1
|
Port H, Coppers B, Tragl S, Manger E, Niemiec LM, Bayat S, Simon D, Fagni F, Corte G, Bay-Jensen AC, Tascilar K, Hueber AJ, Schmidt KG, Schönau V, Sticherling M, Heinrich S, Leyendecker S, Bohr D, Schett G, Kleyer A, Holm Nielsen S, Liphardt AM. Serum extracellular matrix biomarkers in rheumatoid arthritis, psoriatic arthritis and psoriasis and their association with hand function. Sci Rep 2025; 15:13656. [PMID: 40254635 PMCID: PMC12009959 DOI: 10.1038/s41598-025-98395-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 04/11/2025] [Indexed: 04/22/2025] Open
Abstract
Inflammatory arthritis, including rheumatoid arthritis and psoriatic arthritis, is characterized by physical function impairment. This becomes apparent even before arthritis onset, as in psoriasis (PsO). Chronic inflammation triggers an accelerated remodeling of the extracellular matrix (ECM), resulting in released ECM fragments detectable in blood. We aimed to investigate levels of blood-based ECM biomarkers in patients with RA, PsA, PsO, and healthy controls and to explore the association of ECM biomarkers with hand function impairments. Patients with RA (n = 85), PsA (n = 115), PsO (n = 102) and controls (n = 110) were included in this cross-sectional study. ECM catabolic (C1M, C2M, C3M, C4M, PRO-C4, C6M, ARG), formation (PRO-C1, PRO-C3, PRO-C6) and inflammation biomarkers (VICM) were measured in serum from all patients. Objective hand function (fine motor skills (Moberg-Picking-Up Test), isometric grip strength (dynamometer) and patient-perceived hand function (Michigan Hand Questionnaire (MHQ)) were assessed. Patients with RA and PsA received treatment with disease-modifying anti-rheumatic drugs. VICM levels were higher in RA, PsA, and PsO than in controls (p < 0.0001). PsA and PsO showed higher C4M levels compared to controls (p < 0.0001, p < 0.0001), while C6M was lower in patients with RA, PsA and PsO than in controls (p < 0.0001, p < 0.001, p < 0.01). PsO presented with higher levels of C1M compared to controls and to RA (p < 0.001 and p < 0.0001). PRO-C6 correlated negatively with MHQ (ρ = -0.39, p < 0.01) and grip strength (ρ = -0.31, p < 0.05) in PsO, while only weak correlations were observed between biomarkers and hand function scores for RA and PsA patients (all ρ < ± 0.2-0.3). Patients with RA, PsA, and PsO showed significant alterations in ECM remodeling biomarkers. Especially PsA and PsO had higher levels of inflammatory biomarkers compared to RA and controls, likely due to modulation by treatment. Predominantly in PsO, ECM formation biomarkers were associated with hand function impairments.
Collapse
Affiliation(s)
- Helena Port
- University of Copenhagen, Copenhagen, Denmark
- Nordic Bioscience, Herlev, Denmark
| | - Birte Coppers
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sonja Tragl
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Eva Manger
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine, Clinical Division of Internal Medicine II, Medical University Innsbruck/Tirol Kliniken GmbH, Innsbruck, Austria
| | - Lisa M Niemiec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Elisabeth-Krankenhaus Kassel, Kassel, Germany
| | - Sara Bayat
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Simon
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Charité Universitätsmedizin Berlin, Med. Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie Berlin, Berlin, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Giulia Corte
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Koray Tascilar
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Axel J Hueber
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Division of Rheumatology, Klinikum Nürnberg, Paracelsus Medical University, Nürnberg, Germany
| | - Katja G Schmidt
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Verena Schönau
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Sticherling
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Departement of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simon Heinrich
- Institute of Applied Dynamics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sigrid Leyendecker
- Institute of Applied Dynamics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Bohr
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Charité Universitätsmedizin Berlin, Med. Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie Berlin, Berlin, Germany
| | | | - Anna-Maria Liphardt
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
2
|
Li J, Sun H, Guan J, Li B, Jin C, Xie S, Liu Y. Immunogenicity of chondrocyte sheets: a review. Front Immunol 2025; 16:1529384. [PMID: 40124370 PMCID: PMC11926542 DOI: 10.3389/fimmu.2025.1529384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
The chondrocyte sheet is a sheet-like cell structure obtained by separating in vitro expanded and fused autologous chondrocytes from the bottom of the culture dish by physical means. The cell sheet contains autologous chondrocytes, extracellular matrix secreted by chondrocytes, and connective structures established between cells and matrix, and between cells and cells. In cartilage tissue engineering, chondrocyte sheets technology has great potential for the treatment of cartilage defects. Chondrocyte sheets have a low immunogenicity because they avoid the immune reaction caused by scaffolding materials. However, chondrocyte sheets can still cause severe local tissue swelling in the short term after implantation, resulting in a poor patient experience. In individual cases, an inflammatory reaction may even occur, leading to resorption of the chondrocyte sheet. This may be immunogenetically related to chondrocyte membrane surface-associated antigens, components of the extracellular matrix secreted by chondrocytes, and various bioactive components in the culture medium used during in vitro chondrocyte culture. Therefore, in order to investigate the causes of local tissue swelling and immune-inflammatory reactions induced by the implantation of chondrocyte sheets, this article reviews the immunogenicity of chondrocyte-associated antigens, components of the extracellular matrix of cartilage, and the active components of the cell culture medium.
Collapse
Affiliation(s)
- Juncen Li
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Huilin Sun
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaqi Guan
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Bohui Li
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Jin
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Shanhong Xie
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Liu
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Resthetic Biotechnology. Co., Ltd, Shanghai, China
| |
Collapse
|
3
|
Bjerre-Bastos JJ, Sejersen C, Nielsen HB, Boesen M, Secher NH, Distajo G, Flood V, Henrotin Y, Uebelhoer M, Krustrup P, Kitchen CC, Thudium CS, Andersen JR, Bihlet AR. The Impact of Weight-bearing Exercise, Non-Weight-bearing Exercise, and Cardiovascular Stress on Biochemical Markers of Cartilage Turnover in Patients With Mild to Moderate Knee Osteoarthritis: A Sequential, Cross-Over, Clinical Study. Cartilage 2024:19476035241258170. [PMID: 38853398 PMCID: PMC11569697 DOI: 10.1177/19476035241258170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/11/2024] Open
Abstract
OBJECTIVE To investigate how running, cycling, and sedentary cardiovascular stress impact biomarkers of cartilage turnover acutely in subjects with knee osteoarthritis (OA). DESIGN This was a sequential, cross-over, clinical study. Forty subjects with primary knee OA underwent moderate-to-high-intensity cycling, running, and adrenaline infusion on separate days. Blood was sampled before, during, and at 6-time points after intervention. On a control day, similar samples were taken. Biomarkers of type II collagen degradation (C2M, T2CM, Coll2-1, Coll2-1NO2), formation (PRO-C2), and aggrecan degradation (ARGS) were measured. RESULTS Mean age was 60.4 years, 40% were male, 45% had cumulated Kellgren-Lawrence (KL)-grade (Right + Left knee) of 2 to 3 and 55% had 4 to 6. Analyzing overall changes, area under the curve was significantly lower compared with resting values for ARGS and C2M after cycling and for ARGS after running. Considering individual time points, peak changes in biomarker levels showed reduction in C2M shortly following cycling (T20min = -12.3%, 95% confidence interval [CI]: -19.3% to -5.2%). PRO-C2 increased during cycling (T10min = 14.0%, 95% CI = 4.1% to 23.8%) and running (T20min = 16.5%, 95% CI = 4.3% to 28.6%). T2CM decreased after cycling (T50min = -19.9%, 95% CI = -29.2% to -10.6%), running (T50min = -22.8%, 95% CI = -32.1% to -13.5%), and infusion of adrenaline (peak, T50min = -9.8%, 95% CI = -20.0% to 0.4%). A latent increase was seen in Coll2-1 240 minutes after running (T260min = 21.7%, 95% CI = -1.6% to 45.1%). CONCLUSION Exercise had an impact on cartilage markers, but it did not suggest any detrimental effect on cartilage. Changes following adrenaline infusion suggest a sympathomimetic influence on the serological composition of biomarkers.
Collapse
Affiliation(s)
- Jonathan J. Bjerre-Bastos
- Xlab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- NBCD A/S, Herlev, Denmark
| | - Casper Sejersen
- Department of Anaesthesiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henning Bay Nielsen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Anaesthesia and Intensive Care Medicine, Zealand University Hospital, Roskilde, Denmark
- Department of Nutrition, Exercise and Sport, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Boesen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Radiology, Bispebjerg and Frederiksberg Hospitals, Copenhagen, Denmark
| | - Niels H. Secher
- Department of Anaesthesiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Yves Henrotin
- Department of Physical and Rehabilitation, VIVALIA, Marche-en-Famenne, Belgium
- Musculoskeletal Innovative Research Lab (mSKIL), University of Liège, Liège, Belgium
| | | | - Peter Krustrup
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Carl-Christian Kitchen
- Department of Anaesthesiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
4
|
Groen SS, Bay-Jensen AC, Thudium CS, Dziegiel MH, Skougaard M, Thomsen SF, Nielsen SH. Evaluating the inhibition of IL-17A and TNFα in a cartilage explant model cultured with Th17-derived cytokines. J Transl Autoimmun 2024; 8:100231. [PMID: 38292069 PMCID: PMC10826309 DOI: 10.1016/j.jtauto.2024.100231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Introduction T-helper 17 (Th17) cells produce IL-17A playing a critical role in activating the pathogenic chain leading to joint tissue inflammation and destruction. Elevated levels of Th17 cells and IL-17A have been detected in skin lesions, blood, and synovial fluid from patients with psoriatic arthritis (PsA) and ankylosing spondylitis (AS). Moreover, IL-17A inhibitors suppress disease activity in psoriasis, PsA and AS, supporting the evidence of IL-17A contributing to the disease pathogenesis. Although, IL-17A inhibitors are widely approved, it remains unclear how the inhibitory effect of IL-17A alters the extracellular matrix (ECM) of the joint in a Th17-conditioned inflammatory milieu. Therefore, the aim of this study was to establish a cartilage model cultured with conditioned medium from Th17 cells and inhibitors to explore the effect of IL-17A inhibition on joint tissue remodeling. Methods Naïve CD4+ T cells from healthy human buffy coat were differentiated into Th17 cells, followed by Th17 cell activation to secrete Th17-related cytokines and molecules into media. The activated Th17 cells were isolated from the conditioned media (CM) and analyzed using flow cytometry to verify Th17 cell differentiation. The CM were assessed with ELISA to quantify the concentrations of cytokines secreted into the media by the Th17 cells. Healthy bovine cartilage explants were cultured with the Th17-CM and treated with IL-17A and TNFα inhibitors for 21 days. In harvested supernatant from the cartilage cultures, MMP- and ADAMTS-mediated biomarker fragments of type II collagen, aggrecan, and fibronectin were measured by ELISA to investigate the ECM remodeling within the cartilage tissue. Results Th17-CM stimulated a catabolic response in the cartilage. Markers of type II collagen and aggrecan degradation were upregulated, while anabolic marker of type II collagen formation remained on similar levels as the untreated explants. The addition of IL-17A inhibitor to Th17-CM decreased the elevated type II collagen and aggrecan degradation, however, degenerative levels were still elevated compared to untreated group. The addition of TNFα inhibitor completely reduced both type II collagen and aggrecan degradation compared to untreated explants. Moreover, the TNFα inhibitor treatment did not alter the type II collagen formation compared to untreated group. Conclusion This study suggests that inhibition of IL-17A in Th17-conditioned cartilage tissue only partially reduced the MMP-mediated type II collagen degradation and ADAMTS-mediated aggrecan degradation, while the TNFα inhibitor treatment fully reduced both MMP- and ADAMTS-mediated ECM degradation. This exploratory study where ECM biomarkers are combined with Th17-conditioned ex vivo model may hold great potential as output for describing joint disease mechanisms and predicting structural effects of treatment on joint tissue.
Collapse
Affiliation(s)
- Solveig Skovlund Groen
- Immunoscience, Nordic Bioscience, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Morten H. Dziegiel
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Skougaard
- The Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Simon Francis Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark
| | - Signe Holm Nielsen
- Immunoscience, Nordic Bioscience, Herlev, Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
5
|
Hannani MT, Thudium CS, Karsdal MA, Ladel C, Mobasheri A, Uebelhoer M, Larkin J, Bacardit J, Struglics A, Bay-Jensen AC. From biochemical markers to molecular endotypes of osteoarthritis: a review on validated biomarkers. Expert Rev Mol Diagn 2024; 24:23-38. [PMID: 38353446 DOI: 10.1080/14737159.2024.2315282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) affects over 500 million people worldwide. OA patients are symptomatically treated, and current therapies exhibit marginal efficacy and frequently carry safety-risks associated with chronic use. No disease-modifying therapies have been approved to date leaving surgical joint replacement as a last resort. To enable effective patient care and successful drug development there is an urgent need to uncover the pathobiological drivers of OA and how these translate into disease endotypes. Endotypes provide a more precise and mechanistic definition of disease subgroups than observable phenotypes, and a panel of tissue- and pathology-specific biochemical markers may uncover treatable endotypes of OA. AREAS COVERED We have searched PubMed for full-text articles written in English to provide an in-depth narrative review of a panel of validated biochemical markers utilized for endotyping of OA and their association to key OA pathologies. EXPERT OPINION As utilized in IMI-APPROACH and validated in OAI-FNIH, a panel of biochemical markers may uncover disease subgroups and facilitate the enrichment of treatable molecular endotypes for recruitment in therapeutic clinical trials. Understanding the link between biochemical markers and patient-reported outcomes and treatable endotypes that may respond to given therapies will pave the way for new drug development in OA.
Collapse
Affiliation(s)
- Monica T Hannani
- ImmunoScience, Nordic Bioscience A/S, Herlev, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| | | | - Jonathan Larkin
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- SynOA Therapeutics, Philadelphia, PA, USA
| | - Jaume Bacardit
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - André Struglics
- Department of Clinical Sciences, Orthopaedics, Lund University, Lund, Sweden
| | | |
Collapse
|
6
|
Manon-Jensen T, Tangada S, Bager C, Chowdary P, Klamroth R, von Drygalski A, Windyga J, Escobar M, Frederiksen P, Engl W, Ewenstein B, Karsdal M. Evaluation of collagen turnover biomarkers as an objective measure for efficacy of treatment with rurioctocog alfa pegol in patients with hemophilia A: a secondary analysis of a randomized controlled trial. J Thromb Haemost 2024; 22:90-100. [PMID: 37717853 DOI: 10.1016/j.jtha.2023.08.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Patients with hemophilia who have recurrent hemarthroses develop hemophilic arthropathy (HA). Regular prophylaxis with factor (F) VIII (FVIII) can reduce HA, but there is a need for objective outcome measures to evaluate treatment efficacy. OBJECTIVES Evaluate and assess collagen turnover biomarkers in patients with hemophilia A to determine the efficacy of rurioctocog alfa pegol treatment and understand their potential as tools for guiding treatment decisions and monitoring outcomes. METHODS Joint remodeling was assessed by analyzing serum levels of collagen remodeling products at baseline and months 3, 6, 9, and 12 in a 98 patient subset receiving pharmacokinetics-guided prophylaxis with rurioctocog alfa pegol, targeting FVIII trough levels of 1 to 3 International Units (IU)/dL or 8 to 12 IU/dL (PROPEL study, NCT0285960). RESULTS Basement membrane metabolism-related type 4 collagen remodeling products (C4M and PRO-C4) decreased after 3 months at all time points by up to 25% at 1 to 3 IU/dL (P = .049, P < .0001) and 8 to 12 IU/dL FVIII trough levels (P = .0002, P < .0001). Interstitial tissue metabolism-related type 3 (C3M) and 5 (PRO-C5) collagen remodeling products decreased after 3 months, by up to 19% at 1 to 3 IU/dL FVIII trough level (P = .0001, P = .009) and 23% at 8 to 12 IU/dL FVIII trough level (P = .0002, P = .001). An increase of up to 12% was seen for cartilage metabolism-related type 2 collagen product (PRO-C2, not C2M) after 6 months at both trough levels (P = .01, P = .005). When stratified by prior treatment, changes in C3M (P = .03) and C4M (P = .02) levels were observed between trough levels for prior on-demand treatment but not for prophylaxis prior to study entry. CONCLUSION Joint improvement measured by collagen remodeling biomarkers specific to the basement membrane, interstitial matrix, and cartilage was seen with pharmacokinetics-guided prophylaxis. These collagen remodeling biomarkers warrant further exploration as biomarkers to guide treatment toward improvement in HA.
Collapse
Affiliation(s)
| | - Srilatha Tangada
- Takeda Development Center Americas, Inc, Cambridge, Massachusetts, USA
| | | | - Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free Hospital, London, UK
| | - Robert Klamroth
- Vascular Medicine and Haemostaseology, Vivantes Klinikum im Friedrichschain, Berlin, Germany
| | - Annette von Drygalski
- Hemophilia and Thrombosis Treatment Center, UC San Diego Health, San Diego, California, USA
| | - Jerzy Windyga
- Department of Hemostasis Disorders and Internal Medicine, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Miguel Escobar
- University of Texas Health and Science Center at Houston, Houston, Texas, USA
| | | | - Werner Engl
- Baxalta Innovations GmbH, a Takeda company, Vienna, Austria
| | - Bruce Ewenstein
- Takeda Development Center Americas, Inc, Cambridge, Massachusetts, USA
| | | |
Collapse
|
7
|
Bay-Jensen AC, Attur M, Samuels J, Thudium CS, Abramson SB, Karsdal MA. Pathological tissue formation and degradation biomarkers correlate with patient reported pain outcomes: an explorative study. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100379. [PMID: 37342785 PMCID: PMC10277584 DOI: 10.1016/j.ocarto.2023.100379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023] Open
Abstract
Background The lack of disease modifying drugs in Osteoarthritis (OA) may be attributed to the difficulty in robust response based on patient-reported outcomes (PROs) linked to drug mechanism of action. Joint tissue turnover biomarkers are associated with disease progression. A subset of patients has elevated serum levels of CRP metabolite (CRPM). This explorative study investigates the associations between PROs and joint tissue turnover markers in patients with high or low CRPM. Methods Serum of 146 knee OA patients of the New York Inflammation cohort and 21 healthy donors were assessed for biomarkers of collagen degradation (C1M, C2M, C3M, C4M), formation (PRO-C1, PRO-C2, PRO-C3, PRO-C4), and CRPM. Mean (SD) age was 62.5 (10.1); BMI, 26.6 (3.6); 62% women; and, 67.6% had symptomatic OA. WOMAC pain, stiffness, function, and total were recorded at baseline and at two-year follow-up. Associations were adjusted for race, sex, age, BMI, and NSAID. Results There was no difference in markers between donors and patients. C2M correlated with the WOMAC scores in all CRPM groups. Significant correlations were observed between PROs and PRO-C4, C1M, and C3M in the CRPMhigh group. The best predictive models for improvement were found for function and total with AUCs of 0.74 (p < 0.01) and 0.78 (p < 0.01). The best predictive models for worsening were found for function and total with AUCs of 0.84 (p < 0.01) and 0.80 (p < 0.05). Conclusion We hypothesize that collagen markers are prognostic tools for segregating patient populations in clinical trials.
Collapse
Affiliation(s)
| | - Mukundan Attur
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY-10003, USA
| | - Jonathan Samuels
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY-10003, USA
| | | | - Steven B. Abramson
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY-10003, USA
| | | |
Collapse
|
8
|
Port H, Holm Nielsen S, Frederiksen P, Madsen SF, Bay-Jensen AC, Sørensen IJ, Jensen B, Loft AG, Madsen OR, Østergaard M, Pedersen SJ. Extracellular matrix turnover biomarkers reflect pharmacodynamic effects and treatment response of adalimumab in patients with axial spondyloarthritis-results from two randomized controlled trials. Arthritis Res Ther 2023; 25:157. [PMID: 37626399 PMCID: PMC10463764 DOI: 10.1186/s13075-023-03132-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
OBJECTIVE To investigate if extracellular matrix (ECM) blood-based biomarkers reflect the pharmacodynamic effect and response to TNF-α inhibitor therapy (adalimumab, ADA), in patients with axial spondyloarthritis (axSpA). METHODS We investigated ECM biomarkers in two randomized, double-blind, placebo-controlled trials of axSpA patients (DANISH and ASIM, n = 52 and n = 49, respectively) receiving ADA 40 mg or placebo every other week for 12 and 6 weeks, respectively, and thereafter ADA to week 48. Serum concentrations of degraded type I (C1M), II (C2M, T2CM), III (C3M), IV (C4M), VI (C6M), type X (C10C) collagen; metabolite of C-reactive protein (CRPM), prolargin (PROM), citrullinated vimentin (VICM), calprotectin (CPa9-HNE); and formation of type II (PRO‑C2), III (PRO‑C3), and VI (PRO‑C6) turnover of type IV collagen (PRO-C4) were measured at baseline and weeks 6 or 12, 24, and 48. The pharmacodynamic effect and treatment response to ADA was evaluated by linear mixed models, and correlations between biomarkers and clinical scores were assessed by Spearman's correlation. RESULTS C1M, C3M, C4M, C6M, CRP, PRO-C4, and CPa9-HNE levels declined after 6 or 12 weeks in patients receiving ADA compared to placebo (all p < 0.05). Patients with AS Disease Activity Score C-reactive protein (ASDAS CRP) major improvement and/or clinically important improvement had significantly higher C1M, C3M, C4M, C6M, and PRO-C4 levels than patients with no/low improvement at baseline (all p < 0.05). Baseline levels of biomarkers showed weak to moderate correlations with ASDAS and structural damage scores. CONCLUSION ECM metabolites showed a pharmacodynamic effect and were associated with ASDAS response during TNF-α inhibitor treatment in patients with axSpA.
Collapse
Affiliation(s)
- Helena Port
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
- Nordic Bioscience A/S, Immunoscience, Herlev, Denmark.
| | - Signe Holm Nielsen
- Nordic Bioscience A/S, Immunoscience, Herlev, Denmark
- Biomedicine and Biotechnology, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Sofie Falkenløve Madsen
- Nordic Bioscience A/S, Immunoscience, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Inge Juul Sørensen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Rheumatology and Spine Diseases, Righospitalet, Copenhagen, Denmark
| | - Bente Jensen
- Department of Rheumatology and Spine Diseases, Righospitalet, Copenhagen, Denmark
| | - Anne Gitte Loft
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| | - Ole Rintek Madsen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Rheumatology and Spine Diseases, Righospitalet, Copenhagen, Denmark
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Mikkel Østergaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Rheumatology and Spine Diseases, Righospitalet, Copenhagen, Denmark
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Susanne Juhl Pedersen
- Department of Rheumatology and Spine Diseases, Righospitalet, Copenhagen, Denmark
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
9
|
Kwon EY, Abusharkh HA, Ruan X, Du D, Hammond-Pereira E, Van Wie BJ. Pd@Pt nanoparticle-linked immunosorbent assay for quantification of Collagen type II. Anal Chim Acta 2023; 1266:341265. [PMID: 37244654 PMCID: PMC10564550 DOI: 10.1016/j.aca.2023.341265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/01/2023] [Accepted: 04/23/2023] [Indexed: 05/29/2023]
Abstract
The evaluation of specific protein content in engineered tissues provides a gateway for developing regenerative medicine treatments. Since collagen type II, the major component of articular cartilage, is critical for the blossoming field of articular cartilage tissue engineering, the interest in this protein is growing rapidly. Accordingly, the need for quantification of collagen type II is increasing as well. In this study, we provide recent results for a new quantifying nanoparticle sandwich immunoassay technique for collagen type II. Since mesoporous palladium@platinum (Pd@Pt) nanoparticles have peroxidase-like catalytic activities, these nanoparticles were utilized in an enzyme-linked immunosorbent assay (ELISA)-like format to circumvent the need for traditional enzymes. These nanoparticles were easily conjugated with anti-collagen type II antibodies by the natural affinity interaction and used to develop a direct sandwich ELISA-like format for nanoparticle-linked immunosorbent assays. Using this method, we obtained a limit of detection of 1 ng mL-1, a limit of quantification of 9 ng mL-1. and a broad linear range between 1 ng mL-1 and 50 μg mL-1 for collagen type II with an average relative standard deviation of 5.5%, useable over a pH range of 7 - 9 at least. The assay was successfully applied to quantify collagen type II in cartilage tissues and compared with the results of commercial ELISAs and gene expression by reverse transcription-quantitative polymerase chain reaction. This method provides a thermally stable and cost-efficient alternative to traditional ELISAs. It also extends the application of nanoparticle-linked immunosorbent assays, thereby providing the potential to quantify other proteins and apply the technology in the medical, environmental, and biotechnology industry fields.
Collapse
Affiliation(s)
- Eunice Y Kwon
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164-6515, United States
| | - Haneen A Abusharkh
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164-6515, United States
| | - Xiaofan Ruan
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164-2920, United States
| | - Dan Du
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164-2920, United States
| | - Ellis Hammond-Pereira
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164-6515, United States
| | - Bernard J Van Wie
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164-6515, United States.
| |
Collapse
|
10
|
Kloosterman FR, Zwagemaker AF, Bay-Jensen AC, Cnossen MH, Kruip MJHA, Leebeek FWG, Hemke R, Maas M, Fijnvandraat K, Gouw SC, Coppens M. Poor correlation between biomarkers and MRI-detected joint damage in a cross-sectional study of persons with nonsevere hemophilia A (DYNAMO study). J Thromb Haemost 2023; 21:1813-1823. [PMID: 37019364 DOI: 10.1016/j.jtha.2023.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Persons with nonsevere hemophilia A (NSHA) experience less frequent joint bleeding than persons with severe hemophilia A, but may still develop joint damage. Biomarkers of cartilage and synovial remodeling can reflect ongoing pathologic processes that may precede or coincide with damage on joint imaging. If so, biomarkers may be an important diagnostic tool for joint damage in NSHA. OBJECTIVE To assess the correlation between biomarkers and MRI-detected joint damage in persons with NSHA. METHODS In a cross-sectional study, men with NSHA (factor VIII [FVIII], 2-35 IU/dL) were included. Participants underwent magnetic resonance imaging of elbows, knees, and ankles and blood and urine sampling for biomarker analysis on a single visit. The following biomarker(s) were analyzed in urine: CTX-II or serum: cartilage oligomeric matrix protein, chondroitin sulfate 846, vascular cell adhesion molecule 1, osteopontin (OPN), neo-epitope of MMP -mediated degradation of type II collagen, N-terminal propeptide of type II collagen, collagen type IV M, and propetide of type IV collagen. Spearman's rank correlations were calculated between these biomarkers and the total International Prophylaxis Study group (IPSG) score, soft-tissue subscore, and osteochondral subscore. RESULTS In total, 48 persons with NSHA were included. Median age was 43 years (range, 24-55 years) and median FVIII was 10 IU/dL (IQR, 4-16 IU/dL). The median IPSG score was 4 (IQR, 2-9). Median IPSG soft-tissue subscores were 3 (IQR, 2-4) and osteochondral subscores were 0 (IQR, 0-4). No strong correlations were found between the studied biomarkers, total IPSG score, subsequent soft-tissue, and osteochondral subscores. CONCLUSIONS In this study, selected biomarkers indicative of different aspects of hemophilic arthropathy showed no consistent correlation with IPSG scores. This suggests that systemically measured biomarkers are currently not suitable for identifying milder joint damage in NSHA, as observed on magnetic resonance imaging.
Collapse
Affiliation(s)
- Fabienne R Kloosterman
- Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Pediatric Hematology, Amsterdam, the Netherlands
| | - Anne-Fleur Zwagemaker
- Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Pediatric Hematology, Amsterdam, the Netherlands
| | - Anne C Bay-Jensen
- Immunoscience, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - Marjon H Cnossen
- Department of Pediatric Hematology and Oncology, Erasmus MC Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marieke J H A Kruip
- Department of Hematology, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Robert Hemke
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Mario Maas
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Karin Fijnvandraat
- Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Pediatric Hematology, Amsterdam, the Netherlands; Department of Molecular Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Samantha C Gouw
- Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Pediatric Hematology, Amsterdam, the Netherlands; Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Michiel Coppens
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension & Thrombosis, Amsterdam, the Netherlands.
| |
Collapse
|
11
|
Shih PC, Lee YH, Tsou HK, Cheng-Chung Wei J. Recent targets of osteoarthritis research. Best Pract Res Clin Rheumatol 2023; 37:101851. [PMID: 37422344 DOI: 10.1016/j.berh.2023.101851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/14/2023] [Indexed: 07/10/2023]
Abstract
Osteoarthritis is one of the most common diseases and poses a significant medical burden worldwide. Currently, the diagnosis and treatment of osteoarthritis primarily rely on clinical symptoms and changes observed in radiographs or other image modalities. However, identification based on reliable biomarkers would greatly improve early diagnosis, help with precise monitoring of disease progression, and provide aid for accurate treatment. In recent years, several biomarkers for osteoarthritis have been identified, including image modalities and biochemical biomarkers such as collagen degradation products, pro- or anti-inflammatory cytokines, micro RNAs, long non-coding RNAs, and circular RNAs. These biomarkers offer new insights in the pathogenesis of osteoarthritis and provide potential targets for further research. This article reviews the evolution of osteoarthritis biomarkers from the perspective of pathogenesis and emphasizes the importance of continued research to improve the diagnosis, treatment, and management of osteoarthritis.
Collapse
Affiliation(s)
- Po-Cheng Shih
- Department of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yung-Heng Lee
- Department of Orthopedics, Cishan Hospital, Ministry of Health and Welfare, Kaohsiung, Taiwan; Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Senior Services Industry Management, Minghsin University of Science and Technology, Hsinchu, Taiwan; Department of Recreation and Sport Management, Shu-Te University, Kaohsiung, Taiwan
| | - Hsi-Kai Tsou
- Functional Neurosurgery Division, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Houlong, Miaoli County, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Division of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
12
|
Kalogera S, Jansen MP, Bay-Jensen AC, Frederiksen P, Karsdal MA, Thudium CS, Mastbergen SC. Relevance of Biomarkers in Serum vs. Synovial Fluid in Patients with Knee Osteoarthritis. Int J Mol Sci 2023; 24:ijms24119483. [PMID: 37298434 DOI: 10.3390/ijms24119483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
The association between structural changes and pain sensation in osteoarthritis (OA) remains unclear. Joint deterioration in OA leads to the release of protein fragments that can either systemically (serum) or locally (synovial fluid; SF) be targeted as biomarkers and describe structural changes and potentially pain. Biomarkers of collagen type I (C1M), type II (C2M), type III (C3M), type X (C10C), and aggrecan (ARGS) degradation were measured in the serum and SF of knee OA patients. Spearman's rank correlation was used to assess the correlation of the biomarkers' levels between serum and SF. Linear regression adjusted for confounders was used to evaluate the associations between the biomarkers' levels and clinical outcomes. The serum C1M levels were negatively associated with subchondral bone density. The serum C2M levels were negatively associated with KL grade and positively associated with minimum joint space width (minJSW). The C10C levels in SF were negatively associated with minJSW and positively associated with KL grade and osteophyte area. Lastly, the serum C2M and C3M levels were negatively associated with pain outcomes. Most of the biomarkers seemed to mainly be associated with structural outcomes. The overall biomarkers of extracellular matrix (ECM) remodeling in serum and SF may provide different information and reflect different pathogenic processes.
Collapse
Affiliation(s)
- Stefania Kalogera
- Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark
- Department of Drug Design and Pharmacology, Copenhagen University, 1165 Copenhagen, Denmark
| | - Mylène P Jansen
- Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | | | | | - Morten A Karsdal
- Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark
| | | | - Simon C Mastbergen
- Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Regenerative Medicine Center, Utrecht University, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
13
|
Thudium CS, Engstrøm A, Bay-Jensen AC, Frederiksen P, Jansen N, De Zwart A, van der Leeden M, Dekker J, Lems W, Roorda L, van Spil WE, Van der Esch M. Cartilage tissue turnover increases with high- compared to low-intensity resistance training in patients with knee OA. Arthritis Res Ther 2023; 25:22. [PMID: 36765372 PMCID: PMC9912672 DOI: 10.1186/s13075-023-03000-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
OBJECTIVES To investigate cartilage tissue turnover in response to a supervised 12-week exercise-related joint loading training program followed by a 6-month period of unsupervised training in patients with knee osteoarthritis (OA). To study the difference in cartilage tissue turnover between high- and low-resistance training. METHOD Patients with knee OA were randomized into either high-intensity or low-intensity resistance supervised training (two sessions per week) for 3 months and unsupervised training for 6 months. Blood samples were collected before and after the supervised training period and after the follow-up period. Biomarkers huARGS, C2M, and PRO-C2, quantifying cartilage tissue turnover, were measured by ELISA. Changes in biomarker levels over time within and between groups were analyzed using linear mixed models with baseline values as covariates. RESULTS huARGS and C2M levels increased after training and at follow-up in both low- and high-intensity exercise groups. No changes were found in PRO-C2. The huARGS level in the high-intensity resistance training group increased significantly compared to the low-intensity resistance training group after resistance training (p = 0.029) and at follow-up (p = 0.003). CONCLUSION Cartilage tissue turnover and cartilage degradation appear to increase in response to a 3-month exercise-related joint loading training program and at 6-month follow-up, with no evident difference in type II collagen formation. Aggrecan remodeling increased more with high-intensity resistance training than with low-intensity exercise. These exploratory biomarker results, indicating more cartilage degeneration in the high-intensity group, in combination with no clinical outcome differences of the VIDEX study, may argue against high-intensity training.
Collapse
Affiliation(s)
- Christian S. Thudium
- grid.436559.80000 0004 0410 881XImmunoscience, Nordic Bioscience, Herlev, Denmark
| | - Amalie Engstrøm
- grid.436559.80000 0004 0410 881XImmunoscience, Nordic Bioscience, Herlev, Denmark ,grid.5254.60000 0001 0674 042XFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Peder Frederiksen
- grid.436559.80000 0004 0410 881XImmunoscience, Nordic Bioscience, Herlev, Denmark
| | - Nuria Jansen
- grid.418029.60000 0004 0624 3484Reade Centre for Rehabilitation and Rheumatology, Amsterdam, the Netherlands
| | - Arjan De Zwart
- grid.418029.60000 0004 0624 3484Reade Centre for Rehabilitation and Rheumatology, Amsterdam, the Netherlands
| | - Marike van der Leeden
- grid.418029.60000 0004 0624 3484Reade Centre for Rehabilitation and Rheumatology, Amsterdam, the Netherlands ,grid.12380.380000 0004 1754 9227Department of Rehabilitation Medicine, Amsterdam Institute of Public Health, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan, 1117 Amsterdam, the Netherlands
| | - Joost Dekker
- grid.12380.380000 0004 1754 9227Department of Rehabilitation Medicine, Amsterdam Institute of Public Health, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan, 1117 Amsterdam, the Netherlands
| | - Willem Lems
- grid.418029.60000 0004 0624 3484Reade Centre for Rehabilitation and Rheumatology, Amsterdam, the Netherlands ,grid.12380.380000 0004 1754 9227Department of Rheumatology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Leo Roorda
- grid.418029.60000 0004 0624 3484Reade Centre for Rehabilitation and Rheumatology, Amsterdam, the Netherlands
| | - Willem Evert van Spil
- Dijklander Hospital, Hoorn, the Netherlands ,grid.7692.a0000000090126352University Medical Center Utrecht, Utrecht, the Netherlands
| | - Martin Van der Esch
- grid.418029.60000 0004 0624 3484Reade Centre for Rehabilitation and Rheumatology, Amsterdam, the Netherlands ,grid.431204.00000 0001 0685 7679Centre of Expertise Urban Vitality, Faculty of Health, Amsterdam University of Applied Sciences, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Port H, Bay-Jensen AC, He Y, Karsdal MA, Gantzel T, Thudium CS, Holm Nielsen S. A Highly Sensitive Biomarker of Type II Collagen C-Terminal Pro-Peptide Associated with Cartilage Formation. Int J Mol Sci 2022; 24:ijms24010454. [PMID: 36613894 PMCID: PMC9820484 DOI: 10.3390/ijms24010454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
The type II collagen C-terminal pro-peptide is one of the most abundant polypeptides in cartilage. The purpose of this study was to develop a competitive chemiluminescence enzyme-linked immunosorbent assay, CALC2, targeting this pro-peptide as a marker of cartilage formation. Technical assay parameters were evaluated. CALC2 level was measured after in vitro cleavage of recombinant type II collagen with bone morphogenetic protein-1 (BMP-1) and treatment of ex vivo human osteoarthritis (OA) cartilage explant model (HEX) with insulin-like growth factor-1 (IGF-1). Serum CALC2 levels were assessed in 18 patients with rheumatoid arthritis (RA), 19 patients with ankylosing spondylitis (AS), and 18 age- and sex-matched controls in cohort 1 and 8 patients with OA and 14 age- and sex-matched controls in cohort 2. Type II collagen cleavage with BMP-1 increased the CALC2 level. IGF-1 treatment increased the CALC2 levels in HEX compared with the untreated explants (p < 0.05). Results were confirmed using Western blot analysis. CALC2 levels were decreased in the patients with RA and AS compared with the healthy controls (p = 0.01 and p = 0.02, respectively). These findings indicate that CALC2 may be a novel biomarker of type II collagen formation. However, further preclinical and clinical studies are required to validate these findings.
Collapse
Affiliation(s)
- Helena Port
- Biomarkers and Research, Nordic Bioscience, 2730 Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 København, Denmark
- Correspondence: ; Tel.: +45-445-252-52
| | | | - Yi He
- Biomarkers and Research, Nordic Bioscience, 2730 Herlev, Denmark
| | | | - Thorbjørn Gantzel
- Orthopedic Surgery Unit, Gentofte University Hospital, 2820 Gentofte, Denmark
| | | | | |
Collapse
|
15
|
Holm Nielsen S, Sun S, Bay-Jensen AC, Karsdal M, Sørensen IJ, Weber U, Loft AG, Kollerup G, Thamsborg G, Madsen OR, Møller J, Østergaard M, Pedersen SJ. Levels of extracellular matrix metabolites are associated with changes in Ankylosing Spondylitis Disease Activity Score and MRI inflammation scores in patients with axial spondyloarthritis during TNF inhibitor therapy. Arthritis Res Ther 2022; 24:279. [PMID: 36564778 PMCID: PMC9783808 DOI: 10.1186/s13075-022-02967-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/PURPOSE In axial spondyloarthritis (axSpA) inflammation of the sacroiliac joints and spine is associated with local extracellular matrix (ECM) remodeling of affected tissues. We aimed to investigate the association of ECM metabolites with treatment response in axSpA patients treated with TNF-α inhibitory therapy for 46 weeks. METHODS In a prospective clinical study of axSpA patients (n=55) initiating a TNF inhibitor (infliximab, etanercept, or adalimumab), serum concentrations of formation of type I (PRO-C1), type III (PRO-C3), and type VI (PRO-C6) collagen; turnover of type IV collagen (PRO-C4), and matrix-metalloproteinase (MMP)-degraded type III (C3M) collagen, MMP-degraded type IV (C4M), type VI (C6M), and type VII (C7M) collagen, and cathepsin-degraded type X collagen (C10C), MMP-mediated metabolite of C-reactive protein (CRPM), citrullinated vimentin (VICM), and neutrophil elastase-degraded elastin (EL-NE) were measured at baseline, week 2, week 22, and week 46. RESULTS Patients were mostly males (82%), HLA-B27 positive (84%), with a median age of 40 years (IQR: 32-48), disease duration of 5.5 years (IQR: 2-10), and a baseline Ankylosing Spondylitis Disease Activity Score (ASDAS) of 3.9 (IQR: 3.0-4.5). Compared to baseline, PRO-C1 levels were significantly increased after two weeks of treatment, C6M levels were significantly decreased after two and 22 weeks (repeated measures ANOVA, p=0.0014 and p=0.0015, respectively), EL-NE levels were significantly decreased after 2 weeks (p=0.0008), VICM levels were significantly decreased after two and 22 weeks (p=0.0163 and p=0.0374, respectively), and CRP were significantly decreased after two and 22 weeks (both p=0.0001). Baseline levels of PRO-C1, PRO-C3, C6M, VICM, and CRP were all associated with ASDAS clinically important and major improvement after 22 weeks (ΔASDAS ≥1.1) (Mann-Whitney test, p=0.006, p=0.008, p<0.001, <0.001, <0.001, respectively), while C6M, VICM and CRP levels were associated with ASDAS clinically important and major improvement after 46 weeks (ΔASDAS ≥2.0) (p=0.002, p=0.044, and p<0.001, respectively). PRO-C1 and C6M levels were associated with a Bath AS Disease Activity Score (BASDAI) response to TNF-inhibitory therapy after 22 weeks (Mann-Whitney test, p=0.020 and p=0.049, respectively). Baseline levels of PRO-C4 and C6M were correlated with the total SPARCC MRI Spine and Sacroiliac Joint Inflammation score (Spearman's Rho ρ=0.279, p=0.043 and ρ=0.496, p=0.0002, respectively). CONCLUSIONS Extracellular matrix metabolites were associated with ASDAS response, MRI inflammation, and clinical treatment response during TNF-inhibitory treatment in patients with axSpA.
Collapse
Affiliation(s)
- Signe Holm Nielsen
- Nordic Bioscience, Herlev, Denmark.
- Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| | - Shu Sun
- Nordic Bioscience, Herlev, Denmark
| | | | | | - Inge Juul Sørensen
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ulrich Weber
- Danish Hospital for Rheumatic Diseases, University Hospital of Southern Denmark, Sønderborg, Denmark
- Practice Buchsbaum, Rheumatology, Schaffhausen, Switzerland
| | - Anne Gitte Loft
- Departments of Rheumatology, Hospital Lillebælt, Vejle, Denmark
- Aarhus University Hospital, Aarhus, Denmark
| | - Gina Kollerup
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gorm Thamsborg
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ole Rintek Madsen
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Møller
- Department of Radiology, Herlev Hospital, Copenhagen, Denmark
| | - Mikkel Østergaard
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Juhl Pedersen
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Dynamic compression inhibits cytokine-mediated type II collagen degradation. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100292. [DOI: 10.1016/j.ocarto.2022.100292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
|
17
|
Schett G, Loza MJ, Palanichamy A, FitzGerald O, Ritchlin C, Bay-Jensen AC, Nielsen SH, Gao S, Hsia EC, Kollmeier AP, Xu XL, Baribaud F, Sweet K. Collagen Turnover Biomarkers Associate with Active Psoriatic Arthritis and Decrease with Guselkumab Treatment in a Phase 3 Clinical Trial (DISCOVER-2). Rheumatol Ther 2022; 9:1017-1030. [PMID: 35352313 PMCID: PMC9314487 DOI: 10.1007/s40744-022-00444-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Guselkumab, a novel interleukin-23p19 subunit monoclonal antibody, has been shown to effectively improve the diverse manifestations of active psoriatic arthritis (PsA) in two phase 3 trials (DISCOVER-1, DISCOVER-2). Serum concentrations of extracellular matrix (ECM) biomarkers at baseline and following treatment with guselkumab were evaluated in patients with active PsA, and the relationship of these biomarkers with baseline PsA characteristics and clinical response to guselkumab treatment was explored. METHODS Serum samples were collected at weeks 0, 4, 24, and 52 from a selected subset (N = 260) of the 739 biologic-naïve patients with PsA treated with guselkumab 100 mg every 4 or 8 weeks or placebo in DISCOVER-2. Demographically matched healthy controls (N = 76) were used for comparison. The samples were analyzed for ECM biomarkers associated with collagen degradation (C1M, C2M, C3M, C4M, C6M, C10C) and collagen formation (PRO-C1, PRO-C2, PRO-C3, PRO-C4, PRO-C6). RESULTS Baseline concentrations of collagen degradation biomarkers C1M, C3M, C4M, and C6M and collagen formation biomarkers PRO-C3 and PRO-C6 were significantly higher (i.e., ≥ 1.25-fold and false discovery rate adjusted p < 0.05) in PsA patients than in healthy controls. Serum C1M, C3M, C4M, and C6M levels declined from baseline in guselkumab-treated patients in both dosing regimens. In addition, guselkumab-treated ACR20 responders (≥ 20% improvement in American College of Rhematology response criteria) had significantly lower C1M levels than ACR20 nonresponders. CONCLUSION These data demonstrate that serum collagen biomarkers are elevated in patients with PsA compared with healthy controls and that treatment with guselkumab decreases levels of C1M, C3M, C4M, and C6M. Importantly, C1M serves as a biomarker that associates with improvement of joint signs and symptoms. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03158285.
Collapse
Affiliation(s)
- Georg Schett
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.
| | - Matthew J Loza
- Janssen Research & Development LLC, Spring House, PA, USA
| | | | - Oliver FitzGerald
- School of Medicine, Conway Institute for Biomolecular Research, University College Dublin, Dublin 4, Ireland
| | | | | | | | - Sheng Gao
- Janssen Research & Development LLC, Spring House, PA, USA
| | - Elizabeth C Hsia
- Janssen Research & Development LLC, Spring House, PA, USA
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | - Xie L Xu
- Janssen Research & Development, LLC, San Diego, CA, USA
| | | | - Kristen Sweet
- Janssen Research & Development LLC, Spring House, PA, USA
| |
Collapse
|
18
|
Port H, Nielsen SH, Madsen SF, Bay-Jensen AC, Karsdal M, Seven S, Sørensen IJ, Morsel-Carlsen L, Østergaard M, Pedersen SJ. Extracellular matrix protein turnover markers are associated with axial spondyloarthritis-a comparison with postpartum women and other non-axial spondyloarthritis controls with or without back pain. Arthritis Res Ther 2022; 24:152. [PMID: 35739562 PMCID: PMC9219155 DOI: 10.1186/s13075-022-02839-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022] Open
Abstract
Background Axial spondyloarthritis (axSpA) is a common chronic inflammatory disease, associated with extracellular matrix (ECM) remodeling of the cartilage, bone, and connective tissues. The primary symptom of axSpA is back pain, caused by inflammation. However, there is a medical need to truly identify patients with axSpA from other subjects with buttock or low back pain attributable to other reasons. We aimed to investigate circulating biomarkers of ECM/inflammation (MMP-degraded type I (C1M), II (C2M, T2CM), III (C3M), IV (C4M), VI (C6M), and X (C10C, COL10NC) collagens, CRPM, PROM and VICM) and ECM formation of type II (PRO-C2), III (PRO-C3), IV (PRO-C4), and VI (PRO-C6) collagens as potential biomarkers to identify patients with axSpA. Methods We measured biomarkers from a cross-sectional study with 204 participants by enzyme-linked immunosorbent assay (ELISA). The study included axSpA patients (N = 41), women with postpartum buttock/pelvic pain (N = 46), disc herniation (N = 25), and a group of healthy subjects (including women without postpartum pelvic pain (N = 14), subjects with various types of physical strain (cleaning staff (N = 26) long-distance runners (N = 23)), and healthy men (N = 29)). Differences between the groups were calculated by ANCOVA and AUC, while Spearman’s correlations were performed with ECM biomarkers and clinical scores. Results Patients with axSpA expressed significantly higher levels of C1M, C4M, and VICM (p < 0.05-p < 0.0001) compared to all the non-axSpA control groups. Further, C6M and PRO-C4 were significantly higher in patients with axSpA (both p < 0.0001) compared to women with postpartum pelvic pain and healthy subjects, whereas PRO-C3 was significantly lower compared to healthy subjects (p = 0.01). The best ECM common biomarker to differentiate between axSpA and the non-axSpA control groups was PRO-C4 (AUC ≥ 0.75; specificity ≥ 0.79, sensitivity = 0.65). Mild correlations were observed between collagen turnover and inflammation biomarkers and CRP and MRI (ρ ≥ 0.3; p < 0.05-p < 0.001). Conclusions Biomarkers of type I, IV, and VI collagen and biomarkers of inflammation showed an altered turnover in patients with axSpA compared with the non-axSpA control groups. Such biomarkers may be useful in combination with MRI or independently to separate patients with axSpA from other back pain conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02839-1.
Collapse
Affiliation(s)
- Helena Port
- Nordic Bioscience A/S, Immunoscience, Herlev hovedgade 207, 2730, Herlev, Denmark. .,University of Copenhagen, Clinical Medicine, Copenhagen, Denmark.
| | - Signe Holm Nielsen
- Nordic Bioscience A/S, Immunoscience, Herlev hovedgade 207, 2730, Herlev, Denmark.,Technical University of Denmark, Biomedicine and Biotechnology, Kgs. Lyngby, Denmark
| | | | | | - Morten Karsdal
- Nordic Bioscience A/S, Immunoscience, Herlev hovedgade 207, 2730, Herlev, Denmark
| | - Sengül Seven
- Rigshospitalet, Center for Arthritis Research, Glostrup, Denmark
| | - Inge Juul Sørensen
- University of Copenhagen, Clinical Medicine, Copenhagen, Denmark.,Rigshospitalet, Center for Arthritis Research, Glostrup, Denmark
| | | | - Mikkel Østergaard
- University of Copenhagen, Clinical Medicine, Copenhagen, Denmark.,Rigshospitalet, Center for Arthritis Research, Glostrup, Denmark
| | | |
Collapse
|
19
|
Wagner N, Rapp AE, Braun S, Ehnert M, Imhof T, Koch M, Jenei-Lanzl Z, Zaucke F, Meurer A. Generation of Matrix Degradation Products Using an In Vitro MMP Cleavage Assay. Int J Mol Sci 2022; 23:ijms23116245. [PMID: 35682922 PMCID: PMC9181598 DOI: 10.3390/ijms23116245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 02/04/2023] Open
Abstract
Matrix metalloproteinases (MMPs) play crucial roles in tissue homeostasis and pathologies by remodeling the extracellular matrix. Previous studies have demonstrated the biological activities of MMP-derived cleavage products. Furthermore, specific fragments can serve as biomarkers. Therefore, an in vitro cleavage assay to identify substrates and characterize cleavage patterns could provide important insight in disease-relevant mechanisms and the identification of novel biomarkers. In the pathogenesis of osteoarthritis (OA), MMP-2, -8, -9 and -13 are of vital importance. However, it is unclear which protease can cleave which matrix component. To address this question, we established an in vitro cleavage assay using recombinantly expressed MMPs and the two cartilage matrix components, COMP and thrombospondin-4. We found a time- and concentration-dependent degradation and an MMP-specific cleavage pattern for both proteins. Cleavage products can now be enriched and purified to investigate their biological activity. To verify the in vivo relevance, we compared the in vitro cleavage patterns with serum and synovial fluid from OA patients and could indeed detect fragments of similar size in the human samples. The cleavage assay can be adapted to other MMPs and substrates, making it a valuable tool for many research fields.
Collapse
Affiliation(s)
- Niklas Wagner
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (N.W.); (A.E.R.); (Z.J.-L.); (A.M.)
| | - Anna E. Rapp
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (N.W.); (A.E.R.); (Z.J.-L.); (A.M.)
| | - Sebastian Braun
- Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (S.B.); (M.E.)
| | - Markus Ehnert
- Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (S.B.); (M.E.)
| | - Thomas Imhof
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (T.I.); (M.K.)
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (T.I.); (M.K.)
| | - Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (N.W.); (A.E.R.); (Z.J.-L.); (A.M.)
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (N.W.); (A.E.R.); (Z.J.-L.); (A.M.)
- Correspondence:
| | - Andrea Meurer
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (N.W.); (A.E.R.); (Z.J.-L.); (A.M.)
- Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, 60528 Frankfurt am Main, Germany; (S.B.); (M.E.)
| |
Collapse
|
20
|
Angelini F, Widera P, Mobasheri A, Blair J, Struglics A, Uebelhoer M, Henrotin Y, Marijnissen AC, Kloppenburg M, Blanco FJ, Haugen IK, Berenbaum F, Ladel C, Larkin J, Bay-Jensen AC, Bacardit J. Osteoarthritis endotype discovery via clustering of biochemical marker data. Ann Rheum Dis 2022; 81:666-675. [PMID: 35246457 DOI: 10.1136/annrheumdis-2021-221763] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/01/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Osteoarthritis (OA) patient stratification is an important challenge to design tailored treatments and drive drug development. Biochemical markers reflecting joint tissue turnover were measured in the IMI-APPROACH cohort at baseline and analysed using a machine learning approach in order to study OA-dominant phenotypes driven by the endotype-related clusters and discover the driving features and their disease-context meaning. METHOD Data quality assessment was performed to design appropriate data preprocessing techniques. The k-means clustering algorithm was used to find dominant subgroups of patients based on the biochemical markers data. Classification models were trained to predict cluster membership, and Explainable AI techniques were used to interpret these to reveal the driving factors behind each cluster and identify phenotypes. Statistical analysis was performed to compare differences between clusters with respect to other markers in the IMI-APPROACH cohort and the longitudinal disease progression. RESULTS Three dominant endotypes were found, associated with three phenotypes: C1) low tissue turnover (low repair and articular cartilage/subchondral bone turnover), C2) structural damage (high bone formation/resorption, cartilage degradation) and C3) systemic inflammation (joint tissue degradation, inflammation, cartilage degradation). The method achieved consistent results in the FNIH/OAI cohort. C1 had the highest proportion of non-progressors. C2 was mostly linked to longitudinal structural progression, and C3 was linked to sustained or progressive pain. CONCLUSIONS This work supports the existence of differential phenotypes in OA. The biomarker approach could potentially drive stratification for OA clinical trials and contribute to precision medicine strategies for OA progression in the future. TRIAL REGISTRATION NUMBER NCT03883568.
Collapse
Affiliation(s)
| | - Paweł Widera
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.,Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht, The Netherlands.,Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China.,World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Liege, Belgium
| | - Joseph Blair
- ImmunoScience, Nordic Bioscience, Herlev, Denmark
| | - André Struglics
- Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund University, Lund, Sweden
| | | | - Yves Henrotin
- Artialis SA, Liège, Belgium.,Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium
| | | | - Margreet Kloppenburg
- Rheumatology, Leiden Universitair Medisch Centrum, Leiden, The Netherlands.,Department of Clinical Epidemiology, Leiden Universitair Medisch Centrum, Leiden, The Netherlands
| | - Francisco J Blanco
- Servicio de Reumatologia, INIBIC-Hospital Universitario A Coruña, A Coruña, Spain
| | - Ida K Haugen
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Francis Berenbaum
- Institut national de la santé et de la recherche médicale, Sorbonne Université, Paris, France
| | | | | | | | - Jaume Bacardit
- School of Computing, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
21
|
Bay-Jensen AC, Mobasheri A, Thudium CS, Kraus VB, Karsdal MA. Blood and urine biomarkers in osteoarthritis - an update on cartilage associated type II collagen and aggrecan markers. Curr Opin Rheumatol 2022; 34:54-60. [PMID: 34652292 PMCID: PMC8635261 DOI: 10.1097/bor.0000000000000845] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a painful disease for which drug development has proven difficult. One major reason for this is the heterogeneity of the disease and the current lack of operationalized means to distinguish various disease endotypes (molecular subtypes). Biomarkers measured in blood or urine, reflecting joint tissue turnover, have been developed and tested during the last decades. In this narrative review, we provide highlights on biomarkers derived from the two most studied and abundant cartilage proteins - type II collagen and aggrecan. RECENT FINDINGS Multiple biomarkers assessing type II collagen degradation and formation, and aggrecan turnover have been developed. Several markers, such as uCTX-II, have been validated for their association with disease severity and prognosis, as well as pharmacodynamically used to describe the mode of action and efficacy of drugs in development. There is a great need for biomarkers for subdividing patients (i.e., endotyping) and recent scientific advances have not yet come closer to achieving this goal. SUMMARY There is strong support for using biomarkers for understanding OA, reflecting degradation and formation of the joint tissues, focused on type II collagen and aggrecan. There is still a lack of in vitro diagnostics, in all contexts of use.
Collapse
Affiliation(s)
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- University Medical Center Utrecht, Department of Orthopedics, Rheumatology and Clinical Immunology, Utrecht, The Netherlands
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, University of Liege, Liege, Belgium
| | | | - Virginia B. Kraus
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | |
Collapse
|
22
|
Low levels of type II collagen formation (PRO-C2) are associated with response to sprifermin: a pre-defined, exploratory biomarker analysis from the FORWARD study. Osteoarthritis Cartilage 2022; 30:92-99. [PMID: 34737064 DOI: 10.1016/j.joca.2021.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/08/2021] [Accepted: 10/21/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is characterized by the gradual loss of cartilage. Sprifermin, a recombinant FGF18, is being developed as a cartilage anabolic drug. PRO-C2 is a serum marker of type II collagen formation and low levels have been shown to be prognostic of radiographic progression. The aim of the study was to investigate whether the patient groups with either high or low PRO-C2 levels responded differently to sprifermin. DESIGN PRO-C2 was measured in synovial fluid (SF) (n = 59) and serum samples (n = 225) from participants of the FORWARD study, a 2-year phase IIb clinical trial testing the efficacy of intra-articular (IA) sprifermin over placebo. The difference between sprifermin and placebo in respect to in change cartilage thickness (measured by quantitative (q) MRI) was analyzed in groups with either high or low (3rd vs 1st-2nd tertiles) baseline serum PRO-C2 levels. RESULTS SF levels of PRO-C2 increased over time in response to sprifermin, but not to placebo. In the placebo arm, significantly (p = 0.005) more cartilage was lost in the low vs high PRO-C2 group over the 2-year period. The contrast between sprifermin and placebo was significant (p < 0.001), ranging from 0.104 mm at week 26 to 0.229 mm at week 104 in the low PRO-C2 group. This result was not significant in the high PRO-C2 group ranging from -0.034 to 0.142. CONCLUSIONS Patients with low serum PRO-C2 levels lost more cartilage thickness over time and grew more cartilage in response to sprifermin vs a placebo when compared to patients with high PRO-C2 levels.
Collapse
|
23
|
Lorenzo-Gómez R, Casero-Álvarez A, Miranda-Castro R, García-Ocaña M, de Los Toyos JR, de-Los-Santos-Alvarez N, Lobo-Castañón MJ. A competitive assay for the detection of a 16-mer peptide from α1 chain of human collagen XI. Talanta 2021; 240:123196. [PMID: 34998145 DOI: 10.1016/j.talanta.2021.123196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 11/26/2022]
Abstract
Characterization of extracellular matrix (ECM) is becoming more and more important to decipher cancer progression. Constant remodeling results in ECM components degradation or unusual ECM accumulation that releases short fragments to the body fluids. These fragments might be potential cancer biomarkers but to detect them specific receptors are needed. In response to this demand, we present the first electrochemical aptamer-based competitive assay for the minor collagen XI, dysregulated in several carcinomas. It was performed on magnetic beads using enzymatic labeling. First, we selected the most appropriate tag for the aptamer (biotin or 6-carboxyfluorescein). The former yielded higher currents by chronoamperometry and it was used for the competitive assay. The collagen fragment, a 16mer peptide used as the target, was detected from 52 to 1000 nM with an RSD of about 5%. The LOD of the assay was estimated as 24 nM (44 ng/mL). The performance of the assay in serum diluted 1:2 was equivalent to the assay in PBS. The detection of α1 chain of human collagen XI was also possible in cell lysates and confirmed by aptacytofluorescence, which is promising as a new tool to validate this fragment as a cancer biomarker.
Collapse
Affiliation(s)
- Ramón Lorenzo-Gómez
- Departamento de Química Física y Analítica. Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.
| | - Alfonso Casero-Álvarez
- Departamento de Química Física y Analítica. Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain.
| | - Rebeca Miranda-Castro
- Departamento de Química Física y Analítica. Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.
| | - Marcos García-Ocaña
- Unidad de Biotecnología y Ensayos Biomédicos. Servicios Científico Técnicos, Universidad de Oviedo, Oviedo, Spain.
| | - Juan R de Los Toyos
- Instituto de Investigación Sanitaria del Principado de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain; Área de Inmunología, Facultad de Medicina y Ciencias de la Salud, Universidad de Oviedo, Av. Julián Clavería 6, 33006, Oviedo, Spain.
| | - Noemí de-Los-Santos-Alvarez
- Departamento de Química Física y Analítica. Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.
| | - M Jesús Lobo-Castañón
- Departamento de Química Física y Analítica. Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.
| |
Collapse
|
24
|
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
|
25
|
Karsdal MA, Genovese F, Rasmussen DGK, Bay-Jensen AC, Mortensen JH, Holm Nielsen S, Willumsen N, Jensen C, Manon-Jensen T, Jennings L, Reese-Petersen AL, Henriksen K, Sand JM, Bager C, Leeming DJ. Considerations for understanding protein measurements: Identification of formation, degradation and more pathological relevant epitopes. Clin Biochem 2021; 97:11-24. [PMID: 34453894 DOI: 10.1016/j.clinbiochem.2021.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/06/2021] [Accepted: 08/23/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVES There is a need for precision medicine and an unspoken promise of an optimal approach for identification of the right patients for value-based medicine based on big data. However, there may be a misconception that measurement of proteins is more valuable than measurement of fewer selected biomarkers. In population-based research, variation may be somewhat eliminated by quantity. However, this fascination of numbers may limit the attention to and understanding of the single. This review highlights that protein measurements (with collagens as examples) may mean different things depending on the targeted epitope - formation or degradation of tissues, and even signaling potential of proteins. DESIGN AND METHODS PubMed was searched for collagen, neo-epitope, biomarkers. RESULTS Ample examples of assays with specific epitopes, either pathological such as HbA1c, or domain specific such as pro-peptides, which total protein arrays would not have identified were evident. CONCLUSIONS We suggest that big data may be considered as the funnel of data points, in which most important parameters will be selected. If the technical precision is low or the biological accuracy is limited, and we include suboptimal quality of biomarkers, disguised as big data, we may not be able to fulfill the promise of helping patients searching for the optimal treatment. Alternatively, if the technical precision of the total protein quantification is high, but we miss the functional domains with the most considerable biological meaning, we miss the most important and valuable information of a given protein. This review highlights that measurements of the same protein in different ways may provide completely different meanings. We need to understand the pathological importance of each epitope quantified to maximize protein measurements.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark.
| | - F Genovese
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - D G K Rasmussen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - A C Bay-Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - J H Mortensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - S Holm Nielsen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - N Willumsen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - C Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - T Manon-Jensen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | | | | | - K Henriksen
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - J M Sand
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - C Bager
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| | - D J Leeming
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Denmark
| |
Collapse
|
26
|
Bjerre-Bastos JJ, Nielsen HB, Andersen JR, Karsdal M, Bay-Jensen AC, Boesen M, Mackey AL, Byrjalsen I, Bihlet AR. Does moderate intensity impact exercise and non-impact exercise induce acute changes in collagen biochemical markers related to osteoarthritis? - An exploratory randomized cross-over trial. Osteoarthritis Cartilage 2021; 29:986-994. [PMID: 33676014 DOI: 10.1016/j.joca.2021.02.569] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 02/17/2021] [Accepted: 02/25/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate acute changes in biochemical markers of cartilage turnover in response to moderate intensity exercise with and without joint impact in humans with knee osteoarthritis. DESIGN We conducted a randomized, cross-over, exploratory clinical study. Twenty subjects with knee osteoarthritis (OA) were randomized, of which twenty completed 30 min of cycling and 15 completed 30 min of running on days 1 week apart. Fasting blood samples were taken before, immediately after and 1, 2, 3, and 24 h after activity was initiated. Midstream spot urine was sampled before and after activity. Serum samples were analyzed for concentrations of fragment of type II collagen degradation, C2M, fragment of type VI collagen degradation, C6M, cartilage oligomeric matrix protein, COMP, marker of type II collagen formation, PRO-C2, and urine for marker of crosslinked type II collagen degradation, CTX-II. To establish a reference, all subjects had similar samples taken during rest on a separate day. Data was analyzed in a restricted maximum likelihood based random effects linear mixed model. RESULTS C2M trended to increase after cycling compared running (13.49%, 95%CI: -0.36-27.34%) and resting (12.88%, 95%CI: 0.2-25.6%) and the type II collagen formation/degradation ratio switched towards degradation after cycling, but not running. C6M trended to decrease after cycling (-8.1%, 95%CI: -14.8 to -1.4%) and running (-6.8%, 95%CI: -14.16-0.55%). CONCLUSION In persons with knee OA moderate intensity exercise without joint impact may induce acute changes in circulating levels of biochemical markers reflecting type II and VI collagen degradation.
Collapse
Affiliation(s)
- J J Bjerre-Bastos
- Xlab, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Nordic Bioscience Clinical Development, Herlev, Denmark.
| | - H B Nielsen
- Department of Anesthesia, Zealand University Hospital Roskilde, Denmark; Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark.
| | - J R Andersen
- Nordic Bioscience Clinical Development, Herlev, Denmark.
| | | | | | - M Boesen
- Department of Radiology, Bispebjerg Hospital, Copenhagen, Denmark.
| | - A L Mackey
- Institute of Sports Medicine, Bispebjerg Hospital, Copenhagen, Denmark.
| | - I Byrjalsen
- Nordic Bioscience Clinical Development, Herlev, Denmark.
| | - A R Bihlet
- Nordic Bioscience Clinical Development, Herlev, Denmark.
| |
Collapse
|
27
|
Gopal S, Barnes RFW, Cooke EJ, Zhou JY, Levin I, Emery P, Hughes TH, Karsdal MA, Manon-Jensen T, von Drygalski A. Systemic vascular basement membrane markers linked to synovial vascular remodeling are biomarkers of hemarthrosis in patients with hemophilia. J Thromb Haemost 2021; 19:1200-1211. [PMID: 33587779 DOI: 10.1111/jth.15268] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Interstitial, cartilage, and bone collagens have been proposed as biomarkers of joint deterioration in hemophilic arthropathy. The role of basement membrane (type IV and VIII) collagens as biomarkers of endothelial turnover in relation to acute joint bleeding is not understood. METHODS Thirty-one adult patients with hemophilia were studied prospectively for 3 years with musculoskeletal ultrasound/power Doppler (MSKUS/PD) during pain-free intervals and painful events for joint bleed status, synovial vascular flow, and 10 plasma markers of collagen turnover. Joint health was determined using Hemophilia Joint Health Scores and Pettersson scores. In animal studies, bleeding was induced in factor VIII-/- mice by knee joint injury. Synovial vascular remodeling was assessed using MSKUS/PD and histology. Murine plasma samples were analyzed for type IV collagen turnover markers. RESULTS Ninety-one patient visits were compiled. Twenty-five were due to acute painful episodes, with 16 confirmed hemarthroses. Type IV collagen turnover markers (PRO-C4 and C4M), and a type VIII collagen synthesis marker (PRO-C8), were transiently elevated during acute hemarthrosis. Hemarthrosis was accompanied by increased synovial microvascular flow (MSKUS/PD), and levels of type IV collagen markers correlated with PD signals in the joint. In factor VIII-deficient mice, plasma levels of type IV collagen turnover markers correlated negatively with synovial αSMA staining, indicating that reduced type IV collagen turnover was associated with thicker vessels. CONCLUSIONS Our findings suggest that basement membrane turnover markers, closely linked to synovial vascular remodeling, may be systemic biomarkers of acute hemarthrosis. Vascular instability during neovascularization may be involved in the dynamics of hemarthrosis.
Collapse
Affiliation(s)
- Srila Gopal
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Richard F W Barnes
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Esther J Cooke
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Jenny Y Zhou
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ilana Levin
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Pamela Emery
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Tudor H Hughes
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | | | | | - Annette von Drygalski
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
28
|
Location, location, location: how the tissue microenvironment affects inflammation in RA. Nat Rev Rheumatol 2021; 17:195-212. [PMID: 33526927 DOI: 10.1038/s41584-020-00570-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 01/30/2023]
Abstract
Current treatments for rheumatoid arthritis (RA) do not work well for a large proportion of patients, or at all in some individuals, and cannot cure or prevent this disease. One major obstacle to developing better drugs is a lack of complete understanding of how inflammatory joint disease arises and progresses. Emerging evidence indicates an important role for the tissue microenvironment in the pathogenesis of RA. Each tissue is made up of cells surrounded and supported by a unique extracellular matrix (ECM). These complex molecular networks define tissue architecture and provide environmental signals that programme site-specific cell behaviour. In the synovium, a main site of disease activity in RA, positional and disease stage-specific cellular diversity exist. Improved understanding of the architecture of the synovium from gross anatomy to the single-cell level, in parallel with evidence demonstrating how the synovial ECM is vital for synovial homeostasis and how dysregulated signals from the ECM promote chronic inflammation and tissue destruction in the RA joint, has opened up new ways of thinking about the pathogenesis of RA. These new ideas provide novel therapeutic approaches for patients with difficult-to-treat disease and could also be used in disease prevention.
Collapse
|
29
|
Groen SS, Sinkeviciute D, Bay-Jensen AC, Thudium CS, Karsdal MA, Thomsen SF, Schett G, Nielsen SH. Exploring IL-17 in spondyloarthritis for development of novel treatments and biomarkers. Autoimmun Rev 2021; 20:102760. [PMID: 33485992 DOI: 10.1016/j.autrev.2021.102760] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 11/14/2020] [Indexed: 12/17/2022]
Abstract
Spondyloarthritis (SpA) is an umbrella term describing a family of chronic inflammatory rheumatic diseases. These diseases are characterised by inflammation of the axial skeleton, peripheral joints, and entheseal insertion sites throughout the body which can lead to structural joint damage including formation of axial syndesmophytes and peripheral osteophytes. Genetic evidence, preclinical and clinical studies indicate a clear role of interleukin (IL)- 23 and IL-17 as mediators in SpA pathogenesis. Targeting the IL-23/-17 pathways seems an efficient strategy for treatment of SpA patients, and despite the remaining challenges the pathway holds great promise for further advances and improved therapeutic opportunities. Much research is focusing on serological markers and imaging strategies to correctly diagnose patients in the early stages of SpA. Biomarkers may facilitate personalised medicine tailored to each patient's specific disease to optimise treatment efficacy and to monitor therapeutic response. This narrative review focuses on the IL-17 pathway in SpA-related diseases with emphasis on its role in pathogenesis, current approved IL-17 inhibitors, and the need for biomarkers reflecting core disease pathways for early diagnosis and measurement of disease activity, prognosis, and response to therapy.
Collapse
Affiliation(s)
- Solveig Skovlund Groen
- Immunoscience, Nordic Bioscience, Herlev, Denmark; Biomecial Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Dovile Sinkeviciute
- Immunoscience, Nordic Bioscience, Herlev, Denmark; Department of Clinical Sciences Lund, University of Lund, Lund, Sweden
| | | | | | | | - Simon Francis Thomsen
- Biomecial Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Signe Holm Nielsen
- Immunoscience, Nordic Bioscience, Herlev, Denmark; Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
30
|
Drobinski PJ, Bay-Jensen AC, Karsdal MA, Sardar S, Siebuhr AS. Connective tissue remodelling is differently modulated by tocilizumab versus methotrexate monotherapy in patients with early rheumatoid arthritis: the AMBITION study. Arthritis Res Ther 2021; 23:13. [PMID: 33413588 PMCID: PMC7789531 DOI: 10.1186/s13075-020-02378-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/24/2020] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Associations between rheumatoid arthritis (RA) and effect of treatment at the tissue levels are poorly understood. We investigated the scope of released extracellular matrix (ECM) metabolites as a consequence of tissue remodelling in patients treated with methotrexate (MTX) and tocilizumab (TCZ) compared to placebo. METHODS Tissue metabolites from 387 RA patients treated with either TCZ (8 mg/kg) or MTX monotherapy (7.5-20 mg/kg) were measured at baseline and 8 weeks sera by validated ELISA assays. The levels of collagen biomarkers (C1M, C2M, C3M and C4M) together with C-reactive protein (CRP) and CRP metabolite (CRPM) were investigated. Baseline levels of biomarkers have been compared with 72 age- and gender-matched healthy controls. Comparison between treatment and response groups were done by ANCOVA, Spearman's correlation and logistic regression adjusted for age, gender, BMI and disease duration. RESULTS C1M and C3M were significantly (P < 0.05) inhibited by TCZ and C3M by MTX (P < 0.01) compared to placebo. C1M and C3M inhibition with TCZ was respectively 23% and 16% greater than that of MTX (P < 0.01 and P < 0.0001). C4M was inhibited by TCZ and MTX, but the effect of TCZ was 22% greater than MTX (P < 0.0001). TCZ and MTX had minimal effect on C2M levels. MTX had no effect on CRP and CRPM, whereas TCZ reduced their levels to 69% and 27% from baseline. Investigated biomarkers revealed a significant (P < 0.05) difference in biomarker profiles of MTX ACR50 treatment responders and non-responders. Change to week 8 in levels of C3M, C4M, CRP and CRPM in MTX patients correlated significantly (rho = 0.41 to 0.18, P < 0.0001 to 0.039) with change in disease activity (DAS28) at weeks 8, 16 and 24, whereas only CRP in TCZ patients (rho = 0.32 to 0.21, P < 0.0001 to 0.01). CONCLUSION Patients receiving TCZ treatment for 8 weeks had higher suppression of tissue remodelling and inflammatory biomarkers over patients treated with MTX. Measured biomarkers enabled for a discrimination of biomarker profiles of ACR50 treatment responding patients and identification of those who benefit at the early time point. Week 8 change in levels of C3M, C4M, CRP and CRPM significantly predicted clinical response to treatment and correlated with DAS28 at all time points. TRIAL REGISTRATION ClinicalTrials.gov, NCT00109408 . Date of registration: July 2005. Name of the registry: A Study to Assess the Safety and Efficacy of Tocilizumab in Patients with Active Rheumatoid Arthritis.
Collapse
Affiliation(s)
- Patryk J. Drobinski
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Anne C. Bay-Jensen
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Morten A. Karsdal
- Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Samra Sardar
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Anne S. Siebuhr
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| |
Collapse
|
31
|
Nicol L, Srikanth P, Henriksen K, Sun S, Smith R, Karsdal MA, Nagamani SCS, Shapiro J, Lee B, Leder BZ, Orwoll E. Widespread disturbance in extracellular matrix collagen biomarker responses to teriparatide therapy in osteogenesis imperfecta. Bone 2021; 142:115703. [PMID: 33099032 DOI: 10.1016/j.bone.2020.115703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/05/2020] [Accepted: 10/18/2020] [Indexed: 12/30/2022]
Abstract
Osteogenesis imperfecta (OI), a heritable disorder caused by abnormalities in synthesis or processing of type I collagen, is characterized by skeletal fragility. Type I collagen interacts with multiple components of the extracellular matrix (ECM) including other collagens types. Thus, alterations in structure or quantity may broadly affect ECM homeostasis. In fact, while OI is clinically categorized by severity of bone disease, patients can also present with extra-skeletal manifestations, including the pulmonary, muscle and cardiovascular systems. Parathyroid hormone (PTH) is a regulator of skeletal homeostasis but the receptor for PTH/PTH1R is expressed in a variety of other tissues. Given interactions between type I collagen with other collagens in the ECM and the potential for PTH action on tissues beyond the skeleton, we explored whether serum levels of non-type I collagens are altered in response to teriparatide (human parathyroid hormone 1-34). We measured biomarkers of collagens II, III, IV, V, and VI in serum from individuals with type I and types III/IV OI in response to an 18 month course of teriparatide or placebo. These results were compared to similar biomarker measures in postmenopausal (PM) women without OI treated with teriparatide. In type I OI, teriparatide therapy increased concentrations of biomarkers of collagens II, III, IV, V, and VI. In individuals with types III/IV OI these biomarker changes in response to teriparatide were blunted, as we previously reported with collagen I biomarkers during teriparatide therapy. In contrast to OI, in PM women there were no effects of teriparatide on the collagen biomarkers we assessed (II, V, and VI). These findings suggest that in OI teriparatide therapy has abnormal effects on the homeostasis of many ECM collagens likely derived from skeletal as well as extra-skeletal tissues.
Collapse
Affiliation(s)
- Lindsey Nicol
- Department of Pediatrics, Division of Endocrinology, Oregon Health & Science University, Portland, OR, United States of America.
| | - Priya Srikanth
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, United States of America
| | | | - Shu Sun
- Nordic Bioscience, Herlev, Denmark
| | - Rosamund Smith
- Lilly Research Laboratories, Indianapolis, IN, United States of America
| | | | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States of America; Texas Children's Hospital, Houston, TX, United States of America
| | - Jay Shapiro
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America; Dept. Endocrinology and Diabetes, Walter Reed National Military Medical Center, Bethesda, MD, United States of America
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States of America; Texas Children's Hospital, Houston, TX, United States of America
| | - Benjamin Z Leder
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Eric Orwoll
- Department of Medicine, Bone and Mineral Unit, Oregon Health & Science University, Portland, OR, United States of America
| |
Collapse
|
32
|
Bone phenotypes in rheumatology - there is more to bone than just bone. BMC Musculoskelet Disord 2020; 21:789. [PMID: 33248451 PMCID: PMC7700716 DOI: 10.1186/s12891-020-03804-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis, rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis, all have one clear common denominator; an altered turnover of bone. However, this may be more complex than a simple change in bone matrix and mineral turnover. While these diseases share a common tissue axis, their manifestations in the area of pathology are highly diverse, ranging from sclerosis to erosion of bone in different regions. The management of these diseases will benefit from a deeper understanding of the local versus systemic effects, the relation to the equilibrium of the bone balance (i.e., bone formation versus bone resorption), and the physiological and pathophysiological phenotypes of the cells involved (e.g., osteoblasts, osteoclasts, osteocytes and chondrocytes). For example, the process of endochondral bone formation in chondrocytes occurs exists during skeletal development and healthy conditions, but also in pathological conditions. This review focuses on the complex molecular and cellular taxonomy of bone in the context of rheumatological diseases that alter bone matrix composition and maintenance, giving rise to different bone turnover phenotypes, and how biomarkers (biochemical markers) can be applied to potentially describe specific bone phenotypic tissue profiles.
Collapse
|
33
|
Thudium CS, Bay-Jensen AC, Cahya S, Dow ER, Karsdal MA, Koch AE, Zhang W, Benschop RJ. The Janus kinase 1/2 inhibitor baricitinib reduces biomarkers of joint destruction in moderate to severe rheumatoid arthritis. Arthritis Res Ther 2020; 22:235. [PMID: 33046136 PMCID: PMC7552555 DOI: 10.1186/s13075-020-02340-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022] Open
Abstract
Background Tissue released blood-based biomarkers can provide insight into drug mode of action and response. To understand the changes in extracellular matrix turnover, we analyzed biomarkers associated with joint tissue turnover from a phase 3, randomized, placebo-controlled study of baricitinib in patients with active rheumatoid arthritis (RA). Methods Serum biomarkers associated with synovial inflammation (C1M, C3M, and C4M), cartilage degradation (C2M), bone resorption (CTX-I), and bone formation (osteocalcin) were analyzed at baseline, and weeks 4 and 12, from a subgroup of patients (n = 240) randomized to placebo or 2-mg or 4-mg baricitinib (RA-BUILD, NCT01721057). Mixed-model repeated measure was used to identify biomarkers altered by baricitinib. The relationship between changes in biomarkers and clinical measures was evaluated using correlation analysis. Results Treatment arms were well balanced for baseline biomarkers, demographics, and disease activity. At week 4, baricitinib 4-mg significantly reduced C1M from baseline by 21% compared to placebo (p < 0.01); suppression was sustained at week 12 (27%, p < 0.001). Baricitinib 4-mg reduced C3M and C4M at week 4 by 14% and 12% compared to placebo, respectively (p < 0.001); they remained reduced by 16% and 11% at week 12 (p < 0.001). In a pooled analysis including all treatment arms, patients with the largest reduction (upper 25% quartile) in C1M, C3M, and C4M by week 12 had significantly greater clinical improvement in the Simplified Disease Activity Index at week 12 compared to patients with the smallest reduction (lowest 25% quartile). Conclusion Baricitinib treatment resulted in reduced circulating biomarkers associated with joint tissue destruction as well as concomitant RA clinical improvement. Trial registration ClinicalTrials.gov NCT01721057; date of registration: November 1, 2012
Collapse
Affiliation(s)
- Christian S Thudium
- Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark.
| | - Anne C Bay-Jensen
- Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | | | - Ernst R Dow
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Morten A Karsdal
- Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | - Alisa E Koch
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
34
|
Mass Spectrometry Imaging as a Potential Tool to Investigate Human Osteoarthritis at the Tissue Level. Int J Mol Sci 2020; 21:ijms21176414. [PMID: 32899238 PMCID: PMC7503948 DOI: 10.3390/ijms21176414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, predicted to increase in incidence year by year due to an ageing population. Due to the biological complexity of the disease, OA remains highly heterogeneous. Although much work has been undertaken in the past few years, underlying molecular mechanisms leading to joint tissue structural deterioration are not fully understood, with only few validated markers for disease diagnosis and progression being available. Discovery and quantitation of various OA-specific biomarkers is still largely focused on the bodily fluids which does not appear to be reliable and sensitive enough. However, with the advancement of spatial proteomic techniques, several novel peptides and proteins, as well as N-glycans, can be identified and localised in a reliable and sensitive manner. To summarise the important findings from OA biomarker studies, papers published between 2000 and 2020 were searched via Google Scholar and PubMed. Medical subject heading (MeSH) terms ‘osteoarthritis’, ‘biomarker’, ‘synovial fluid’, ‘serum’, ‘urine’, ’matrix-assisted laser desorption/ionisation’, ‘mass spectrometry imaging’, ‘proteomic’, ‘glycomic’, ‘cartilage’, ‘synovium’ AND ‘subchondral bone’ were selectively used. The literature search was restricted to full-text original research articles and written only in English. Two main areas were reviewed for OA biomarker studies: (1) an overview of disease-specific markers detected from different types of OA bio-samples, and (2) an up-to-date summary of the tissue-specific OA studies that have utilised matrix-assisted laser desorption/ionisation mass spectrometry imaging (MALDI-MSI). Overall, these OA biomarkers could provide clinicians with information for better the diagnosis, and prognosis of individual patients, and ultimately help facilitate the development of disease-modifying treatments.
Collapse
|
35
|
The Anti-ADAMTS-5 Nanobody ® M6495 Protects Cartilage Degradation Ex Vivo. Int J Mol Sci 2020; 21:ijms21175992. [PMID: 32825512 PMCID: PMC7503673 DOI: 10.3390/ijms21175992] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/28/2020] [Accepted: 08/02/2020] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is associated with cartilage breakdown, brought about by ADAMTS-5 mediated aggrecan degradation followed by MMP-derived aggrecan and type II collagen degradation. We investigated a novel anti-ADAMTS-5 inhibiting Nanobody® (M6495) on cartilage turnover ex vivo. Bovine cartilage (BEX, n = 4), human osteoarthritic - (HEX, n = 8) and healthy—cartilage (hHEX, n = 1) explants and bovine synovium and cartilage were cultured up to 21 days in medium alone (w/o), with pro-inflammatory cytokines (oncostatin M (10 ng/mL) + TNFα (20 ng/mL) (O + T), IL-1α (10 ng/mL) or oncostatin M (50 ng/mL) + IL-1β (10 ng/mL)) with or without M6495 (1000−0.46 nM). Cartilage turnover was assessed in conditioned medium by GAG (glycosaminoglycan) and biomarkers of ADAMTS-5 driven aggrecan degradation (huARGS and exAGNxI) and type II collagen degradation (C2M) and formation (PRO-C2). HuARGS, exAGNxI and GAG peaked within the first culture week in pro-inflammatory stimulated explants. C2M peaked from day 14 by O + T and day 21 in co-culture experiments. M6495 dose dependently decreased huARGS, exAGNxI and GAG after pro-inflammatory stimulation. In HEX C2M was dose-dependently reduced by M6495. M6495 showed no effect on PRO-C2. M6495 showed cartilage protective effects by dose-dependently inhibiting ADAMTS-5 mediated cartilage degradation and inhibiting overall cartilage deterioration in ex vivo cartilage cultures.
Collapse
|
36
|
Sinkeviciute D, Aspberg A, He Y, Bay-Jensen AC, Önnerfjord P. Characterization of the interleukin-17 effect on articular cartilage in a translational model: an explorative study. BMC Rheumatol 2020; 4:30. [PMID: 32426694 PMCID: PMC7216541 DOI: 10.1186/s41927-020-00122-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/06/2020] [Indexed: 12/29/2022] Open
Abstract
Background Osteoarthritis (OA) is a progressive, chronic disease characterized by articular cartilage destruction. The pro-inflammatory cytokine IL-17 levels have been reported elevated in serum and synovial fluid of OA patients and correlated with increased cartilage defects and bone remodeling. The aim of this study was to characterize an IL-17-mediated articular cartilage degradation ex-vivo model and to investigate IL-17 effect on cartilage extracellular matrix protein turnover. Methods Full-depth bovine femoral condyle articular cartilage explants were cultured in serum-free medium for three weeks in the absence, or presence of cytokines: IL-17A (100 ng/ml or 25 ng/ml), or 10 ng OSM combined with 20 ng/ml TNFα (O + T). RNA isolation and PCR analysis were performed on tissue lysates to confirm IL-17 receptor expression. GAG and ECM-turnover biomarker release into conditioned media was assessed with dimethyl methylene blue and ELISA assays, respectively. Gelatin zymography was used for matrix metalloproteinase (MMP) 2 and MMP9 activity assessment in conditioned media, and shotgun LC-MS/MS for identification and label-free quantification of proteins and protein fragments in conditioned media. Western blotting was used to validate MS results. Results IL-17RA mRNA was expressed in bovine full-depth articular cartilage and the treatment with IL-17A did not interfere with metabolic activity of the model. IL-17A induced cartilage breakdown; conditioned media GAG levels were 3.6-fold-elevated compared to untreated. IL-17A [100 ng/ml] induced ADAMTS-mediated aggrecan degradation fragment release (14-fold increase compared to untreated) and MMP-mediated type II collagen fragment release (6-fold-change compared to untreated). MS data analysis revealed 16 differentially expressed proteins in IL-17A conditioned media compared to untreated, and CHI3L1 upregulation in conditioned media in response to IL-17 was confirmed by Western blotting. Conclusions We showed that IL-17A has cartilage modulating potential. It induces collagen and aggrecan degradation indicating an upregulation of MMPs. This was confirmed by zymography and mass spectrometry data. We also showed that the expression of other cytokines is induced by IL-17A, which provide further insight to the pathways that are active in response to IL-17A. This exploratory study confirms that IL-17A may play a role in cartilage pathology and that the applied model may be a good tool to further investigate it.
Collapse
Affiliation(s)
- Dovile Sinkeviciute
- 1Nordic Bioscience, Biomarkers & Research, Herlev, Denmark.,2Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Anders Aspberg
- 2Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Yi He
- 1Nordic Bioscience, Biomarkers & Research, Herlev, Denmark
| | | | - Patrik Önnerfjord
- 2Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
37
|
Sharma N, Drobinski P, Kayed A, Chen Z, Kjelgaard-Petersen CF, Gantzel T, Karsdal MA, Michaelis M, Ladel C, Bay-Jensen AC, Lindemann S, Thudium CS. Inflammation and joint destruction may be linked to the generation of cartilage metabolites of ADAMTS-5 through activation of toll-like receptors. Osteoarthritis Cartilage 2020; 28:658-668. [PMID: 31734268 DOI: 10.1016/j.joca.2019.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/30/2019] [Accepted: 11/04/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Links between pain and joint degradation are poorly understood. We investigated the role of activation of Toll-like receptors (TLR) by cartilage metabolites in initiating and maintaining the inflammatory loop in OA causing joint destruction. METHODS Synovial membrane explants (SMEs) were prepared from OA patients' synovial biopsies. SMEs were cultured for 10 days under following conditions: culture medium alone, OSM + TNFα, TLR2 agonist - Pam2CSK4, Pam3CSK4 or synthetic aggrecan 32-mer, TLR4 agonist - Lipid A. Release of pro-inflammatory and degradation biomarkers (acMMP3 and C3M) were measured by ELISA in conditioned media along with IL-6. Additionally, human cartilage was digested with ADAMTS-5, with or without the ADAMTS-5 inhibiting nanobody - M6495. Digested cartilage solution (DCS) and synthetic 32-mer were tested for TLR activation in SEAP based TLR reporter assay. RESULTS Western blotting confirmed TLR2 and TLR4 in untreated OA synovial biopsies. TLR agonists showed an increase in release of biomarkers - acMMP3 and C3M in SME. Synthetic 32-mer showed no activation in the TLR reporter assay. ADAMTS-5 degraded cartilage fragments activated TLR2 in vitro. Adding M6495 - an anti-ADAMTS-5 inhibiting nanobody®, blocked ADAMTS-5-mediated DCS TLR2 activation. CONCLUSION TLR2 is expressed in synovium of OA patients and their activation by synthetic ligands causes increased tissue turnover. ADAMTS-5-mediated cartilage degradation leads to release of aggrecan fragments which activates the TLR2 receptor in vitro. M6495 suppressed cartilage degradation by ADAMTS-5, limiting the activation of TLR2. In conclusion, pain and joint destruction may be linked to generation of ADAMTS-5 cartilage metabolites.
Collapse
Affiliation(s)
- N Sharma
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| | - P Drobinski
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| | - A Kayed
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| | - Z Chen
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| | | | - T Gantzel
- Orthopaedic Surgery Unit, Gentofte University Hospital, Gentofte, Denmark.
| | - M A Karsdal
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark.
| | - M Michaelis
- Head of Osteoarthritis Research and Early Clinical Development, Merck KGaA, Darmstadt, Germany.
| | - C Ladel
- Clinical Biomarker & Diagnostics Lead, Merck KGaA, Darmstadt, Germany.
| | - A C Bay-Jensen
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark.
| | - S Lindemann
- Head of Exploratory Osteoarthritis, Merck KGaA, Darmstadt, Germany.
| | - C S Thudium
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark.
| |
Collapse
|
38
|
Nagy EE, Nagy-Finna C, Popoviciu H, Kovács B. Soluble Biomarkers of Osteoporosis and Osteoarthritis, from Pathway Mapping to Clinical Trials: An Update. Clin Interv Aging 2020; 15:501-518. [PMID: 32308378 PMCID: PMC7152733 DOI: 10.2147/cia.s242288] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/02/2020] [Indexed: 12/20/2022] Open
Abstract
Serum biomarkers of osteoarticular diseases have been in the limelight of current clinical research trends. Laboratory validation of defined and candidate biomarkers for both osteoarthritis and osteoporosis is of key importance for future decisional algorithms in the diagnosis, monitoring, and prognosis of these diseases. The current guidelines recommend the use of collagen degradation remnants, eg, CTX-I and CTX-II, in the complementary diagnosis of both osteoporosis and osteoarthritis. Besides the collagen degradation markers, enzymes that regulate bone and articular metabolism are useful in the clinical evaluation of osteoarticular pathologies. Along these, several other recommended and new nominee molecules have been recently studied. Wnts and Wnt-related molecules have a cardinal role in the bone-joint homeostasis, making them a promising target not only for pharmaceutical modulation, but also to be considered as soluble biomarkers. Sclerostin and dickkopf, two inhibitor molecules of the Wnt/β-catenin signaling, might have a dual role in the assessment of the clinical manifestations of the osteoarticular unit. In osteoarthritis, besides fragments of collagen type II many pathway-related molecules have been studied and proposed for biomarker validation. The most serious limitation is that a significant proportion of studies lack statistical power due to the reduced number of cases enrolled. Serum biomarkers of bone and joint turnover markers represent an encouraging possibility for the diagnosis and prognosis of osteoarticular diseases, although further studies and laboratory validations should be carried out as to solely rely on them.
Collapse
Affiliation(s)
- Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș, Romania
| | - Csilla Nagy-Finna
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș, Romania
- Department M4, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș, Romania; Rheumatology Clinic, Clinical Emergency Hospital, Târgu Mureș, Romania
| | - Horațiu Popoviciu
- Department M4, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș, Romania; Rheumatology Clinic, Clinical Emergency Hospital, Târgu Mureș, Romania
| | - Béla Kovács
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș, Romania
| |
Collapse
|
39
|
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
|
40
|
Hardy E, Fernandez-Patron C. Destroy to Rebuild: The Connection Between Bone Tissue Remodeling and Matrix Metalloproteinases. Front Physiol 2020; 11:47. [PMID: 32116759 PMCID: PMC7013034 DOI: 10.3389/fphys.2020.00047] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
Bone is a dynamic organ that undergoes constant remodeling, an energetically costly process by which old bone is replaced and localized bone defects are repaired to renew the skeleton over time, thereby maintaining skeletal health. This review provides a general overview of bone’s main players (bone lining cells, osteocytes, osteoclasts, reversal cells, and osteoblasts) that participate in bone remodeling. Placing emphasis on the family of extracellular matrix metalloproteinases (MMPs), we describe how: (i) Convergence of multiple protease families (including MMPs and cysteine proteinases) ensures complexity and robustness of the bone remodeling process, (ii) Enzymatic activity of MMPs affects bone physiology at the molecular and cellular levels and (iii) Either overexpression or deficiency/insufficiency of individual MMPs impairs healthy bone remodeling and systemic metabolism. Today, it is generally accepted that proteolytic activity is required for the degradation of bone tissue in osteoarthritis and osteoporosis. However, it is increasingly evident that inactivating mutations in MMP genes can also lead to bone pathology including osteolysis and metabolic abnormalities such as delayed growth. We argue that there remains a need to rethink the role played by proteases in bone physiology and pathology.
Collapse
Affiliation(s)
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
41
|
Bay-Jensen AC, Engstroem A, Sharma N, Karsdal MA. Blood and urinary collagen markers in osteoarthritis: markers of tissue turnover and disease activity. Expert Rev Mol Diagn 2019; 20:57-68. [PMID: 31847627 DOI: 10.1080/14737159.2020.1704257] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Introduction: The need for diagnostic markers in osteoarthritis (OA) is acute and immediate, as sensitive and precise tools that monitor disease activity and treatment response are lacking. Collagens - types I, II, and III - are the skeleton of the extracellular matrix of joint tissues. Joint collagens are generally turned over at a low rate, but the balance between formation and degradation is disturbed, leading to the loss of, for example, cartilage.Areas covered: We discuss the markers reflecting collagen turnover and provide examples of how they have been applied in OA research, as well as how we believe these should be used in the future. We have searched PubMed for full-text articles written in English using different combinations of the following terms: OA, biomarker, and collagen. The result is a narrative review that gives examples from the literature.Expert opinion: Collagen markers show promise, as they are direct measures of tissue balance. Until now, collagen markers have mainly been tested in observational cohorts, which may provide insights into the association between the candidate marker and clinical variables; however, these do not advance the development of qualified markers that can be used for drug development or in clinical practice.
Collapse
Affiliation(s)
| | - Amalie Engstroem
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark.,Biomedical institute, University of Copenhagen, Copenhagen, Denmark
| | - Neha Sharma
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark.,Biomedical institute, University of Copenhagen, Copenhagen, Denmark
| | - Morten Asser Karsdal
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| |
Collapse
|
42
|
Sofat N, Ejindu V, Heron C, Harrison A, Koushesh S, Assi L, Kuttapitiya A, Whitley GS, Howe FA. Biomarkers in Painful Symptomatic Knee OA Demonstrate That MRI Assessed Joint Damage and Type II Collagen Degradation Products Are Linked to Disease Progression. Front Neurosci 2019; 13:1016. [PMID: 31680799 PMCID: PMC6803383 DOI: 10.3389/fnins.2019.01016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/06/2019] [Indexed: 01/01/2023] Open
Abstract
Background Osteoarthritis (OA) is the most prevalent arthritis worldwide, but the evolution of pain in relation to joint damage and biochemical markers are not well understood. We evaluated the relation between clinical pain measures and evoked pain in relation to structural damage and biochemical biomarkers in knee OA. Methods A cross-sectional study in people with knee OA and healthy controls was conducted. A total of 130 participants with advanced OA requiring total knee replacement (TKR) (n = 78), mild OA having standard care (n = 42) and non-OA controls (n = 6), with four drop-outs were assessed. Pain scoring was performed by the Western Ontario and McMaster Universities OA Index (WOMAC_P) and the Visual Analog Scale (VAS). Pain sensitization was assessed by pain pressure thresholds (PPTs). Knee magnetic resonance imaging (MRI) assessed joint damage using the MRI Knee OA Score (MOAKS). Overall MOAKS scores were created for bone marrow lesions (BMLs), cartilage degradation (CD), and effusion/Hoffa synovitis (tSyn). Type II collagen cleavage products (CTX-II) were determined by ELISA. Results The advanced OA group had a mean age of 68.9 ± 7.7 years and the mild group 63.1 ± 9.6. The advanced OA group had higher levels of pain, with mean WOMAC_P of 58.8 ± 21.7 compared with the mild OA group of 40.6 ± 26.0. All OA subjects had pain sensitization by PPT compared with controls (p < 0.05). WOMAC_P correlated with the total number of regions with cartilage damage (nCD) (R = 0.225, p = 0.033) and total number of BMLs (nBML) (R = 0.195, p = 0.065) using body mass index (BMI), age, and Hospital Anxiety and Depression Scale (HADS) as covariates. Levels of CTX-II correlated with tSyn (R = 0.313, p = 0.03), nBML (R = 0.252, p = 0.019), number of osteophytes (R = 0.33, p = 0.002), and nCD (R = 0.218, p = 0.042), using BMI and age as covariates. A multivariate analysis indicated that BMI and HADS were the most significant predictors of pain scores (p < 0.05). Conclusion People with both mild and advanced OA show features of pain sensitization. We found that increasing MRI-detected joint damage was associated with higher levels of CTX-II, suggesting that increasing disease severity can be assessed by MRI and CTX-II biomarkers to evaluate OA disease progression.
Collapse
Affiliation(s)
- Nidhi Sofat
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom.,St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Vivian Ejindu
- St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Christine Heron
- St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Abiola Harrison
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| | - Soraya Koushesh
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| | - Lena Assi
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| | - Anasuya Kuttapitiya
- Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| | - Guy S Whitley
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Franklyn A Howe
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| |
Collapse
|
43
|
Metabolites of type I, II, III, and IV collagen may serve as markers of disease activity in axial spondyloarthritis. Sci Rep 2019; 9:11218. [PMID: 31375691 PMCID: PMC6677742 DOI: 10.1038/s41598-019-47502-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 07/12/2019] [Indexed: 12/26/2022] Open
Abstract
Local inflammation in axial spondyloarthritis (axSpA) leads to the release of collagen metabolites from the disease-affected tissue. We investigated whether collagen metabolites were associated with disease activity and could distinguish non-radiographic(nr)-axSpA from ankylosing spondylitis (AS). A total of 193 axSpA patients (nr-axSpA, n = 121 and AS, n = 72) and asymptomatic controls (n = 100) were included. Serum levels of metalloproteinase (MMP)-degraded collagen type I (C1M), type II (C2M), type III (C3M) and type IV (C4M2) were quantified by enzyme-linked immunosorbent assay (ELISA). All metabolites were higher in axSpA than in controls (all p < 0.001). Serum levels of C1M, C3M, and C4M2 were increased in AS compared to nr-axSpA (43.4 ng/mL vs. 34.6; p < 0.001, 15.4 vs. 12.8; p = 0.001, and 27.8 vs. 22.4; p < 0.001). The best metabolite to differentiate between axSpA and controls was C3M (AUC 0.95; specificity 92.0, sensitivity 83.4). C1M correlated with ASDAS-CRP in nr-axSpA (ρ = 0.37; p < 0.001) and AS (ρ = 0.57; p < 0.001). C1M, C3M, and C4M2 were associated with ASDAS-CRP in AS and nr-axSpA after adjustment for age, gender, and disease duration. Serum levels of collagen metabolites were significantly higher in AS and nr-axSpA than in controls. Moreover, the present study indicates that collagen metabolites reflect disease activity and are useful biomarkers of axSpA.
Collapse
|
44
|
Blair JPM, Bager C, Platt A, Karsdal M, Bay-Jensen AC. Identification of pathological RA endotypes using blood-based biomarkers reflecting tissue metabolism. A retrospective and explorative analysis of two phase III RA studies. PLoS One 2019; 14:e0219980. [PMID: 31339920 PMCID: PMC6655687 DOI: 10.1371/journal.pone.0219980] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022] Open
Abstract
There is an increasing demand for accurate endotyping of patients according to their pathogenesis to allow more targeted treatment. We explore a combination of blood-based joint tissue metabolites (neoepitopes) to enable patient clustering through distinct disease profiles. We analysed data from two RA studies (LITHE (N = 574, follow-up 24 and 52 weeks), OSKIRA-1 (N = 131, follow-up 24 weeks)). Two osteoarthritis (OA) studies (SMC01 (N = 447), SMC02 (N = 81)) were included as non-RA comparators. Specific tissue-derived neoepitopes measured at baseline, included: C2M (cartilage degradation); CTX-I and PINP (bone turnover); C1M and C3M (interstitial matrix degradation); CRPM (CRP metabolite) and VICM (macrophage activity). Clustering was performed to identify putative endotypes. We identified five clusters (A-E). Clusters A and B were characterized by generally higher levels of biomarkers than other clusters, except VICM which was significantly higher in cluster B than in cluster A (p<0.001). Biomarker levels in Cluster C were all close to the median, whilst Cluster D was characterised by low levels of all biomarkers. Cluster E also had low levels of most biomarkers, but with significantly higher levels of CTX-I compared to cluster D. There was a significant difference in ΔSHP score observed at 52 weeks (p<0.05). We describe putative RA endotypes based on biomarkers reflecting joint tissue metabolism. These endotypes differ in their underlining pathogenesis, and may in the future have utility for patient treatment selection.
Collapse
Affiliation(s)
- J. P. M. Blair
- ProScion, Herlev, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
- * E-mail:
| | | | - A. Platt
- Target & Translational Science, Respiratory, Inflammation and Autoimmunity (RIA), IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - M. Karsdal
- Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - A. -C. Bay-Jensen
- Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| |
Collapse
|
45
|
Löfvall H, Katri A, Dąbrowska A, Karsdal MA, Luo Y, He Y, Manon-Jensen T, Dziegiel MH, Bay-Jensen AC, Thudium CS, Henriksen K. GPDPLQ 1237-A Type II Collagen Neo-Epitope Biomarker of Osteoclast- and Inflammation-Derived Cartilage Degradation in vitro. Sci Rep 2019; 9:3050. [PMID: 30816326 PMCID: PMC6395810 DOI: 10.1038/s41598-019-39803-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 01/31/2019] [Indexed: 01/21/2023] Open
Abstract
C-telopeptide of type II collagen (CTX-II) has been shown to be a highly relevant biomarker of cartilage degradation in human rheumatic diseases, if measured in synovial fluid or urine. However, serum or plasma CTX-II have not been demonstrated to have any clinical utility to date. Here, we describe the GPDPLQ1237 ELISA which targets the EKGPDPLQ↓ neo-epitope, an elongated version of the CTX-II neo-epitope (EKGPDP↓), speculated to be a blood-precursor of CTX-II generated by the cysteine protease cathepsin K. Human osteoclast cartilage resorption cultures as well as oncostatin M and tumour necrosis factor α-stimulated bovine cartilage explant cultures were used to validate GPDPLQ1237 biologically by treating the cultures with the cysteine protease inhibitor E-64 and/or the matrix metalloproteinase (MMP) inhibitor GM6001 to assess the potential contributions of these two protease classes to GPDPLQ1237 release. Cartilage resorption-derived GPDPLQ1237 release was inhibited by E-64 (72.1% inhibition), GM6001 (75.5%), and E-64/GM6001 (91.5%), whereas CTX-II release was inhibited by GM6001 (87.0%) but not by E-64 (5.5%). Cartilage explant GPDPLQ1237 and CTX-II release were both fully inhibited by GM6001 but were not inhibited by E-64. No clinically relevant GPDPLQ1237 reactivity was identified in human serum, plasma, or urine from healthy donors or arthritis patients. In conclusion, the GPDPLQ1237 biomarker is released during osteoclast-derived cysteine protease- and MMP-mediated cartilage degradation in vitro, whereas CTX-II release is mediated by MMPs and not by cysteine proteases, as well as from MMP-mediated cartilage degradation under a pro-inflammatory stimulus. These findings suggest that GPDPLQ1237 may be relevant in diseases with pathological osteoclast activity and cartilage degradation. Further studies are required to validate the neo-epitope in human samples.
Collapse
Affiliation(s)
- Henrik Löfvall
- Nordic Bioscience, Herlev, Denmark.,Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | - Anna Katri
- Nordic Bioscience, Herlev, Denmark.,Department of Drug Design and Pharmacology, Copenhagen University, Copenhagen, Denmark
| | | | | | | | - Yi He
- Nordic Bioscience, Herlev, Denmark
| | | | - Morten H Dziegiel
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | |
Collapse
|
46
|
Luo Y, He Y, Reker D, Gudmann NS, Henriksen K, Simonsen O, Ladel C, Michaelis M, Mobasheri A, Karsdal M, Bay-Jensen AC. A Novel High Sensitivity Type II Collagen Blood-Based Biomarker, PRO-C2, for Assessment of Cartilage Formation. Int J Mol Sci 2018; 19:ijms19113485. [PMID: 30404167 PMCID: PMC6275061 DOI: 10.3390/ijms19113485] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 01/07/2023] Open
Abstract
N-terminal propeptide of type II collagen (PIINP) is a biomarker reflecting cartilage formation. PIINP exists in two main splice variants termed as type IIA and type IIB collagen NH₂-propeptide (PIIANP, PIIBNP). PIIANP has been widely recognized as a cartilage formation biomarker. However, the utility of PIIBNP as a marker in preclinical and clinical settings has not been fully investigated yet. In this study, we aimed to characterize an antibody targeting human PIIBNP and to develop an immunoassay assessing type II collagen synthesis in human blood samples. A high sensitivity electrochemiluminescence immunoassay, hsPRO-C2, was developed using a well-characterized antibody against human PIIBNP. Human cartilage explants from replaced osteoarthritis knees were cultured for ten weeks in the presence of growth factors, insulin-like growth factor 1 (IGF-1) or recombinant human fibroblast growth factor 18 (rhFGF-18). The culture medium was changed every seven days, and levels of PIIBNP, PIIANP, and matrix metalloproteinase 9-mediated degradation of type II collagen (C2M) were analyzed herein. Serum samples from a cross-sectional knee osteoarthritis cohort, as well as pediatric and rheumatoid arthritis samples, were assayed for PIIBNP and PIIANP. Western blot showed that the antibody recognized PIIBNP either as a free fragment or attached to the main molecule. Immunohistochemistry demonstrated that PIIBNP was predominately located in the extracellular matrix of the superficial and deep zones and chondrocytes in both normal and osteoarthritic articular cartilage. In addition, the hsPRO-C2 immunoassay exhibits acceptable technical performances. In the human cartilage explants model, levels of PIIBNP, but not PIIANP and C2M, were increased (2 to 7-fold) time-dependently in response to IGF-1. Moreover, there was no significant correlation between PIIBNP and PIIANP levels when measured in knee osteoarthritis, rheumatoid arthritis, and pediatric serum samples. Serum PIIBNP was significantly higher in controls (KL0/1) compared to OA groups (KL2/3/4, p = 0.012). The hsPRO-C2 assay shows completely different biological and clinical patterns than PIIANP ELISA, suggesting that it may be a promising biomarker of cartilage formation.
Collapse
Affiliation(s)
- Yunyun Luo
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark.
- Faculty of Health and Medical Sciences, University of Copenhagen, 2200 København, Denmark.
| | - Yi He
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark.
| | - Ditte Reker
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark.
| | - Natasja Stæhr Gudmann
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark.
| | - Kim Henriksen
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark.
| | - Ole Simonsen
- Department Orthopedic Surgery, Aalborg University Hospital, 9000 Aalborg, Denmark.
| | | | | | - Ali Mobasheri
- D-BOARD EU Consortium for Biomarker Discovery, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, Surrey, UK.
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Queen's Medical Centre, Nottingham NG7 2UH, Nottinghamshire, UK.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania.
| | - Morten Karsdal
- Department of Rheumatology, Nordic Bioscience, Biomarkers and Research, 2730 Herlev, Denmark.
| | | |
Collapse
|
47
|
Bay-Jensen A, Kjelgaard-Petersen C, Petersen K, Arendt-Nielsen L, Quasnichka H, Mobasheri A, Karsdal M, Leeming D. Aggrecanase degradation of type III collagen is associated with clinical knee pain. Clin Biochem 2018; 58:37-43. [DOI: 10.1016/j.clinbiochem.2018.04.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 04/17/2018] [Accepted: 04/23/2018] [Indexed: 11/16/2022]
|
48
|
Kjelgaard-Petersen CF, Sharma N, Kayed A, Karsdal MA, Mobasheri A, Hägglund P, Bay-Jensen AC, Thudium CS. Tofacitinib and TPCA-1 exert chondroprotective effects on extracellular matrix turnover in bovine articular cartilage ex vivo. Biochem Pharmacol 2018; 165:91-98. [PMID: 30059674 DOI: 10.1016/j.bcp.2018.07.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/25/2018] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Currently, there are no disease-modifying osteoarthritis drugs (DMOADs) approved for osteoarthritis. It is hypothesized that a subtype of OA may be driven by inflammation and may benefit from treatment with anti-inflammatory small molecule inhibitors adopted from treatments of rheumatoid arthritis. This study aimed to investigate how small molecule inhibitors of intracellular signaling modulate cartilage degradation and formation as a pre-clinical model for structural effects. DESIGN Bovine cartilage explants were cultured with oncostatin M (OSM) and tumour necrosis factor α (TNF-α) either alone or combined with the small molecule inhibitors: SB203580 (p38 inhibitor), R406 (Spleen tyrosine kinase (Syk) inhibitor), TPCA-1 (Inhibitor of κB kinase (Ikk) inhibitor), or Tofacitinib (Tofa) (Janus kinases (Jak) inhibitor). Cartilage turnover was assessed with the biomarkers of degradation (AGNx1 and C2M), and type II collagen formation (PRO-C2) using ELISA. Explant proteoglycan content was assessed by Safranin O/Fast Green staining. RESULTS R406, TPCA-1 and Tofa reduced the cytokine-induced proteoglycan loss and decreased AGNx1 release 3.7-, 43- and 32-fold, respectively. SB203580 showed no effect. All inhibitors suppressed C2M at a concentration of 3 µM. TPCA-1 and Tofa increased the cytokine reduced PRO-C2 3.5 and 3.7-fold, respectively. CONCLUSION Using a pre-clinical model we found that the inhibitors TPCA-1 and Tofa inhibited cartilage degradation and rescue formation of type II collagen under inflammatory conditions, while R406 and SB203580 only inhibited cartilage degradation, and SB203580 only partially. These pre-clinical data suggest that TPCA-1 and Tofa preserve and help maintain cartilage ECM under inflammatory conditions and could be investigated further as DMOADs for inflammation-driven osteoarthritis.
Collapse
Affiliation(s)
- Cecilie F Kjelgaard-Petersen
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark; Department of Bioengineering and Biomedicine, Technical University of Denmark, Søltofts Plads Building 221, DK-2800 Kgs. Lyngby, Denmark
| | - Neha Sharma
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark.
| | - Ashref Kayed
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark.
| | - Morten A Karsdal
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark.
| | - Ali Mobasheri
- Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences University of Surrey, Guildford GU2 7AL, United Kingdom.
| | - Per Hägglund
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark.
| | | | - Christian S Thudium
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark.
| |
Collapse
|
49
|
Kjelgaard-Petersen CF, Platt A, Braddock M, Jenkins MA, Musa K, Graham E, Gantzel T, Slynn G, Weinblatt ME, Karsdal MA, Thudium CS, Bay-Jensen AC. Translational Biomarkers and Ex Vivo Models of Joint Tissues as a Tool for Drug Development in Rheumatoid Arthritis. Arthritis Rheumatol 2018; 70:1419-1428. [PMID: 29669391 PMCID: PMC6174937 DOI: 10.1002/art.40527] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 04/10/2018] [Indexed: 12/15/2022]
Abstract
Objective Rheumatoid arthritis (RA) is a chronic and degenerative autoimmune joint disease that leads to disability, reduced quality of life, and increased mortality. Although several synthetic and biologic disease‐modifying antirheumatic drugs are available, there is still a medical need for novel drugs that control disease progression. As only 10% of experimental drug candidates for treatment of RA that enter phase I trials are eventually registered by the Food and Drug Administration, there is an immediate need for translational tools to facilitate early decision‐making in drug development. In this study, we aimed to determine if the inability of fostamatinib (a small molecule inhibitor of Syk) to demonstrate sufficient efficacy in phase III of a previous clinical study could have been predicted earlier in the development process. Methods Biomarkers of bone, cartilage, and interstitial matrix turnover (C‐telopeptide of type I collagen [CTX‐I], matrix metalloproteinase–derived types I, II, and III collagen neoepitopes [C1M, C2M, and C3M]) were measured in 450 serum samples from the Oral Syk Inhibition in Rheumatoid Arthritis 1 study (OSKIRA‐1, a phase III clinical study of the efficacy of fostamatinib in RA) at baseline and follow‐up. Additionally, the same biomarkers were subsequently measured in conditioned media from osteoclast, cartilage, and synovial membrane cultured with the active metabolite of fostamatinib, R406, to assess the level of suppression induced by the drug. Results In OSKIRA‐1 serum samples and osteoclast and cartilage cultures, fostamatinib suppressed the levels of CTX‐I and C2M. In OSKIRA‐1 serum samples and synovial membrane cultures, fostamatinib did not mediate any clinical or preclinical effect on either C1M or C3M, which have previously been associated with disease response and efficacy. Conclusion These data demonstrate that translational biomarkers are a potential tool for early assessment and decision‐making in drug development for RA treatment.
Collapse
|
50
|
Löfvall H, Newbould H, Karsdal MA, Dziegiel MH, Richter J, Henriksen K, Thudium CS. Osteoclasts degrade bone and cartilage knee joint compartments through different resorption processes. Arthritis Res Ther 2018; 20:67. [PMID: 29636095 PMCID: PMC5894194 DOI: 10.1186/s13075-018-1564-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/12/2018] [Indexed: 01/13/2023] Open
Abstract
Background Osteoclasts have been strongly implicated in osteoarthritic cartilage degradation, at least indirectly via bone resorption, and have been shown to degrade cartilage in vitro. The osteoclast resorption processes required to degrade subchondral bone and cartilage—the remodeling of which is important in the osteoarthritic disease process—have not been previously described, although cathepsin K has been indicated to participate. In this study we profile osteoclast-mediated degradation of bovine knee joint compartments in a novel in vitro model using biomarkers of extracellular matrix (ECM) degradation to assess the potential of osteoclast-derived resorption processes to degrade different knee joint compartments. Methods Mature human osteoclasts were cultured on ECMs isolated from bovine knees—articular cartilage, cortical bone, and osteochondral junction ECM (a subchondral bone-calcified cartilage mixture)—in the presence of inhibitors: the cystein protease inhibitor E-64, the matrix metalloproteinase (MMP) inhibitor GM6001, or the vacuolar-type H+-ATPase (V-ATPase) inhibitor diphyllin. Biomarkers of bone (calcium and C-terminal type I collagen (CTX-I)) and cartilage (C2M) degradation were measured in the culture supernatants. Cultures without osteoclasts were used as background samples. Background-subtracted biomarker levels were normalized to the vehicle condition and were analyzed using analysis of variance with Tukey or Dunnett’s T3 post hoc test, as applicable. Results Osteochondral CTX-I release was inhibited by E-64 (19% of vehicle, p = 0.0008), GM6001 (51% of vehicle, p = 0.013), and E-64/GM6001 combined (4% of vehicle, p = 0.0007)—similarly to bone CTX-I release. Diphyllin also inhibited osteochondral CTX-I release (48% of vehicle, p = 0.014), albeit less than on bone (4% of vehicle, p < 0.0001). Osteochondral C2M release was only inhibited by E-64 (49% of vehicle, p = 0.07) and GM6001 (14% of vehicle, p = 0.006), with complete abrogation when combined (0% of vehicle, p = 0.004). Cartilage C2M release was non-significantly inhibited by E-64 (69% of vehicle, p = 0.98) and was completely abrogated by GM6001 (0% of vehicle, p = 0.16). Conclusions Our study supports that osteoclasts can resorb non-calcified and calcified cartilage independently of acidification. We demonstrated both MMP-mediated and cysteine protease-mediated resorption of calcified cartilage. Osteoclast functionality was highly dependent on the resorbed substrate, as different ECMs required different osteoclast processes for degradation. Our novel culture system has potential to facilitate drug and biomarker development aimed at rheumatic diseases, e.g. osteoarthritis, where pathological osteoclast processes in specific joint compartments may contribute to the disease process. Electronic supplementary material The online version of this article (10.1186/s13075-018-1564-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Henrik Löfvall
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark.,Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | - Hannah Newbould
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Morten A Karsdal
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Morten H Dziegiel
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Johan Richter
- Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | - Kim Henriksen
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | | |
Collapse
|