1
|
Roemer FW, Jansen MP, Maschek S, Mastbergen SC, Marijnissen AK, Wisser A, Heiss R, Weinans HH, Blanco FJ, Berenbaum F, Kloppenburg M, Haugen IK, Eckstein F, Hunter DJ, Guermazi A, Wirth W. Fluctuation of Bone Marrow Lesions and Inflammatory MRI Markers over 2 Years and Concurrent Associations with Quantitative Cartilage Loss. Cartilage 2024:19476035241287694. [PMID: 39460605 DOI: 10.1177/19476035241287694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVE To assess whether change of semiquantitatively magnetic resonance imaging (MRI)-defined bone marrow lesions (BMLs) and inflammatory markers is associated with change in quantitatively-assessed cartilage loss in the femorotibial joint (FTJ) in knees with radiographic osteoarthritis (OA) over 24 months. DESIGN Participants were included from the IMI-APPROACH and the Osteoarthritis Initiative FNIH studies. Semiquantitative MRI assessment was performed for BMLs, Hoffa- and effusion-synovitis. Quantitative cartilage thickness measurements were performed manually. Definitions of change included number of subregions with BMLs, change in sum and change in maximum increase in size. Change in Hoffa-synovitis and effusion-synovitis was categorized in addition. Between-group comparisons regarding cartilage loss in the FTJ, medial and lateral compartments were performed using analysis of variance (ANOVA). RESULTS A total of 629 participants were included. Knees without any BMLs at baseline (BL) and follow-up (FU) had significantly less cartilage loss compared to the other subgroups. Change in both directions in the sum score of BMLs was associated with increased rates of cartilage loss. Maximum increase in size of BMLs was associated with increased rates of cartilage loss (FTJ increase by 2 grades -0.183 mm, 95% CI [-0.335, -0.031], by 3 grades -0.306 mm, [-0.511, -0.101]). Worsening of Hoffa-synovitis was associated with increased rates of cartilage loss. CONCLUSION Knees without BMLs at BL and FU showed lowest rates of cartilage loss. Knees with an increase in BML size showed increased rates of concurrent cartilage loss. Approaches with the aim to inhibit BML development, avoidance of increase in size and avoidance of Hoffa-synovitis worsening may have beneficial effects on cartilage loss.
Collapse
Affiliation(s)
- Frank W Roemer
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
- Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | | | | | - Anna Wisser
- Chondrometrics GmbH, Freilassing, Germany
- Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| | - Rafael Heiss
- Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | - Francis Berenbaum
- AP-HP Saint- Antoine Hospital, Paris, France
- Sorbonne University, Paris, France
| | | | | | - Felix Eckstein
- Chondrometrics GmbH, Freilassing, Germany
- Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| | - David J Hunter
- Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling Institute, University of Sydney, St. Leonards, NSW, Australia
| | - Ali Guermazi
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
- VA Boston Healthcare System, West Roxbury, MA, USA
| | - Wolfgang Wirth
- Chondrometrics GmbH, Freilassing, Germany
- Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| |
Collapse
|
2
|
Zhang Y, Zhou Y. Advances in targeted therapies for age-related osteoarthritis: A comprehensive review of current research. Biomed Pharmacother 2024; 179:117314. [PMID: 39167845 DOI: 10.1016/j.biopha.2024.117314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that disproportionately impacts the elderly population on a global scale. As aging is a significant risk factor for OA, there is a growing urgency to develop specific therapies that target the underlying mechanisms of aging associated with this condition. This summary seeks to offer a thorough introduction of ongoing research efforts aimed at developing therapies to combat senescence in the context of OA. Cellular senescence plays a pivotal role in both the deterioration of cartilage integrity and the perpetuation of chronic inflammation and tissue remodeling. Consequently, targeting SnCs has emerged as a promising therapeutic approach to alleviate symptoms and hinder the progression of OA. This review examines a range of approaches, including senolytic drugs targeting SnCs, senomorphics that modulate the senescence-associated secretory phenotype (SASP), and interventions that enhance immune system clearance of SnCs. Novel methodologies, such as utilizing novel materials for exosome delivery and administering anti-aging medications with precision, offer promising avenues for the precise treatment of OA. Accumulating evidence underscores the potential of targeting senescence in OA management, potentially facilitating the development of more effective and personalized therapeutic interventions.
Collapse
Affiliation(s)
- Yantao Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuan 430060, China
| | - Yan Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuan 430060, China.
| |
Collapse
|
3
|
Freitag J, Chamberlain M, Wickham J, Shah K, Cicuttini F, Wang Y, Solterbeck A. Safety and efficacy of an allogeneic adipose-derived mesenchymal stem cell preparation in the treatment of knee osteoarthritis: A Phase I/IIa randomised controlled trial. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100500. [PMID: 39161739 PMCID: PMC11331931 DOI: 10.1016/j.ocarto.2024.100500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/25/2024] [Indexed: 08/21/2024] Open
Abstract
Objectives To assess the safety and efficacy of an allogeneic adipose-derived mesenchymal stem cell preparation (MAG200) in the treatment of knee osteoarthritis over 12 months. Design A single-centre, double-blind, ascending dose, randomised controlled trial. 40 participants with moderate knee osteoarthritis were randomised to receive a single intra-articular injection of MAG200 (dose cohorts:10, 20, 50, 100 × 106 cells) or placebo. Primary objectives were safety and efficacy according to a compound responder analysis of minimal clinically important difference in pain (numerical pain rating scale [NPRS]) and function (Knee Injury and Osteoarthritis Outcome Score - Function in Daily Living subscale [KOOSADL]) at month 12. Secondary efficacy outcomes included changes from baseline in patient reported outcome measures and evaluation of disease-modification using quantitative MRI. Results Treatment was well tolerated with no treatment-related serious adverse events. MAG200 cohorts reported a greater proportion of responders than placebo and demonstrated clinical and statistically significant improvement in pain and clinically relevant improvement in all KOOS subscales. MAG200 demonstrated a reproducible treatment effect over placebo, which was clinically relevant for pain in the 10 × 106 dose cohort (mean difference NPRS:-2.25[95%CI:-4.47,-0.03, p = 0.0468]) and for function in the 20 × 106 and 100 × 106 dose cohorts (mean difference KOOSADL:10.12[95%CI:-1.51,21.76, p = 0.0863] and 10.81[95%CI:-1.42,23.04, p = 0.0810] respectively). A trend in disease-modification was observed with improvement in total knee cartilage volume in MAG200 10, 20, and 100 × 106 dose cohorts, with progression of osteoarthritis in placebo, though this was not statistically significant. No clear dose response was observed. Conclusion This early-phase study provides supportive safety and efficacy evidence to progress MAG200 to later-stage trial development. Trial registration ACTRN12617001095358/ACTRN12621000622808.
Collapse
Affiliation(s)
- Julien Freitag
- School of Rural Medicine, Charles Sturt University, Orange, NSW, 2800, Australia
- Melbourne Stem Cell Centre Research, Box Hill, VIC, 3128, Australia
- Magellan Stem Cells, 9A Sugar Gum Court, Braeside, VIC, 3195, Australia
| | | | - James Wickham
- School of Dentistry & Medical Sciences, Charles Sturt University, Orange, NSW, 2800, Australia
| | - Kiran Shah
- Magellan Stem Cells, 9A Sugar Gum Court, Braeside, VIC, 3195, Australia
| | - Flavia Cicuttini
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
- Department of Rheumatology, Alfred Hospital, Melbourne, VIC, 3004, Australia
| | - Yuanyuan Wang
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Ann Solterbeck
- Statistical Revelations Pty Ltd, Ocean Grove, VIC, 3226, Australia
| |
Collapse
|
4
|
Oo WM. Prospects of Disease-Modifying Osteoarthritis Drugs. Rheum Dis Clin North Am 2024; 50:483-518. [PMID: 38942581 DOI: 10.1016/j.rdc.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Osteoarthritis (OA) causes a massive disease burden with a global prevalence of nearly 23% in 2020 and an unmet need for adequate treatment, given a lack of disease-modifying drugs (DMOADs). The author reviews the prospects of active DMOAD candidates in the phase 2/3 clinical trials of drug development pipeline based on key OA pathogenetic mechanisms directed to inflammation-driven, bone-driven, and cartilage-driven endotypes. The challenges and possible research opportunities are stated in terms of the formulation of a research question known as the PICO approach: (1) population, (2) interventions, (3) comparison or placebo, and (4) outcomes.
Collapse
Affiliation(s)
- Win Min Oo
- Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar; Rheumatology Department, Royal North Shore Hospital, Institute of Bone and Joint Research, Kolling Institute, The University of Sydney, Sydney, Australia.
| |
Collapse
|
5
|
Abbasifard M, Khorramdelazad H. Harmonizing hope: navigating the osteoarthritis melody through the CCL2/CCR2 axis for innovative therapeutic avenues. Front Immunol 2024; 15:1387651. [PMID: 39076996 PMCID: PMC11284107 DOI: 10.3389/fimmu.2024.1387651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Osteoarthritis (OA) is characterized by a complex interplay of molecular signals orchestrated by the CCL2/CCR2 axis. The pathogenesis of OA has been revealed to be influenced by a multifaceted effect of CCL2/CCR2 signaling on inflammation, cartilage degradation, and joint homeostasis. The CCL2/CCR2 axis promotes immune cell recruitment and tips the balance toward degeneration by influencing chondrocyte behavior. Insights into these intricate pathways will offer novel therapeutic approaches, paving the way for targeted interventions that may redefine OA management in the future. This review article explores the molecular symphony through the lens of the CCL2/CCR2 axis, providing a harmonious blend of current knowledge and future directions on OA treatment. Furthermore, in this study, through a meticulous review of recent research, the key players and molecular mechanisms that amplify the catabolic cascade within the joint microenvironment are identified, and therapeutic approaches to targeting the CCL2/CCR axis are discussed.
Collapse
Affiliation(s)
- Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
6
|
Zhang M, Wang Z, Ding C. Pharmacotherapy for osteoarthritis-related pain: current and emerging therapies. Expert Opin Pharmacother 2024; 25:1209-1227. [PMID: 38938057 DOI: 10.1080/14656566.2024.2374464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/26/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) related pain has affected millions of people worldwide. However, the current pharmacological options for managing OA-related pain have not achieved a satisfactory effect. AREAS COVERED This narrative review provides an overview of the current and emerging drugs for OA-related pain. It covers the drugs' mechanism of action, safety, efficacy, and limitations. The National Library of Medicine (PubMed) database was primarily searched from 2000 to 2024. EXPERT OPINION Current treatment options are limited and suboptimal for OA pain management. Topical nonsteroidal anti-inflammatory drugs (NSAIDs) are the recognized and first-line treatment in the management of OA-related pain, and other drugs are inconsistent recommendations by guidelines. Emerging treatment options are promising for OA-related pain, including nerve growth factor (NGF) inhibitors, ion channel inhibitors, and calcitonin gene-related peptide (CGRP) antagonists. Besides, drugs repurposing from antidepressants and antiepileptic analgesics are shedding light on the management of OA-related pain. The management of OA-related pain is challenging as pain is heterogeneous and subjective. A more comprehensive strategy combined with non-pharmacological therapy needs to be considered, and tailored management options to individualized patients.
Collapse
Affiliation(s)
- Mengdi Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiqiang Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Chapman JH, Ghosh D, Attari S, Ude CC, Laurencin CT. Animal Models of Osteoarthritis: Updated Models and Outcome Measures 2016-2023. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:127-146. [PMID: 38983776 PMCID: PMC11233113 DOI: 10.1007/s40883-023-00309-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2024]
Abstract
Purpose Osteoarthritis (OA) is a global musculoskeletal disorder that affects primarily the knee and hip joints without any FDA-approved disease-modifying therapies. Animal models are essential research tools in developing therapies for OA; many animal studies have provided data for the initiation of human clinical trials. Despite this, there is still a need for strategies to recapitulate the human experience using animal models to better develop treatments and understand pathogenesis. Since our last review on animal models of osteoarthritis in 2016, there have been exciting updates in OA research and models. The main purpose of this review is to update the latest animal models and key features of studies in OA research. Method We used our existing classification method and screened articles in PubMed and bibliographic search for animal OA models between 2016 and 2023. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Recent studies were analyzed and classified. We also identified ex vivo models as an area of ongoing research. Each animal model offers its own benefit in the study of OA and there are a full range of outcome measures that can be assessed. Despite the vast number of models, each has its drawbacks that have limited translating approved therapies for human use. Conclusion Depending on the outcome measures and objective of the study, researchers should pick the best model for their work. There have been several exciting studies since 2016 that have taken advantage of regenerative engineering techniques to develop therapies and better understand OA. Lay Summary Osteoarthritis (OA) is a chronic debilitating disease without any cure that affects mostly the knee and hip joints and often results in surgical joint replacement. Cartilage protects the joint from mechanical forces and degrades with age or in response to injury. The many contributing causes of OA are still being investigated, and animals are used for preclinical research and to test potential new treatments. A single consensus OA animal model for preclinical studies is non-existent. In this article, we review the many animal models for OA and provide a much-needed update on studies and model development since 2016.
Collapse
Affiliation(s)
- James H. Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Seyyedmorteza Attari
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
8
|
Joshi N, Yan J, Dang M, Slaughter K, Wang Y, Wu D, Ung T, Pandya V, Chen MX, Kaur S, Bhagchandani S, Alfassam HA, Joseph J, Gao J, Dewani M, Yip RCS, Weldon E, Shah P, Shukla C, Sherman NE, Luo JN, Conway T, Eickhoff JP, Botelho L, Alhasan AH, Karp JM, Ermann J. A Mechanically Resilient Soft Hydrogel Improves Drug Delivery for Treating Post-Traumatic Osteoarthritis in Physically Active Joints. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594611. [PMID: 38826308 PMCID: PMC11142096 DOI: 10.1101/2024.05.16.594611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Intra-articular delivery of disease-modifying osteoarthritis drugs (DMOADs) is likely to be most effective in early post-traumatic osteoarthritis (PTOA) when symptoms are minimal and patients are physically active. DMOAD delivery systems therefore must withstand repeated mechanical loading without affecting the drug release kinetics. Although soft materials are preferred for DMOAD delivery, mechanical loading can compromise their structural integrity and disrupt drug release. Here, we report a mechanically resilient soft hydrogel that rapidly self-heals under conditions resembling human running while maintaining sustained release of the cathepsin-K inhibitor L-006235 used as a proof-of-concept DMOAD. Notably, this hydrogel outperformed a previously reported hydrogel designed for intra-articular drug delivery, used as a control in our study, which neither recovered nor maintained drug release under mechanical loading. Upon injection into mouse knee joints, the hydrogel showed consistent release kinetics of the encapsulated agent in both treadmill-running and non-running mice. In a mouse model of aggressive PTOA exacerbated by treadmill running, L-006235 hydrogel markedly reduced cartilage degeneration. To our knowledge, this is the first hydrogel proven to withstand human running conditions and enable sustained DMOAD delivery in physically active joints, and the first study demonstrating reduced disease progression in a severe PTOA model under rigorous physical activity, highlighting the hydrogel's potential for PTOA treatment in active patients.
Collapse
|
9
|
Oo WM, Linklater J, Siddiq MAB, Fu K, Hunter DJ. Comparison of ultrasound guidance with landmark guidance for symptomatic benefits in knee, hip and hand osteoarthritis: Systematic review and meta-analysis of randomised controlled trials. Australas J Ultrasound Med 2024; 27:97-105. [PMID: 38784696 PMCID: PMC11109994 DOI: 10.1002/ajum.12386] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Introduction More than half of the patients with moderate and severe osteoarthritis (OA) report unsatisfactory pain relief, requiring consideration of intra-articular (IA) injections as the second-line management. Ultrasound-guided IA injection has proven evidence of higher accuracy in administering IA injectates into the joints than landmark-guided or blind IA injections. However, questions remain about translating higher accuracy rates of ultrasound-guided injection into better clinical improvements. Therefore, we examined the symptomatic benefits (pain, function and patient satisfaction) of ultrasound-guided injection in knee, hip and hand OA compared with blind injections by synthesising a systematic review and meta-analysis of randomised controlled trials (RCT). Methods PubMed, Medline and Embase databases were searched for eligible studies from their inception to August 28, 2023. Results Out of 295 records, our meta-analysis included four RCTs (338 patients with knee OA), demonstrating significant improvement in procedural pain [-0.89 (95% CI -1.25, -0.53)], pain at follow-up [-0.51 (95% CI -0.98, -0.04)] and function [1.30 (95% CI 0.86, 1.73)], favouring ultrasound guidance. One single study showed higher patient satisfaction with ultrasound guidance. Conclusion Ultrasound-guided IA injection provided superior clinical outcomes compared with landmark-guided IA injection.
Collapse
Affiliation(s)
- Win Min Oo
- Department of Physical Medicine and Rehabilitation, Mandalay General HospitalUniversity of MedicineMandalayMyanmar
- Rheumatology Department, Royal North Shore Hospital, and Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | | | - Md Abu Bakar Siddiq
- Rheumatology Department, Royal North Shore Hospital, and Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Kai Fu
- Rheumatology Department, Royal North Shore Hospital, and Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - David J. Hunter
- Rheumatology Department, Royal North Shore Hospital, and Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
10
|
Coppola C, Greco M, Munir A, Musarò D, Quarta S, Massaro M, Lionetto MG, Maffia M. Osteoarthritis: Insights into Diagnosis, Pathophysiology, Therapeutic Avenues, and the Potential of Natural Extracts. Curr Issues Mol Biol 2024; 46:4063-4105. [PMID: 38785519 PMCID: PMC11119992 DOI: 10.3390/cimb46050251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Osteoarthritis (OA) stands as a prevalent and progressively debilitating clinical condition globally, impacting joint structures and leading to their gradual deterioration through inflammatory mechanisms. While both non-modifiable and modifiable factors contribute to its onset, numerous aspects of OA pathophysiology remain elusive despite considerable research strides. Presently, diagnosis heavily relies on clinician expertise and meticulous differential diagnosis to exclude other joint-affecting conditions. Therapeutic approaches for OA predominantly focus on patient education for self-management alongside tailored exercise regimens, often complemented by various pharmacological interventions primarily targeting pain alleviation. However, pharmacological treatments typically exhibit short-term efficacy and local and/or systemic side effects, with prosthetic surgery being the ultimate resolution in severe cases. Thus, exploring the potential integration or substitution of conventional drug therapies with natural compounds and extracts emerges as a promising frontier in enhancing OA management. These alternatives offer improved safety profiles and possess the potential to target specific dysregulated pathways implicated in OA pathogenesis, thereby presenting a holistic approach to address the condition's complexities.
Collapse
Affiliation(s)
- Chiara Coppola
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy; (C.C.); (A.M.)
| | - Marco Greco
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Anas Munir
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy; (C.C.); (A.M.)
| | - Debora Musarò
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Stefano Quarta
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy;
| | - Maria Giulia Lionetto
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Michele Maffia
- Department of Experimental Medicine, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| |
Collapse
|
11
|
Bihlet AR, Balchen T, Goteti K, Sonne J, Ladel C, Karsdal MA, Ona V, Moreau F, Waterhouse R, Bay‐Jensen A, Guehring H. Safety, Tolerability, and Pharmacodynamics of the ADAMTS-5 Nanobody M6495: Two Phase 1, Single-Center, Double-Blind, Randomized, Placebo-Controlled Studies in Healthy Subjects and Patients With Osteoarthritis. ACR Open Rheumatol 2024; 6:205-213. [PMID: 38311369 PMCID: PMC11016567 DOI: 10.1002/acr2.11610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVE To assess the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of single and multiple injections of M6495, a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS-5) nanobody, in healthy volunteers and patients with osteoarthritis. METHODS Two randomized, placebo-controlled, double-blind studies were performed. Study 1 enrolled 54 healthy male volunteers who received one subcutaneous (s.c.) injection of M6495 (1-300 mg) or placebo (ratio 2:1), evaluating safety, PK, and PD as changes in the serum aggrecan fragment alanine-arginine-glycine-serine (ARGS). Study 2 enrolled 32 patients with osteoarthritis with Kellgren-Lawrence grades 2 to 4 and pain greater than or equal to 40 on the Western Ontario and McMaster Universities Arthritis Index pain subscale at screening and evaluated the safety, PK, and PD of three doses every two weeks (75-300 mg per dose) or six once-weekly M6495 s.c. doses (300 mg) or placebo (ratio 3:1) over 106 days' follow-up. RESULTS M6495 in single and multiple doses of less than or equal to 300 mg s.c. weekly was well tolerated with no clinically significant changes in any safety parameter. Adverse events more frequently reported in the M6495 groups were mostly mild cases of injection site reactions, myalgia, and nausea, which resolved after treatment cessation. The elimination half-life of single s.c. doses of M6495 ranged from 79 to 267 hours. M6495 administration substantially reduced serum ARGS levels, indicative of target engagement and indicating disease-modifying potential of M6495. CONCLUSION Treatment with M6495 in single and multiple doses up to and including 300 mg s.c. was found to be well tolerated and adequately safe for further clinical evaluation of potential disease-modifying effects.
Collapse
Affiliation(s)
| | | | | | - Jesper Sonne
- Bispebjerg Hospital and Copenhagen UniversityCopenhagenDenmark
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Vlashi R, Zhang X, Li H, Chen G. Potential therapeutic strategies for osteoarthritis via CRISPR/Cas9 mediated gene editing. Rev Endocr Metab Disord 2024; 25:339-367. [PMID: 38055160 DOI: 10.1007/s11154-023-09860-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
Osteoarthritis (OA) is an incapacitating and one of the most common physically degenerative conditions with an assorted etiology and a highly complicated molecular mechanism that to date lacks an efficient treatment. The capacity to design biological networks and accurately modify existing genomic sites holds an apt potential for applications across medical and biotechnological sciences. One of these highly specific genomes editing technologies is the CRISPR/Cas9 mechanism, referred to as the clustered regularly interspaced short palindromic repeats, which is a defense mechanism constituted by CRISPR associated protein 9 (Cas9) directed by small non-coding RNAs (sncRNA) that bind to target DNA through Watson-Crick base pairing rules where subsequent repair of the target DNA is initiated. Up-to-date research has established the effectiveness of the CRISPR/Cas9 mechanism in targeting the genetic and epigenetic alterations in OA by suppressing or deleting gene expressions and eventually distributing distinctive anti-arthritic properties in both in vitro and in vivo osteoarthritic models. This review aims to epitomize the role of this high-throughput and multiplexed gene editing method as an analogous therapeutic strategy that could greatly facilitate the clinical development of OA-related treatments since it's reportedly an easy, minimally invasive technique, and a comparatively less painful method for osteoarthritic patients.
Collapse
Affiliation(s)
- Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, China.
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
13
|
Ye J, Xie D, Li X, Lu N, Zeng C, Lei G, Wei J, Li J. Phenotypes of osteoarthritis-related knee pain and their transition over time: data from the osteoarthritis initiative. BMC Musculoskelet Disord 2024; 25:173. [PMID: 38402384 PMCID: PMC10893610 DOI: 10.1186/s12891-024-07286-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/16/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Identification of knee osteoarthritis (OA) pain phenotypes, their transition patterns, and risk factors for worse phenotypes, may guide prognosis and targeted treatment; however, few studies have described them. We aimed to investigate different pain phenotypes, their transition patterns, and potential risk factors for worse pain phenotypes. METHODS Utilizing data from the Osteoarthritis Initiative (OAI), pain severity was assessed using the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain subscale. We identified the activity-related pain phenotypes and estimated the transition probabilities of pain phenotypes from baseline to the 24-month using latent transition analysis. We examined the risk factors at baseline with the 24-month pain phenotypes and the transition of pain phenotypes. RESULTS In 4796 participants, we identified four distinct knee pain phenotypes at both baseline and 24-month follow-up: no pain, mild pain during activity (Mild P-A), mild pain during both rest and activity (Mild P-R-A), and moderate pain during both rest and activity (Mod P-R-A). 82.9% knees with no pain at baseline stayed the same at 24-month follow-up, 17.1% progressed to worse pain phenotypes. Among "Mild P-A" at baseline, 32.0% converted to no-pain, 12.8% progressed to "Mild P-R-A", and 53.2% remained. Approximately 46.1% of "Mild P-R-A" and 54.5% of "Mod P-R-A" at baseline experienced remission by 24-month. Female, non-whites, participants with higher depression score, higher body mass index (BMI), higher Kellgren and Lawrence (KL) grade, and knee injury history were more likely to be in the worse pain phenotypes, while participants aged 65 years or older and with higher education were less likely to be in worse pain phenotypes at 24-month follow-up visit. Risk factors for greater transition probability to worse pain phenotypes at 24-month included being female, non-whites, participants with higher depression score, higher BMI, and higher KL grade. CONCLUSIONS We identified four distinct knee pain phenotypes. While the pain phenotypes remained stable in the majority of knees over 24 months period, substantial proportion of knees switched to different pain phenotypes. Several socio-demographics as well as radiographic lesions at baseline are associated with worse pain phenotypes at 24-month follow-up visit and transition of pain phenotypes.
Collapse
Affiliation(s)
- Jing Ye
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Dongxing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiao Li
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Na Lu
- Arthritis Research Canada, Richmond, Canada
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jiatian Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
14
|
Cao J, Wang D, Yuan J, Hu F, Wu Z. Exploration of the potential mechanism of Duhuo Jisheng Decoction in osteoarthritis treatment by using network pharmacology and molecular dynamics simulation. Comput Methods Biomech Biomed Engin 2024; 27:251-265. [PMID: 37830364 DOI: 10.1080/10255842.2023.2268232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2023]
Abstract
In this study, the active ingredients of 15 Chinese herbal medicines of Duhuo Jisheng Decoction and their corresponding targets were obtained from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database. The microarray data of Osteoarthritis (OA) were obtained through the GEO database for differential analysis and then a drug target-OA-related gene protein-protein interaction (PPI) network was established. The potential targets of Duhuo Jisheng Decoction in the treatment of OA were acquired by intersecting the OA-associated genes with the target genes of active ingredients. Random walk with restart (RWR) analysis of PPI networks was performed using potential targets as seed, and the top 50 genes of affinity coefficients were used as key action genes of Duhuo Jisheng Decoction in the treatment of OA. A drug-active ingredient-gene interaction network was established. AKT1, a key target of Duhuo Jisheng Decoction in the treatment of OA, was obtained by topological analysis of the gene interaction network. Molecular docking and molecular dynamics verified the binding of AKT1 to its corresponding drug active ingredients. CETSA assay demonstrated that the combination of luteolin and AKT1 increased the stability of AKT1, and the combination efficiency was high. In conclusion, the molecular mechanism of Duhuo Jisheng Decoction in treating OA featured by multiple components, targets, and pathways had been further investigated in this study, which is of significance for discovering as well as developing new drugs for this disease. The findings can also offer personalized diagnosis and treatment strategies for patients with OA in clinical practice.
Collapse
Affiliation(s)
- Jin Cao
- Department of Orthopedics, First People's Hospital of Linping District, Hangzhou, China
| | - Dayong Wang
- Department of Orthopedics, First People's Hospital of Linping District, Hangzhou, China
| | - Jianhua Yuan
- Department of Orthopedics, First People's Hospital of Linping District, Hangzhou, China
| | - Fenggen Hu
- Department of Orthopedics, First People's Hospital of Linping District, Hangzhou, China
| | - Zhen Wu
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
15
|
Copp ME, Shine J, Brown HL, Nimmala KR, Hansen OB, Chubinskaya S, Collins JA, Loeser RF, Diekman BO. Sirtuin 6 activation rescues the age-related decline in DNA damage repair in primary human chondrocytes. Aging (Albany NY) 2023; 15:13628-13645. [PMID: 38078876 PMCID: PMC10756124 DOI: 10.18632/aging.205394] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023]
Abstract
While advanced age is widely recognized as the greatest risk factor for osteoarthritis (OA), the biological mechanisms behind this connection remain unclear. Previous work has demonstrated that chondrocytes from older cadaveric donors have elevated levels of DNA damage as compared to chondrocytes from younger donors. The purpose of this study was to determine whether a decline in DNA repair efficiency is one explanation for the accumulation of DNA damage with age, and to quantify the improvement in repair with activation of Sirtuin 6 (SIRT6). After acute damage with irradiation, DNA repair was shown to be more efficient in chondrocytes from young (≤45 years old) as compared to middle-aged (50-65 years old) or older (>70 years old) cadaveric donors. Activation of SIRT6 with MDL-800 improved the repair efficiency, while inhibition with EX-527 reduced the rate of repair and increased the percentage of cells that retain high levels of damage. In addition to affecting repair after acute damage, treating chondrocytes from older donors with MDL-800 for 48 hours significantly reduced the amount of baseline DNA damage. Chondrocytes isolated from the knees of mice between 4 months and 22 months of age revealed both an increase in DNA damage with aging, and a decrease in DNA damage following MDL-800 treatment. Lastly, treating murine cartilage explants with MDL-800 lowered the percentage of chondrocytes with high p16 promoter activity, which supports the concept that using SIRT6 activation to maintain low levels of DNA damage may prevent the initiation of senescence.
Collapse
Affiliation(s)
- Michaela E. Copp
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hannon L. Brown
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| | - Kirti R. Nimmala
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| | - Oliver B. Hansen
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL 60612, USA
| | - John A. Collins
- Department of Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA 19144, USA
| | - Richard F. Loeser
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian O. Diekman
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| |
Collapse
|
16
|
Bordon G, Berenbaum F, Distler O, Luciani P. Harnessing the multifunctionality of lipid-based drug delivery systems for the local treatment of osteoarthritis. Biomed Pharmacother 2023; 168:115819. [PMID: 37939613 DOI: 10.1016/j.biopha.2023.115819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023] Open
Abstract
Osteoarthritis (OA) is a widespread joint condition affecting millions globally, presenting a growing socioeconomic burden thus making the development of more effective therapeutic strategies crucial. This review emphasizes recent advancements in lipid-based drug delivery systems (DDSs) for intra-articular administration of OA therapeutics, encompassing non-steroidal anti-inflammatory drugs, corticosteroids, small molecule disease-modifying OA drugs, and RNA therapeutics. Liposomes, lipid nanoparticles, lipidic mesophases, extracellular vesicles and composite systems exhibit enhanced stability, targeted delivery, and extended joint retention, which contribute to improved therapeutic outcomes and minimized systemic drug exposure. Although active targeting strategies hold promise, further research is needed to assess their targeting efficiency in physiologically relevant conditions. Simultaneously, multifunctional DDSs capable of delivering combinations of distinct therapeutic classes offer synergistic effects and superior OA treatment outcomes. The development of such long-acting systems that resist rapid clearance from the joint space is crucial, where particle size and targeting capabilities emerge as vital factors. Additionally, combining cartilage lubrication properties with sustained drug delivery has demonstrated potential in animal models, meriting further investigation in human clinical trials. This review highlights the crucial need for direct, head-to-head comparisons of novel DDSs with standard treatments, particularly within the same drug class. These comparisons are essential in accurately evaluating their effectiveness, safety, and clinical applicability, and are set to significantly shape the future of OA therapy.
Collapse
Affiliation(s)
- Gregor Bordon
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Francis Berenbaum
- Sorbonne University, INSERM CRSA, AP-HP Saint-Antoine Hospital, Paris, France
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
17
|
Ward RJ, Driban JB, MacKay JW, McAlindon TE, Lu B, Eaton CB, Lo GH, Barbe MF, Harkey MS. Meniscal degeneration is prognostic of destabilzing meniscal tear and accelerated knee osteoarthritis: Data from the Osteoarthritis Initiative. J Orthop Res 2023; 41:2418-2423. [PMID: 37094976 PMCID: PMC10592659 DOI: 10.1002/jor.25575] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/03/2023] [Accepted: 04/17/2023] [Indexed: 04/26/2023]
Abstract
The objective of this study was to assess the prognostic potential of magnetic resonance (MR)-detected meniscal degeneration in relation to incident destabilizing meniscal tears (radial, complex, root, or macerated) or accelerated knee osteoarthritis (AKOA). We used existing MR data from a case-control study of three groups from the Osteoarthritis Initiative without radiographic KOA at baseline: AKOA, typical KOA, and no KOA. From these groups, we included people without medial and lateral meniscal tear at baseline (n = 226) and 48-month meniscal data (n = 221). Intermediate-weighted fat-suppressed MR images annually from baseline to the 48-month visit were graded using a semiquantitative meniscal tear classification criterion. Incident destabilizing meniscal tear was defined as progressing from an intact meniscus to a destabilizing tear by the 48-month visit. We used two logistic regression models to assess whether: (1) presence of medial meniscal degeneration was associated with an incident medial destabilizing meniscal tear, and (2) presence of meniscal degeneration in either meniscus was associated with incident AKOA over the next 4 years. People with the presence of a medial meniscal degeneration had three times the odds of developing an incident destabilizing medial meniscal tear within 4 years compared with a person without medial meniscus degeneration (odds ratio [OR]: 3.03; 95% confidence interval [CI]: 1.40-6.59). People with meniscal degeneration had five times the odds of developing incident AKOA within 4 years compared with a person without meniscal degeneration in either meniscus (OR: 5.04; 95% CI: 2.57-9.89). Meniscal degeneration on MR is clinically meaningful as it relates to future poor outcomes.
Collapse
Affiliation(s)
- Robert J. Ward
- Department of Radiology, Saint Georges University, Grenada
WI, USA; Sullivan’s Island Imaging, Sullivan’s Island SC, USA
| | - Jeffrey B. Driban
- Division of Rheumatology, Allergy, & Immunology, Tufts
Medical Center, Boston MA, USA
| | - James W. MacKay
- Norwich Medical School, University of East Anglia, Norwich,
UK & Department of Radiology, University of Cambridge, Cambridge, UK
| | - Timothy E. McAlindon
- Division of Rheumatology, Allergy, & Immunology, Tufts
Medical Center, Boston MA, USA
| | - Bing Lu
- Division of Rheumatology, Immunology & Allergy, Brigham
& Women’s Hospital and Harvard Medical School, Boston MA, USA
| | - Charles B. Eaton
- Center for Primary Care and Prevention, Alpert Medical
School of Brown University, Pawtucket RI, USA
| | - Grace H. Lo
- Medical Care Line and Research Care Line, Houston VA
HSR&D Center for Innovations in Quality, Effectiveness and Safety, Michael E.
DeBakey Medical Center, Houston TX, USA
- Section of Immunology, Allergy, and Rheumatology, Baylor
College of Medicine, Houston TX, USA
| | - Mary F. Barbe
- Center for Translational Medicine, Temple University School
of Medicine, Philadelphia PA, USA
| | - Matthew S. Harkey
- Department of Kinesiology, Michigan State University, East
Lansing MI, USA
| |
Collapse
|
18
|
Golovach I, Rekalov D, Akimov OY, Kostenko H, Kostenko V, Mishchenko A, Solovyova N, Kostenko V. Molecular mechanisms and potential applications of chondroitin sulphate in managing post-traumatic osteoarthritis. Reumatologia 2023; 61:395-407. [PMID: 37970120 PMCID: PMC10634410 DOI: 10.5114/reum/172211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/06/2023] [Indexed: 11/17/2023] Open
Abstract
Post-traumatic osteoarthritis (PTOA), a disorder of the synovium, subchondral bone, and cartilage that affects the entire joint, constitutes approximately 12% of all cases of symptomatic osteoarthritis. This review summarizes the pathogenetic mechanisms that underlie the positive influence of chondroitin sulphates (CSs) on PTOA as means of preventive and therapeutic treatment. Mechanisms of PTOA development involve chondrocytes undergoing various forms of cell death (apoptosis, pyroptosis, necroptosis, ferroptosis and/or necrosis). Chondroitin sulphates are a class of glycosaminoglycans that improve the structure and function of cartilage and subchondral bone, which is associated with their ability to decrease the activation of NF-κB and p38 MAPK, and up-regulate Nrf2. Standardized small fish extract (SSFE) is an example of the drugs that can attenuate NF-κB-mediated systemic inflammation, potentially helping to reduce joint inflammation and cartilage degradation, improve joint function, and alleviate pain and disability in patients with these conditions.
Collapse
Affiliation(s)
- Iryna Golovach
- Centre for Rheumatology, Osteoporosis and Immunobiological Therapy, Feofania Clinical Hospital of the State Affairs Administration, Kyiv, Ukraine
| | - Dmytro Rekalov
- Department of Internal Diseases No 3, Zaporizhzhia State Medical and Pharmaceutical University, Ukraine
| | - Oleh Ye Akimov
- Department of Pathophysiology, Poltava State Medical University, Ukraine
| | - Heorhii Kostenko
- Department of Pathophysiology, Poltava State Medical University, Ukraine
| | - Viktoriia Kostenko
- Department of Foreign Languages with Latin and Medical Terminology, Poltava State Medical University, Ukraine
| | - Artur Mishchenko
- Department of Pathophysiology, Poltava State Medical University, Ukraine
| | - Natalia Solovyova
- Department of Pathophysiology, Poltava State Medical University, Ukraine
| | - Vitalii Kostenko
- Department of Pathophysiology, Poltava State Medical University, Ukraine
| |
Collapse
|
19
|
Makaram NS, Simpson AHRW. Disease-modifying agents in osteoarthritis: where are we now and what does the future hold? Bone Joint Res 2023; 12:654-656. [PMID: 37839796 PMCID: PMC10577043 DOI: 10.1302/2046-3758.1210.bjr-2023-0237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
Cite this article: Bone Joint Res 2023;12(10):654–656.
Collapse
Affiliation(s)
- Navnit S. Makaram
- Edinburgh Orthopaedics, Royal Infirmary of Edinburgh, Edinburgh, UK
- University of Edinburgh, Edinburgh, UK
| | - A. H. R. W. Simpson
- Edinburgh Orthopaedics, Royal Infirmary of Edinburgh, Edinburgh, UK
- University of Edinburgh, Edinburgh, UK
| |
Collapse
|
20
|
De Roover A, Escribano-Núñez A, Monteagudo S, Lories R. Fundamentals of osteoarthritis: Inflammatory mediators in osteoarthritis. Osteoarthritis Cartilage 2023; 31:1303-1311. [PMID: 37353140 DOI: 10.1016/j.joca.2023.06.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/25/2023]
Abstract
OBJECTIVES As more has become known of the pathophysiology of osteoarthritis (OA), evidence that inflammation plays a critical role in its development and progression has accumulated. Here, we aim to review current knowledge of the complex inflammatory network in the OA joint. DESIGN This narrative review is presented in three main sections: local inflammation, systemic inflammation, and therapeutic implications. We focused on inflammatory mediators and their link to OA structural changes in the joint. RESULTS OA is characterized by chronic and low-grade inflammation mediated mostly by the innate immune system, which results in cartilage degradation, bone remodeling and synovial changes. Synovitis is regarded as an OA characteristic and associated with increased severity of symptoms and joint dysfunction. However, the articular cartilage and the subchondral bone also produce several pro-inflammatory mediators thus establishing a complex interplay between the different tissues of the joint. In addition, systemic low-grade inflammation induced by aging, obesity and metabolic syndrome can contribute to OA development and progression. The main inflammatory mediators associated with OA include cytokines, chemokines, growth factors, adipokines, and neuropeptides. CONCLUSIONS Future research is needed to deeper understand the molecular pathways mediating the inflammation in OA to provide new therapeutics that target these pathways, or to repurpose existing drugs.
Collapse
Affiliation(s)
- Astrid De Roover
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Ana Escribano-Núñez
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Silvia Monteagudo
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Rik Lories
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
21
|
Ku SK, Kim JK, Chun YS, Song CH. Anti-Osteoarthritic Effects of Antarctic Krill Oil in Primary Chondrocytes and a Surgical Rat Model of Knee Osteoarthritis. Mar Drugs 2023; 21:513. [PMID: 37888448 PMCID: PMC10608626 DOI: 10.3390/md21100513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/07/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Osteoarthritis (OA) is characterized by progressive cartilage destruction and synovitis; however, there are no approved disease-modifying OA drugs. Krill oil (KO) has been reported to possess anti-inflammatory properties and alleviate joint pain in knee OA, indicating its potential to target the inflammatory mechanism of OA. Therefore, the anti-OA effects of KO were investigated in primary chondrocytes and a surgical rat model of knee OA. The oral administration of KO at 200 and 100 mg/kg for 8 weeks improved joint swelling and mobility in the animal model and led to increased bone mineral density and compressive strength in the cartilage. The oral KO doses upregulated chondrogenic genes (type 2 collagen, aggrecan, and Sox9), with inhibition of inflammation markers (5-lipoxygenase and prostaglandin E2) and extracellular matrix (ECM)-degrading enzymes (MMP-2 and MMP-9) in the cartilage and synovium. Consistently, KO treatments increased the viability of chondrocytes exposed to interleukin 1α, accompanied by the upregulation of the chondrogenic genes and the inhibition of the ECM-degrading enzymes. Furthermore, KO demonstrated inhibitory effects on lipopolysaccharide-induced chondrocyte inflammation. Histopathological and immunohistochemical analyses revealed that KO improved joint destruction and synovial inflammation, probably due to the anti-inflammatory, anti-apoptotic, and chondrogenic effects. These findings suggest the therapeutic potential of KO for knee OA.
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea;
| | - Jong-Kyu Kim
- AriBnC Co., Ltd., Yongin 16914, Republic of Korea; (J.-K.K.); (Y.-S.C.)
| | - Yoon-Seok Chun
- AriBnC Co., Ltd., Yongin 16914, Republic of Korea; (J.-K.K.); (Y.-S.C.)
| | - Chang-Hyun Song
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea;
| |
Collapse
|
22
|
Oo WM, Linklater J, Bennell KL, Yu SP, Duong V, Hunter DJ. Inflammatory ultrasound features as prognostic factors of pain and functional outcomes following intra-articular platelet-rich plasma in knee osteoarthritis. Int J Rheum Dis 2023; 26:1540-1546. [PMID: 37309612 PMCID: PMC10946528 DOI: 10.1111/1756-185x.14781] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 05/05/2023] [Accepted: 05/26/2023] [Indexed: 06/14/2023]
Abstract
AIM To explore inflammatory ultrasound predictors of improvements in pain and function over 2, 6, and 12 months following administration of intra-articular platelet-rich plasma (PRP) in knee osteoarthritis (OA). METHOD Patients with painful mild-moderate radiographic knee OA from a subset of the RESTORE RCT underwent ultrasound assessment according to the standardized OMERACT scanning protocol to detect inflammatory features such as synovitis, synovial hypertrophy, and effusion with power Doppler. The study knee was treated with 3 once-weekly PRP injections obtained after centrifugation at 1500 g for 5 min. Numerical Rating Score (NRS), Intermittent and Constant Osteoarthritis Pain (ICOAP) questionnaire, and the Western Ontario and McMaster Universities Arthritis Index (WOMAC) function sub-score were used to measure pain and functional severity. Separate linear regression models were performed to determine whether baseline ultrasound-detected features of inflammation predicted the improvement in pain and function following PRP injection in both unadjusted and adjusted models for confounders. RESULTS Forty-four participants were included, with 25 (56.8%) being female. In an unadjusted model, higher OMERACT scores for inflammatory features such as global synovitis and/or effusion were significantly associated with greater improvement in all outcomes measured at 2 months but not at 6 and 12 months for pain measures. Only global synovitis showed significant association with functional improvement at 2 and 12 months. Similar findings were observed in the adjusted model. CONCLUSION Ultrasound indices of knee inflammation predicted short-term improvements in pain severity and both short- and longer-term improvements in function following intra-articular PRP injection.
Collapse
Affiliation(s)
- Win Min Oo
- Rheumatology Department, Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling InstituteUniversity of SydneySydneyNew South WalesAustralia
- Department of Physical Medicine and Rehabilitation, Mandalay General HospitalUniversity of Medicine, MandalayMandalayMyanmar
| | - James Linklater
- Department of Musculoskeletal ImagingCastlereagh Sports Imaging CentreSt. Leonards, SydneyNew South WalesAustralia
| | - Kim L. Bennell
- Department of Physiotherapy, Centre for Health, Exercise and Sports Medicine, School of Health Sciences, Faculty of Medicine Dentistry & Health SciencesThe University of MelbourneMelbourneVictoriaAustralia
| | - Shirley P. Yu
- Rheumatology Department, Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling InstituteUniversity of SydneySydneyNew South WalesAustralia
| | - Vicky Duong
- Rheumatology Department, Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling InstituteUniversity of SydneySydneyNew South WalesAustralia
| | - David J. Hunter
- Rheumatology Department, Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling InstituteUniversity of SydneySydneyNew South WalesAustralia
| |
Collapse
|
23
|
Papathanasiou I, Balis C, Destounis D, Mourmoura E, Tsezou A. NEAT1-mediated miR-150-5p downregulation regulates b-catenin expression in OA chondrocytes. Funct Integr Genomics 2023; 23:246. [PMID: 37468759 DOI: 10.1007/s10142-023-01139-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023]
Abstract
We investigated the role of miR-150-5p in osteoarthritic (OA) chondrocytes, as well as the possible regulatory role of long non-coding RNAs (lncRNAs) in miR-150-5p expression. TargetScan, StarBase, DIANA-LncBase, and Open Targets databases were used to predict miR-150-5p target genes, lncRNAs/miR-150-5p interactions, and OA-related genes. Protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). Gene ontology (GO) and pathway analysis were performed using Enrichr database. A publicly available RNA-seq dataset was retrieved to identify differentially expressed lncRNAs in damaged vs intact cartilage. We re-analyzed the retrieved RNA-seq data and revealed 177 differentially expressed lncRNAs in damage vs intact cartilage, including Nuclear Paraspeckle Assembly Transcript 1(NEAT1). MiR-150-5p, NEAT1, b-catenin, matrix metallopeptidase 13 (MMP-13), and ADAM metallopeptidase with thrombospondin type 1 motif 5 (ADAMTS-5) expressions were assessed by reverse transcription-quantitative PCR (RT-qPCR) and western blot assay. Knockout and transfection experiments were conducted to investigate the role of NEAT1/miR-150-5p/b-catenin in cartilage degradation. Bioinformatics analysis revealed that b-catenin was an OA-related miR-150-5p target. MiR-150-5p overexpression in OA chondrocytes resulted in decreased expression of b-catenin, as well as MMP-13 and ADAMTS-5, both being Wnt/b-catenin downstream target genes. NEAT1/miR-150-5p interaction was predicted by bioinformatics analysis, while NEAT1 knockout led to increased expression of miR-150-5p in OA chondrocytes. Moreover, inhibition of miR-150-5p reversed the repressive effects of NEAT1 silencing in b-catenin expression in OA chondrocytes. Our results support a possible catabolic role of NEAT1/miR-150-5p interaction in OA progression by regulating b-catenin expression.
Collapse
Affiliation(s)
- Ioanna Papathanasiou
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece
- Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Charalampos Balis
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Dimitrios Destounis
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Evanthia Mourmoura
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Aspasia Tsezou
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece.
- Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece.
| |
Collapse
|
24
|
Li X, Shen L, Deng Z, Huang Z. New treatment for osteoarthr: pbad014itis: Gene therapy. PRECISION CLINICAL MEDICINE 2023; 6:pbad014. [PMID: 37333626 PMCID: PMC10273835 DOI: 10.1093/pcmedi/pbad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Osteoarthritis is a complex degenerative disease that affects the entire joint tissue. Currently, non-surgical treatments for osteoarthritis focus on relieving pain. While end-stage osteoarthritis can be treated with arthroplasty, the health and financial costs associated with surgery have forced the search for alternative non-surgical treatments to delay the progression of osteoarthritis and promote cartilage repair. Unlike traditional treatment, the gene therapy approach allows for long-lasting expression of therapeutic proteins at specific sites. In this review, we summarize the history of gene therapy in osteoarthritis, outlining the common expression vectors (non-viral, viral), the genes delivered (transcription factors, growth factors, inflammation-associated cytokines, non-coding RNAs) and the mode of gene delivery (direct delivery, indirect delivery). We highlight the application and development prospects of the gene editing technology CRISPR/Cas9 in osteoarthritis. Finally, we identify the current problems and possible solutions in the clinical translation of gene therapy for osteoarthritis.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Leyao Shen
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
25
|
Mobasheri A, Thudium CS, Bay-Jensen AC, Maleitzke T, Geissler S, Duda GN, Winkler T. Biomarkers for osteoarthritis: Current status and future prospects. Best Pract Res Clin Rheumatol 2023; 37:101852. [PMID: 37620236 DOI: 10.1016/j.berh.2023.101852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/14/2023] [Indexed: 08/26/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis globally and a major cause of pain, physical disability, and loss of economic productivity, with currently no causal treatment available. This review article focuses on current research on OA biomarkers and the potential for using biomarkers in future clinical practice and clinical trials of investigational drugs. We discuss how biomarkers, specifically soluble ones, have a long path to go before reaching clinical standards of care. We also discuss how biomarkers can help in phenotyping and subtyping to achieve enhanced stratification and move toward better-designed clinical trials. We also describe how biomarkers can be used for molecular endotyping and for determining the clinical outcomes of investigational cell-based therapies. Biomarkers have the potential to be developed as surrogate end points in clinical trials and help private-public consortia and the biotechnology and pharmaceutical industries develop more effective and targeted personalized treatments and enhance clinical care for patients with OA.
Collapse
Affiliation(s)
- Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Belgium.
| | | | | | - Tazio Maleitzke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Sven Geissler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany; Berlin Center for Advanced Therapies (BECAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Georg N Duda
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany; Berlin Center for Advanced Therapies (BECAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Tobias Winkler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
26
|
Li S, Cao P, Chen T, Ding C. Latest insights in disease-modifying osteoarthritis drugs development. Ther Adv Musculoskelet Dis 2023; 15:1759720X231169839. [PMID: 37197024 PMCID: PMC10184265 DOI: 10.1177/1759720x231169839] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 03/29/2023] [Indexed: 05/19/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent and severely debilitating disease with an unmet medical need. In order to alleviate OA symptoms or prevent structural progression of OA, new drugs, particularly disease-modifying osteoarthritis drugs (DMOADs), are required. Several drugs have been reported to attenuate cartilage loss or reduce subchondral bone lesions in OA and thus potentially be DMOADs. Most biologics (including interleukin-1 (IL-1) and tumor necrosis factor (TNF) inhibitors), sprifermin, and bisphosphonates failed to yield satisfactory results when treating OA. OA clinical heterogeneity is one of the primary reasons for the failure of these clinical trials, which can require different therapeutic approaches based on different phenotypes. This review describes the latest insights into the development of DMOADs. We summarize in this review the efficacy and safety profiles of various DMOADs targeting cartilage, synovitis, and subchondral bone endotypes in phase 2 and 3 clinical trials. To conclude, we summarize the reasons for clinical trial failures in OA and suggest possible solutions.
Collapse
Affiliation(s)
- Shengfa Li
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peihua Cao
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tianyu Chen
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, 261 Industry Road, Guangzhou 510515, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Clinical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
27
|
Higa S, Nakata K, Karasawa Y, Ohwaki K. Comparative effectiveness of early initiation of oral nonsteroidal anti-inflammatory drug and oral acetaminophen therapies on the time to knee replacement in patients with knee osteoarthritis in Japan. BMC Musculoskelet Disord 2023; 24:297. [PMID: 37060072 PMCID: PMC10103473 DOI: 10.1186/s12891-023-06415-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 04/10/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Although disease-modifying properties of nonsteroidal anti-inflammatory drugs (NSAIDs) for osteoarthritis (OA) have been reported, the effects of NSAIDs on OA progression remain controversial. The purpose of this study was to investigate the effect of early initiation of oral NSAID therapy on the progression of knee OA. METHODS In this retrospective cohort study, we extracted data of patients newly diagnosed with knee OA between November 2007 and October 2018 from a Japanese claims database. The primary outcome was the time to knee replacement (KR), and the secondary outcome was the time to composite event including joint lavage and debridement, osteotomy, or arthrodesis in addition to KR. Weighted Cox regression analysis with standardized mortality/morbidity ratio (SMR) weight was performed to compare the outcomes between patients prescribed oral NSAID (NSAID group) and those prescribed oral acetaminophen (APAP) (APAP group) early after a diagnosis of knee OA. Propensity scores were calculated using logistic regression conditioned on potential confounding factors, and SMR weights were calculated using the propensity scores. RESULTS The study population comprised 14,261 patients, who were divided into two groups as follows: 13,994 in the NSAID group and 267 in the APAP group. The mean ages of patients in the NSAID and APAP groups were 56.9 and 56.1 years, respectively. Furthermore, 62.01% and 68.16% patients in the NSAID and APAP groups, respectively, were female. The NSAID group had a reduced risk of KR compared with the APAP group in the analysis using SMR weighting (SMR-weighted hazard ratio, 0.19; 95% confidence interval, 0.05-0.78). While no statistically significant difference was found for the risk of composite event between the two groups (SMR-weighted hazard ratio, 0.56; 95% confidence interval, 0.16-1.91). CONCLUSIONS The risk of KR in the NSAID group was significantly lower than that in the APAP group after accounting for residual confounding using SMR weighting. This finding suggests that oral NSAID therapy early after the initial diagnosis is associated with a reduced risk of KR in patients with symptomatic knee OA.
Collapse
Affiliation(s)
- Shingo Higa
- Teikyo University Graduate School of Public Health, Tokyo, Japan.
- Medical Affairs, Viatris Pharmaceuticals Japan Inc., Tokyo, Japan.
| | - Ken Nakata
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Karasawa
- Medical Affairs, Viatris Pharmaceuticals Japan Inc., Tokyo, Japan
| | - Kazuhiro Ohwaki
- Teikyo University Graduate School of Public Health, Tokyo, Japan
| |
Collapse
|
28
|
Roemer F, Maschek S, Wisser A, Guermazi A, Hunter D, Eckstein F, Wirth W. Worsening of Articular Tissue Damage as Defined by Semi-Quantitative MRI Is Associated With Concurrent Quantitative Cartilage Loss Over 24 Months. Cartilage 2023; 14:39-47. [PMID: 36624993 PMCID: PMC10076901 DOI: 10.1177/19476035221147677] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE To assess the association of worsening of magnetic resonance imaging (MRI) semi-quantitative (SQ) tissue features with concurrent change in quantitative (Q) cartilage thickness measurements over 24 months within the Foundation for the National Institutes of Health (FNIH) Biomarker Consortium study. METHODS In all, 599 participants were included. SQ assessment included cartilage damage, meniscal extrusion and damage, osteophytes, bone marrow lesions (BMLs), and effusion- and Hoffa-synovitis. Change in medial compartment Q cartilage thickness was stratified by concurrent ipsicompartmental SQ changes. Between-group comparisons were performed using analysis of covariance (ANCOVA) with adjustment for age, sex, and body mass index (BMI). Results were presented as adjusted mean difference. RESULTS Knees with any increase in SQ cartilage scores in the medial compartment (n = 268) showed more Q cartilage loss compared to knees that remained stable (mean adjusted difference [MAD] = -0.16 mm, 95% confidence interval [CI]: [-0.19, -0.13] mm). Knees with any increase in meniscal extrusion in the medial compartment (n = 98) showed more Q cartilage loss than knees without (MAD = -0.18 mm, 95% CI: [-0.22, -0.14] mm. Comparable findings were seen for meniscal damage worsening. Regarding BMLs, an increase by one subregion resulted in a MAD of Q cartilage loss of -0.10 mm, 95% CI: [-0.14, -0.06] mm, while this effect almost tripled for change in two or more subregions. Increase in either effusion- and/or Hoffa-synovitis by one grade resulted in a MAD of -0.07 mm, 95% CI: [-0.10, -0.03] mm. CONCLUSION Worsening of SQ cartilage damage, meniscal extrusion and damage, number of subregions affected by BML, maximum size of BMLs and worsening of effusion- and/or Hoffa synovitis is associated with increased Q cartilage loss.
Collapse
Affiliation(s)
- Frank Roemer
- Friedrich-Alexander-Universitat Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Maschek
- Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | - Anna Wisser
- Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | | | - David Hunter
- The University of Sydney, Sydney, NSW, Australia
| | - Felix Eckstein
- Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | - Wolfgang Wirth
- Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| |
Collapse
|
29
|
Copp ME, Shine J, Brown HL, Nimmala KR, Chubinskaya S, Collins JA, Loeser RF, Diekman BO. SIRT6 activation rescues the age-related decline in DNA damage repair in primary human chondrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530205. [PMID: 36909504 PMCID: PMC10002640 DOI: 10.1101/2023.02.27.530205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
While advanced age has long been recognized as the greatest risk factor for osteoarthritis (OA), the biological mechanisms behind this connection remain unclear. Previous work has demonstrated that chondrocytes from older cadaveric donors have elevated levels of DNA damage as compared to chondrocytes from younger donors. The purpose of this study was to determine whether a decline in DNA repair efficiency is one explanation for the accumulation of DNA damage with age, and to quantify the improvement in repair with activation of Sirtuin 6 (SIRT6). Using an acute irradiation model to bring the baseline level of all donors to the same starting point, this study demonstrates a decline in repair efficiency during aging when comparing chondrocytes from young (≤45 years old), middle-aged (50-65 years old), or older (>70 years old) cadaveric donors with no known history of OA or macroscopic cartilage degradation at isolation. Activation of SIRT6 in middle-aged chondrocytes with MDL-800 (20 μM) improved the repair efficiency, while inhibition with EX-527 (10 μM) inhibited the rate of repair and the increased the percentage of cells that retained high levels of damage. Treating chondrocytes from older donors with MDL-800 for 48 hours significantly reduced the amount of DNA damage, despite this damage having accumulated over decades. Lastly, chondrocytes isolated from the proximal femurs of mice between 4 months and 22 months of age revealed both an increase in DNA damage with aging, and a decrease in DNA damage following MDL-800 treatment.
Collapse
Affiliation(s)
- Michaela E Copp
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Hannon L Brown
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC
| | - Kirti R Nimmala
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL
| | - John A Collins
- Department of Orthopedic Surgery, Thomas Jefferson University
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC
| |
Collapse
|
30
|
Franco-Trepat E, Alonso-Pérez A, Guillán-Fresco M, López-Fagúndez M, Pazos-Pérez A, Crespo-Golmar A, Belén Bravo S, López-López V, Jorge-Mora A, Cerón-Carrasco JP, Lois Iglesias A, Gómez R. β Boswellic Acid Blocks Articular Innate Immune Responses: An In Silico and In Vitro Approach to Traditional Medicine. Antioxidants (Basel) 2023; 12:371. [PMID: 36829930 PMCID: PMC9952103 DOI: 10.3390/antiox12020371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is hallmarked as a silent progressive rheumatic disease of the whole joint. The accumulation of inflammatory and catabolic factors such as IL6, TNFα, and COX2 drives the OA pathophysiology into cartilage degradation, synovia inflammation, and bone destruction. There is no clinical available OA treatment. Although traditional ayurvedic medicine has been using Boswellia serrata extracts (BSE) as an antirheumatic treatment for a millennium, none of the BSE components have been clinically approved. Recently, β boswellic acid (BBA) has been shown to reduce in vivo OA-cartilage loss through an unknown mechanism. We used computational pharmacology, proteomics, transcriptomics, and metabolomics to present solid evidence of BBA therapeutic properties in mouse and primary human OA joint cells. Specifically, BBA binds to the innate immune receptor Toll-like Receptor 4 (TLR4) complex and inhibits both TLR4 and Interleukin 1 Receptor (IL1R) signaling in OA chondrocytes, osteoblasts, and synoviocytes. Moreover, BBA inhibition of TLR4/IL1R downregulated reactive oxygen species (ROS) synthesis and MAPK p38/NFκB, NLRP3, IFNαβ, TNF, and ECM-related pathways. Altogether, we present a solid bulk of evidence that BBA blocks OA innate immune responses and could be transferred into the clinic as an alimentary supplement or as a therapeutic tool after clinical trial evaluations.
Collapse
Affiliation(s)
- Eloi Franco-Trepat
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Ana Alonso-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - María Guillán-Fresco
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Miriam López-Fagúndez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Andrés Pazos-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Antía Crespo-Golmar
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Susana Belén Bravo
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Verónica López-López
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Alberto Jorge-Mora
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - José P. Cerón-Carrasco
- Centro Universitario de la Defensa, Universidad Politécnica de Cartagena, C/Coronel López Peña S/N, Base Aérea de San Javier, Santiago de La Ribera, 30720 Murcia, Spain
| | - Ana Lois Iglesias
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| | - Rodolfo Gómez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain
| |
Collapse
|
31
|
Niemann M, Ort M, Lauterbach L, Streitz M, Wilhelm A, Grütz G, Fleckenstein FN, Graef F, Blankenstein A, Reinke S, Stöckle U, Perka C, Duda GN, Geißler S, Winkler T, Maleitzke T. Individual immune cell and cytokine profiles determine platelet-rich plasma composition. Arthritis Res Ther 2023; 25:6. [PMID: 36627721 PMCID: PMC9830842 DOI: 10.1186/s13075-022-02969-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 12/02/2022] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE Platelet-rich plasma (PRP) therapy is increasingly popular to treat musculoskeletal diseases, including tendinopathies and osteoarthritis (OA). To date, it remains unclear to which extent PRP compositions are determined by the immune cell and cytokine profile of individuals or by the preparation method. To investigate this, we compared leukocyte and cytokine distributions of different PRP products to donor blood samples and assessed the effect of pro-inflammatory cytokines on chondrocytes. DESIGN For each of three PRP preparations (ACP®, Angel™, and nSTRIDE® APS), products were derived using whole blood samples from twelve healthy donors. The cellular composition of PRP products was analyzed by flow cytometry using DURAClone antibody panels (DURAClone IM Phenotyping Basic and DURAClone IM T Cell Subsets). The MESO QuickPlex SQ 120 system was used to assess cytokine profiles (V-PLEX Proinflammatory Panel 1 Human Kit, Meso Scale Discovery). Primary human chondrocyte 2D and 3D in vitro cultures were exposed to recombinant IFN-γ and TNF-α. Proliferation and chondrogenic differentiation were quantitatively assessed. RESULTS All three PRP products showed elevated portions of leukocytes compared to baseline levels in donor blood. Furthermore, the pro-inflammatory cytokines IFN-γ and TNF-α were significantly increased in nSTRIDE® APS samples compared to donor blood and other PRP products. The characteristics of all other cytokines and immune cells from the donor blood, including pro-inflammatory T cell subsets, were maintained in all PRP products. Chondrocyte proliferation was impaired by IFN-γ and enhanced by TNF-α treatment. Differentiation and cartilage formation were compromised upon treatment with both cytokines, resulting in altered messenger ribonucleic acid (mRNA) expression of collagen type 1A1 (COL1A1), COL2A1, and aggrecan (ACAN) as well as reduced proteoglycan content. CONCLUSIONS Individuals with elevated levels of cells with pro-inflammatory properties maintain these in the final PRP products. The concentration of pro-inflammatory cytokines strongly varies between PRP products. These observations may help to unravel the previously described heterogeneous response to PRP in OA therapy, especially as IFN-γ and TNF-α impacted primary chondrocyte proliferation and their characteristic gene expression profile. Both the individual's immune profile and the concentration method appear to impact the final PRP product. TRIAL REGISTRATION This study was prospectively registered in the Deutsches Register Klinischer Studien (DRKS) on 4 November 2021 (registration number DRKS00026175).
Collapse
Affiliation(s)
- Marcel Niemann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Melanie Ort
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany.
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany.
| | - Luis Lauterbach
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Mathias Streitz
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Andreas Wilhelm
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Gerald Grütz
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Florian N Fleckenstein
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Diagnostic and Interventional Radiology, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Frank Graef
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Antje Blankenstein
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Simon Reinke
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Ulrich Stöckle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Carsten Perka
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Georg N Duda
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Sven Geißler
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Tobias Winkler
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Tazio Maleitzke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| |
Collapse
|
32
|
Dhanabalan KM, Dravid AA, Agarwal S, Sharath RK, Padmanabhan AK, Agarwal R. Intra-articular injection of rapamycin microparticles prevent senescence and effectively treat osteoarthritis. Bioeng Transl Med 2023; 8:e10298. [PMID: 36684078 PMCID: PMC9842044 DOI: 10.1002/btm2.10298] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 01/25/2023] Open
Abstract
Trauma to the knee joint is associated with significant cartilage degeneration and erosion of subchondral bone, which eventually leads to osteoarthritis (OA), resulting in substantial morbidity and healthcare burden. With no disease-modifying drugs in clinics, the current standard of care focuses on symptomatic relief and viscosupplementation. Modulation of autophagy and targeting senescence pathways are emerging as potential treatment strategies. Rapamycin has shown promise in OA disease amelioration by autophagy upregulation, yet its clinical use is hindered by difficulties in achieving therapeutic concentrations, necessitating multiple weekly injections. Rapamycin-loaded in poly(lactic-co-glycolic acid) microparticles (RMPs) induced autophagy, prevented senescence, and sustained sulphated glycosaminoglycans production in primary human articular chondrocytes from OA patients. RMPs were potent, nontoxic, and exhibited high retention time (up to 35 days) in mice joints. Intra-articular delivery of RMPs effectively mitigated cartilage damage and inflammation in surgery-induced OA when administered as a prophylactic or therapeutic regimen. Together, the study demonstrates the feasibility of using RMPs as a potential clinically translatable therapy to prevent the progression of post-traumatic OA.
Collapse
Affiliation(s)
- Kaamini M. Dhanabalan
- Centre for BioSystems Science and EngineeringIndian Institute of ScienceBengaluruIndia
| | - Ameya A. Dravid
- Centre for BioSystems Science and EngineeringIndian Institute of ScienceBengaluruIndia
| | - Smriti Agarwal
- Centre for BioSystems Science and EngineeringIndian Institute of ScienceBengaluruIndia
| | | | | | - Rachit Agarwal
- Centre for BioSystems Science and EngineeringIndian Institute of ScienceBengaluruIndia
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Imaging plays a pivotal role for diagnosis, follow-up and stratification of osteoarthritis patients in clinical trials and research. We aim to present an overview of currently available and emerging imaging techniques for osteoarthritis assessment and provide insight into relevant benefits and pitfalls of the different modalities. RECENT FINDINGS Although radiography is considered sufficient for a structural diagnosis of osteoarthritis and is commonly used to define eligibility of patients for participation in clinical trials, it has inherent limitations based on the projectional nature of the technique and inherent challenges regarding reproducibility in longitudinal assessment. MRI has changed our understanding of the disease from 'wear and tear' of cartilage to a whole organ disorder. MRI assessment of structural changes of osteoarthritis includes semi-quantitative, quantitative and compositional evaluation. Ultrasound is helpful in evaluating the degree of synovitis and has value in the assessment particularly of the patella-femoral joint. Recent development of computed tomography technology including weight-bearing systems has led to broader application of this technology in a research context. SUMMARY Advances in MRI technology have resulted in a significant improvement in understanding osteoarthritis as a multitissue disease.
Collapse
Affiliation(s)
- Majid Chalian
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Frank W Roemer
- Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine
| | - Ali Guermazi
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine
- Department of Radiology, VA Boston Healthcare System, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Ding L, Liao T, Yang N, Wei Y, Xing R, Wu P, Li X, Mao J, Wang P. Chrysin ameliorates synovitis and fibrosis of osteoarthritic fibroblast-like synoviocytes in rats through PERK/TXNIP/NLRP3 signaling. Front Pharmacol 2023; 14:1170243. [PMID: 37021049 PMCID: PMC10067567 DOI: 10.3389/fphar.2023.1170243] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
Objective: Synovitis and fibrosis are common pathological features of knee osteoarthritis (KOA). The interaction of synovitis and fibrosis can promote KOA progression. Chrysin (CHR), a natural flavonoid, may treat inflammation and prevent fibrosis. However, the effect and mechanism of CHR in KOA synovitis and fibrosis remains unclear. Methods: The KOA model was established in male SD rats by anterior cruciate ligament transection (ACLT), and histological analysis was used to evaluate synovitis and fibrosis. IL-6, IL-1β and TNF-α mRNA expression in synovial tissue was measured by qRT‒PCR. Immunohistochemistry (IHC) was performed to detect GRP78, ATF-6 and TXNIP expression in vivo. Synovial fibroblasts (SFs) were treated with TGF-β1 to stimulate the inflammatory response and fibrosis. CCK-8 assays were used to detect the viability of CHR-treated SFs. The IL-1β level was detected by immunofluorescence analysis. Coimmunoprecipitation (Co-IP) and double immunofluorescence colocalization were used to detect the physiological interaction between TXNIP and NLRP3. The expression of fibrosis-related mediators and PERK/TXNIP/NLRP3 signaling molecules was detected by western blotting and qRT-PCR. Results: Four weeks after CHR treatment, pathological sections and associated scores showed that CHR improved synovitis and fibrosis in the ACLT model. In vitro, CHR attenuated the TGF-β1-induced inflammatory response and fibrosis in SFs. Moreover, CHR suppressed the expression of synovial fibrosis markers and PERK/TXNIP/NLRP3 signaling molecules in the synovial tissue of rats with ACLT and cultured SFs. More importantly, we found that CHR inhibited TXNIP-NLRP3 interactions in TGF-β-induced SFs. Conclusion: Our findings indicate that CHR can ameliorate synovitis and fibrosis in KOA. The underlying mechanism may be related to the PERK/TXNIP/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Liang Ding
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Taiyang Liao
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nan Yang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yibao Wei
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Runlin Xing
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Peng Wu
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaochen Li
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jun Mao
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- *Correspondence: Jun Mao, ; Peimin Wang,
| | - Peimin Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- *Correspondence: Jun Mao, ; Peimin Wang,
| |
Collapse
|
35
|
Peng JW, Gu YY, Wei J, Sun Y, Zhu CL, Zhang L, Song Y, Chen L, Chen X, Wang Q, Zhang HL. LncRNA MEG3-TRPV1 signaling regulates chronic inflammatory pain in rats. Mol Pain 2022; 18:17448069221144246. [PMID: 36424837 PMCID: PMC9726848 DOI: 10.1177/17448069221144246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Osteoarthritis (OA) is a common osteoarthropathy with chronic inflammatory pain as the core symptom in middle-aged and elderly people. LncRNA MEG3 (Maternally expressed gene 3) is involved in the development of OA via regulation of angiogenesis, which causes the activation and overexpression of transient receptor potential vanilloid type-1 (TRPV1). In this study, we investigated the mechanism of MEG3-TRPV1 signaling in chronic inflammatory pain (CIP) of rat model. Chronic inflammatory pain was modeled using subcutaneous microinjection of complete Freund's adjuvant (CFA) into the left hind paw of rats. We showed that TRPV1 mRNA and protein were significantly increased, while MEG3 mRNA was significantly decreased, in the DRG and SDH of CFA-induced rats. In addition, intrathecal injection of MEG3-overexpressing lentivirus significantly downregulated TRPV1 expression and alleviated chronic inflammatory pain in CFA-induced rats. Treatment with a TRPV1 antagonist also significantly relieved chronic inflammatory pain in CFA-induced rats. In general, our results reveal that MEG3 alleviates chronic inflammatory pain by downregulating TRPV1 expression. These findings may provide new therapeutic targets in the treatment of patients with OA.
Collapse
Affiliation(s)
- Jing-Wei Peng
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China,Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Yin-Yin Gu
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Jia Wei
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Ye Sun
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Chun-Long Zhu
- Department of Traditional Chinese Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Ling Zhang
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Yu Song
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Long Chen
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Xia Chen
- Department of Anesthesiology, Children’s Hospital of Soochow University, Suzhou, China
| | - Qian Wang
- Department of Anesthesiology, Children’s Hospital of Soochow University, Suzhou, China,Qian Wang, Department of Anesthesiology, Children’s Hospital of Soochow University, Suzhou 215123, China.
| | - Hai-Long Zhang
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China,Hai-Long Zhang, Center for Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
36
|
Roemer FW, Jansen M, Marijnissen ACA, Guermazi A, Heiss R, Maschek S, Lalande A, Blanco FJ, Berenbaum F, van de Stadt LA, Kloppenburg M, Haugen IK, Ladel CH, Bacardit J, Wisser A, Eckstein F, Lafeber FPJG, Weinans HH, Wirth W. Structural tissue damage and 24-month progression of semi-quantitative MRI biomarkers of knee osteoarthritis in the IMI-APPROACH cohort. BMC Musculoskelet Disord 2022; 23:988. [PMID: 36397054 PMCID: PMC9670371 DOI: 10.1186/s12891-022-05926-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The IMI-APPROACH cohort is an exploratory, 5-centre, 2-year prospective follow-up study of knee osteoarthritis (OA). Aim was to describe baseline multi-tissue semiquantitative MRI evaluation of index knees and to describe change for different MRI features based on number of subregion-approaches and change in maximum grades over a 24-month period. METHODS MRIs were acquired using 1.5 T or 3 T MRI systems and assessed using the semi-quantitative MRI OA Knee Scoring (MOAKS) system. MRIs were read at baseline and 24-months for cartilage damage, bone marrow lesions (BML), osteophytes, meniscal damage and extrusion, and Hoffa- and effusion-synovitis. In descriptive fashion, the frequencies of MRI features at baseline and change in these imaging biomarkers over time are presented for the entire sample in a subregional and maximum score approach for most features. Differences between knees without and with structural radiographic (R) OA are analyzed in addition. RESULTS Two hundred eighty-nine participants had readable baseline MRI examinations. Mean age was 66.6 ± 7.1 years and participants had a mean BMI of 28.1 ± 5.3 kg/m2. The majority (55.3%) of included knees had radiographic OA. Any change in total cartilage MOAKS score was observed in 53.1% considering full-grade changes only, and in 73.9% including full-grade and within-grade changes. Any medial cartilage progression was seen in 23.9% and any lateral progression on 22.1%. While for the medial and lateral compartments numbers of subregions with improvement and worsening of BMLs were very similar, for the PFJ more improvement was observed compared to worsening (15.5% vs. 9.0%). Including within grade changes, the number of knees showing BML worsening increased from 42.2% to 55.6%. While for some features 24-months change was rare, frequency of change was much more common in knees with vs. without ROA (e.g. worsening of total MOAKS score cartilage in 68.4% of ROA knees vs. 36.7% of no-ROA knees, and 60.7% vs. 21.8% for an increase in maximum BML score per knee). CONCLUSIONS A wide range of MRI-detected structural pathologies was present in the IMI-APPROACH cohort. Baseline prevalence and change of features was substantially more common in the ROA subgroup compared to the knees without ROA. TRIAL REGISTRATION Clinicaltrials.gov identification: NCT03883568.
Collapse
Grants
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- 115770 The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007-2013) and EFPIA companies' in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- The research leading to these results have received support from the Innovative Medicines Initiative Joint Undertaking under Grant Agreement no 115770, resources of which are composed of financial contribution from the European Union’s Seventh Framework Programme (FP7/2007-2013) and EFPIA companies’ in kind contribution. See www.imi.europa.eu and www.approachproject.eu.
- Friedrich-Alexander-Universität Erlangen-Nürnberg (1041)
Collapse
Affiliation(s)
- Frank W. Roemer
- Department of Radiology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine, Boston, MA USA
| | - Mylène Jansen
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Ali Guermazi
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine, Boston, MA USA
- Department of Radiology, VA Boston Healthcare System, West Roxbury, MA USA
| | - Rafael Heiss
- Department of Radiology, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 3, 91054 Erlangen, Germany
| | - Susanne Maschek
- Chondrometrics GmbH, Freilassing, Germany
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| | - Agnes Lalande
- Institut de Recherches Internationales Servier, Suresnes, France
| | | | - Francis Berenbaum
- Department of Rheumatology, AP-HP Saint-Antoine Hospital, Paris, France
- INSERM, Sorbonne University, Paris, France
| | - Lotte A. van de Stadt
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Margreet Kloppenburg
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
- Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ida K. Haugen
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | | | - Jaume Bacardit
- School of Computing, Newcastle University, Newcastle, UK
| | - Anna Wisser
- Chondrometrics GmbH, Freilassing, Germany
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Ludwig Boltzmann Inst. for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| | - Felix Eckstein
- Chondrometrics GmbH, Freilassing, Germany
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Ludwig Boltzmann Inst. for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| | | | | | - Wolfgang Wirth
- Chondrometrics GmbH, Freilassing, Germany
- Department of Imaging & Functional Musculoskeletal Research, Institute of Anatomy & Cell Biology, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
- Ludwig Boltzmann Inst. for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg & Nuremberg, Salzburg, Austria
| |
Collapse
|
37
|
Rios JL, Sapède D, Djouad F, Rapp AE, Lang A, Larkin J, Ladel C, Mobasheri A. Animal Models of Osteoarthritis Part 1-Preclinical Small Animal Models: Challenges and Opportunities for Drug Development. Curr Protoc 2022; 2:e596. [PMID: 36342311 DOI: 10.1002/cpz1.596] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis and a major source of pain and disability in the adult population. There is a significant unmet medical need for the development of effective pharmacological therapies for the treatment of OA. In addition to spontaneously occurring animal models of OA, many experimental animal models have been developed to provide insights into mechanisms of pathogenesis and progression. Many of these animal models are also being used in the drug development pipeline. Here, we provide an overview of commonly used and emerging preclinical small animal models of OA and highlight the strengths and limitations of small animal models in the context of translational drug development. There is limited information in the published literature regarding the technical reliability of these small animal models and their ability to accurately predict clinical drug development outcomes. The cost and complexity of the available models however is an important consideration for pharmaceutical companies, biotechnology startups, and contract research organizations wishing to incorporate preclinical models in target validation, discovery, and development pipelines. Further considerations relevant to industry include timelines, methods of induction, the key issue of reproducibility, and appropriate outcome measures needed to objectively assess outcomes of experimental therapeutics. Preclinical small animal models are indispensable tools that will shine some light on the pathogenesis of OA and its molecular endotypes in the context of drug development. This paper will focus on small animal models used in preclinical OA research. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Jaqueline Lourdes Rios
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Percuros BV, Leiden, The Netherlands
| | - Dora Sapède
- IRMB, Université de Montpellier, INSERM, Montpellier, France
| | - Farida Djouad
- IRMB, Université de Montpellier, INSERM, Montpellier, France
| | - Anna E Rapp
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopaedics (Friedrichsheim), University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Annemarie Lang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | | | | | - Ali Mobasheri
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Departments of Orthopaedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Liege, Belgium
| |
Collapse
|
38
|
Patterns of progression differ between Kellgren-Lawrence 2 and 3 knees fulfilling different definitions of a cartilage-meniscus phenotype in the Foundation for National Institutes of Health Osteoarthritis Biomarkers study (FNIH). OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100284. [DOI: 10.1016/j.ocarto.2022.100284] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/23/2022] [Accepted: 06/02/2022] [Indexed: 01/18/2023] Open
|
39
|
Li D, Ruan G, Zhang Y, Zhao Y, Zhu Z, Ou Q, Huang H, Chen J, Han W, Tang S, Li J, Wang L, Chen T, Bai X, Cai D, Ding C. Metformin attenuates osteoarthritis by targeting chondrocytes, synovial macrophages and adipocytes. Rheumatology (Oxford) 2022; 62:1652-1661. [PMID: 35984286 DOI: 10.1093/rheumatology/keac467] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To investigate the therapeutic effect and mechanism of metformin on knee osteoarthritis (OA) in normal diet (ND) mice or high-fat diet (HFD)-induced obese mice. METHODS Destabilization of the medial meniscus surgery was performed in ND mice or HFD mice, and metformin was administrated in drinking water or not. The changes of OA joint structure, infiltration and polarization of synovial macrophages and circulating and local levels of leptin and adiponectin were evaluated. In vitro, the effects of metformin on chondrocytes and macrophages, and of conditioned mediums derived from mouse abdominal fat on murine chondrogenic cell line ATDC5 and murine macrophage cell line RAW264.7, were detected. RESULTS Metformin showed protective effects on OA, characterized by reductions on OARSI score (2.00, 95% CI [1.15-2.86] for ND mice and 3.17, 95% CI [2.37-3.96] for HFD mice) and synovitis score (1.17, 95% CI [0.27-2.06] for ND mice and 2.50, 95% CI [1.49-3.51] for HFD mice) after 10 weeks of treatment, and the effects were more significant in HFD mice than in ND mice. Mechanistically, in addition to decreasing apoptosis and matrix-degrading enzymes expression in chondrocytes as well as infiltration and pro-inflammatory differentiation of synovial macrophages, metformin reduced leptin secretion by adipose tissue in HFD mice. CONCLUSIONS Metformin protects against knee OA which could be through reducing apoptosis and catabolism of chondrocytes, and suppressing infiltration and pro-inflammatory polarization of synovial macrophages. For obese mice, metformin has a greater protective effect in knee OA additionally through reducing leptin secretion from adipose tissue.
Collapse
Affiliation(s)
- Delong Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.,Department of Orthopedics, Academy of Orthopedics Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Guangfeng Ruan
- Clinical Research Centre, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yang Zhao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qianhua Ou
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Hong Huang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jieli Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Weiyu Han
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Su'an Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jia Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Liang Wang
- Department of Orthopedics, Academy of Orthopedics Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Tianyu Chen
- Department of Orthopedics, Academy of Orthopedics Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Xiaochun Bai
- Department of Orthopedics, Academy of Orthopedics Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China.,Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Daozhang Cai
- Department of Orthopedics, Academy of Orthopedics Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.,Menzies Institute for Medical Research, University of Tasmania, Hobart, 7000, Australia
| |
Collapse
|
40
|
Roemer FW, Guermazi A, Demehri S, Wirth W, Kijowski R. Imaging in Osteoarthritis. Osteoarthritis Cartilage 2022; 30:913-934. [PMID: 34560261 DOI: 10.1016/j.joca.2021.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/22/2021] [Accepted: 04/28/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is the most frequent form of arthritis with major implications on both individual and public health care levels. The field of joint imaging, and particularly magnetic resonance imaging (MRI), has evolved rapidly due to the application of technical advances to the field of clinical research. This narrative review will provide an introduction to the different aspects of OA imaging aimed at an audience of scientists, clinicians, students, industry employees, and others who are interested in OA but who do not necessarily focus on OA. The current role of radiography and recent advances in measuring joint space width will be discussed. The status of cartilage morphology assessment and evaluation of cartilage biochemical composition will be presented. Advances in quantitative three-dimensional morphologic cartilage assessment and semi-quantitative whole-organ assessment of OA will be reviewed. Although MRI has evolved as the most important imaging method used in OA research, other modalities such as ultrasound, computed tomography, and metabolic imaging play a complementary role and will also be discussed.
Collapse
Affiliation(s)
- F W Roemer
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine, FGH Building, 3rd Floor, 820 Harrison Ave, Boston, MA, 02118, USA; Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Maximiliansplatz 3, Erlangen, 91054, Germany.
| | - A Guermazi
- Quantitative Imaging Center, Department of Radiology, Boston University School of Medicine, FGH Building, 3rd Floor, 820 Harrison Ave, Boston, MA, 02118, USA; Department of Radiology, VA Boston Healthcare System, 1400 VFW Pkwy, Suite 1B105, West Roxbury, MA, 02132, USA
| | - S Demehri
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 600 N. Wolf Street, Park 311, Baltimore, MD, 21287, USA
| | - W Wirth
- Institute of Anatomy, Paracelsus Medical University Salzburg, Salzburg, Austria, Nüremberg, Germany; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University Salzburg, Strubergasse 21, 5020, Salzburg, Austria; Chondrometrics, GmbH, Freilassing, Germany
| | - R Kijowski
- Department of Radiology, New York University Grossmann School of Medicine, 550 1st Avenue, 3nd Floor, New York, NY, 10016, USA
| |
Collapse
|
41
|
Shahid A, Inam‐Ur‐Raheem M, Iahtisham‐Ul‐Haq , Nawaz MY, Rashid MH, Oz F, Proestos C, Aadil RM. Diet and lifestyle modifications: An update on non‐pharmacological approach in the management of osteoarthritis. J FOOD PROCESS PRES 2022. [DOI: 10.1111/jfpp.16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Arashi Shahid
- National Institute of Food Science and Technology University of Agriculture Faisalabad Pakistan
| | - Muhammad Inam‐Ur‐Raheem
- National Institute of Food Science and Technology University of Agriculture Faisalabad Pakistan
| | - Iahtisham‐Ul‐Haq
- Kauser Abdulla Malik School of Life Sciences Forman Christian College (A Chartered University) Punjab Pakistan
| | - Muhammad Yasir Nawaz
- Department of Pathology Faculty of Veterinary Science, University of Agriculture Faisalabad Faisalabad Pakistan
| | - Muhammad Hamdan Rashid
- National Institute of Food Science and Technology University of Agriculture Faisalabad Pakistan
| | - Fatih Oz
- Department of Food Engineering, Faculty of Agriculture Ataturk University Erzurum Turkey
| | - Charalampos Proestos
- Laboratory of Food Chemistry, Department of Chemistry National and Kapodistrian University of Athens Zografou Athens Greece
| | - Rana Muhammad Aadil
- National Institute of Food Science and Technology University of Agriculture Faisalabad Pakistan
| |
Collapse
|
42
|
Ruan G, Yuan S, Lou A, Mo Y, Qu Y, Guo D, Guan S, Zhang Y, Lan X, Luo J, Mei Y, Zhang H, Wu W, Dai L, Yu Q, Cai X, Ding C. Can metformin relieve tibiofemoral cartilage volume loss and knee symptoms in overweight knee osteoarthritis patients? Study protocol for a randomized, double-blind, and placebo-controlled trial. BMC Musculoskelet Disord 2022; 23:486. [PMID: 35598008 PMCID: PMC9124394 DOI: 10.1186/s12891-022-05434-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 05/13/2022] [Indexed: 01/07/2023] Open
Abstract
Background Osteoarthritis (OA) is the most common joint disease, and is most frequently seen in the knees. However, there is no effective therapy to relieve the progression of knee OA. Metformin is a safe, well-tolerated oral medication that is extensively used as first-line therapy for type 2 diabetes. Previous observational studies and basic researches suggested that metformin may have protective effects on knee OA, which needs to be verified by clinical trials. This study, therefore, aims to examine the effects of metformin versus placebo on knee cartilage volume loss and knee symptoms in overweight knee OA patients by a randomized controlled trial over 24 months. Methods This protocol describes a multicenter, randomized, double-blind, and placebo-controlled clinical trial aiming to recruit 262 overweight knee OA patients. Participants will be randomly allocated to the two arms of the study, receiving metformin hydrochloride sustained-release tablets or identical inert placebo for 24 months (start from 0.5 g/day for the first 2 weeks, and increase to 1 g/day for the second 2 weeks, and further increase to 2 g/day for the remaining period if tolerated). Primary outcomes will be changes in tibiofemoral cartilage volume and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score over 24 months. Secondary outcomes will be changes in visual analogue scale (VAS) knee pain, tibiofemoral cartilage defects, effusion-synovitis volume, and tibiofemoral bone marrow lesions maximum size over 24 months. The primary analyses will be intention-to-treat analyses of primary and secondary outcomes. Per-protocol analyses will be performed as the secondary analyses. Discussion If metformin is proved to slow knee cartilage volume loss and to relieve knee symptoms among overweight knee OA patients, it will have the potential to become a disease modifying drug for knee OA. Metformin is a convenient intervention with low cost, and its potential effects on slowing down the structural progression and relieving the symptoms of knee OA would effectively reduce the disease burden worldwide. Trial registration ClinicalTrials. gov NCT05034029. Registered on 30 Sept 2021.
Collapse
Affiliation(s)
- Guangfeng Ruan
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shiwen Yuan
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Aiju Lou
- Department of Rheumatology and Immunology, Liwan Central Hospital of Guangzhou, Guangzhou, Guangdong, China
| | - Yingqian Mo
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuan Qu
- Rheumatology and Clinical Immunology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Dongmei Guo
- Department of Rheumatology, Foshan First People's Hospital, Foshan, Guangdong, China
| | - Shangqi Guan
- Department of Rheumatology, Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoyong Lan
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jun Luo
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yifang Mei
- Department of Rheumatology, Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Hongwei Zhang
- Department of Rheumatology, Foshan First People's Hospital, Foshan, Guangdong, China
| | - Weirong Wu
- Department of Rheumatology and Immunology, Liwan Central Hospital of Guangzhou, Guangzhou, Guangdong, China
| | - Lie Dai
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qinghong Yu
- Rheumatology and Clinical Immunology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoyan Cai
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| | - Changhai Ding
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China. .,Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
43
|
Oo WM, Hunter DJ. Repurposed and investigational disease-modifying drugs in osteoarthritis (DMOADs). Ther Adv Musculoskelet Dis 2022; 14:1759720X221090297. [PMID: 35619876 PMCID: PMC9128067 DOI: 10.1177/1759720x221090297] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
In spite of a major public health burden with increasing prevalence, current osteoarthritis (OA) management is largely palliative with an unmet need for effective treatment. Both industry and academic researchers have invested a vast amount of time and financial expense to discover the first diseasing-modifying osteoarthritis drugs (DMOADs), with no regulatory success so far. In this narrative review, we discuss repurposed drugs as well as investigational agents which have progressed into phase II and III clinical trials based on three principal endotypes: bone-driven, synovitis-driven and cartilage-driven. Then, we will briefly describe the recent failures and lessons learned, promising findings from predefined post hoc analyses and insights gained, novel methodologies to enhance future success and steps underway to overcome regulatory hurdles.
Collapse
Affiliation(s)
- Win Min Oo
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar
| | - David J. Hunter
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia
| |
Collapse
|
44
|
Dual functions of microRNA-17 in maintaining cartilage homeostasis and protection against osteoarthritis. Nat Commun 2022; 13:2447. [PMID: 35508470 PMCID: PMC9068604 DOI: 10.1038/s41467-022-30119-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/14/2022] [Indexed: 12/16/2022] Open
Abstract
Damaged hyaline cartilage has no capacity for self-healing, making osteoarthritis (OA) "difficult-to-treat". Cartilage destruction is central to OA patho-etiology and is mediated by matrix degrading enzymes. Here we report decreased expression of miR-17 in osteoarthritic chondrocytes and its deficiency contributes to OA progression. Supplementation of exogenous miR-17 or its endogenous induction by growth differentiation factor 5, effectively prevented OA by simultaneously targeting pathological catabolic factors including matrix metallopeptidase-3/13 (MMP3/13), aggrecanase-2 (ADAMTS5), and nitric oxide synthase-2 (NOS2). Single-cell RNA sequencing of hyaline cartilage revealed two distinct superficial chondrocyte populations (C1/C2). C1 expressed physiological catabolic factors including MMP2, and C2 carries synovial features, together with C3 in the middle zone. MiR-17 is highly expressed in both superficial and middle chondrocytes under physiological conditions, and maintains the physiological catabolic and anabolic balance potentially by restricting HIF-1α signaling. Together, this study identified dual functions of miR-17 in maintaining cartilage homeostasis and prevention of OA.
Collapse
|
45
|
Roemer FW, Felson DT, Stefanik JJ, Rabasa G, Wang N, Crema MD, Neogi T, Nevitt MC, Torner J, Lewis CE, Peloquin C, Guermazi A. Heterogeneity of cartilage damage in Kellgren and Lawrence grade 2 and 3 knees: the MOST study. Osteoarthritis Cartilage 2022; 30:714-723. [PMID: 35202808 PMCID: PMC9433455 DOI: 10.1016/j.joca.2022.02.614] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Eligibility for clinical trials in osteoarthritis (OA) is usually limited to Kellgren-Lawrence (KL) grades 2 and 3 knees. Our aim was to describe the prevalence and severity of cartilage damage in KL 2 and 3 knees by compartment and articular subregion. DESIGN The Multicenter Osteoarthritis (MOST) study is a cohort study of individuals with or at risk for knee OA. All baseline MRIs with radiographic disease severity KL2 and 3 were included. Knee MRIs were read for cartilage damage in 14 subregions. We determined the frequencies of no, any and widespread full-thickness cartilage damage by knee compartment, and the prevalence of any cartilage damage in 14 articular subregions. RESULTS 665 knees from 665 participants were included (mean age 63.8 ± 7.9 years, 66.5% women). 372 knees were KL2 and 293 knees were KL3. There was no cartilage damage in 78 (21.0%) medial tibio-femoral joint (TFJ), 157 (42.2%) lateral TFJ and 62 (16.7%) patello-femoral joint (PFJ) compartments of KL2 knees, and 17 (5.8%), 115 (39.3%) and 35 (12.0%) compartments, respectively, of KL3 knees. There was widespread full-thickness damage in 94 (25.3%) medial TFJ, 36 (9.7%) lateral TFJ and 176 (47.3%) PFJ compartments of KL2 knees, and 217 (74.1%), 70 (23.9%) and 104 (35.5%) compartments, respectively, of KL3 knees. The subregions most likely to have any damage were central medial femur (80.5%), medial patella (69.8%) and central medial tibia (69.9). CONCLUSIONS KL2 and KL3 knees vary greatly in cartilage morphology. Heterogeneity in the prevalence, severity and location of cartilage damage in in KL2 and 3 knees should be considered when planning disease modifying trials for knee OA.
Collapse
Affiliation(s)
- F W Roemer
- Quantitative Imaging Center (QIC), Department of Radiology, Boston University School of Medicine, Boston, MA, USA; Department of Radiology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| | - D T Felson
- Department of Medicine, Section of Rheumatology, Boston University School of Medicine, Boston, MA, USA
| | - J J Stefanik
- Department of Physical Therapy, Movement and Rehabilitation Sciences, Northeastern University, Boston, MA, USA
| | - G Rabasa
- Department of Medicine, Section of Rheumatology, Boston University School of Medicine, Boston, MA, USA
| | - N Wang
- Department of Medicine, Section of Rheumatology, Boston University School of Medicine, Boston, MA, USA
| | - M D Crema
- Quantitative Imaging Center (QIC), Department of Radiology, Boston University School of Medicine, Boston, MA, USA; Institute of Sports Imaging, French National Institute of Sports (INSEP), Paris, France
| | - T Neogi
- Department of Medicine, Section of Rheumatology, Boston University School of Medicine, Boston, MA, USA
| | - M C Nevitt
- Department of Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, CA, USA
| | - J Torner
- Department of Epidemiology, University of Iowa, Iowa City, IA, USA
| | - C E Lewis
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - C Peloquin
- Department of Medicine, Section of Rheumatology, Boston University School of Medicine, Boston, MA, USA
| | - A Guermazi
- Quantitative Imaging Center (QIC), Department of Radiology, Boston University School of Medicine, Boston, MA, USA; Department of Radiology, VA Boston Healthcare System, West Roxbury, MA, USA
| |
Collapse
|
46
|
|
47
|
Gao J, Xia Z, Mary HB, Joseph J, Luo JN, Joshi N. Overcoming barriers for intra-articular delivery of disease-modifying osteoarthritis drugs. Trends Pharmacol Sci 2022; 43:171-187. [PMID: 35086691 PMCID: PMC8840969 DOI: 10.1016/j.tips.2021.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/27/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023]
Abstract
Despite four decades of research in intra-articular drug delivery systems (DDS) and two decades of advances in disease-modifying osteoarthritis drugs (DMOADs), there is still no clinically available disease-modifying therapy for osteoarthritis (OA). Multiple barriers compromise intra-articular DMOAD delivery. Although multiple exciting approaches have been developed to overcome these barriers, there are still outstanding questions. We make several recommendations that can help in fully overcoming these barriers. Considering OA heterogeneity, we also propose a patient-centered, bottom-up workflow to guide preclinical development of DDS-based intra-articular DMOAD therapies. Overall, we expect this review to inspire paradigm-shifting innovations for developing next-generation DDS that can enable clinical translation of intra-articular DMOADs.
Collapse
Affiliation(s)
- Jingjing Gao
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Ziting Xia
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Helna B Mary
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - John Joseph
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - James N Luo
- Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Nitin Joshi
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Kim BH, Bae HC, Wang SY, Jang BS, Chang JH, Chie EK, Yi HS, Kwon J, Han HS, Kim HJ. Low-dose irradiation could mitigate osteoarthritis progression via anti-inflammatory action that modulates mitochondrial function. Radiother Oncol 2022; 170:231-241. [DOI: 10.1016/j.radonc.2022.02.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 12/15/2022]
|
49
|
Lee SY, Wong PF, Jamal J, Roebuck MM. Naturally-derived endoplasmic reticulum stress inhibitors for osteoarthritis? Eur J Pharmacol 2022; 922:174903. [DOI: 10.1016/j.ejphar.2022.174903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/22/2022] [Accepted: 03/17/2022] [Indexed: 01/15/2023]
|
50
|
Kim SE, Choi SJ, Park K, Kim HJ, Song GG, Jung JH. Intra-Articular Injection of Rebamipide-Loaded Nanoparticles Attenuate Disease Progression and Joint Destruction in Osteoarthritis Rat Model: A Pilot Study. Cartilage 2022; 13:19476035211069250. [PMID: 35118894 PMCID: PMC9137306 DOI: 10.1177/19476035211069250] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Rebamipide has antioxidant effects and is a drug with a local rather than systemic mechanism of action. Oxidative stress and inflammation in chondrocytes are the major factors contributing to the development and progression of osteoarthritis (OA). Since OA is mainly developed in weight bearing or overused joints, the locally sustained therapy is effective for targeting inflammatory component of OA. We investigated the effects of intra-articular injection of rebamipide loaded nanoparticles (NPs) in OA rat model. DESIGN We fabricated rebamipide-loaded methoxy poly(ethylene glycol)-b-poly(D,L-lactide) (mPEG-PDLLA) and poly(D, L-lactide-co-glycolide) (PLGA) NPs that allow the sustained release of rebamipide. In vitro, chondrocytes from rat were used to investigate the cytotoxicity and anti-inflammatory effect of rebamipide-loaded NPs. In vivo, monosodium iodoacetate (MIA)-induced OA rats were divided into 7 groups, consisting of healthy control rats and rats injected with MIA alone or in combination with NPs, rebamipide (1 mg)/NPs, rebamipide (10 mg)/NPs, rebamipide (10 mg) solution, or oral administration. RESULTS In vitro, rebamipide/NPs dose-dependently suppressed the mRNA levels of pro-inflammatory mediators, including interleukin (IL)-1β, IL-6, tumor necrosis factor-α, matrix metalloproteinase (MMP)-3, MMP-13, and cyclo-oxygenase-2. In vivo, the mRNA levels of pro-inflammatory components most markedly decreased in the intra-articularly injected rebamipide (10 mg)/NP group compared to other groups. Macroscopic, radiographic, and histological evaluations showed that the intra-articular injection of rebamipide/NPs inhibited cartilage degeneration more than rebamipide solution or rebamipide administration. CONCLUSIONS Using a chemically induced rat model of OA, intra-articular delivery of rebamipide was associated with decreased local and systemic inflammatory response decreased joint degradation and arthritic progression.
Collapse
Affiliation(s)
- Sung Eun Kim
- Department of Orthopedic Surgery and Rare Diseases Institute, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Sung Jae Choi
- Korea University College of Medicine, Seoul, Republic of Korea,Division of Rheumatology, Department of Internal Medicine, Korea University Ansan Hospital, Ansan-si, Republic of Korea
| | - Kyeongsoon Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, Republic of Korea
| | - Hak-Jun Kim
- Department of Orthopedic Surgery and Rare Diseases Institute, Korea University Guro Hospital, Seoul, Republic of Korea,Korea University College of Medicine, Seoul, Republic of Korea
| | - Gwan Gyu Song
- Korea University College of Medicine, Seoul, Republic of Korea,Division of Rheumatology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Jae Hyun Jung
- Korea University College of Medicine, Seoul, Republic of Korea,Division of Rheumatology, Department of Internal Medicine, Korea University Ansan Hospital, Ansan-si, Republic of Korea,Jae Hyun Jung, Division of Rheumatology, Department of Internal Medicine, Korea University Ansan Hospital, 123 Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Gyeonggi-do, Republic of Korea.
| |
Collapse
|