of moniliformin and selenium on human articular cartilage metabolism and their potential relationships to the pathogenesis of Kashin-Beck disease. J Zhejiang Univ Sci B 2010; 11:200-8. [PMID: 20205306 DOI: 10.1631/jzus.b0900074] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To investigate the effects of mycotoxin moniliformin (MON) on the metabolism of aggrecan and type II collagen in human chondrocytes in vitro and the relationship between MON and Kashin-Beck disease (KBD). METHODS Human chondrocytes were isolated and cultured on bone matrix gelatin to form an artificial cartilage model in vitro with or without MON toxin. Cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The expression of aggrecan and type II collagen in the cartilage was determined using immunocytochemical staining. RESULTS MON toxin inhibited chondrocyte viability in dose-dependent and time-dependent manners. MON reduced aggrecan and type II collagen syntheses in the tissue-engineered cartilage. MON also increased the expression of matrix metalloproteinase-1 (MMP-1), MMP-13, BC4 epitopes, and CD44 in cartilages. However, the expression of 3B3(-) epitopes in cartilages was inhibited by MON. Selenium partially alleviated the damage of aggrecan induced by MON toxin. CONCLUSION MON toxin promoted the catabolism of aggrecan and type II collagen in human chondrocytes.
Collapse
Affiliation(s)
- An Zhang
- Institute of Endemic Diseases, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bay-Jensen AC, Sondergaard BC, Christiansen C, Karsdal MA, Madsen SH, Qvist P. Biochemical markers of joint tissue turnover. Assay Drug Dev Technol 2010; 8:118-24. [PMID: 19715454 DOI: 10.1089/adt.2009.0199] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recent disappointments in late stage developments of anti-osteoarthritic drugs have reinforced efforts to develop better biomarkers for application in both the drug development process as well as in the routine management of these patients. Here we provide a brief review of biochemical tests available for the study of tissue turnover in each of the three compartments of the articular joint, that is the bone, the cartilage, and the synovium. Finally, we provide some perspective to future developments in biomarker discovery and discuss the potential impact such technologies could have on the drug development process.
Collapse
|
32
|
Garvican ER, Vaughan-Thomas A, Redmond C, Gabriel N, Clegg PD. MMP-mediated collagen breakdown induced by activated protein C in equine cartilage is reduced by corticosteroids. J Orthop Res 2010; 28:370-8. [PMID: 19777546 DOI: 10.1002/jor.21001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The plasma serine protease activated protein C (APC) is synthesized by human chondrocytes at sites of pathological cartilage fibrillation. APC levels are increased in osteoarthritis (OA) synovial fluid, and in vitro APC has been shown to synergize with interleukin-1beta (IL-1) to promote degradation from ovine cartilage. A model of equine cartilage degradation was established and used to explore corticosteroid activities. Intraarticular corticosteroids are a commonly prescribed treatment for joint disease, however their role in disease modification remains unclear. APC synergized with IL-1 or tumor necrosis factor-alpha (TNFalpha), promoting significant collagen degradation from equine cartilage explants within 4 days, but did not augment glycoaminoglycan (GAG) release. APC activated pro-matrix metalloproteinases (MMP)-2 but not pro-MMP-9, as assessed by gelatin zymography. APC did not directly activate pro-MMP-13. Dexamethasone, triamcinolone, and methylprednisolone acetate (MPA) were evaluated at concentrations between 10(- 5)M and 10(-10)M. High concentrations significantly increased GAG release from IL-1+APC-treated explants. With the exception of MPA at 10(-10)M, all concentrations of corticosteroids caused significant decreases in IL-1+APC-driven hydroxyproline loss. Treatment with corticosteroids suppressed expression of MMP-1, -3, and -13 mRNA. The collagenolysis associated with IL-1+APC synergy, and the inhibition of this effect by corticosteroids may involve gelatinase activation and downregulation of MMP expression, respectively.
Collapse
Affiliation(s)
- Elaine R Garvican
- Musculoskeletal Research Group, University of Liverpool Veterinary Teaching Hospital, Leahurst, Neston, Wirral CH647TE, United Kingdom.
| | | | | | | | | |
Collapse
|
33
|
Sondergaard BC, Schultz N, Madsen SH, Bay-Jensen AC, Kassem M, Karsdal MA. MAPKs are essential upstream signaling pathways in proteolytic cartilage degradation--divergence in pathways leading to aggrecanase and MMP-mediated articular cartilage degradation. Osteoarthritis Cartilage 2010; 18:279-88. [PMID: 19932675 DOI: 10.1016/j.joca.2009.11.005] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 10/28/2009] [Accepted: 11/04/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Matrix metalloproteinases (MMPs) and aggrecanases are essential players in cartilage degradation. However, the signaling pathways that results in MMP and/or aggrecanase synthesis and activation are not well understood. We investigated the molecular events leading to MMP- and aggrecanase-mediated cartilage degradation. METHODS Cartilage degradation was induced in bovine articular cartilage explants by oncostatin M (OSM) and tumor necrosis factor (TNF), in the presence or absence of specific inhibitors of the mitogen-activated protein kinases (MAPKs) P38, P44/42 and Src family. Toxicity was followed by the AlamarBlue colorimetric assay. MMP-activity was assessed using a fluorescent substrate assay and MMP-9 and -2 activities by gelatinase zymography. MMP-mediated collagen type II degradation and MMP as well as aggrecanase-mediated aggrecan degradation was investigated with specific ELISA and hydroxyproline release by standard methods. The findings were verified by immunohistochemistry and histology. RESULTS Stimulation of cartilage degradation by OSM+TNF resulted in 100-fold induction of CTX-II release (P<0.01). This was dose-dependently inhibited by MAPK P38 inhibitors and by the MAPK P44/42 inhibitors. MMP-activity and expression was significantly decreased, as evaluated by cleavage of fluorescence MMP-substrate and zymography. Immunohistochemistry confirmed these findings. Interestingly, only the P44/42 inhibitors abrogated aggrecanase-mediated aggrecan degradation. CONCLUSION We found that inhibition of MAPK P38, P44/42 and Src family abrogated proteolytic cartilage degradation by blocking MMP synthesis and activity. However, only MAPK P44/42 was essential for aggrecanase-mediated aggrecan degradation. These data suggest that various aspects of cartilage degradation can be targeted independently by inhibiting specific upstream signaling pathway.
Collapse
|
34
|
Naito K, Watari T, Muta T, Furuhata A, Iwase H, Igarashi M, Kurosawa H, Nagaoka I, Kaneko K. Low-intensity pulsed ultrasound (LIPUS) increases the articular cartilage type II collagen in a rat osteoarthritis model. J Orthop Res 2010; 28:361-9. [PMID: 19810106 DOI: 10.1002/jor.20995] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In this study, the effect of low-intensity pulsed ultrasound (LIPUS) on cartilage was evaluated in a rat osteoarthritis (OA) model using serum biomarkers such as CTX-II (type II collagen degradation) and CPII (type II collagen synthesis) as well as histological criteria (Mankin score and immunohistochemical type II collagen staining). OA was surgically induced in the knee joint of rats by anterior cruciate/medial collateral ligament transection and medial meniscus resection (ACLT + MMx). Animals were divided into three groups: sham-operated group (Sham), ACLT + MMx group without LIPUS (-LIPUS), and ACLT + MMx group with LIPUS (+LIPUS; 30 mW/cm(2), 20 min/day for 28 days). CTX-II levels were elevated in both -LIPUS and +LIPUS groups compared to that in the Sham group after the operation, but there was no significant difference between +LIPUS and -LIPUS groups, suggesting that LIPUS does not affect the degradation of type II collagen in this model. In contrast, CPII was significantly increased in +LIPUS group compared to -LIPUS and Sham. Moreover, histological damage on the cartilage (Mankin score) was ameliorated by LIPUS, and type II collagen was immunohistochemically increased by LIPUS in the cartilage of an OA model. Of interest, mRNA expression of type II collagen was enhanced by LIPUS in chondrocytes. Together these observations suggest that LIPUS is likely to increase the type II collagen synthesis in articular cartilage, possibly via the activation of chondrocytes and induction of type II collagen mRNA expression, thereby exhibiting chondroprotective action in a rat OA model.
Collapse
Affiliation(s)
- Kiyohito Naito
- Department of Orthopaedic Surgery, Juntendo University Shizuoka Hospital, 1129 Nagaoka, Izunokuni, Shizuoka 410-2295, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Torzilli PA, Bhargava M, Park S, Chen CC. Mechanical load inhibits IL-1 induced matrix degradation in articular cartilage. Osteoarthritis Cartilage 2010; 18:97-105. [PMID: 19747586 PMCID: PMC2818235 DOI: 10.1016/j.joca.2009.07.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 07/17/2009] [Accepted: 06/22/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis is a disease process of cellular degradation of articular cartilage caused by mechanical loads and inflammatory cytokines. We studied the cellular response in native cartilage subjected to a mechanical load administered simultaneously with an inflammatory cytokine interleukin-1 (IL-1), hypothesizing that the combination of load and cytokine would result in accelerated extracellular matrix (ECM) degradation. METHODS Mature bovine articular cartilage was loaded for 3 days (stimulation) with 0.2 and 0.5 MPa stresses, with and without IL-1 (IL-1alpha, 10 ng/ml), followed by 3 days of no stimulation (recovery). Aggrecan and collagen loss were measured as well as aggrecan cleavage using monoclonal antibodies AF-28 and BC-3 for cleavage by aggrecanases (ADAMTS) and matrix metalloproteinases (MMPs), respectively. RESULTS Incubation with IL-1 caused aggrecan cleavage by aggrecanases and MMPs during the 3 days of stimulation. A load of 0.5 MPa inhibited the IL-1-induced aggrecan loss while no inhibition was found for the 0.2 MPa stress. There was no collagen loss during the treatments but upon load and IL-1 removal proteoglycan and collagen loss increased. Load itself under these conditions was found to have no effect when compared to the unloaded controls. CONCLUSIONS A mechanical load of sufficient magnitude can inhibit ECM degradation by chondrocytes when stimulated by IL-1. The molecular mechanisms involved in this process are not clear but probably involve altered mechanochemical signal transduction between the ECM and chondrocyte.
Collapse
Affiliation(s)
| | | | - Seonghun Park
- School of Mechanical Engineering Pusan National University Busan, Republic of Korea (South Korea)
| | | |
Collapse
|
36
|
Struglics A, Larsson S. A comparison of different purification methods of aggrecan fragments from human articular cartilage and synovial fluid. Matrix Biol 2010; 29:74-83. [DOI: 10.1016/j.matbio.2009.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 07/18/2009] [Accepted: 08/28/2009] [Indexed: 10/20/2022]
|
37
|
Madsen SH, Sumer EU, Bay-Jensen AC, Sondergaard BC, Qvist P, Karsdal MA. Aggrecanase- and matrix metalloproteinase-mediated aggrecan degradation is associated with different molecular characteristics of aggrecan and separated in timeex vivo. Biomarkers 2009; 15:266-76. [DOI: 10.3109/13547500903521810] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
38
|
Embree MC, Kilts TM, Ono M, Inkson CA, Syed-Picard F, Karsdal MA, Oldberg A, Bi Y, Young MF. Biglycan and fibromodulin have essential roles in regulating chondrogenesis and extracellular matrix turnover in temporomandibular joint osteoarthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:812-26. [PMID: 20035055 DOI: 10.2353/ajpath.2010.090450] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The temporomandibular joint is critical for jaw movements and allows for mastication, digestion of food, and speech. Temporomandibular joint osteoarthritis is a degenerative disease that is marked by permanent cartilage destruction and loss of extracellular matrix (ECM). To understand how the ECM regulates mandibular condylar chondrocyte (MCC) differentiation and function, we used a genetic mouse model of temporomandibular joint osteoarthritis that is deficient in two ECM proteins, biglycan and fibromodulin (Bgn(-/0)Fmod(-/-)). Given the unavailability of cell lines, we first isolated primary MCCs and found that they were phenotypically unique from hyaline articular chondrocytes isolated from the knee joint. Using Bgn(-/0) Fmod(-/-) MCCs, we discovered the early basis for temporomandibular joint osteoarthritis arises from abnormal and accelerated chondrogenesis. Transforming growth factor (TGF)-beta1 is a growth factor that is critical for chondrogenesis and binds to both biglycan and fibromodulin. Our studies revealed the sequestration of TGF-beta1 was decreased within the ECM of Bgn(-/0) Fmod(-/-) MCCs, leading to overactive TGF-beta1 signal transduction. Using an explant culture system, we found that overactive TGF-beta1 signals induced chondrogenesis and ECM turnover in this model. We demonstrated for the first time a comprehensive study revealing the importance of the ECM in maintaining the mandibular condylar cartilage integrity and identified biglycan and fibromodulin as novel key players in regulating chondrogenesis and ECM turnover during temoporomandibular joint osteoarthritis pathology.
Collapse
Affiliation(s)
- Mildred C Embree
- Craniofacial and Skeletal Diseases Branch, National Institutes of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang B, Chen P, Jensen ACB, Karsdal MA, Madsen SH, Sondergaard BC, Zheng Q, Qvist P. Suppression of MMP activity in bovine cartilage explants cultures has little if any effect on the release of aggrecanase-derived aggrecan fragments. BMC Res Notes 2009; 2:259. [PMID: 20021645 PMCID: PMC2803187 DOI: 10.1186/1756-0500-2-259] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Accepted: 12/18/2009] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Progressive loss of articular cartilage is a central hallmark in many joint disease, however, the relative importance of individual proteolytic pathways leading to cartilage erosion is at present unknown. We therefore investigated the time-dependant release ex vivo of MMP- and aggrecanase-derived fragments of aggrecan and type II collagen into the supernatant of bovine cartilage explants cultures using neo-epitope specific immunoassays, and to associate the release of these fragments with the activity of proteolytic enzymes using inhibitors. FINDINGS Bovine cartilage explants were cultured in the presence or absence of the catabolic cytokines oncostatin M (OSM) and tumor necrosis factor alpha (TNFalpha). In parallel, explants were co-cultured with protease inhibitors such as GM6001, TIMP1, TIMP2 and TIMP3. Fragments released into the supernatant were determined using a range of neo-epitope specific immunoassays; (1) sandwich (342)FFGVG-G2 ELISA, (2) competition NITEGE(373)ELISA (3) sandwich G1-NITEGE(373 )ELISA (4) competition (374)ARGSV ELISA, and (5) sandwich (374)ARGSV-G2 ELISA all detecting aggrecan fragments, and (6) sandwich CTX-II ELISA, detecting C-telopeptides of type II collagen. We found that (1) aggrecanase-derived aggrecan fragments are released in the early (day 2-7) and mid phase (day 9-14) into the supernatant from bovine explants cultures stimulated with catabolic cytokines, (2) the release of NITEGE(373 )neo-epitopes are delayed compared to the corresponding (374)ARGSV fragments, (3) the MMP inhibitor GM6001 did not reduce the release of aggrecanase-derived fragment, but induced a further delay in the release of these fragments, and finally (4) the MMP-derived aggrecan and type II collagen fragments were released in the late phase (day 16-21) only. CONCLUSION Our data support the model, that aggrecanases and MMPs act independently in the processing of the aggrecan molecules, and furthermore that suppression of MMP-activity had little if any effect on the quantity of aggrecanase-derived fragments released from explants cultures.
Collapse
Affiliation(s)
- Bijue Wang
- Nordic Bioscience A/S, Zhongguancun Life Science Park, Beijing 102206, PR China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Qvist P, Christiansen C, Karsdal MA, Madsen SH, Sondergaard BC, Bay-Jensen AC. Application of biochemical markers in development of drugs for treatment of osteoarthritis. Biomarkers 2009; 15:1-19. [DOI: 10.3109/13547500903295873] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Larsson S, Lohmander LS, Struglics A. Synovial fluid level of aggrecan ARGS fragments is a more sensitive marker of joint disease than glycosaminoglycan or aggrecan levels: a cross-sectional study. Arthritis Res Ther 2009; 11:R92. [PMID: 19545413 PMCID: PMC2714148 DOI: 10.1186/ar2735] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2009] [Revised: 05/19/2009] [Accepted: 06/22/2009] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Aggrecanase cleavage at the 392Glu-393Ala bond in the interglobular domain (IGD) of aggrecan, releasing N-terminal 393ARGS fragments, is an early key event in arthritis and joint injuries. Here, we use a quantitative immunoassay of aggrecan ARGS neoepitope fragments in human synovial fluid to determine if this cleavage-site specific method better identifies joint pathology than previously available less specific aggrecan assays. METHODS Synovial fluid (SF) from 26 people with healthy knees (reference) and 269 patients were analyzed in a cross-sectional study. Patient groups were acute inflammatory arthritis, acute knee injury, chronic knee injury and knee osteoarthritis (OA). Aggrecan ARGS fragments were assayed by ELISA using the monoclonal antibody OA-1. Total aggrecan content was analyzed by an ELISA using the monoclonal antibody 1-F21, and sulfated glycosaminoglycan by Alcian blue precipitation. RESULTS Aggrecan ARGS fragment concentrations in all groups differed from the reference group (P < 0.001). The acute inflammatory arthritis group had the highest median level, 177-fold greater than that of the reference group. Median levels (in pmol ARGS/ml SF) were: reference 0.5, acute inflammatory arthritis 88.5, acute knee injury 53.9, chronic knee injury 0.5 and OA 4.6. In contrast, aggrecan and sulfated glycosaminoglycan concentrations varied much less between groups, and only acute inflammatory arthritis and acute knee injury were found to have a two-fold increase in median levels compared to the reference. CONCLUSIONS Levels of aggrecan ARGS fragments in human synovial fluid are increased in human arthritis, OA and after knee injury, likely reflecting an enhanced cleavage at the 392Glu-393Ala bond in the IGD by aggrecanase. An assay that specifically quantified these fragments better distinguished samples from joints with pathology than assays monitoring aggrecan or glycosaminoglycan concentrations. The newly developed ARGS fragment assay can be used to monitor aggrecanase activity in human joint disease and experimental models.
Collapse
Affiliation(s)
- Staffan Larsson
- Department of Orthopaedics, Clinical Sciences Lund, Lund University, SE-221 85 Lund, Sweden.
| | | | | |
Collapse
|
42
|
Karsdal M, Henriksen K, Leeming D, Mitchell P, Duffin K, Barascuk N, Klickstein L, Aggarwal P, Nemirovskiy O, Byrjalsen I, Qvist P, Bay-Jensen A, Dam E, Madsen S, Christiansen C. Biochemical markers and the FDA Critical Path: How biomarkers may contribute to the understanding of pathophysiology and provide unique and necessary tools for drug development. Biomarkers 2009; 14:181-202. [DOI: 10.1080/13547500902777608] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
43
|
Struglics A, Larsson S, Hansson M, Lohmander LS. Western blot quantification of aggrecan fragments in human synovial fluid indicates differences in fragment patterns between joint diseases. Osteoarthritis Cartilage 2009; 17:497-506. [PMID: 19095471 DOI: 10.1016/j.joca.2008.09.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Accepted: 09/30/2008] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To develop a Western blot method for quantification of multiple aggrecan fragments in human synovial fluids (SFs). METHOD SF aggrecan fragments were prepared from knee healthy (reference), knee injury and arthritis subjects by CsCl gradient centrifugations collecting D1 fractions. Samples were analyzed by Western blot, using antibodies against the N-terminal epitope ARGS and the G3 domain, and fragments were quantified using a digital luminescence image analyzer. RESULTS The method had a coefficients of variation of 10-30%, and a high correlation (r(S)=0.86) with a corresponding enzyme-linked immunosorbent assay (ELISA). The SFs from reference, knee injured and arthritic subjects contained two major ARGS fragments, ARGS-SELE and ARGS-CS1, and three major G3 fragments (GRGT-G3, GLGS-G3 and AGEG-G3). Compared to the reference, the acute arthritis and acute joint injury groups had a 30-fold elevated concentration of ARGS fragments, and both groups had a higher proportion of the aggrecan in joint fluid as ARGS fragments compared to the other groups. The reference and chronic injury groups had an excess of ARGS-CS1 fragments over ARGS-SELE fragments, while subjects with acute arthritis or osteoarthritis had a more even distribution between these fragments. CONCLUSIONS We have developed a novel Western blot quantification method for quantification of SF aggrecan fragments which can differentiate fragments of different sizes sharing the same epitope. The anti-ARGS and anti-G3 quantitative Western blots provided information important for a better understanding of the proteolytic pathways in aggrecan breakdown, information that discriminates between different joint diseases, and may aid in identification of new biomarkers.
Collapse
Affiliation(s)
- A Struglics
- Department of Orthopaedics, Clinical Sciences Lund, Lund University, Sweden.
| | | | | | | |
Collapse
|
44
|
Karsdal MA, Madsen SH, Christiansen C, Henriksen K, Fosang AJ, Sondergaard BC. Cartilage degradation is fully reversible in the presence of aggrecanase but not matrix metalloproteinase activity. Arthritis Res Ther 2008; 10:R63. [PMID: 18513402 PMCID: PMC2483454 DOI: 10.1186/ar2434] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 05/11/2008] [Accepted: 05/30/2008] [Indexed: 12/16/2022] Open
Abstract
Introduction Physiological and pathophysiological cartilage turnover may coexist in articular cartilage. The distinct enzymatic processes leading to irreversible cartilage damage, compared with those needed for continuous self-repair and regeneration, remain to be identified. We investigated the capacity of repair of chondrocytes by analyzing their ability to initiate an anabolic response subsequent to three different levels of catabolic stimulation. Methods Cartilage degradation was induced by oncostatin M and tumour necrosis factor in articular cartilage explants for 7, 11, or 17 days. The catabolic period was followed by 2 weeks of anabolic stimulation (insulin growth factor-I). Cartilage formation was assessed by collagen type II formation (PIINP). Cartilage degradation was measured by matrix metalloproteinase (MMP) mediated type II collagen degradation (CTX-II), and MMP and aggrecanase mediated aggrecan degradation by detecting the 342FFGVG and 374ARGSV neoepitopes. Proteoglycan turnover, content, and localization were assessed by Alcian blue. Results Catabolic stimulation resulted in increased levels of cartilage degradation, with maximal levels of 374ARGSV (20-fold induction), CTX-II (150-fold induction), and 342FFGVG (30-fold induction) (P < 0.01). Highly distinct protease activities were found with aggrecanase-mediated aggrecan degradation at early stages, whereas MMP-mediated aggrecan and collagen degradation occurred during later stages. Anabolic treatment increased proteoglycan content at all time points (maximally, 250%; P < 0.001). By histology, we found a complete replenishment of glycosaminoglycan at early time points and pericellular localization at an intermediate time point. In contrast, only significantly increased collagen type II formation (200%; P < 0.01) was observed at early time points. Conclusion Cartilage degradation was completely reversible in the presence of high levels of aggrecanase-mediated aggrecan degradation. After induction of MMP-mediated aggrecan and collagen type II degradation, the chondrocytes had impaired repair capacity.
Collapse
Affiliation(s)
- Morten A Karsdal
- Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Denmark.
| | | | | | | | | | | |
Collapse
|
45
|
Patients with rheumatoid arthritis have an altered circulatory aggrecan profile. BMC Musculoskelet Disord 2008; 9:74. [PMID: 18507823 PMCID: PMC2426686 DOI: 10.1186/1471-2474-9-74] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Accepted: 05/28/2008] [Indexed: 11/11/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic auto-immune disease with extensive articular cartilage destruction. Aggrecan depletion, mediated by aggrecanases is one of the first signs of early cartilage erosion. We investigated, whether measurement of aggrecan and fragments thereof in serum, could be used as biomarkers for joint-disease in RA patients and furthermore characterized the fragments found in the circulation. Methods The study consisted of 38 patients, 12 males (62.2 ± 16.0 years) and 26 females (59.8 ± 20.7 years) diagnosed with RA: 41.5 ± 27.5 mm/h erythrocyte sedimentation rate (ESR), 38.4 ± 34.7 mg/ml C-reactive protein (CRP) and 4.8 ± 1.7 disease activity score (DAS) and 108 healthy age-matched controls. Aggrecan levels were measured using two immunoassays, i.e. the 374ARGSVI-G2 sandwich ELISA measuring aggrecanase-mediated aggrecan degradation and the G1/G2 sandwich assay, detecting aggrecan molecules containing G1 and/or G2 (total aggrecan) We further characterized serum samples by western blots, by using monoclonal antibodies F-78, binding to G1 and G2, or by BC-3, detecting the aggrecanase-generated N-terminal 374ARGSVI neo-epitope. Results Total aggrecan levels in RA patients were significantly decreased from 824.8 ± 31 ng/ml in healthy controls to 570.5 ± 30 ng/ml (31% decrease, P < 0.0001), as measured by the G1/G2 ELISA. Western blot analysis with F-78 showed one strong band at 10 kDa, and weaker bands at 25 and 45 kDa in both healthy controls and RA patients. In contrast, staining for aggrecanase-activity revealed only one strong band in RA patients of 45 kDa. Conclusion This is the first study, which characterizes different aggrecan fragments in human serum. The data strongly suggests that total aggrecan levels, i.e. aggrecan molecules containing G1 and/or G2 are lower in RA patients, and that RA patients have at least one specific subpopulation of aggrecan fragments, namely aggrecanse generated 374ARGSVI fragments. Further clinical studies are needed to investigate the potential of G1/G2 as a structure-related biochemical marker in destructive joint-diseases.
Collapse
|
46
|
Abstract
Treatment strategies for osteoarthritis most commonly involve the removal or replacement of damaged joint tissue. Relatively few treatments attempt to arrest, slow down or reverse the disease process. Such options include peri-articular osteotomy around the hip or knee, and treatment of femoro-acetabular impingement, where early intervention may potentially alter the natural history of the disease. A relatively small proportion of patients with osteoarthritis have a clear predisposing factor that is both suitable for modification and who present early enough for intervention to be deemed worthwhile. This paper reviews recent advances in our understanding of the pathology, imaging and progression of early osteoarthritis.
Collapse
Affiliation(s)
- T. C. B. Pollard
- Nuffield Department of Orthopaedic Surgery, University of Oxford, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford OX3 7LD, UK
| | - S. E. Gwilym
- Nuffield Department of Orthopaedic Surgery, University of Oxford, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford OX3 7LD, UK
| | - A. J. Carr
- Nuffield Department of Orthopaedic Surgery, University of Oxford, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford OX3 7LD, UK
| |
Collapse
|
47
|
Abstract
In arthritic diseases, the stability of the extracellular matrix of articular cartilage is compromised by extensive proteolytic breakdown associated with alterations of synthesis of the proteins of the tissue leading to cartilage loss. This article reviews developments in assays of biochemical markers of cartilage matrix turnover and studies investigating their use. Because type II collagen and aggrecan are the most abundant proteins of the cartilage matrix, current biochemical markers are based mainly on immunologic reagents detecting their synthesis and degradation. Clinical studies indicate that some markers of type II collagen may be useful to predict disease progression in osteoarthritis and rheumatoid arthritis. Conversely, major achievements have been made in the development of immunoassays detecting the various fragments of aggrecan released by matrix metalloproteases or aggrecanases, but their use has been limited mostly to investigating cartilage turnover in ex vivo experiments. Because of the complexity of the mechanisms involved in arthritic joint damage, only a combination of different biochemical markers reflecting the various aspects of synthesis and degradation of matrix molecules will likely provide efficient cartilage turnover monitoring.
Collapse
|
48
|
Sumer EU, Qvist P, Tankó LB. Matrix metalloproteinase and aggrecanase generated aggrecan fragments: implications for the diagnostics and therapeutics of destructive joint diseases. Drug Dev Res 2007. [DOI: 10.1002/ddr.20166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|