51
|
Trabecular structural changes in the mandibular condyle caused by degenerative osteoarthritis: a comparative study by cone-beam computed tomography imaging. Oral Radiol 2018; 35:51-58. [DOI: 10.1007/s11282-018-0324-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 01/27/2018] [Indexed: 10/17/2022]
|
52
|
Biochemical marker discovery, testing and evaluation for facilitating OA drug discovery and development. Drug Discov Today 2018; 23:349-358. [DOI: 10.1016/j.drudis.2017.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 09/18/2017] [Accepted: 10/06/2017] [Indexed: 01/25/2023]
|
53
|
Xu X, Zheng L, Yuan Q, Zhen G, Crane JL, Zhou X, Cao X. Transforming growth factor-β in stem cells and tissue homeostasis. Bone Res 2018; 6:2. [PMID: 29423331 PMCID: PMC5802812 DOI: 10.1038/s41413-017-0005-4] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/12/2017] [Accepted: 11/15/2017] [Indexed: 02/05/2023] Open
Abstract
TGF-β 1-3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Gehua Zhen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Janet L. Crane
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
54
|
Savic D, Pedoia V, Seo Y, Yang J, Bucknor M, Franc BL, Majumdar S. Imaging Bone-Cartilage Interactions in Osteoarthritis Using [ 18F]-NaF PET-MRI. Mol Imaging 2018; 15:1-12. [PMID: 28654417 PMCID: PMC5470142 DOI: 10.1177/1536012116683597] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Purpose: Simultaneous positron emission tomography–magnetic resonance imaging (PET-MRI) is an emerging technology providing both anatomical and functional images without increasing the scan time. Compared to the traditional PET/computed tomography imaging, it also exposes the patient to significantly less radiation and provides better anatomical images as MRI provides superior soft tissue characterization. Using PET-MRI, we aim to study interactions between cartilage composition and bone function simultaneously, in knee osteoarthritis (OA). Procedures: In this article, bone turnover and remodeling was studied using [18F]-sodium fluoride (NaF) PET data. Quantitative MR-derived T1ρ relaxation times characterized the biochemical cartilage degeneration. Sixteen participants with early signs of OA of the knee received intravenous injections of [18F]-NaF at the onset of PET-MR image acquisition. Regions of interest were identified, and kinetic analysis of dynamic PET data provided the rate of uptake (Ki) and the normalized uptake (standardized uptake value) of [18F]-NaF in the bone. Morphological MR images and quantitative voxel-based T1ρ maps of cartilage were obtained using an atlas-based registration technique to segment cartilage automatically. Voxel-by-voxel statistical parameter mapping was used to investigate the relationship between bone and cartilage. Results: Increases in cartilage T1ρ, indicating degenerative changes, were associated with increased turnover in the adjoining bone but reduced turnover in the nonadjoining compartments. Associations between pain and increased bone uptake were seen in the absence of morphological lesions in cartilage, but the relationship was reversed in the presence of incident cartilage lesions. Conclusion: This study shows significant cartilage and bone interactions in OA of the knee joint using simultaneous [18F]-NaF PET-MR, the first in human study. These observations highlight the complex biomechanical and biochemical interactions in the whole knee joint in OA, which potentially could help assess therapeutic targets in treating OA.
Collapse
Affiliation(s)
- Dragana Savic
- 1 Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.,2 Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Valentina Pedoia
- 1 Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Youngho Seo
- 1 Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Jaewon Yang
- 1 Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Matt Bucknor
- 1 Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin L Franc
- 1 Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Sharmila Majumdar
- 1 Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
55
|
Yang X, He H, Gao Q, He C. Pulsed electromagnetic field improves subchondral bone microstructure in knee osteoarthritis rats through a Wnt/β-catenin signaling-associated mechanism. Bioelectromagnetics 2017; 39:89-97. [PMID: 29251361 DOI: 10.1002/bem.22106] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 11/22/2017] [Indexed: 02/05/2023]
Affiliation(s)
- Xiaotian Yang
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Hongchen He
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Qiang Gao
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Chengqi He
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| |
Collapse
|
56
|
Reina N, Cavaignac E, Pailhé R, Pailliser A, Bonnevialle N, Swider P, Laffosse JM. BMI-related microstructural changes in the tibial subchondral trabecular bone of patients with knee osteoarthritis. J Orthop Res 2017; 35:1653-1660. [PMID: 27747928 DOI: 10.1002/jor.23459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 10/07/2016] [Indexed: 02/04/2023]
Abstract
Overweight is a risk factor for osteoarthritis on the knees. Subchondral trabecular bone (SCTB) densification has been shown to be associated with cartilage degeneration. This study analyzed the microarchitectural changes in the SCTB of tibial plateaus to validate the hypothesis that the degree of remodeling is correlated with a patient's body weight. Twenty-one tibial plateaus were collected during total knee arthroplasty from 21 patients (15 women and 6 men). These patients had a mean age of 70.4 years (49-81), mean weight of 74.7 kg (57-93) and mean body mass index (BMI) of 28.4 kg/m2 (21.3-40.8). One cylindrical plug was harvested in the center of each tibial plateau (medial and lateral). Micro-CT parameters (7.4 μm resolution) were determined to describe the SCTB structure. On the medial plateau, there were significant correlations between BMI and bone volume fraction BV/TV (r = 0.595, p = 0.004), structure model index SMI (r = -0.704 p = 0.0002), trabecular space Tb.Sp (r = 0.600, p = 0.04) and trabecular number Tb.N (r = 0.549, p = 0.01). SCTB densification during osteoarthritis is associated with a reduction in its elastic modulus, which could increase cartilage stress, and accelerate cartilage loss. SCTB densification has been shown to precede cartilage degeneration. The correlation of SCTB microarchitecture and body weight may explain why knee osteoarthritis is more common in overweight or obese patients. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1653-1660, 2017.
Collapse
Affiliation(s)
- Nicolas Reina
- Department of Orthopaedic and Trauma Surgery, Hôpital Pierre Paul Riquet, Centre Hospitalier Universitaire de Toulouse, Place du Docteur Baylac, TSA 40031, 31059 Toulouse cedex 9, France.,Hominid Evolutionary Biology, AMIS-UMR 5288 CNRS, University of Toulouse, Toulouse, France
| | - Etienne Cavaignac
- Department of Orthopaedic and Trauma Surgery, Hôpital Pierre Paul Riquet, Centre Hospitalier Universitaire de Toulouse, Place du Docteur Baylac, TSA 40031, 31059 Toulouse cedex 9, France.,Hominid Evolutionary Biology, AMIS-UMR 5288 CNRS, University of Toulouse, Toulouse, France
| | - Régis Pailhé
- Biomechanics Laboratory, IMFT UMR 5502, University of Toulouse, Toulouse, France.,Department of Orthopaedic and Trauma Surgery, CHU Grenoble, Toulouse, France
| | - Aymeric Pailliser
- Biomechanics Laboratory, IMFT UMR 5502, University of Toulouse, Toulouse, France
| | - Nicolas Bonnevialle
- Department of Orthopaedic and Trauma Surgery, Hôpital Pierre Paul Riquet, Centre Hospitalier Universitaire de Toulouse, Place du Docteur Baylac, TSA 40031, 31059 Toulouse cedex 9, France.,Biomechanics Laboratory, IMFT UMR 5502, University of Toulouse, Toulouse, France
| | - Pascal Swider
- Biomechanics Laboratory, IMFT UMR 5502, University of Toulouse, Toulouse, France
| | - Jean-Michel Laffosse
- Department of Orthopaedic and Trauma Surgery, Hôpital Pierre Paul Riquet, Centre Hospitalier Universitaire de Toulouse, Place du Docteur Baylac, TSA 40031, 31059 Toulouse cedex 9, France.,Biomechanics Laboratory, IMFT UMR 5502, University of Toulouse, Toulouse, France
| |
Collapse
|
57
|
Contribution of Circulatory Disturbances in Subchondral Bone to the Pathophysiology of Osteoarthritis. Curr Rheumatol Rep 2017; 19:49. [DOI: 10.1007/s11926-017-0660-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
58
|
Aho OM, Finnilä M, Thevenot J, Saarakkala S, Lehenkari P. Subchondral bone histology and grading in osteoarthritis. PLoS One 2017; 12:e0173726. [PMID: 28319157 PMCID: PMC5358796 DOI: 10.1371/journal.pone.0173726] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/24/2017] [Indexed: 11/26/2022] Open
Abstract
Objective Osteoarthritis (OA) has often regarded as a disease of articular cartilage only. New evidence has shifted the paradigm towards a system biology approach, where also the surrounding tissue, especially bone is studied more vigorously. However, the histological features of subchondral bone are only poorly characterized in current histological grading scales of OA. The aim of this study is to specifically characterize histological changes occurring in subchondral bone at different stages of OA and propose a simple grading system for them. Design 20 patients undergoing total knee replacement surgery were randomly selected for the study and series of osteochondral samples were harvested from the tibial plateaus for histological analysis. Cartilage degeneration was assessed using the standardized OARSI grading system, while a novel four-stage grading system was developed to illustrate the changes in subchondral bone. Subchondral bone histology was further quantitatively analyzed by measuring the thickness of uncalcified and calcified cartilage as well as subchondral bone plate. Furthermore, internal structure of calcified cartilage-bone interface was characterized utilizing local binary patterns (LBP) based method. Results The histological appearance of subchondral bone changed drastically in correlation with the OARSI grading of cartilage degeneration. As the cartilage layer thickness decreases the subchondral plate thickness and disorientation, as measured with LBP, increases. Calcified cartilage thickness was highest in samples with moderate OA. Conclusion The proposed grading system for subchondral bone has significant relationship with the corresponding OARSI grading for cartilage. Our results suggest that subchondral bone remodeling is a fundamental factor already in early stages of cartilage degeneration.
Collapse
Affiliation(s)
- Olli-Matti Aho
- Department of Anatomy and Cell Biology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- * E-mail:
| | - Mikko Finnilä
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Jerome Thevenot
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Petri Lehenkari
- Department of Anatomy and Cell Biology, Institute of Biomedicine, University of Oulu, Oulu, Finland
- Division of Orthopaedic and Trauma Surgery, Department of Surgery, Medical Research Center, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
59
|
Wu J, Ma L, Wu L, Jin Q. Wnt-β-catenin signaling pathway inhibition by sclerostin may protect against degradation in healthy but not osteoarthritic cartilage. Mol Med Rep 2017; 15:2423-2432. [PMID: 28259981 PMCID: PMC5428759 DOI: 10.3892/mmr.2017.6278] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 10/04/2016] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to determine the regulation of sclerostin (SOST) in osteoarthritis (OA) and its effect on articular cartilage degradation. Human cartilage samples from healthy and OA subjects were assessed by Safranin O staining and immunohistochemistry. Primary chondrocytes were pre‑incubated with 250 ng/ml SOST, 10 ng/ml interleukin‑1‑α (IL‑1α) or a combination of the two. The effects of treatment on the Wnt-β-catenin signaling pathway and cartilage degradation were examined by reverse transcription‑quantitative polymerase chain reaction and western blotting. SOST was detected in the cartilage focal area, demonstrating secretion by osteocytes and chondrocytes. SOST has been identified to inhibit the Wnt-β-catenin signaling pathway by binding to low‑density lipoprotein‑related receptors 5 and 6, and catabolic factors were decreased in healthy chondrocytes. However, SOST did not influence human OA chondrocytes. IL‑1α activated the Wnt-β-catenin signaling pathway and promoted cartilage degradation, which was inhibited by SOST in healthy and OA cartilage. The results of the present study suggested that SOST is important in maintaining the integrity of healthy, but not end‑stage OA, cartilage.
Collapse
Affiliation(s)
- Jiang Wu
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Long Ma
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Long Wu
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Qunhua Jin
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
60
|
Yang X, He H, Zhou Y, Zhou Y, Gao Q, Wang P, He C. Pulsed electromagnetic field at different stages of knee osteoarthritis in rats induced by low-dose monosodium iodoacetate: Effect on subchondral trabecular bone microarchitecture and cartilage degradation. Bioelectromagnetics 2016; 38:227-238. [PMID: 28026095 DOI: 10.1002/bem.22028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 12/03/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Xiaotian Yang
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Hongchen He
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Yuan Zhou
- Institute for Disaster Management and Reconstruction of Sichuan University and Hong Kong Polytechnic University; Chengdu China
| | - Yujing Zhou
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Qiang Gao
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Pu Wang
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
| | - Chengqi He
- Department of Rehabilitation Medicine; West China Hospital; Sichuan University; Chengdu China
- Key Laboratory of Rehabilitation Medicine in Sichuan; Chengdu China
- Institute for Disaster Management and Reconstruction of Sichuan University and Hong Kong Polytechnic University; Chengdu China
| |
Collapse
|
61
|
Karsdal MA, Michaelis M, Ladel C, Siebuhr AS, Bihlet AR, Andersen JR, Guehring H, Christiansen C, Bay-Jensen AC, Kraus VB. Disease-modifying treatments for osteoarthritis (DMOADs) of the knee and hip: lessons learned from failures and opportunities for the future. Osteoarthritis Cartilage 2016; 24:2013-2021. [PMID: 27492463 DOI: 10.1016/j.joca.2016.07.017] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 06/30/2016] [Accepted: 07/26/2016] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is the biggest unmet medical need among the many musculoskeletal conditions and the most common form of arthritis. It is a major cause of disability and impaired quality of life in the elderly. We review several ambitious but failed attempts to develop joint structure-modifying treatments for OA. Insights gleaned from these attempts suggest that these failures arose from unrealistic hypotheses, sub-optimal selection of patient populations or drug dose, and/or inadequate sensitivity of the trial endpoints. The long list of failures has prompted a paradigm shift in OA drug development with redirection of attention to: (1) consideration of the benefits of localized vs systemic pharmacological agents, as indicated by the increasing number of intra-articularly administered compounds entering clinical development; (2) recognition of OA as a complex disease with multiple phenotypes, that may each require somewhat different approaches for optimizing treatment; and (3) trial enhancements based on guidance regarding biomarkers provided by regulatory agencies, such as the Food and Drug Administration (FDA), that could be harnessed to help turn failures into successes.
Collapse
Affiliation(s)
| | | | - C Ladel
- Merck KGaA, Darmstadt, Germany
| | | | | | | | | | | | | | - V B Kraus
- Duke Molecular Physiology Institute and Division of Rheumatology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
62
|
Wu L, Guo H, Sun K, Zhao X, Ma T, Jin Q. Sclerostin expression in the subchondral bone of patients with knee osteoarthritis. Int J Mol Med 2016; 38:1395-1402. [PMID: 27665782 PMCID: PMC5065295 DOI: 10.3892/ijmm.2016.2741] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 08/30/2016] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to assess the expression of β-catenin, transcription factor-4 (TCF-4) and sclerostin in the subchondral bone of patients with primary knee osteoarthritis (OA). Tibial plateau specimens from patients with OA who underwent total knee arthroplasty were classified into the early stage (n=15), intermediate stage (n=13) and late stage (n=17) groups using the Mankin score. Structural parameters, including total articular cartilage (TAC), subchondral bone plate (SCP) thickness and trabecular bone volume (BV/TV), were assessed using Image-Pro Plus 6.0 analysis software. Subsequently, β-catenin and sclerostin expression levels in subchondral bone were determined by immunohistochemistry. In addition, the mRNA and protein levels of β-catenin, TCF-4 and sclerostin were evaluated by RT-qPCR and western blot analysis, respectively. As regards the cartilage and subchondral bone structural parameters, TAC was reduced, while SCP thickness and BV/TV were increased due to OA, with significant differences observed among the different stages (all P<0.05). The results of immunohistochemistry revealed that the β-catenin levels in the intermediate- and late-stage samples were significantly increased, while the levels of sclerostin were markedly decreased compared with the values in the early-stage samples (all P<0.05). Compared with the intermediate-stage samples, the sclerostin levels were decreased, and SCP thickness and the β-catenin levels were increased in the late-stage samples (all P<0.05). The results of RT-qPCR and western blot analysis revealed that the β-catenin and TCF-4 mRNA and protein levels in the intermediate- and late-stage samples were significantly increased, while sclerostin expression was significantly decreased compared with the early-stage samples; a similar trend was observed between the intermediate- and late-stage samples (all P<0.05). Finally, the β-catenin and TCF-4 levels positively correlated with the Mankin scores, while there was a negative correlation with sclerostin expression. Our findings demonstrate that sclerostin expression is closely associated with the degree of joint damage in patients with OA, confirming its involvement in the development of OA.
Collapse
Affiliation(s)
- Long Wu
- Department of Orthopedics, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Haohui Guo
- Department of Orthopedics, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Kening Sun
- Department of Orthopedics, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xin Zhao
- Department of Orthopedics, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Tao Ma
- Department of Orthopedics, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Qunhua Jin
- Department of Orthopedics, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
63
|
Tarquini C, Mattera R, Mastrangeli F, Agostinelli S, Ferlosio A, Bei R, Orlandi A, Tarantino U. Comparison of tissue transglutaminase 2 and bone biological markers osteocalcin, osteopontin and sclerostin expression in human osteoporosis and osteoarthritis. Amino Acids 2016; 49:683-693. [PMID: 27357308 DOI: 10.1007/s00726-016-2290-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/22/2016] [Indexed: 01/18/2023]
Abstract
Osteoporosis (OP) and osteoarthritis (OA) are the most common joint diseases, with a high incidence in the elderly population. OP is characterized by trabecular bone remodeling and reabsorption, whereas articular cartilage and subchondral bone remodeling are major features of OA. Although classically considered as independent or even conflicting processes, clinical coexistence of OP and OA was recently described. Transglutaminase 2 (TG2) expression is considered a biomarker of OA, but its role in osteoporotic bone remodeling is still uncertain. We investigated TG2 and bone biological markers (Osteocalcin, Osteopontin, and Sclerostin) in osteoporotic and osteoarthritic osteocartilagineous tissue (n = 54) and human chondrocyte cultures in vitro by immunohistochemistry, immunofluorescence and RT-PCR. Histomorphometric evaluation of bone trabecular remodeling was also performed. In cartilage, TG2 expression was faint in control and OP and significantly less than in OA and OP + OA chondrocytes; the opposite was found for Osteocalcin, whereas Osteopontin and Sclerostin expression was similar. In the subchondral trabecular bone, osteocytes/osteoblasts TG2 expression was slight and similar comparing control, OP, OA, and OP + OA group, whereas Osteocalcin and Osteopontin expression was lower in OP compared to control, OA and OP + OA. Increased TG2 and reduced Osteocalcin expression were maintained in human osteoarthritic chondrocytes in vitro. Histomorphometric analysis confirmed reduced trabecular bone mass in OP and OP + OA compared with OA patients. TG2 represented a suitable biomarker of osteoarthritic chondrocyte activation, whereas osteocalcin and osteopontin characterized osteoporotic osteocyte/osteoblast changes; differences were lost in OP + OA patients, suggesting careful consideration when coexistence of the two diseases occurs.
Collapse
Affiliation(s)
- Chiara Tarquini
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy.,Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy
| | - Rosanna Mattera
- General Pathology, Department of Clinical Sciences and Translational Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Francesca Mastrangeli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy.,Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy
| | - Sara Agostinelli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
| | - Amedeo Ferlosio
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
| | - Roberto Bei
- General Pathology, Department of Clinical Sciences and Translational Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy. .,Policlinic of Tor Vergata of Rome, Rome, Italy.
| | - Umberto Tarantino
- Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy.,Policlinic of Tor Vergata of Rome, Rome, Italy
| |
Collapse
|
64
|
Poulet B, Staines KA. New developments in osteoarthritis and cartilage biology. Curr Opin Pharmacol 2016; 28:8-13. [DOI: 10.1016/j.coph.2016.02.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 01/05/2023]
|
65
|
Yu DG, Nie SB, Liu FX, Wu CL, Tian B, Wang WG, Wang XQ, Zhu ZA, Mao YQ. Dynamic Alterations in Microarchitecture, Mineralization and Mechanical Property of Subchondral Bone in Rat Medial Meniscal Tear Model of Osteoarthritis. Chin Med J (Engl) 2016; 128:2879-86. [PMID: 26521785 PMCID: PMC4756896 DOI: 10.4103/0366-6999.168045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background: The properties of subchondral bone influence the integrity of articular cartilage in the pathogenesis of osteoarthritis (OA). However, the characteristics of subchondral bone alterations remain unresolved. The present study aimed to observe the dynamic alterations in the microarchitecture, mineralization, and mechanical properties of subchondral bone during the progression of OA. Methods: A medial meniscal tear (MMT) operation was performed in 128 adult Sprague Dawley rats to induce OA. At 2, 4, 8, and 12 weeks following the MMT operation, cartilage degeneration was evaluated using toluidine blue O staining, whereas changes in the microarchitecture indices and tissue mineral density (TMD), mineral-to-collagen ratio, and intrinsic mechanical properties of subchondral bone plates (BPs) and trabecular bones (Tbs) were measured using micro-computed tomography scanning, confocal Raman microspectroscopy and nanoindentation testing, respectively. Results: Cartilage degeneration occurred and worsened progressively from 2 to 12 weeks after OA induction. Microarchitecture analysis revealed that the subchondral bone shifted from bone resorption early (reduced trabecular BV/TV, trabecular number, connectivity density and trabecular thickness [Tb.Th], and increased trabecular spacing (Tb.Sp) at 2 and 4 weeks) to bone accretion late (increased BV/TV, Tb.Th and thickness of subchondral bone plate, and reduced Tb.Sp at 8 and 12 weeks). The TMD of both the BP and Tb displayed no significant changes at 2 and 4 weeks but decreased at 8 and 12 weeks. The mineral-to-collagen ratio showed a significant decrease from 4 weeks for the Tb and from 8 weeks for the BP after OA induction. Both the elastic modulus and hardness of the Tb showed a significant decrease from 4 weeks after OA induction. The BP showed a significant decrease in its elastic modulus from 8 weeks and its hardness from 4 weeks. Conclusion: The microarchitecture, mineralization and mechanical properties of subchondral bone changed in a time-dependent manner as OA progressed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yuan-Qing Mao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
66
|
Muttigi MS, Han I, Park HK, Park H, Lee SH. Matrilin-3 Role in Cartilage Development and Osteoarthritis. Int J Mol Sci 2016; 17:ijms17040590. [PMID: 27104523 PMCID: PMC4849044 DOI: 10.3390/ijms17040590] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/10/2016] [Accepted: 04/13/2016] [Indexed: 11/16/2022] Open
Abstract
The extracellular matrix (ECM) of cartilage performs essential functions in differentiation and chondroprogenitor cell maintenance during development and regeneration. Here, we discuss the vital role of matrilin-3, an ECM protein involved in cartilage development and potential osteoarthritis pathomechanisms. As an adaptor protein, matrilin-3 binds to collagen IX to form a filamentous network around cells. Matrilin-3 is an essential component during cartilage development and ossification. In addition, it interacts directly or indirectly with transforming growth factor β (TGF-β), and bone morphogenetic protein 2 (BMP2) eventually regulates chondrocyte proliferation and hypertrophic differentiation. Interestingly, matrilin-3 increases interleukin receptor antagonists (IL-Ra) in chondrocytes, suggesting its role in the suppression of IL-1β-mediated inflammatory action. Matrilin-3 downregulates the expression of matrix-degrading enzymes, such as a disintegrin metalloproteinase with thrombospondin motifs 4 (ADAMTS4) and ADAMTS5, matrix metalloproteinase 13 (MMP13), and collagen X, a hypertrophy marker during development and inflammatory conditions. Matrilin-3 essentially enhances collagen II and aggrecan expression, which are required to maintain the tensile strength and elasticity of cartilage, respectively. Interestingly, despite these attributes, matrilin-3 induces osteoarthritis-associated markers in chondrocytes in a concentration-dependent manner. Existing data provide insights into the critical role of matrilin-3 in inflammation, matrix degradation, and matrix formation in cartilage development and osteoarthritis.
Collapse
Affiliation(s)
- Manjunatha S Muttigi
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Korea.
- Department of Biomedical Science, CHA University, Seongnam-Si 13488, Korea.
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Korea.
| | - Hun-Kuk Park
- Department of Biomedical Engineering, Collage of Medicine, Kyung Hee University, Seoul 02447, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-Si 13488, Korea.
| |
Collapse
|
67
|
Subchondral and epiphyseal bone remodeling following surgical transection and noninvasive rupture of the anterior cruciate ligament as models of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2016; 24:698-708. [PMID: 26620090 DOI: 10.1016/j.joca.2015.11.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/03/2015] [Accepted: 11/06/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Animal models are frequently used to study post-traumatic osteoarthritis (PTOA). A common anterior cruciate ligament (ACL) injury model is surgical transection, which may introduce confounding factors from surgery. Noninvasive models could model human injury more closely. The purpose of this study was to compare subchondral and epiphyseal trabecular bone remodeling after surgical transection and noninvasive rupture of the ACL. METHODS Thirty-six rats were randomized to an uninjured control, surgical transection (Transection), or noninvasive rupture (Rupture). Animals were randomized to 4 or 10 week time points (n = 6 per group). Micro computed tomography (μCT) imaging was performed with an isotropic voxel size of 12 μm. Subchondral and epiphyseal bone was segmented semi-automatically, and morphometric analysis was performed. RESULTS Transection caused a greater decrease in subchondral bone volume fraction (BV/TV) than Rupture in the femur and tibia. Rupture had greater subchondral bone tissue mineral density (TMD) at 4 and 10 weeks in the femur and tibia. Subchondral bone thickness (SCB.Th) was decreased in the femur in Transection only. Epiphyseal BV/TV was decreased in Transection only, and Rupture exhibited increased femoral epiphyseal TMD compared to both Control and Transection. Rupture exhibited greater femoral epiphyseal trabecular thickness (Tb.Th.) compared to Control and Transection at 4 weeks, and both Rupture and Transection had increased femoral epiphyseal Tb.Th. at 10 weeks. Epiphyseal trabecular number (Tb.N) was decreased in both injury groups at both time points. Femoral and tibial epiphyseal structure model index (SMI) increased in both groups. CONCLUSIONS The two injury models cause differences in post-injury bone morphometry, and surgical transection may be introducing confounding factors that affect downstream bony remodeling.
Collapse
|
68
|
Bruyère O, Cooper C, Arden N, Branco J, Brandi ML, Herrero-Beaumont G, Berenbaum F, Dennison E, Devogelaer JP, Hochberg M, Kanis J, Laslop A, McAlindon T, Reiter S, Richette P, Rizzoli R, Reginster JY. Can we identify patients with high risk of osteoarthritis progression who will respond to treatment? A focus on epidemiology and phenotype of osteoarthritis. Drugs Aging 2016; 32:179-87. [PMID: 25701074 PMCID: PMC4366553 DOI: 10.1007/s40266-015-0243-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Osteoarthritis is a syndrome affecting a variety of patient profiles. A European Society for Clinical and Economic Aspects of Osteoporosis and Osteoarthritis and the European Union Geriatric Medicine Society working meeting explored the possibility of identifying different patient profiles in osteoarthritis. The risk factors for the development of osteoarthritis include systemic factors (e.g., age, sex, obesity, genetics, race, and bone density) and local biomechanical factors (e.g., obesity, sport, joint injury, and muscle weakness); most also predict disease progression, particularly joint injury, malalignment, and synovitis/effusion. The characterization of patient profiles should help to better orientate research, facilitate trial design, and define which patients are the most likely to benefit from treatment. There are a number of profile candidates. Generalized, polyarticular osteoarthritis and local, monoarticular osteoarthritis appear to be two different profiles; the former is a feature of osteoarthritis co-morbid with inflammation or the metabolic syndrome, while the latter is more typical of post-trauma osteoarthritis, especially in cases with severe malalignment. Other biomechanical factors may also define profiles, such as joint malalignment, loss of meniscal function, and ligament injury. Early- and late-stage osteoarthritis appear as separate profiles, notably in terms of treatment response. Finally, there is evidence that there are two separate profiles related to lesions in the subchondral bone, which may determine benefit from bone-active treatments. Decisions on appropriate therapy should be made considering clinical presentation, underlying pathophysiology, and stage of disease. Identification of patient profiles may lead to more personalized healthcare, with more targeted treatment for osteoarthritis.
Collapse
Affiliation(s)
- Olivier Bruyère
- Department of Public Health, Epidemiology and Health Economics, University of Liège, CHU Sart-Tilman B23, 4000, Liège, Belgium,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
AMD3100 Attenuates Matrix Metalloprotease-3 and -9 Expressions and Prevents Cartilage Degradation in a Monosodium Iodo-Acetate-Induced Rat Model of Temporomandibular Osteoarthritis. J Oral Maxillofac Surg 2016; 74:927.e1-927.e13. [PMID: 26851314 DOI: 10.1016/j.joms.2015.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 12/25/2015] [Indexed: 01/02/2023]
Abstract
PURPOSE Temporomandibular joint osteoarthritis (TMJOA) is an important subtype of temporomandibular disorder. This study investigated the inflammatory role of the stromal cell-derived factor-1 (SDF-1) and C-X-C chemokine receptor-4 (CXCR4) axis and the probable signaling pathway involved in matrix metalloprotease (MMP)-3 and MMP-9 productions stimulated by the SDF-1-CXCR4 axis in an experimental rat model of TMJOA. MATERIALS AND METHODS Rats were randomly divided into a control group, a pathologic model group, and an AMD3100 group. Effects of the bicyclam derivative AMD3100 (the specific antagonist of SDF-1-CXCR4 axis) were studied in TMJOA experimentally induced by monosodium iodo-acetate. Productions of SDF-1 and CXCR4 were compared in the normal and pathologic model groups, and cartilage changes and expressions of MMP-3, MMP-9, and phosphorylated extracellular signal-regulated kinase (p-ERK) were compared in the control, pathologic model, and AMD3100 groups. RESULTS Expressions of SDF-1 and CXCR4 in the pathologic model group were increased compared with the control group (P < .05). Releases of MMP-3, MMP-9, and p-ERK and cartilage changes were downregulated in the AMD3100 group compared with the pathologic model group (P < .05), and these changes occurred in a dose-dependent manner with AMD3100 concentrations. Moreover, there were strong predictive relations between the expression of p-ERK with MMP-3 (r(2) = 0.419; P < .001) and with MMP-9 (r(2) = 0.542; P < .001). CONCLUSIONS The SDF-1-CXCR4 signaling pathway plays a proinflammatory role in experimental TMJOA, the bicyclam derivative AMD3100 can alleviate the severity of experimental TMJOA, and there might be a potential relation between the SDF-1-CXCR4 axis and the ERK signaling pathway.
Collapse
|
70
|
Kartha S, Zhou T, Granquist EJ, Winkelstein BA. Development of a Rat Model of Mechanically Induced Tunable Pain and Associated Temporomandibular Joint Responses. J Oral Maxillofac Surg 2016; 74:54.e1-10. [DOI: 10.1016/j.joms.2015.09.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 11/24/2022]
|
71
|
Bagi CM, Berryman E, Zakur DE, Wilkie D, Andresen CJ. Effect of antiresorptive and anabolic bone therapy on development of osteoarthritis in a posttraumatic rat model of OA. Arthritis Res Ther 2015; 17:315. [PMID: 26542671 PMCID: PMC4635572 DOI: 10.1186/s13075-015-0829-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/20/2015] [Indexed: 12/15/2022] Open
Abstract
Introduction Osteoarthritis (OA) is a leading cause of disability, but despite the high unmet clinical need and extensive research seeking dependable therapeutic interventions, no proven disease-modifying treatment for OA is currently available. Due to the close interaction and interplay between the articular cartilage and the subchondral bone plate, it has been hypothesized that antiresorptive drugs can also reduce cartilage degradation, inhibit excessive turnover of the subchondral bone plate, prevent osteophyte formation, and/or that bone anabolic drugs might also stimulate cartilage synthesis by chondrocytes and preserve cartilage integrity. The benefit of intensive zoledronate (Zol) and parathyroid hormone (PTH) therapy for bone and cartilage metabolism was evaluated in a rat model of OA. Methods Medial meniscectomy (MM) was used to induce OA in male Lewis rats. Therapy with Zol and human PTH was initiated immediately after surgery. A dynamic weight-bearing (DWB) system was deployed to evaluate the weight-bearing capacity of the front and hind legs. At the end of the 10-week study, the rats were euthanized and the cartilage pathology was evaluated by contrast (Hexabrix)-enhanced μCT imaging and traditional histology. Bone tissue was evaluated at the tibial metaphysis and epiphysis, including the subchondral bone. Histological techniques and dynamic histomorphometry were used to evaluate cartilage morphology and bone mineralization. Results The results of this study highlight the complex changes in bone metabolism in different bone compartments influenced by local factors, including inflammation, pain and mechanical loads. Surgery caused severe and extensive deterioration of the articular cartilage at the medial tibial plateau, as evidenced by contrast-enhanced μCT and histology. The study results showed the negative impact of MM surgery on the weight-bearing capacity of the operated limb, which was not corrected by treatment. Although both Zol and PTH improved subchondral bone mass and Zol reduced serum CTX-II level, both treatments failed to prevent or correct cartilage deterioration, osteophyte formation and mechanical incapacity. Conclusions The various methods utilized in this study showed that aggressive treatment with Zol and PTH did not have the capacity to prevent or correct the deterioration of the hyaline cartilage, thickening of the subchondral bone plate, osteophyte formation or the mechanical incapacity of the osteoarthritic knee.
Collapse
Affiliation(s)
- Cedo M Bagi
- Global Science and Technology, Pfizer Global Research and Development, Pfizer Inc., 100 Eastern Point Road, Groton, CT, 06340, USA.
| | - Edwin Berryman
- Comparative Medicine, Global Science and Technology, Pfizer Global Research and Development, Pfizer Inc., 100 Eastern Point Road, Groton, CT, 06340, USA.
| | - David E Zakur
- Comparative Medicine, Global Science and Technology, Pfizer Global Research and Development, Pfizer Inc., 100 Eastern Point Road, Groton, CT, 06340, USA. david.zakur.@pfizer.com
| | - Dean Wilkie
- Investigative Pathology, Drug Safety Research and Development, Pfizer Inc., 100 Eastern Point Road, Groton, CT, 06340, USA.
| | - Catharine J Andresen
- Comparative Medicine, Global Science and Technology, Pfizer Global Research and Development, Pfizer Inc., 100 Eastern Point Road, Groton, CT, 06340, USA.
| |
Collapse
|
72
|
Wang K, Xu J, Hunter DJ, Ding C. Investigational drugs for the treatment of osteoarthritis. Expert Opin Investig Drugs 2015; 24:1539-1556. [PMID: 26429673 DOI: 10.1517/13543784.2015.1091880] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Osteoarthritis (OA) is a common joint disease with multiple pathophysiological processes, affecting the whole joint. Current therapeutic options such as NSAIDs can provide a palliative effect on symptoms but have limited effect on disease progression. New drugs targeting OA structures may retard disease progression at an earlier stage and delay the need for joint replacement. AREAS COVERED Some drugs have entered into clinical trials and a few, such as strontium ranelate, do have improvements in both pain and structure changes. However, most of them have failed in clinical trials largely due to increased side effects or the failure to identify the right OA phenotype for the right drug in clinical design. This review describes various investigational drugs developed for the treatment of OA covering those at stages from preclinical experiments to early phase clinical trials. They include drugs for slowing cartilage degradation, regulating cartilage metabolism, targeting subchondral bone, controlling inflammation and relieving pain. EXPERT OPINION Treatment options for OA remain limited. However, with the emergence of sensitive tools to detect early disease progression and identification of different OA phenotypes, disease-modifying anti-OA drugs with increased benefit and reduced risks will become available for OA treatment in the near future.
Collapse
Affiliation(s)
- Kang Wang
- a 1 The First Affiliated Hospital of Anhui Medical University, Arthritis Research Institute, Department of Rheumatology and Immunology , Hefei, China
| | - Jianhua Xu
- a 1 The First Affiliated Hospital of Anhui Medical University, Arthritis Research Institute, Department of Rheumatology and Immunology , Hefei, China
| | - David J Hunter
- b 2 University of Sydney, Kolling Institute, Institute of Bone and Joint Research, Royal North Shore Hospital, Rheumatology Department , Sydney, NSW, Australia
| | - Changhai Ding
- a 1 The First Affiliated Hospital of Anhui Medical University, Arthritis Research Institute, Department of Rheumatology and Immunology , Hefei, China
- c 3 University of Tasmania, Menzies Institute for Medical Research , Private Bag 23, Hobart, Tasmania 7000, Australia +61 3 62 26 77 30 ; +61 3 62 26 77 04 ;
| |
Collapse
|
73
|
[OVERLOAD of joints and its role in osteoarthritis. Towards understanding and preventing progression of primary osteoarthritis]. Z Rheumatol 2015; 74:618-21. [PMID: 26334971 DOI: 10.1007/s00393-015-1649-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
74
|
Lu S, Lam J, Trachtenberg JE, Lee EJ, Seyednejad H, van den Beucken JJJP, Tabata Y, Kasper FK, Scott DW, Wong ME, Jansen JA, Mikos AG. Technical Report: Correlation Between the Repair of Cartilage and Subchondral Bone in an Osteochondral Defect Using Bilayered, Biodegradable Hydrogel Composites. Tissue Eng Part C Methods 2015; 21:1216-25. [PMID: 26177155 DOI: 10.1089/ten.tec.2015.0117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The present work investigated correlations between cartilage and subchondral bone repair, facilitated by a growth factor-delivering scaffold, in a rabbit osteochondral defect model. Histological scoring indices and microcomputed tomography morphological parameters were used to evaluate cartilage and bone repair, respectively, at 6 and 12 weeks. Correlation analysis revealed significant associations between specific cartilage indices and subchondral bone parameters that varied with location in the defect (cortical vs. trabecular region), time point (6 vs. 12 weeks), and experimental group (insulin-like growth factor-1 only, bone morphogenetic protein-2 only, or both growth factors). In particular, significant correlations consistently existed between cartilage surface regularity and bone quantity parameters. Overall, correlation analysis between cartilage and bone repair provided a fuller understanding of osteochondral repair and can help drive informed studies for future osteochondral regeneration strategies.
Collapse
Affiliation(s)
- Steven Lu
- 1 Department of Bioengineering, Rice University , Houston, Texas
| | - Johnny Lam
- 1 Department of Bioengineering, Rice University , Houston, Texas
| | | | - Esther J Lee
- 1 Department of Bioengineering, Rice University , Houston, Texas
| | - Hajar Seyednejad
- 1 Department of Bioengineering, Rice University , Houston, Texas
| | | | - Yasuhiko Tabata
- 3 Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University , Kyoto, Japan
| | - F Kurtis Kasper
- 4 Department of Orthodontics, University of Texas School of Dentistry at Houston , Houston, Texas
| | - David W Scott
- 5 Department of Statistics, Rice University , Houston, Texas
| | - Mark E Wong
- 6 Department of Oral and Maxillofacial Surgery, University of Texas School of Dentistry at Houston , Houston, Texas
| | - John A Jansen
- 2 Department of Biomaterials, Radboudumc , Nijmegen, The Netherlands
| | - Antonios G Mikos
- 1 Department of Bioengineering, Rice University , Houston, Texas
| |
Collapse
|
75
|
Willie BM, Pap T, Perka C, Schmidt CO, Eckstein F, Arampatzis A, Hege HC, Madry H, Vortkamp A, Duda GN. OVERLOAD of joints and its role in osteoarthritis : Towards understanding and preventing progression of primary osteoarthritis. English version. Z Rheumatol 2015. [PMID: 26224533 DOI: 10.1007/s00393-014-1561-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- B M Willie
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - T Pap
- Institute of Experimental Musculoskeletal Medicine, Westfalian Wilhelms-University Münster, Münster, Germany
| | - C Perka
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany.,Orthopädische Klinik, Centrum für Musculoskeletale Chirurgie, Berlin, Germany
| | - C O Schmidt
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - F Eckstein
- Institute of Anatomy, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| | - A Arampatzis
- Department of Training and Movement Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - H-C Hege
- Zuse Institute Berlin (ZIB), Berlin, Germany
| | - H Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - A Vortkamp
- Department of Developmental Biology and Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - G N Duda
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
76
|
Xia P, Ren S, Lin Q, Cheng K, Shen S, Gao M, Li X. Low-Intensity Pulsed Ultrasound Affects Chondrocyte Extracellular Matrix Production via an Integrin-Mediated p38 MAPK Signaling Pathway. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:1690-1700. [PMID: 25736607 DOI: 10.1016/j.ultrasmedbio.2015.01.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/26/2014] [Accepted: 01/17/2015] [Indexed: 06/04/2023]
Abstract
Although low-intensity pulsed ultrasound (LIPUS) regulates p38 mitogen-activated protein kinase (MAPK) and promotes cartilage repair in osteoarthritis, the role of integrin-mediated p38 MAPK in the effect of LIPUS on extracellular matrix (ECM) production of normal and OA chondrocytes remains unknown. The aim of this study was to investigate whether LIPUS affects ECM production in normal and OA rabbit chondrocytes through an integrin-p38 signaling pathway. A rabbit model of OA was established by anterior cruciate ligament transection, and chondrocytes were isolated from normal or OA cartilage and cultured in vitro. Chondrocytes were treated with LIPUS and then pre-incubated with the integrin inhibitor GRGDSP or the p38 inhibitor SB203580. Expression of type II collagen, MMP-13, integrin β1, p38 and phosphorylated p38 was assessed by Western blot analysis. We found that type II collagen and integrin β1 were upregulated (p < 0.05), whereas MMP-13 was downregulated (p < 0.05) in normal and OA chondrocytes. Furthermore, phosphorylated p38 was upregulated (p < 0.05) in normal chondrocytes, but downregulated (p < 0.05) in OA chondrocytes after LIPUS stimulation. Pre-incubation of chondrocytes with the integrin inhibitor disrupted the effects of LIPUS on normal and OA chondrocytes. Pre-incubation of chrondocytes with the p38 inhibitor reduced the effects of LIPUS on normal chondrocytes, but had no impact on OA chondrocytes. Our findings suggest that the integrin-p38 MAPK signaling pathway plays an important role in LIPUS-mediated ECM production in chondrocytes.
Collapse
Affiliation(s)
- Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shasha Ren
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qiang Lin
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kai Cheng
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shihao Shen
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Mingxia Gao
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
77
|
Dyke JP, Synan M, Ezell P, Ballon D, Racine J, Aaron RK. Characterization of bone perfusion by dynamic contrast-enhanced magnetic resonance imaging and positron emission tomography in the Dunkin-Hartley guinea pig model of advanced osteoarthritis. J Orthop Res 2015; 33:366-72. [PMID: 25410523 PMCID: PMC4346481 DOI: 10.1002/jor.22768] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/11/2014] [Indexed: 02/04/2023]
Abstract
This study characterizes changes in subchondral bone circulation in OA and examines relationships to bone structure and cartilage degeneration in Dunkin-Hartley guinea pigs. We have used dynamic contrast-enhanced MRI (DCE-MRI) and PET, with pharmacokinetic modeling, to characterize subchondral bone perfusion. Assessments are made of perfusion kinetics and vascular permeability by MRI, and blood volume and flow, and radionuclide incorporation into bone, by PET. These parameters are compared to cartilage lesion severity and bone histomorphometry. Assessments of intraosseous thrombi are made morphologically. Prolonged signal enhancement during the clearance phase of MRI correlated with OA severity and suggested venous stasis. Vascular permeability was not increased indicating that transvascular migration of contrast agent was not responsible for signal enhancement. Intraosseous thrombi were not observed. Decreased perfusion associated with severe OA was confirmed by PET and was associated with reduced radionuclide incorporation and osteoporosis. MRI and PET can be used to characterize kinetic parameters of circulation in OA and correlate them with subchondral bone metabolism of interest to the pathophysiology of OA. The significance of these observations may lie in alterations induced in the expression of cytokines by OA osteoblasts that are related to bone remodeling and cartilage breakdown.
Collapse
Affiliation(s)
- Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medical College, New York, NY,Citigroup Biomedical Imaging Center, Weill Cornell Medical College, New York, NY
| | - Michael Synan
- Citigroup Biomedical Imaging Center, Weill Cornell Medical College, New York, NY
| | - Paula Ezell
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC
| | - Douglas Ballon
- Department of Radiology, Weill Cornell Medical College, New York, NY,Citigroup Biomedical Imaging Center, Weill Cornell Medical College, New York, NY
| | - Jennifer Racine
- Department of Orthopedics, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Roy K. Aaron
- Department of Orthopedics, The Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
78
|
Lv Y, Xia JY, Chen JY, Zhao H, Yan HC, Yang HS, Li Q, Fan YX, Guo KJ, Chen XY. Effects of pamidronate disodium on the loss of osteoarthritic subchondral bone and the expression of cartilaginous and subchondral osteoprotegerin and RANKL in rabbits. BMC Musculoskelet Disord 2014; 15:370. [PMID: 25377946 PMCID: PMC4240862 DOI: 10.1186/1471-2474-15-370] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/20/2014] [Indexed: 12/03/2022] Open
Abstract
Background Osteoarthritis (OA) is a major health problem in the increasingly elderly population. Therefore, it is crucial to prevent and treat OA at an early stage. The present study investigated whether pamidronate disodium (PAM), a bone-loss inhibitor, can significantly prevent or reverse the progression of early anterior cruciate ligament transection (ACLT)-induced OA. Whether therapeutic intervention is associated with regulation of the expression of osteoprotegerin (OPG), receptor activator of nuclear factor-κB ligand (RANKL), metalloproteinase-9 (MMP-9) or Toll-like receptor-4 (TLR-4) in cartilage and/or subchondral bone was also investigated. Methods 60 New Zealand rabbits were randomized into four groups: Sham-operated (n = 20); ACLT (n = 20); short-term treatment with PAM (PAM-S, n = 10) and long-term treatment with PAM (PAM-L, n = 10). For cartilage and subchondral bone testing, rabbits from Sham and ACLT groups were harvested at 2, 4, 6, and 14 weeks. Rabbits were given PAM from the 4th week after ACLT operation in PAM-S and PAM-L group, and were harvested at 6 and 14 weeks, respectively. Trabecular characteristics and cartilage changes were detected using Micro-CT, safranin O and rapid green staining, respectively. Immunohistochemical staining for OPG and RANKL were also performed. OPG, RANKL, MMP-9 and TLR-4 expression was evaluated by western blot analysis. Results Micro-CT and histology analyses indicated that PAM treatment for 2 or 10 weeks could completely prevent or reverse osteoarthritic subchondral bone loss and cartilage surface erosion. Immunohistochemistry and western blot analysis indicated that expression of OPG and RANKL increased, although RANKL expression increased more significantly than that of OPG. Therefore the ratio of OPG to RANKL was lower in the ACLT group. However, the ratio of OPG to RANKL in the PAM group was significantly higher than that in the ACLT group. Additionally, expression of MMP-9 and TLR-4 were upregulated in the ACLT group and downregulated in the PAM treated groups. Conclusions PAM can significantly inhibit and even reverse early osteoarthritic subchondral bone loss, thus alleviating the process of cartilaginous degeneration. The mechanisms involved may be associated with the upregulation of OPG expression, and downregulation of RANKL, MMP-9 and TLR-4 expression. Electronic supplementary material The online version of this article (doi:10.1186/1471-2474-15-370) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kai-jin Guo
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou 221002, Jiangsu, China.
| | | |
Collapse
|
79
|
Li G, Yin J, Gao J, Cheng TS, Pavlos NJ, Zhang C, Zheng MH. Subchondral bone in osteoarthritis: insight into risk factors and microstructural changes. Arthritis Res Ther 2014; 15:223. [PMID: 24321104 PMCID: PMC4061721 DOI: 10.1186/ar4405] [Citation(s) in RCA: 535] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 12/02/2013] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is a major cause of disability in the adult population. As a
progressive degenerative joint disorder, OA is characterized by cartilage damage,
changes in the subchondral bone, osteophyte formation, muscle weakness, and
inflammation of the synovium tissue and tendon. Although OA has long been viewed as a
primary disorder of articular cartilage, subchondral bone is attracting increasing
attention. It is commonly reported to play a vital role in the pathogenesis of OA.
Subchondral bone sclerosis, together with progressive cartilage degradation, is
widely considered as a hallmark of OA. Despite the increase in bone volume fraction,
subchondral bone is hypomineralized, due to abnormal bone remodeling. Some
histopathological changes in the subchondral bone have also been detected, including
microdamage, bone marrow edema-like lesions and bone cysts. This review summarizes
basic features of the osteochondral junction, which comprises subchondral bone and
articular cartilage. Importantly, we discuss risk factors influencing subchondral
bone integrity. We also focus on the microarchitectural and histopathological changes
of subchondral bone in OA, and provide an overview of their potential contribution to
the progression of OA. A hypothetical model for the pathogenesis of OA is
proposed.
Collapse
|
80
|
Genome-wide expression profiles of subchondral bone in osteoarthritis. Arthritis Res Ther 2014; 15:R190. [PMID: 24229462 PMCID: PMC3979015 DOI: 10.1186/ar4380] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 11/01/2013] [Indexed: 12/31/2022] Open
Abstract
Introduction The aim of this study was to evaluate, for the first time, the differences in gene expression profiles of normal and osteoarthritic (OA) subchondral bone in human subjects. Methods Following histological assessment of the integrity of overlying cartilage and the severity of bone abnormality by micro-computed tomography, we isolated total RNA from regions of interest from human OA (n = 20) and non-OA (n = 5) knee lateral tibial (LT) and medial tibial (MT) plateaus. A whole-genome profiling study was performed on an Agilent microarray platform and analyzed using Agilent GeneSpring GX11.5. Confirmatory quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis was performed on samples from 9 OA individuals to confirm differential expression of 85 genes identified by microarray. Ingenuity Pathway Analysis (IPA) was used to investigate canonical pathways and immunohistochemical staining was performed to validate protein expression levels in samples. Results A total of 972 differentially expressed genes were identified (fold change ≥ ± 2, P ≤0.05) between LT (minimal degeneration) and MT (significant degeneration) regions from OA samples; these data implicated 279 canonical pathways in IPA. The qRT-PCR data strongly confirmed the accuracy of microarray results (R2 = 0.58, P <0.0001). Novel pathways were identified in this study including Periostin (POSTN) and Leptin (LEP), which are implicated in bone remodeling by osteoblasts. Conclusions To the best of our knowledge, this study represents the most comprehensive direct assessment to date of gene expression profiling in OA subchondral bone. This study provides insights that could contribute to the development of new biomarkers and therapeutic strategies for OA.
Collapse
|
81
|
Huebner JL, Bay-Jensen AC, Huffman KM, He Y, Leeming DJ, McDaniel GE, Karsdal MA, Kraus VB. Alpha C-telopeptide of type I collagen is associated with subchondral bone turnover and predicts progression of joint space narrowing and osteophytes in osteoarthritis. Arthritis Rheumatol 2014; 66:2440-9. [PMID: 24909851 DOI: 10.1002/art.38739] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 06/03/2014] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To evaluate joint tissue remodeling using the urinary collagen biomarkers urinary α-C-telopeptide of type I collagen (α-CTX) and urinary C-telopeptide of type II collagen (CTX-II) and to determine the association of these biomarkers with osteoarthritis (OA) severity, progression, and localized knee bone turnover. METHODS Participants (n = 149) with symptomatic and radiographic knee OA underwent fixed-flexion knee radiography at baseline and 3 years, and late-phase bone scintigraphy of both knees at baseline, which were scored semiquantitatively for osteophyte and joint space narrowing (JSN) severity and uptake intensity, with scores summed across knees. Urinary concentrations of α-CTX and CTX-II were determined by enzyme-linked immunosorbent assay. Immunohistochemical analysis of human OA knees was performed to localize the joint tissue origin of the biomarker epitopes. RESULTS Urinary α-CTX concentrations correlated strongly with the intensity of bone scintigraphic uptake and with JSN progression (risk ratio 13.2) and osteophyte progression (risk ratio 3). Urinary CTX-II concentrations were strongly associated with intensity of bone scintigraphic uptake, with JSN and osteophyte severity, and with OA progression based on osteophyte score. Urinary α-CTX localized primarily to high bone turnover areas in subchondral bone. CTX-II localized to the bone-cartilage interface, the tidemark, and damaged articular cartilage. CONCLUSION Baseline urinary α-CTX, which was localized to high turnover areas of subchondral bone, was associated with dynamic bone turnover of knees, as signified by scintigraphy, and progression of both osteophytes and JSN. Urinary CTX-II correlated with JSN and osteophyte severity and progression of osteophytes. To our knowledge, this represents the first report of serologic markers reflecting subchondral bone turnover. These collagen markers may be useful for noninvasive detection and quantification of active subchondral bone turnover and joint remodeling in knee OA.
Collapse
|
82
|
Leyh M, Seitz A, Dürselen L, Schaumburger J, Ignatius A, Grifka J, Grässel S. Subchondral bone influences chondrogenic differentiation and collagen production of human bone marrow-derived mesenchymal stem cells and articular chondrocytes. Arthritis Res Ther 2014; 16:453. [PMID: 25296561 PMCID: PMC4209060 DOI: 10.1186/s13075-014-0453-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 09/05/2014] [Indexed: 11/30/2022] Open
Abstract
Introduction Osteoarthritis (OA) is characterized by an imbalance in cartilage and underlying subchondral bone homeostasis. We hypothesized that signals from the subchondral bone may modulate production of matrix components, alter chondrogenic differentiation potential of cocultured bone marrow-derived mesenchymal stem cells (BMSC) and induce a phenotypic shift in differentiated OA chondrocytes. Methods We established a novel coculture model between BMSC, mixed cultures (BMSC and chondrocytes) and chondrocytes embedded in fibrin gel with OA and normal subchondral bone explants (OAB and NB). Tissues and cells were either derived from OA or trauma patients. In addition, we used adipose-derived stem cells (ASC) from liposuction. With gene expression analysis, biochemical assays, immunofluorescence and biomechanical tests we characterized the properties of newly generated extracellular matrix (ECM) from chondrocytes and chondrogenically differentiating BMSC cocultured with OAB or NB in comparison with monocultures (cultures without bone explants). Results Overall, gene expression of collagens of OAB and NB cocultured cells was reduced compared to monocultures. Concomitantly, we observed significantly lower collagen I, II and III and glycosaminoglycan (GAG) production in OAB cocultured cell lysates. In parallel, we detected increased concentrations of soluble GAGs and basic fibroblast growth factor (bFGF), interleukin (IL)-6 and IL-8 in supernatants of OAB and NB cocultures mainly at early time points. IL-1ß concentration was increased in supernatants of OAB cocultures, but not in NB cocultures. Cell-free NB or OAB explants released different amounts of IL-1ß, bFGF and soluble GAG into cell culture supernatants. In comparison to cocultures, monocultures exhibited higher Young’s modulus and equilibrium modulus. Stimulation of monocultures with IL-1ß led to a downregulation of aggrecan (ACAN) gene expression and in general to induced matrix metalloprotease (MMP)2, MMP3 and MMP-13 gene expression while IL-6 and IL-8 stimulation partly reduced ACAN, MMP3 and MMP-13 gene expression. Conclusions Our results suggest an alteration of molecular composition and mechanical properties of the newly formed ECM in subchondral bone cocultures. We suggest that soluble factors, that is interleukins and bFGF, released in cocultures exert inhibitory effects on collagen and temporary effects on proteoglycan production, which finally results in a reduction of mechanical strength of newly formed fibrillar networks. Electronic supplementary material The online version of this article (doi:10.1186/s13075-014-0453-9) contains supplementary material, which is available to authorized users.
Collapse
|
83
|
Lorenz J, Seebach E, Hackmayer G, Greth C, Bauer RJ, Kleinschmidt K, Bettenworth D, Böhm M, Grifka J, Grässel S. Melanocortin 1 receptor-signaling deficiency results in an articular cartilage phenotype and accelerates pathogenesis of surgically induced murine osteoarthritis. PLoS One 2014; 9:e105858. [PMID: 25191747 PMCID: PMC4156302 DOI: 10.1371/journal.pone.0105858] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 07/25/2014] [Indexed: 11/18/2022] Open
Abstract
Proopiomelanocortin-derived peptides exert pleiotropic effects via binding to melanocortin receptors (MCR). MCR-subtypes have been detected in cartilage and bone and mediate an increasing number of effects in diathrodial joints. This study aims to determine the role of MC1-receptors (MC1) in joint physiology and pathogenesis of osteoarthritis (OA) using MC1-signaling deficient mice (Mc1re/e). OA was surgically induced in Mc1re/e and wild-type (WT) mice by transection of the medial meniscotibial ligament. Histomorphometry of Safranin O stained articular cartilage was performed with non-operated controls (11 weeks and 6 months) and 4/8 weeks past surgery. µCT-analysis for assessing epiphyseal bone architecture was performed as a longitudinal study at 4/8 weeks after OA-induction. Collagen II, ICAM-1 and MC1 expression was analysed by immunohistochemistry. Mc1re/e mice display less Safranin O and collagen II stained articular cartilage area compared to WT prior to OA-induction without signs of spontaneous cartilage surface erosion. This MC1-signaling deficiency related cartilage phenotype persisted in 6 month animals. At 4/8 weeks after OA-induction cartilage erosions were increased in Mc1re/e knees paralleled by weaker collagen II staining. Prior to OA-induction, Mc1re/e mice do not differ from WT with respect to bone parameters. During OA, Mc1re/e mice developed more osteophytes and had higher epiphyseal bone density and mass. Trabecular thickness was increased while concomitantly trabecular separation was decreased in Mc1re/e mice. Numbers of ICAM-positive chondrocytes were equal in non-operated 11 weeks Mc1re/e and WT whereas number of positive chondrocytes decreased during OA-progression. Unchallenged Mc1re/e mice display smaller articular cartilage covered area without OA-related surface erosions indicating that MC1-signaling is critical for proper cartilage matrix integrity and formation. When challenged with OA, Mc1re/e mice develop a more severe OA-pathology. Our data suggest that MC1-signaling protects against cartilage degradation and subchondral bone sclerosis in OA indicating a beneficial role of the POMC system in joint pathophysiology.
Collapse
Affiliation(s)
- Julia Lorenz
- Experimental Orthopedics, University Hospital of Regensburg, Regensburg, Bavaria, Germany
- Orthopedic Surgery, University Hospital of Regensburg, Bad Abbach, Bavaria, Germany
| | - Elisabeth Seebach
- Research Centre for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Baden-Württemberg, Germany
| | - Gerit Hackmayer
- Experimental Orthopedics, University Hospital of Regensburg, Regensburg, Bavaria, Germany
- Dermatology, University Hospital of Münster, Münster, North Rhine-Westphalia, Germany
| | - Carina Greth
- Research Centre for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Baden-Württemberg, Germany
| | - Richard J. Bauer
- Oral and Maxillofacial Surgery, University Hospital of Regensburg, Regensburg, Bavaria, Germany
| | - Kerstin Kleinschmidt
- TIP Immunology, Merck Serono Global Research & Development, Darmstadt, Hessen, Germany
| | - Dominik Bettenworth
- Medical Hospital B, University Hospital of Münster, Münster, North Rhine-Westphalia, Germany
| | - Markus Böhm
- Dermatology, University Hospital of Münster, Münster, North Rhine-Westphalia, Germany
| | - Joachim Grifka
- Orthopedic Surgery, University Hospital of Regensburg, Bad Abbach, Bavaria, Germany
| | - Susanne Grässel
- Experimental Orthopedics, University Hospital of Regensburg, Regensburg, Bavaria, Germany
- Orthopedic Surgery, University Hospital of Regensburg, Bad Abbach, Bavaria, Germany
| |
Collapse
|
84
|
Soto SA, Chiappe Barbará A. Bisphosphonates: Pharmacology and Clinical Approach to Their Use in Equine Osteoarticular Diseases. J Equine Vet Sci 2014. [DOI: 10.1016/j.jevs.2014.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
85
|
Karsdal MA, Bay-Jensen AC, Lories RJ, Abramson S, Spector T, Pastoureau P, Christiansen C, Attur M, Henriksen K, Goldring SR, Kraus V. The coupling of bone and cartilage turnover in osteoarthritis: opportunities for bone antiresorptives and anabolics as potential treatments? Ann Rheum Dis 2014; 73:336-48. [PMID: 24285494 DOI: 10.1136/annrheumdis-2013-204111] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Osteoarthritis (OA) is the most common form of arthritic disease, and a major cause of disability and impaired quality of life in the elderly. OA is a complex disease of the entire joint, affecting bone, cartilage and synovium that thereby presents multiple targets for treatment. This manuscript will summarise emerging observations from cell biology, preclinical and preliminary clinical trials that elucidate interactions between the bone and cartilage components in particular. Bone and cartilage health are tightly associated. Ample evidence has been found for bone changes during progression of OA including, but not limited to, increased turnover in the subchondral bone, undermineralisation of the trabecular structure, osteophyte formation, bone marrow lesions and sclerosis of the subchondral plate. Meanwhile, a range of investigations has shown positive effects on cartilage health when bone resorption is suppressed, or deterioration of the cartilage when resorption is increased. Known bone therapies, namely oestrogens, selective oestrogen receptor modifiers (SERMs), bisphosphonates, strontium ranelate, calcitonin and parathyroid hormone, might prove useful for treating two critical tissue components of the OA joint, the bone and the cartilage. An optimal treatment for OA likely targets at least these two tissue components. The patient subgroups for whom these therapies are most appropriate have yet to be fully defined but would likely include, at a minimum, those with high bone turnover.
Collapse
|
86
|
Karsdal MA, Christiansen C, Ladel C, Henriksen K, Kraus VB, Bay-Jensen AC. Osteoarthritis--a case for personalized health care? Osteoarthritis Cartilage 2014; 22:7-16. [PMID: 24216058 DOI: 10.1016/j.joca.2013.10.018] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/25/2013] [Accepted: 10/29/2013] [Indexed: 02/02/2023]
Abstract
For both economic and ethical reasons, identification of the optimal treatment for each individual patient is a pressing concern, not only for the patients and their physician, but also health care payers and the pharmaceutical industry. In the field of osteoarthritis (OA) this is of particular relevance, due to the heterogeneity of the disease and the very large number of affected individuals. There is a need to pair the right patients with the right therapeutic modes of action. At present, the clinical trial failures in OA may be a consequence of both bona fide treatment failures and trial failures due to clinical design deficiencies. Tools are needed for characterization and segregation of patients with OA. Key lessons may be learned from advances with another form of arthritis, namely rheumatoid arthritis (RA). Personalized health care (PHC) may be more advantageous for a number of specific indications which are characterized by costly therapy, low response rates and significant problems associated with trial and error prescription, including the risk of serious side effects. We discuss the use of diagnostic practices guiding RA treatment, which may serve as a source of key insights for diagnostic practices in OA. We discuss the emerging concept of PHC, and outline the opportunities and current successes and failures across the RA field, as the OA field collects further data to support the hypothesis. We attempt to outline a possible path forward to assist patients, physicians, payers and the pharmaceutical industry in assuring the 'right' patients are treated with the 'right drug' in OA. Finally we highlight methods for possible segregation of OA patients that would allow identification of patient subtypes, such as OA driven by inflammation that may be ideally suited for PHC and for targeted therapies.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience, Herlev Hovedgade, DK-2730 Herlev, Denmark.
| | - C Christiansen
- Nordic Bioscience, Herlev Hovedgade, DK-2730 Herlev, Denmark
| | - C Ladel
- Merck-Serono Research, Merck KGaA, Darmstadt, Germany
| | - K Henriksen
- Nordic Bioscience, Herlev Hovedgade, DK-2730 Herlev, Denmark
| | - V B Kraus
- Division of Rheumatology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - A C Bay-Jensen
- Nordic Bioscience, Herlev Hovedgade, DK-2730 Herlev, Denmark
| |
Collapse
|
87
|
Mistry JB, Bukhari M, Taylor AM. Alkaptonuria. Rare Dis 2013; 1:e27475. [PMID: 25003018 DOI: 10.4161/rdis.27475] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 11/26/2013] [Accepted: 12/09/2013] [Indexed: 12/15/2022] Open
Abstract
Alkaptonuria (AKU) is a rare disorder of autosomal recessive inheritance. It is caused by a mutation in a gene that results in the accumulation of homogentisic acid (HGA). Characteristically, the excess HGA means sufferers pass dark urine, which upon standing turns black. This is a feature present from birth. Over time patients develop other manifestations of AKU, due to deposition of HGA in collagenous tissues, namely ochronosis and ochronotic osteoarthropathy. Although this condition does not reduce life expectancy, it significantly affects quality of life. The natural history of this condition is becoming better understood, despite gaps in knowledge. Clinical assessment of the condition has also improved along with the development of a potentially disease-modifying therapy. Furthermore, recent developments in AKU research have led to new understanding of the disease, and further study of the AKU arthropathy has the potential to influence therapy in the management of osteoarthritis.
Collapse
Affiliation(s)
- Jemma B Mistry
- Lancaster Medical School; Faculty of Health & Medicine; Lancaster, UK
| | - Marwan Bukhari
- University Hospitals of Morecambe Bay NHS Foundation Trust; Royal Lancaster Infirmary; Lancaster, UK
| | - Adam M Taylor
- Lancaster Medical School; Faculty of Health & Medicine; Lancaster, UK
| |
Collapse
|
88
|
Reginster JY, Beaudart C, Neuprez A, Bruyère O. Strontium ranelate in the treatment of knee osteoarthritis: new insights and emerging clinical evidence. Ther Adv Musculoskelet Dis 2013; 5:268-76. [PMID: 24101948 DOI: 10.1177/1759720x13500862] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis is a primary cause of disability and functional incapacity. Pharmacological treatment is currently limited to symptomatic management, and in advanced stages, surgery remains the only solution. The therapeutic armamentarium for osteoarthritis remains poor in treatments with an effect on joint structure, that is, disease-modifying osteoarthritis drugs (DMOADs). Glucosamine sulfate and chondroitin sulfate are the only medications for which some conclusive evidence for a disease-modifying effect is available. Strontium ranelate is currently indicated for the prevention of fracture in severe osteoporosis. Its efficacy and safety as a DMOAD in knee osteoarthritis has recently been explored in the SEKOIA trial, a 3-year randomized, double-blind, placebo-controlled trial. Outpatients with knee osteoarthritis, Kellgren and Lawrence grade 2 or 3, and joint space width (JSW) of 2.5-5 mm received strontium ranelate 1 g/day (n = 558) or 2 g/day (n = 566), or placebo (n = 559). This sizable population was aged 62.9 years and had a JSW of 3.50 ± 0.84 mm. Treatment with strontium ranelate led to significantly less progression of knee osteoarthritis: estimates for annual difference in joint space narrowing versus placebo were 0.14 mm [95% confidence interval (CI) 0.05-0.23 mm; p < 0.001] for 1 g/day and 0.10 mm (95% CI 0.02-0.19 mm; p = 0.018) for 2 g/day, with no difference between strontium ranelate groups. Radiological progression was less frequent with strontium ranelate (22% with 1 g/day and 26% with 2 g/day versus 33% with placebo, both p < 0.05), as was radioclinical progression (8% and 7% versus 12%, both p < 0.05). Symptoms also improved with strontium ranelate 2 g/day only in terms of total WOMAC (Western Ontario and McMaster Universities Osteoarthritis Index) score (p = 0.045), and its components for pain (p = 0.028) and physical function (p = 0.099). Responder analyses using a range of criteria for symptoms indicated that the effect of strontium ranelate 2 g/day on pain and physical function was clinically meaningful. Strontium ranelate was well tolerated. The observation of both structure and symptom modification with strontium ranelate 2 g/day makes SEKOIA a milestone in osteoarthritis research and treatment.
Collapse
Affiliation(s)
- Jean-Yves Reginster
- Department of Public Health and Health Economics, University of Liege, 4020 Liege, Belgium
| | | | | | | |
Collapse
|
89
|
Interplay between cartilage and subchondral bone contributing to pathogenesis of osteoarthritis. Int J Mol Sci 2013; 14:19805-30. [PMID: 24084727 PMCID: PMC3821588 DOI: 10.3390/ijms141019805] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/17/2013] [Accepted: 09/23/2013] [Indexed: 12/23/2022] Open
Abstract
Osteoarthritis (OA) is a common debilitating joint disorder, affecting large sections of the population with significant disability and impaired quality of life. During OA, functional units of joints comprising cartilage and subchondral bone undergo uncontrolled catabolic and anabolic remodeling processes to adapt to local biochemical and biological signals. Changes in cartilage and subchondral bone are not merely secondary manifestations of OA but are active components of the disease, contributing to its severity. Increased vascularization and formation of microcracks in joints during OA have suggested the facilitation of molecules from cartilage to bone and vice versa. Observations from recent studies support the view that both cartilage and subchondral bone can communicate with each other through regulation of signaling pathways for joint homeostasis under pathological conditions. In this review we have tried to summarize the current knowledge on the major signaling pathways that could control the cartilage-bone biochemical unit in joints and participate in intercellular communication between cartilage and subchondral bone during the process of OA. An understanding of molecular communication that regulates the functional behavior of chondrocytes and osteoblasts in both physiological and pathological conditions may lead to development of more effective strategies for treating OA patients.
Collapse
|
90
|
Zhu S, Chen K, Lan Y, Zhang N, Jiang R, Hu J. Alendronate protects against articular cartilage erosion by inhibiting subchondral bone loss in ovariectomized rats. Bone 2013; 53:340-9. [PMID: 23291609 DOI: 10.1016/j.bone.2012.12.044] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 11/21/2012] [Accepted: 12/21/2012] [Indexed: 02/05/2023]
Abstract
Osteoporosis (OP) and osteoarthritis (OA) are major health problems in the increasing elderly population, particularly in postmenopausal women, but their relationship remains unclear. The present study investigated whether alendronate (ALN), a potent inhibitor of bone resorption, could protect articular cartilage from degeneration in a combined animal model of OP and OA induced by ovariectomy (OVX). Seventy-eight seven-month-old female Sprague-Dawley rats were assigned into five experimental groups: (1) sham-operated with vehicle treatment, (2) sham-operated with ALN treatment, (3) OVX with vehicle treatment, (4) ALN treatment starting at OVX, and (5) ALN treatment starting at eight weeks after OVX. Histological and micro-CT analyses, together with urine collagen degradation markers, indicated that early ALN treatment completely prevented both subchondral bone loss and cartilage surface erosion induced by OVX. Although late ALN treatment also inhibited subchondral bone loss and significantly reduced cartilage erosion in the OVX rats, these tissues did not completely recover even after 10-weeks of ALN treatment. Quantitative RT-PCR analyses showed that the protective effect of ALN correlated with increased ratio of OPG/RANKL in both subchondral bone and cartilage. Moreover, whereas OVX caused upregulation of expression of matrix metalloproteinases MMP-13 and MMP-9 in the articular cartilage and chondrocytes in the interface between the articular cartilage and subchondral bone, respectively, early ALN treatment blocked whereas late ALN treatment attenuated the upregulation of these catabolic enzymes in the corresponding tissues. Together, these data indicate that the subchondral bone loss plays an important role in OA pathogenesis in the combined OP and OA model and suggest that treatment timing is an important factor for the effectiveness of anti-resorptive drug therapy of combined OP and OA.
Collapse
Affiliation(s)
- Songsong Zhu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | |
Collapse
|
91
|
Thomsen JS, Straarup TS, Danielsen CC, Oxlund H, Brüel A. No effect of risedronate on articular cartilage damage in the Dunkin Hartley guinea pig model of osteoarthritis. Scand J Rheumatol 2013; 42:408-16. [PMID: 23527881 DOI: 10.3109/03009742.2013.774046] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES To investigate whether treatment with a bisphosphonate would influence the subchondral bone plate stiffness and the development of cartilage damage in Dunkin Hartley guinea pigs, which develop osteoarthritis (OA) spontaneously. METHOD Fifty-six 3-month-old male Dunkin Hartley guinea pigs were randomized into a baseline group and six groups receiving either the bisphosphonate risedronate (30 µg/kg) or vehicle five times a week for 6, 12, or 24 weeks. The medial condyle of the right stifle joint was investigated by histology, using the Osteoarthritis Research Society International (OARSI) score, along with static and dynamic histomorphometry. The subchondral bone plate of the left tibia was tested mechanically with indentation testing. Degradation products of C-terminal telopeptides of type II collagen (CTX-II) were measured in serum. RESULTS The OARSI score did not differ between risedronate-treated and control animals at any time point. The fraction of bone surfaces covered with osteoclasts (Oc.S/BS) was significantly suppressed in risedronate-treated animals at all time points, as were the fractions of mineralizing surfaces (MS/BS) and osteoid-covered surfaces (OS/BS), and also serum CTX-II. This was accompanied by a significant increase in the epiphyseal content of calcified tissue and in the thickness of the subchondral bone plate. However, this did not result in a stiffer subchondral bone at any time point. DISCUSSION The risedronate treatment inhibited osteoclastic resorption of calcified cartilage in the primary spongiosa under the epiphyseal growth plate, explaining the risedronate-mediated decrease in CTX-II. Moreover, the serum CTX-II level was not related to the OA-induced articular cartilage degradation seen in this model. CONCLUSIONS Risedronate did not influence the OARSI score and subchondral plate stiffness, but decreased serum CTX-II in Dunkin Hartley guinea pigs.
Collapse
Affiliation(s)
- J S Thomsen
- Department of Biomedicine - Anatomy, Aarhus University , Aarhus , Denmark
| | | | | | | | | |
Collapse
|
92
|
Karsdal MA, Byrjalsen I, Henriksen K, Riis BJ, Christiansen C. A Pharmacokinetic and Pharmacodynamic Comparison of Synthetic and Recombinant Oral Salmon Calcitonin. J Clin Pharmacol 2013; 49:229-34. [DOI: 10.1177/0091270008329552] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
93
|
Yu DG, Ding HF, Mao YQ, Liu M, Yu B, Zhao X, Wang XQ, Li Y, Liu GW, Nie SB, Liu S, Zhu ZA. Strontium ranelate reduces cartilage degeneration and subchondral bone remodeling in rat osteoarthritis model. Acta Pharmacol Sin 2013; 34:393-402. [PMID: 23334238 DOI: 10.1038/aps.2012.167] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIM To investigate whether strontium ranelate (SR), a new antiosteoporotic agent, could attenuate cartilage degeneration and subchondral bone remodeling in osteoarthritis (OA). METHODS Medial meniscal tear (MMT) operation was performed in adult SD rats to induce OA. SR (625 or 1800 mg·kg(-1)·d(-1)) was administered via gavage for 3 or 6 weeks. After the animals were sacrificed, articular cartilage degeneration was evaluated using toluidine blue O staining, SOX9 immunohistochemistry and TUNEL assay. The changes in microarchitecture indices and tissue mineral density (TMD), chemical composition (mineral-to-collagen ratio), and intrinsic mechanical properties of the subchondral bones were measured using micro-CT scanning, confocal Raman microspectroscopy and nanoindentation testing, respectively. RESULTS The high-dose SR significantly attenuated cartilage matrix and chondrocyte loss at 6 weeks, and decreased chondrocyte apoptosis, improved the expression of SOX9, a critical transcription factor responsible for the expression of anabolic genes type II collagen and aggrecan, at both 3 and 6 weeks. Meanwhile, the high-dose SR also significantly attenuated the subchondral bone remodeling at both 3 and 6 weeks, as shown by the improved microarchitecture indices, TMD, mineral-to-collagen ratio and intrinsic mechanical properties. In contrast, the low-dose SR did not significantly change all the detection indices of cartilage and bone at both 3 and 6 weeks. CONCLUSION The high-dose SR treatment can reduce articular cartilage degeneration and subchondral bone remodeling in the rat MMT model of OA.
Collapse
|
94
|
Lafeber FPJG, van Laar JM. Strontium ranelate: ready for clinical use as disease-modifying osteoarthritis drug? Ann Rheum Dis 2013; 72:157-61. [PMID: 23292449 DOI: 10.1136/annrheumdis-2012-202453] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
95
|
Armagan O, Serin DK, Calisir C, Dokumacioglu A, Ozgen M, Oner S, Alatas O. Inhalation therapy of calcitonin relieves osteoarthritis of the knee. J Korean Med Sci 2012; 27:1405-10. [PMID: 23166425 PMCID: PMC3492678 DOI: 10.3346/jkms.2012.27.11.1405] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 08/27/2012] [Indexed: 01/15/2023] Open
Abstract
This study was conducted to determine if nasal salmon calcitonin has additional beneficial effects on clinical symptoms, serum NO, IL-1β, matrix metalloproteinase 3, urinary C-terminal telopeptide type II collagen (CTX-II) levels and MRI findings in knee osteoarthritis (OA) when used concomitantly with exercise therapy. Fifty female patients with knee OA were randomized into two groups. The first group (n = 30) received 200 IU/day nasal salmon calcitonin and a home exercise program; the second group (n = 20) received a home exercise program for 6 months. Compared with baseline,while significant improvements were observed in visual analogue scale (VAS), WOMAC pain, physical function scores, 20-m walking time (P < 0.001) and WOMAC stiffness score (P = 0.041) in the first group, walking and resting VAS, and WOMAC physical function scores were improved (P = 0.029) in the second group after treatment. Significantly increased levels of serum NO and urinary CTX-II (P < 0.001) and significant improvements in the area of medial femoral condyle (P < 0.05) were noted only in the first group. There were significant differences in VAS activation values (P = 0.032) and NO levels (P < 0.001) in the favor of the first group. In conclusion, nasal salmon calcitonin may have possible chondroprotective effects besides its known effects on symptoms in patients with knee OA.
Collapse
Affiliation(s)
- Onur Armagan
- Department of Physical Therapy and Rehabilitation, Eskisehir Osmangazi University Faculty of Medicine, Eskisehir, Turkey.
| | | | | | | | | | | | | |
Collapse
|
96
|
Karsdal MA, Nielsen MJ, Sand JM, Henriksen K, Genovese F, Bay-Jensen AC, Smith V, Adamkewicz JI, Christiansen C, Leeming DJ. Extracellular matrix remodeling: the common denominator in connective tissue diseases. Possibilities for evaluation and current understanding of the matrix as more than a passive architecture, but a key player in tissue failure. Assay Drug Dev Technol 2012; 11:70-92. [PMID: 23046407 DOI: 10.1089/adt.2012.474] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Increased attention is paid to the structural components of tissues. These components are mostly collagens and various proteoglycans. Emerging evidence suggests that altered components and noncoded modifications of the matrix may be both initiators and drivers of disease, exemplified by excessive tissue remodeling leading to tissue stiffness, as well as by changes in the signaling potential of both intact matrix and fragments thereof. Although tissue structure until recently was viewed as a simple architecture anchoring cells and proteins, this complex grid may contain essential information enabling the maintenance of the structure and normal functioning of tissue. The aims of this review are to (1) discuss the structural components of the matrix and the relevance of their mutations to the pathology of diseases such as fibrosis and cancer, (2) introduce the possibility that post-translational modifications (PTMs), such as protease cleavage, citrullination, cross-linking, nitrosylation, glycosylation, and isomerization, generated during pathology, may be unique, disease-specific biochemical markers, (3) list and review the range of simple enzyme-linked immunosorbent assays (ELISAs) that have been developed for assessing the extracellular matrix (ECM) and detecting abnormal ECM remodeling, and (4) discuss whether some PTMs are the cause or consequence of disease. New evidence clearly suggests that the ECM at some point in the pathogenesis becomes a driver of disease. These pathological modified ECM proteins may allow insights into complicated pathologies in which the end stage is excessive tissue remodeling, and provide unique and more pathology-specific biochemical markers.
Collapse
|
97
|
Progression of cartilage degradation, bone resorption and pain in rat temporomandibular joint osteoarthritis induced by injection of iodoacetate. PLoS One 2012; 7:e45036. [PMID: 22984604 PMCID: PMC3439407 DOI: 10.1371/journal.pone.0045036] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 08/11/2012] [Indexed: 12/18/2022] Open
Abstract
Background Osteoarthritis (OA) is an important subtype of temporomandibular disorders. A simple and reproducible animal model that mimics the histopathologic changes, both in the cartilage and subchondral bone, and clinical symptoms of temporomandibular joint osteoarthritis (TMJOA) would help in our understanding of its process and underlying mechanism. Objective To explore whether injection of monosodium iodoacetate (MIA) into the upper compartment of rat TMJ could induce OA-like lesions. Methods Female rats were injected with varied doses of MIA into the upper compartment and observed for up to 12 weeks. Histologic, radiographic, behavioral, and molecular changes in the TMJ were evaluated by light and electron microscopy, MicroCT scanning, head withdrawal threshold test, real-time PCR, immunohistochemistry, and TUNEL assay. Results The intermediate zone of the disc loosened by 1 day post-MIA injection and thinned thereafter. Injection of an MIA dose of 0.5 mg or higher induced typical OA-like lesions in the TMJ within 4 weeks. Condylar destruction presented in a time-dependent manner, including chondrocyte apoptosis in the early stages, subsequent cartilage matrix disorganization and subchondral bone erosion, fibrosis, subchondral bone sclerosis, and osteophyte formation in the late stages. Nociceptive responses increased in the early stages, corresponding to severe synovitis. Furthermore, chondrocyte apoptosis and an imbalance between anabolism and catabolism of cartilage and subchondral bone might account for the condylar destruction. Conclusions Multi-level data demonstrated a reliable and convenient rat model of TMJOA could be induced by MIA injection into the upper compartment. The model might facilitate TMJOA related researches.
Collapse
|
98
|
Abstract
Calcitonin is a hormone secreted by the C-cells of the thyroid gland in response to elevations of the plasma calcium level. It reduces bone resorption by inhibiting mature active osteoclasts and increases renal calcium excretion. It is used in the management of postmenopausal osteoporosis, Paget’s disease of bone, and malignancy-associated hypercalcemia. Synthetic and recombinant calcitonin preparations are available; both have similar pharmacokinetic and pharmacodynamic profiles. As calcitonin is a peptide, the traditional method of administration has been parenteral or intranasal. This hinders its clinical use: adherence with therapy is notoriously low, and withdrawal from clinical trials has been problematic. An oral formulation would be more attractive, practical, and convenient to patients. In addition to its effect on active osteoclasts and renal tubules, calcitonin has an analgesic action, possibly mediated through β-endorphins and the central modulation of pain perception. It also exerts a protective action on cartilage and may be useful in the management of osteoarthritis and possibly rheumatoid arthritis. Oral formulations of calcitonin have been developed using different techniques. The most studied involves drug-delivery carriers such as Eligen® 8-(N-2hydroxy-5-chloro-benzoyl)-amino-caprylic acid (5-CNAC) (Emisphere Technologies, Cedar Knolls, NJ). Several factors affect the bioavailability and efficacy of orally administered calcitonin, including amount of water used to take the tablet, time of day the tablet is taken, and proximity to intake of a meal. Preliminary results looked promising. Unfortunately, in two Phase III studies, oral calcitonin (0.8 mg with 200 mg 5-CNAC, once a day for postmenopausal osteoporosis and twice a day for osteoarthritis) failed to meet key end points, and in December 2011, Novartis Pharma AG announced that it would not pursue further clinical development of oral calcitonin for postmenopausal osteoporosis or osteoarthritis. A unique feature of calcitonin is that it is able to uncouple bone turnover, reducing bone resorption without affecting bone formation and therefore increasing bone mass and improving bone quality. This effect, however, may be dose-dependent, with higher doses inhibiting both resorption and formation. Because so many factors affect the pharmacokinetics and pharmacodynamics of calcitonin, especially orally administered calcitonin, much work remains to be done to explore the full pharmacologic spectrum and potential of calcitonin and determine the optimum dose and timing of administration, as well as water and food intake.
Collapse
Affiliation(s)
- Ronald C Hamdy
- Osteoporosis Center, College of Medicine, East Tennessee State University, Johnson City, TN, USA ; Veterans Affairs Medical Center, Johnson City, TN, USA
| | | |
Collapse
|
99
|
Abstract
The classical view of the pathogenesis of osteoarthritis (OA) is that subchondral sclerosis is associated with, and perhaps causes, age-related joint degeneration. Recent observations have demonstrated that OA is associated with early loss of bone owing to increased bone remodelling, followed by slow turnover leading to densification of the subchondral plate and complete loss of cartilage. Subchondral densification is a late event in OA that involves only the subchondral plate and calcified cartilage; the subchondral cancellous bone beneath the subchondral plate may remain osteopenic. In experimental models, inducing subchondral sclerosis without allowing the prior stage of increased bone remodelling to occur does not lead to progressive OA. Therefore, both early-stage increased remodelling and bone loss, and the late-stage slow remodelling and subchondral densification are important components of the pathogenetic process that leads to OA. The apparent paradoxical observations that OA is associated with both increased remodelling and osteopenia, as well as decreased remodelling and sclerosis, are consistent with the spatial and temporal separation of these processes during joint degeneration. This Review provides an overview of current knowledge on OA and discusses the role of subchondral bone in the initiation and progression of OA. A hypothetical model of OA pathogenesis is proposed.
Collapse
Affiliation(s)
- David B Burr
- Department of Anatomy and Cell Biology, MS 5035, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | |
Collapse
|
100
|
Baker-LePain JC, Lane NE. Role of bone architecture and anatomy in osteoarthritis. Bone 2012; 51:197-203. [PMID: 22401752 PMCID: PMC3372683 DOI: 10.1016/j.bone.2012.01.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 12/17/2011] [Accepted: 01/12/2012] [Indexed: 12/24/2022]
Abstract
When considering the pathogenesis of osteoarthritis (OA), it is important to review the contribution of bone in addition to the contribution of cartilage and synovium. Although bone clearly plays a role in determining the distribution of biomechanical forces across joints, which in turn plays a role in the initiation of OA, it has also more recently been appreciated that bone may contribute in a biological sense to the pathogenesis of OA. Far from being a static structure, bone is a dynamic tissue undergoing constant remodeling, and it is clear from a number of radiographic and biochemical studies that bone and cartilage degradation occurs hand in hand. Whether the initial instigating event in OA occurs in cartilage or bone is not known, but it is clear that bony changes occur very early in the pathogenesis of OA and often predate radiographic appearance of the disease. This review focuses on the structural variants of both hip and knee that have been associated with OA and the ultrastructural bone changes in these sites occurring in early OA pathogenesis. This article is part of a Special Issue entitled "Osteoarthritis".
Collapse
Affiliation(s)
| | - Nancy E. Lane
- Department of Internal Medicine, Center for Healthy Aging, UC Davis Medical Center, Sacramento, CA 95817, Telephone: 916-734-0763, Fax: 916-734-4773
| |
Collapse
|