1
|
Jiang P, Zhou X, Yang Y, Bai L. Pectolinarigenin targeting FGFR3 alleviates osteoarthritis progression by regulating the NF-κB/NLRP3 inflammasome pyroptotic pathway. Int Immunopharmacol 2024; 140:112741. [PMID: 39094365 DOI: 10.1016/j.intimp.2024.112741] [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: 05/08/2024] [Revised: 07/08/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE Osteoarthritis (OA) is a chronic degenerative disease characterized by cartilage degeneration, involving inflammation, pyroptosis, and degeneration of the extracellular matrix (ECM). Pectolinarigenin (PEC) is a natural flavonoid with antioxidant, anti-inflammatory and anti-tumor properties. This study aims to explore the potential of PEC in ameliorating OA progression and its underlying mechanisms. METHODS Chondrocytes were exposed to 10 ng/mL IL-1β to simulate OA-like changes. The effect of PEC on IL-1β-treated chondrocytes was assessed using ELISA, western blot, and immunofluorescence. The mRNA sequencing (mRNA-seq) was employed to explore the possible targets of PEC in delaying OA progression. The OA mouse model was induced through anterior cruciate ligament transection (ACLT) and divided into sham, ACLT, ACLT+5 mg/kg PEC, and ACLT+10 mg/kg PEC groups. Micro-computed tomography and histological analysis were conducted to confirm the beneficial effects of PEC on OA in vivo. RESULTS PEC mitigated chondrocyte pyroptosis, as evidenced by reduced levels of pyroptosis-related proteins. Additionally, PEC attenuated IL-1β-mediated chondrocyte ECM degradation and inflammation. Mechanistically, mRNA-seq showed that FGFR3 was a downstream target of PEC. FGFR3 silencing reversed the beneficial effects of PEC on IL-1β-exposed chondrocytes. PEC exerted anti-pyroptotic, anti-ECM degradative, and anti-inflammatory effects through upregulating FGFR3 to inhibit the NF-κB/NLRP3 pyroptosis-related pathway. Consistently, in vivo experiments demonstrated the chondroprotective effects of PEC in OA mice. CONCLUSION PEC alleviate OA progression by FGFR3/NF-κB/NLRP3 pathway mediated chondrocyte pyroptosis, ECM degradation and inflammation, suggesting the potential of PEC as a therapeutic agent for OA.
Collapse
Affiliation(s)
- Peng Jiang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaonan Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
2
|
Zhou F, Chen M, Qian Y, Yuan K, Han X, Wang W, Guo JJ, Chen Q, Li B. Enhancing Endogenous Hyaluronic Acid in Osteoarthritic Joints with an Anti-Inflammatory Supramolecular Nanofiber Hydrogel Delivering HAS2 Lentivirus. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400542. [PMID: 38593309 DOI: 10.1002/smll.202400542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/24/2024] [Indexed: 04/11/2024]
Abstract
Osteoarthritis (OA) management remains challenging because of its intricate pathogenesis. Intra-articular injections of drugs, such as glucocorticoids and hyaluronic acid (HA), have certain limitations, including the risk of joint infection, pain, and swelling. Hydrogel-based therapeutic strategies have attracted considerable attention because of their enormous therapeutic potential. Herein, a supramolecular nanofiber hydrogel is developed using dexamethasone sodium phosphate (DexP) as a vector to deliver lentivirus-encoding hyaluronan synthase 2 (HAS2) (HAS2@DexP-Gel). During hydrogel degradation, HAS2 lentivirus and DexP molecules are slowly released. Intra-articular injection of HAS2@DexP-Gel promotes endogenous HA production and suppresses synovial inflammation. Additionally, HAS2@DexP-Gel reduces subchondral bone resorption in the anterior cruciate ligament transection-induced OA mice, attenuates cartilage degeneration, and delays OA progression. HAS2@DexP-Gel exhibited good biocompatibility both in vitro and in vivo. The therapeutic mechanisms of the HAS2@DexP-Gel are investigated using single-cell RNA sequencing. HAS2@DexP-Gel optimizes the microenvironment of the synovial tissue by modulating the proportion of synovial cell subpopulations and regulating the interactions between synovial fibroblasts and macrophages. The innovative nanofiber hydrogel, HAS2@DexP-Gel, effectively enhances endogenous HA production while reducing synovial inflammation. This comprehensive approach holds promise for improving joint function, alleviating pain, and slowing OA progression, thereby providing significant benefits to patients.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Medical 3D Printing Center, Orthopedic Institute, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Muchao Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Yufan Qian
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Medical 3D Printing Center, Orthopedic Institute, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Kai Yuan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xuequan Han
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Center for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Weishan Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832099, P. R. China
| | - Jiong Jiong Guo
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Medical 3D Printing Center, Orthopedic Institute, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Bin Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Medical 3D Printing Center, Orthopedic Institute, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| |
Collapse
|
3
|
Ilyas S, Lee J, Hwang Y, Choi Y, Lee D. Deciphering Cathepsin K inhibitors: a combined QSAR, docking and MD simulation based machine learning approaches for drug design. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:771-793. [PMID: 39382544 DOI: 10.1080/1062936x.2024.2405626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
Cathepsin K (CatK), a lysosomal cysteine protease, contributes to skeletal abnormalities, heart diseases, lung inflammation, and central nervous system and immune disorders. Currently, CatK inhibitors are associated with severe adverse effects, therefore limiting their clinical utility. This study focuses on exploring quantitative structure-activity relationships (QSAR) on a dataset of CatK inhibitors (1804) compiled from the ChEMBL database to predict the inhibitory activities. After data cleaning and pre-processing, a total of 1568 structures were selected for exploratory data analysis which revealed physicochemical properties, distributions and statistical significance between the two groups of inhibitors. PubChem fingerprinting with 11 different machine-learning classification models was computed. The comparative analysis showed the ET model performed well with accuracy values for the training set (0.999), cross-validation (0.970) and test set (0.977) in line with OECD guidelines. Moreover, to gain structural insights on the origin of CatK inhibition, 15 diverse molecules were selected for molecular docking. The CatK inhibitors (1 and 2) exhibited strong binding energies of -8.3 and -7.2 kcal/mol, respectively. MD simulation (300 ns) showed strong structural stability, flexibility and interactions in selected complexes. This synergy between QSAR, docking, MD simulation and machine learning models strengthen our evidence for developing novel and resilient CatK inhibitors.
Collapse
Affiliation(s)
- S Ilyas
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| | - J Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| | - Y Hwang
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| | - Y Choi
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| | - D Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, Seongnam-si, Korea
| |
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
|
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
|
6
|
Wang H, Yuan T, Wang Y, Liu C, Li D, Li Z, Sun S. Osteoclasts and osteoarthritis: Novel intervention targets and therapeutic potentials during aging. Aging Cell 2024; 23:e14092. [PMID: 38287696 PMCID: PMC11019147 DOI: 10.1111/acel.14092] [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: 10/17/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
Osteoarthritis (OA), a chronic degenerative joint disease, is highly prevalent among the aging population, and often leads to joint pain, disability, and a diminished quality of life. Although considerable research has been conducted, the precise molecular mechanisms propelling OA pathogenesis continue to be elusive, thereby impeding the development of effective therapeutics. Notably, recent studies have revealed subchondral bone lesions precede cartilage degeneration in the early stage of OA. This development is marked by escalated osteoclast-mediated bone resorption, subsequent imbalances in bone metabolism, accelerated bone turnover, and a decrease in bone volume, thereby contributing significantly to the pathological changes. While the role of aging hallmarks in OA has been extensively elucidated from the perspective of chondrocytes, their connection with osteoclasts is not yet fully understood. There is compelling evidence to suggest that age-related abnormalities such as epigenetic alterations, proteostasis network disruption, cellular senescence, and mitochondrial dysfunction, can stimulate osteoclast activity. This review intends to systematically discuss how aging hallmarks contribute to OA pathogenesis, placing particular emphasis on the age-induced shifts in osteoclast activity. It also aims to stimulate future studies probing into the pathological mechanisms and therapeutic approaches targeting osteoclasts in OA during aging.
Collapse
Affiliation(s)
- Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Yi Wang
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Changxing Liu
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Dengju Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Ziqing Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|
7
|
Zhou H, Zhang YF, Zhang QQ, Liu F, Zhang JY, Chen Y. Cathepsin K inhibition alleviates periodontal bone resorption by promoting type H vessel formation through PDGF-BB/PDGFR-β axis. Oral Dis 2024. [PMID: 38462960 DOI: 10.1111/odi.14920] [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: 12/13/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVES To explore the effects of cathepsin K (CTSK) inhibition on type H vessel formation and alveolar bone resorption within periodontitis. METHODS Conditioned media derived from preosteoclasts pretreated with the CTSK inhibitor odanacatib (ODN), ODN supplemented small interfering RNA targeting PDGF-BB (si-PDGF-BB), or PBS were prepared, to assess their proangiogenic effects on endothelial cells (HUVECs). A series of angiogenic-related assays were conducted to evaluate HUVEC proliferation, migration, and tube formation abilities in vitro. In addition, qRT-PCR and Western blot assays were employed to examine the expression levels of genes/proteins related to PDGF-BB/PDGFR-β axis components. A mouse periodontitis model was established to evaluate the effects of CTSK inhibition on type H vessel formation. RESULTS CTSK inhibition promoted PDGF-BB secretion from preosteoclasts and proliferation, migration, and tube formation activities of HUVECs in vitro. However, the conditioned medium from preosteoclasts pretreated by si-PDGF-BB impaired the angiogenic activities of HUVECs. This promoted angiogenesis function by CTSK inhibition may be mediated by the PDGF-BB/PDGFR-β axis. Functionally, in vivo studies demonstrated that CTSK inhibition significantly accelerated type H vessel formation and alleviated bone loss within periodontitis. CONCLUSION CTSK inhibition promotes type H vessel formation and attenuates alveolar bone resorption within periodontitis via PDGF-BB/PDGFR-β axis.
Collapse
Affiliation(s)
- Huan Zhou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yi-Fan Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Qian-Qian Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Fen Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Jia-Yu Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yue Chen
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Li H, Yuan Y, Zhang L, Xu C, Xu H, Chen Z. Reprogramming Macrophage Polarization, Depleting ROS by Astaxanthin and Thioketal-Containing Polymers Delivering Rapamycin for Osteoarthritis Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305363. [PMID: 38093659 PMCID: PMC10916582 DOI: 10.1002/advs.202305363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/22/2023] [Indexed: 03/07/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by synovitis and joint cartilage destruction. The severity of OA is highly associated with the imbalance between M1 and M2 synovial macrophages. In this study, a novel strategy is designed to modulate macrophage polarization by reducing intracellular reactive oxygen species (ROS) levels and regulating mitochondrial function. A ROS-responsive polymer is synthesized to self-assemble with astaxanthin and autophagy activator rapamycin to form nanoparticles (NP@PolyRHAPM ). In vitro experiments show that NP@PolyRHAPM significantly reduced intracellular ROS levels. Furthermore, NP@PolyRHAPM restored mitochondrial membrane potential, increased glutathione (GSH) levels, and promoted intracellular autophagy, hence successfully repolarizing M1 macrophages into the M2 phenotype. This repolarization enhanced chondrocyte proliferation and vitality while inhibiting apoptosis. In vivo experiments utilizing an anterior cruciate ligament transection (ACLT)-induced OA mouse model revealed the anti-inflammatory and cartilage-protective effects of NP@PolyRHAPM , effectively mitigating OA progression. Consequently, the findings suggest that intra-articular delivery of ROS-responsive nanocarrier systems holds significant promise as a potential and effective therapeutic strategy for OA treatment.
Collapse
Affiliation(s)
- Huiyun Li
- Department of Orthopedic SurgeryThe First Affiliated Hospital of University of South ChinaHengyangHunan421001China
| | - Yusong Yuan
- Department of Orthopaedic SurgeryChina‐Japan Friendship HospitalNo.2 Yinghuayuan East StreetBeijing100029China
| | - Lingpu Zhang
- Beijing National Laboratory for Molecular ScienceState Key Laboratory of Polymer Physics and ChemistryInstitute of ChemistryChinese Academy of ScienceBeijing100190China
| | - Chun Xu
- School of DentistryThe University of QueenslandBrisbane4006Australia
| | - Hailin Xu
- Department of Trauma and OrthopedicsPeking University People's Hospital Diabetic Foot Treatment CenterPeking University People's Hospital11th XizhimenSouth StreetBeijing100044China
| | - Zhiwei Chen
- Department of Orthopedic SurgeryThe First Affiliated Hospital of University of South ChinaHengyangHunan421001China
| |
Collapse
|
9
|
Qian Y, Chu G, Zhang L, Wu Z, Wang Q, Guo JJ, Zhou F. M2 macrophage-derived exosomal miR-26b-5p regulates macrophage polarization and chondrocyte hypertrophy by targeting TLR3 and COL10A1 to alleviate osteoarthritis. J Nanobiotechnology 2024; 22:72. [PMID: 38374072 PMCID: PMC10877765 DOI: 10.1186/s12951-024-02336-4] [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: 12/08/2023] [Accepted: 02/09/2024] [Indexed: 02/21/2024] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent chronic musculoskeletal diseases among the elderly population. In this study, macrophage-derived exosomes were isolated and identified. Exosomes were subjected to microRNA (miRNA) sequencing and bioinformatic analysis, and differentially expressed miRNAs were verified. miR-26b-5p target genes were confirmed through target-site mutation combined with a dual-luciferase reporter assay. The effects of miR-26b-5p on macrophage polarization and chondrocyte hypertrophy were assessed in vitro. miR-26b-5p agomir was applied to mice with OA induced by anterior cruciate ligament transection (ACLT). The therapeutic effects of miR-26b-5p were evaluated via pain behavior experiments and histological observations. In vitro, miR-26b-5p repolarized M1 macrophages to an anti-inflammatory M2 type by targeting the TLR3 signaling pathway. miR-26b-5p could target COL10A1, further inhibiting chondrocyte hypertrophy induced by M1 macrophage-conditioned medium (M1-CM). In vivo, miR-26b-5p agomir ameliorated gait abnormalities and mechanical allodynia in OA mice. miR-26b-5p treatment attenuated synovitis and cartilage degeneration, thereby delaying OA progression. In conclusion, M2 macrophage-derived exosomal miR-26b-5p could protect articular cartilage and ameliorate gait abnormalities in OA mice by targeting TLR3 and COL10A1. miR-26b-5p further affected macrophage polarization and chondrocyte hypertrophy. Thus, this exosomal miR-26b-5p-based strategy might be a potential method for OA treatment.
Collapse
Affiliation(s)
- Yufan Qian
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
| | - Genglei Chu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Lei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
| | - Zhikai Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
| | - Qiuyuan Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
| | - Jiong Jiong Guo
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China.
| | - Feng Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China.
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
10
|
Kim GM, Kim J, Lee JY, Park MC, Lee SY. IgSF11 deficiency alleviates osteoarthritis in mice by suppressing early subchondral bone changes. Exp Mol Med 2023; 55:2576-2585. [PMID: 38036734 PMCID: PMC10767117 DOI: 10.1038/s12276-023-01126-6] [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: 05/24/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 12/02/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease. While it is classically characterized by articular cartilage destruction, OA affects all tissues in the joints and is thus also accompanied by local inflammation, subchondral bone changes, and persistent pain. However, our understanding of the underlying subchondral bone dynamics during OA progression is poor. Here, we demonstrate the contribution of immunoglobulin superfamily 11 (IgSF11) to OA subchondral bone remodeling by using a murine model. In particular, IgSF11 was quickly expressed by differentiating osteoclasts and upregulated in subchondral bone soon after destabilization-of-the-medial-meniscus (DMM)-induced OA. In mice, IgSF11 deficiency not only suppressed subchondral bone changes in OA but also blocked cartilage destruction. The IgSF11-expressing cells in OA subchondral bone were found to be involved in osteoclast maturation and bone resorption and colocalized with receptor-activator of nuclear-factor κ-B (RANK), the key osteoclast differentiation factor. Thus, our study shows that blocking early subchondral bone changes in OA can ameliorate articular cartilage destruction in OA.
Collapse
Affiliation(s)
- Gyeong Min Kim
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jihee Kim
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - June-Yong Lee
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Soo Young Lee
- Department of Life Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, Republic of Korea.
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
11
|
Kasaeian A, Roemer FW, Ghotbi E, Ibad HA, He J, Wan M, Zbijewski WB, Guermazi A, Demehri S. Subchondral bone in knee osteoarthritis: bystander or treatment target? Skeletal Radiol 2023; 52:2069-2083. [PMID: 37646795 DOI: 10.1007/s00256-023-04422-4] [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] [Received: 11/14/2022] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 09/01/2023]
Abstract
The subchondral bone is an important structural component of the knee joint relevant for osteoarthritis (OA) incidence and progression once disease is established. Experimental studies have demonstrated that subchondral bone changes are not simply the result of altered biomechanics, i.e., pathologic loading. In fact, subchondral bone alterations have an impact on joint homeostasis leading to articular cartilage loss already early in the disease process. This narrative review aims to summarize the available and emerging imaging techniques used to evaluate knee OA-related subchondral bone changes and their potential role in clinical trials of disease-modifying OA drugs (DMOADs). Radiographic fractal signature analysis has been used to quantify OA-associated changes in subchondral texture and integrity. Cross-sectional modalities such as cone-beam computed tomography (CT), contrast-enhanced cone beam CT, and micro-CT can also provide high-resolution imaging of the subchondral trabecular morphometry. Magnetic resonance imaging (MRI) has been the most commonly used advanced imaging modality to evaluate OA-related subchondral bone changes such as bone marrow lesions and altered trabecular bone texture. Dual-energy X-ray absorptiometry can provide insight into OA-related changes in periarticular subchondral bone mineral density. Positron emission tomography, using physiological biomarkers of subchondral bone regeneration, has provided additional insight into OA pathogenesis. Finally, artificial intelligence algorithms have been developed to automate some of the above subchondral bone measurements. This paper will particularly focus on semiquantitative methods for assessing bone marrow lesions and their utility in identifying subjects at risk of symptomatic and structural OA progression, and evaluating treatment responses in DMOAD clinical trials.
Collapse
Affiliation(s)
- Arta Kasaeian
- Musculoskeletal Radiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frank W Roemer
- Department of Radiology, Boston University School of Medicine, Boston, MA, USA
- Department of Radiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Elena Ghotbi
- Musculoskeletal Radiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hamza Ahmed Ibad
- Musculoskeletal Radiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jianwei He
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mei Wan
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wojciech B Zbijewski
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ali Guermazi
- Department of Radiology, Boston University School of Medicine, Boston, MA, USA
| | - Shadpour Demehri
- Musculoskeletal Radiology, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Zhao T, Li X, Li H, Deng H, Li J, Yang Z, He S, Jiang S, Sui X, Guo Q, Liu S. Advancing drug delivery to articular cartilage: From single to multiple strategies. Acta Pharm Sin B 2023; 13:4127-4148. [PMID: 37799383 PMCID: PMC10547919 DOI: 10.1016/j.apsb.2022.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022] Open
Abstract
Articular cartilage (AC) injuries often lead to cartilage degeneration and may ultimately result in osteoarthritis (OA) due to the limited self-repair ability. To date, numerous intra-articular delivery systems carrying various therapeutic agents have been developed to improve therapeutic localization and retention, optimize controlled drug release profiles and target different pathological processes. Due to the complex and multifactorial characteristics of cartilage injury pathology and heterogeneity of the cartilage structure deposited within a dense matrix, delivery systems loaded with a single therapeutic agent are hindered from reaching multiple targets in a spatiotemporal matched manner and thus fail to mimic the natural processes of biosynthesis, compromising the goal of full cartilage regeneration. Emerging evidence highlights the importance of sequential delivery strategies targeting multiple pathological processes. In this review, we first summarize the current status and progress achieved in single-drug delivery strategies for the treatment of AC diseases. Subsequently, we focus mainly on advances in multiple drug delivery applications, including sequential release formulations targeting various pathological processes, synergistic targeting of the same pathological process, the spatial distribution in multiple tissues, and heterogeneous regeneration. We hope that this review will inspire the rational design of intra-articular drug delivery systems (DDSs) in the future.
Collapse
Affiliation(s)
- Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Hao Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Haoyuan Deng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jianwei Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhen Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China
| | - Songlin He
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuangpeng Jiang
- Department of Joint Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
13
|
Shanmugasundaram S, Solanki K, Saseendar S, Chavada VK, D'Ambrosi R. Role of Doxycycline as an Osteoarthritis Disease-Modifying Drug. J Clin Med 2023; 12:jcm12082927. [PMID: 37109263 PMCID: PMC10145123 DOI: 10.3390/jcm12082927] [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] [Received: 02/04/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Doxycycline is a drug that has been proposed to modify osteoarthritis (OA) progression, in addition to its role as an antibiotic. However, available evidence thus far comprises sporadic reports, with no consensus on its benefits. Hence, this review attempts to analyze the evidence available thus far on the role of doxycycline as a disease-modifying osteoarthritis drug (DMOAD) in knee osteoarthritis. The earliest evidence of doxycycline in OA appeared in 1991 when doxycycline was found to inhibit the type XI collagenolytic activity of extracts from the human osteoarthritic cartilage, and gelatinase and tetracycline were found to inhibit this metalloproteinase activity in articular cartilage in vivo, which could modify cartilage breakdown in osteoarthritis. Apart from the inhibition of cartilage damage by metalloproteinases (MMPs) and other cartilage-related mechanisms, doxycycline also affects the bone and interferes with many enzyme systems. The most significant finding after reviewing various studies was that doxycycline has a definitive role in structural changes in osteoarthritis progression and radiological joint space width, but its role in the improvement of clinical outcomes as a DMOAD has not been established. However, there is much of a gap and lack of evidence in this regard. Doxycycline, as an MMP inhibitor, has theoretical advantages for clinical outcomes, but the present studies reveal only beneficial structural changes in osteoarthritis and very minimal or nonexistent advantages in clinical outcomes. Current evidence does not favor the regular use of doxycycline for the treatment of osteoarthritis as an individual treatment option or in combination with others. However, multicenter large cohort studies are warranted to determine the long-term benefits of doxycycline.
Collapse
Affiliation(s)
| | | | | | - Vijay K Chavada
- Department of Community Medicine, Indira Gandhi Medical College and Research Institute, Puducherry 605009, India
| | - Riccardo D'Ambrosi
- IRCCS Ospedale Galeazzi-Sant'Ambrogio, 20161 Milan, Italy
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
14
|
Liu D, Li X, Zhang L, Hu B, Hu S, Zhang X, Hu J. Small molecule inhibitors of osteoarthritis: Current development and future perspective. Front Physiol 2023; 14:1156913. [PMID: 37089415 PMCID: PMC10119395 DOI: 10.3389/fphys.2023.1156913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Osteoarthritis (OA) is one of the common degenerative joint diseases in clinic. It mainly damages articular cartilage, causing pain, swelling and stiffness around joints, and is the main cause of disability of the elderly. Due to the unclear pathogenesis of osteoarthritis and the poor self-healing ability of articular cartilage, the treatment options for this disease are limited. At present, NSAIDs, Glucocorticoid and Duloxetine are the most commonly used treatment choice for osteoarthritis. Although it is somewhat effective, the adverse reactions are frequent and serious. The development of safer and more effective anti-osteoarthritis drugs is essential and urgent. This review summarizes recent advances in the pharmacological treatment of OA, focusing on small molecule inhibitors targeting cartilage remodeling in osteoarthritis as well as the research idea of reducing adverse effects by optimizing the dosage form of traditional drugs for the treatment of osteoarthritis. It should provide a reference for exploration of new potential treatment options.
Collapse
Affiliation(s)
- Dan Liu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Xingxing Li
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Lin Zhang
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Bin Hu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Sang Hu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiao Zhang
- Institute of Pathology, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing, China
| | - Jing Hu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
15
|
Mo H, Wang Z, He Z, Wan J, Lu R, Wang C, Chen A, Cheng P. Decreased Peli1 expression attenuates osteoarthritis by protecting chondrocytes and inhibiting M1-polarization of macrophages. Bone Joint Res 2023; 12:121-132. [PMID: 36718653 PMCID: PMC9950670 DOI: 10.1302/2046-3758.122.bjr-2022-0214.r1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
AIMS Pellino1 (Peli1) has been reported to regulate various inflammatory diseases. This study aims to explore the role of Peli1 in the occurrence and development of osteoarthritis (OA), so as to find new targets for the treatment of OA. METHODS After inhibiting Peli1 expression in chondrocytes with small interfering RNA (siRNA), interleukin (IL)-1β was used to simulate inflammation, and OA-related indicators such as synthesis, decomposition, inflammation, and apoptosis were detected. Toll-like receptor (TLR) and nuclear factor-kappa B (NF-κB) signalling pathway were detected. After inhibiting the expression of Peli1 in macrophages Raw 264.7 with siRNA and intervening with lipopolysaccharide (LPS), the polarization index of macrophages was detected, and the supernatant of macrophage medium was extracted as conditioned medium to act on chondrocytes and detect the apoptosis index. The OA model of mice was established by destabilized medial meniscus (DMM) surgery, and adenovirus was injected into the knee cavity to reduce the expression of Peli1. The degree of cartilage destruction and synovitis were evaluated by haematoxylin and eosin (H&E) staining, Safranin O/Fast Green staining, and immunohistochemistry. RESULTS In chondrocytes, knockdown of Peli1 produced anti-inflammatory and anti-apoptotic effects by targeting the TLR and NF-κB signalling pathways. We found that in macrophages, knockdown of Peli1 can inhibit M1-type polarization of macrophages. In addition, the corresponding conditioned culture medium of macrophages applied to chondrocytes can also produce an anti-apoptotic effect. During in vivo experiments, the results have also shown that knockdown Peli1 reduces cartilage destruction and synovial inflammation. CONCLUSION Knockdown of Peli1 has a therapeutic effect on OA, which therefore makes it a potential therapeutic target for OA.Cite this article: Bone Joint Res 2023;12(2):121-132.
Collapse
Affiliation(s)
- Haokun Mo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenggang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Zhiyi He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junlai Wan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Lu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenwen Wang
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, Anmin Chen. E-mail:
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Biasizzo M, Javoršek U, Vidak E, Zarić M, Turk B. Cysteine cathepsins: A long and winding road towards clinics. Mol Aspects Med 2022; 88:101150. [PMID: 36283280 DOI: 10.1016/j.mam.2022.101150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022]
Abstract
Biomedical research often focuses on properties that differentiate between diseased and healthy tissue; one of the current focuses is elevated expression and altered localisation of proteases. Among these proteases, dysregulation of cysteine cathepsins can frequently be observed in inflammation-associated diseases, which tips the functional balance from normal physiological to pathological manifestations. Their overexpression and secretion regularly exhibit a strong correlation with the development and progression of such diseases, making them attractive pharmacological targets. But beyond their mostly detrimental role in inflammation-associated diseases, cysteine cathepsins are physiologically highly important enzymes involved in various biological processes crucial for maintaining homeostasis and responding to different stimuli. Consequently, several challenges have emerged during the efforts made to translate basic research data into clinical applications. In this review, we present both physiological and pathological roles of cysteine cathepsins and discuss the clinical potential of cysteine cathepsin-targeting strategies for disease management and diagnosis.
Collapse
Affiliation(s)
- Monika Biasizzo
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Miki Zarić
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
17
|
Wu Z, Yuan K, Zhang Q, Guo JJ, Yang H, Zhou F. Antioxidant PDA-PEG nanoparticles alleviate early osteoarthritis by inhibiting osteoclastogenesis and angiogenesis in subchondral bone. J Nanobiotechnology 2022; 20:479. [DOI: 10.1186/s12951-022-01697-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 11/02/2022] [Indexed: 11/17/2022] Open
Abstract
AbstractAccumulating evidence suggests that osteoclastogenesis and angiogenesis in subchondral bone are critical destructive factors in the initiation and progression of osteoarthritis (OA). Herein, methoxypolyethylene glycol amine (mPEG-NH2) modified polydopamine nanoparticles (PDA-PEG NPs) were synthesized for treating early OA. The cytotoxicity and reactive oxygen species (ROS) scavenging ability of PDA-PEG NPs were evaluated. The effects of PDA-PEG NPs on osteoclast differentiation and vessel formation were then evaluated. Further, PDA-PEG NPs were administrated to anterior cruciate ligament transection (ACLT)-induced OA mice. Results demonstrated that PDA-PEG NPs had low toxicity both in vitro and in vivo. PDA-PEG NPs could inhibit osteoclastogenesis via regulating nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. Moreover, PDA-PEG NPs suppressed osteoclast-related angiogenesis via down-regulating platelet-derived growth factor-BB (PDGF-BB). In vivo, PDA-PEG NPs inhibited subchondral bone resorption and angiogenesis, further rescuing cartilage degradation in OA mice. In conclusion, we demonstrated that PDA-PEG NPs deployment could be a potential therapy for OA.
Graphical Abstract
Collapse
|
18
|
Jin Y, Zhang Q, Qin X, Liu Z, Li Z, Zhong X, Xia L, He J, Fang B. Carbon dots derived from folic acid attenuates osteoarthritis by protecting chondrocytes through NF-κB/MAPK pathway and reprogramming macrophages. J Nanobiotechnology 2022; 20:469. [PMCID: PMC9632154 DOI: 10.1186/s12951-022-01681-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022] Open
Abstract
Background Osteoarthritis (OA) is a common joint disorder worldwide which causes great health and economic burden. However, there remains an unmet goal to develop an effective therapeutic method to prevent or delay OA. Chondrocytes, as the major cells involved in OA progression, may serve as a promising therapeutic target. Results A kind of carbon dots (CDs) with excellent biocompatibility was fabricated from folic acid via hydrothermal method and could effectively attenuate osteoarthritis. It was demonstrated that CDs treatment could rescue IL1β-induced proinflammatory responses, oxidative stress, cartilage degeneration and extracellular matrix degradation. Moreover, CDs reprogrammed lipopolysaccharide (LPS)-induced macrophage inflammation and polarization. Conditioned medium (CM) from CDs-treated macrophages could attenuate IL1β-induced chondrocyte injury. Also, CM from CDs-treated chondrocytes had immunoregulatory functions on macrophages. Mechanistically, CDs inhibited the activation of nuclear factor-κB (NF-κB) and mitogen-activated protein kinases (MAPK) signaling pathways in IL1β-stimulated chondrocytes. In vivo, anterior cruciate ligament transection (ACLT) mice model was adopted and it was indicated that intra-articular injection of CDs effectively delays OA pathogenesis. Conclusions Taken together, these findings indicated CDs could mediate OA via promoting cartilage repair and immunomodulating macrophages within local microenvironment, which may provide evidences for utilizing CDs as a novel nanomaterial for OA treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01681-6.
Collapse
Affiliation(s)
- Yu Jin
- grid.412523.30000 0004 0386 9086Department of Orthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, 500 Quxi Road, Shanghai, 200011 China
| | - Qing Zhang
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Advanced Optical Communication Systems and Networks, Key Laboratory for Laser Plasmas (Ministry of Education), School of Physics and Astronomy, Shanghai Jiao Tong University, No. 800, Dongchuan Road, Minhang District, Shanghai, 200240 China
| | - Xing Qin
- grid.16821.3c0000 0004 0368 8293Department of Oral and Maxillofacial-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Zhen Liu
- grid.412523.30000 0004 0386 9086Department of Orthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, 500 Quxi Road, Shanghai, 200011 China
| | - Zhenxia Li
- grid.412523.30000 0004 0386 9086Department of Orthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, 500 Quxi Road, Shanghai, 200011 China
| | - Xiaoxia Zhong
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Advanced Optical Communication Systems and Networks, Key Laboratory for Laser Plasmas (Ministry of Education), School of Physics and Astronomy, Shanghai Jiao Tong University, No. 800, Dongchuan Road, Minhang District, Shanghai, 200240 China
| | - Lunguo Xia
- grid.412523.30000 0004 0386 9086Department of Orthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, 500 Quxi Road, Shanghai, 200011 China
| | - Jie He
- grid.16821.3c0000 0004 0368 8293Department of Oral and Maxillofacial-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011 China
| | - Bing Fang
- grid.412523.30000 0004 0386 9086Department of Orthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, 500 Quxi Road, Shanghai, 200011 China
| |
Collapse
|
19
|
Zhu H, Liu H, Chen X, Xu X, Zhang S, Xie D. Enhancing autophagy and energy metabolism in the meniscus can delay the occurrence of PTOA in ACLT rat. Front Cell Dev Biol 2022; 10:971736. [PMID: 36120586 PMCID: PMC9479128 DOI: 10.3389/fcell.2022.971736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative joint disease characterized by the destruction of the articular cartilage, meniscus and the like. Autophagy and cellular energy metabolism are the mechanisms by which cells maintain homeostasis. However, little is known about the effects of autophagy and cellular energy metabolism on meniscus degeneration, and the pathogenesis of posttraumatic osteoarthritis (PTOA) after the meniscal injury is rarely reported. Therefore, this study aimed to investigate the relationship between changes in autophagy and cellular energy metabolism in the meniscus following anterior cruciate ligament transection (ACLT) and PTOA induced by subsequent articular cartilage injury. In this study, we use a combination of cell experiments in vitro and animal experiments in vivo. On the one hand, cell experiment results show that inhibiting the mTORC1 signaling pathway by inhibiting the phosphorylation of S6K and AKT proteins in meniscal cells will lead to the increase of Beclin1, LC-3B, ATG12, ULK1, P62, and activate autophagy-related signaling pathways, which in turn protects the extracellular matrix component COL1 of meniscal cells from degradation by catabolic factor MMP13. In addition, it increased the generation of mitochondrial membrane potential in meniscal cells, increased the expression of anti-apoptotic factor BCL-XL, decreased the expression of pro-apoptotic factors BAD and BAX, and reduced the apoptosis of meniscal cells. More importantly, under the stimulation of inflammatory factor IL-1β, the secretion of meniscus cells can reduce the elevated levels of MMP13 and Adamts5 caused by chondrocytes affected by IL-1β. On the other hand, the results of animal experiments in vivo further proved the validity of the results of the cell experiments, and also proved that the meniscus injury did prior to the articular cartilage degeneration after ACLT. In conclusion, this study suggests that the meniscus prior to articular cartilage damage during the development of PTOA after ACLT, and that promoting autophagy and energy metabolism of meniscal cells may be a potential therapeutic target for delaying PTOA.
Collapse
Affiliation(s)
- Huangrong Zhu
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics), Guangzhou, China
- Orthopedic Hospital of Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Hai Liu
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics), Guangzhou, China
- Orthopedic Hospital of Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xizhong Chen
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics), Guangzhou, China
- Orthopedic Hospital of Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xin Xu
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics), Guangzhou, China
- Orthopedic Hospital of Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shuqin Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Denghui Xie
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics), Guangzhou, China
- Orthopedic Hospital of Guangdong, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- *Correspondence: Denghui Xie,
| |
Collapse
|
20
|
Ginnetti AT, Paone DV, Nanda KK, Li J, Busuek M, Johnson SA, Lu J, Soisson SM, Robinson R, Fisher J, Webber A, Wesolowski G, Ma B, Duong L, Carroll S, Burgey CS, Stachel SJ. Lead Optimization of Cathepsin K Inhibitors for the Treatment of Osteoarthritis. Bioorg Med Chem Lett 2022; 74:128927. [DOI: 10.1016/j.bmcl.2022.128927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022]
|
21
|
Khoshdel A, Forootan M, Afsharinasab M, Rezaian M, Abbasifard M. Assessment of the circulatory concentrations of cathepsin D, cathepsin K, and alpha-1 antitrypsin in patients with knee osteoarthritis. Ir J Med Sci 2022:10.1007/s11845-022-03061-3. [PMID: 35749030 DOI: 10.1007/s11845-022-03061-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Evidence has shown that cysteine protease enzymes, such as cathepsin D, cathepsin A, cathepsin K, and alpha-1 antitrypsin (AAT) are involved in the chronic degenerative joint process. This study aimed to determine the potential involvement of cathepsin K, cathepsin D, and AAT in patients with osteoarthritis (OA). METHODS This study was performed on 31 patients with knee OA and 29 age- and sex-matched healthy subjects (both with Fars ethnicity from Iran). American College of Rheumatology (ACR) criteria were used to diagnose OA patients. The clinical status of the patients was scored by Western Ontario McMaster Universities Osteoarthritis (WOMAC), and pain intensity was measured by the Visual Analog Scale (VAS). The serum level of AAT was measured using high-resolution cellulose acetate electrophoresis. Additionally, serum levels of cathepsin D and cathepsin K were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS The findings showed that the serum level of cathepsin K was significantly increased in OA patients compared to healthy subjects (P = 0.01), while there was no significant difference between serum level of cathepsin D in study groups (P = 0.2). In addition, the serum concentration of AAT was significantly decreased in OA patients compared to healthy subjects (P = 0.003). There was a significant correlation between WOMAC score and age (r = 0.644, P = 0.0001) and VAS (r = 0.866, P < 0.0001) in OA patients. CONCLUSIONS The decreased level of AAT in OA patients and a rise in serum level of cathepsin K are involved in the pathogenesis of OA via stimulation of bone resorption and cartilage degradation.
Collapse
Affiliation(s)
- Alireza Khoshdel
- Nervous System Stem Cells Research Center , Semnan University of Medical Sciences, Semnan, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mohammad Forootan
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mehdi Afsharinasab
- Department of Clinical Biochemistry, Schoolof Medicine, Tehran University of Medical Sciences, Rafsanjan, Iran
| | - Mohsen Rezaian
- Epidemiology and Biostatistics Department, Occupational Environmental Research Center, Rafsanjan Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mitra Abbasifard
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
22
|
|
23
|
Hu W, Cai C, Li Y, Kang F, Chu T, Dong S. Farnesoid X receptor agonist attenuates subchondral bone osteoclast fusion and osteochondral pathologies of osteoarthritis via suppressing JNK1/2/NFATc1 pathway. FASEB J 2022; 36:e22243. [PMID: 35224782 DOI: 10.1096/fj.202101717r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/04/2022] [Accepted: 02/17/2022] [Indexed: 11/11/2022]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease of the joint, featured by articular cartilage destruction and subchondral bone marrow lesions. Articular cartilage and subchondral bone constitute an osteochondral unit that guarantees joint homeostasis. During OA initiation, activated osteoclasts in subchondral bone ultimately result in impaired capacities of the subchondral bone in response to mechanical stress, followed by the degradation of overlying articular cartilage. Thus, targeting osteoclasts could be a potential therapeutic option for treating OA. Here, we observed that farnesoid X receptor (FXR) expression and osteoclast fusion and activity in subchondral bone were concomitantly changed during early-stage OA in the OA mouse model established by anterior cruciate ligament transection (ACLT). Then, we explored the therapeutic effects of FXR agonist GW4064 on the osteochondral pathologies in ACLT mice. We showed that GW4064 obviously ameliorated subchondral bone deterioration, associated with reduction in tartrate-resistant acid phosphatase (TRAP) positive multinuclear osteoclast number, as well as articular cartilage degradation, which were blocked by the treatment with FXR antagonist Guggulsterone. Mechanistically, GW4064 impeded osteoclastogenesis through inhibiting subchondral bone osteoclast fusion via suppressing c-Jun N-terminal kinase (JNK) 1/2/nuclear factor of activated T-cells 1 (NFATc1) pathway. Taken together, our results present evidence for the protective effects of GW4064 against OA by blunting osteoclast-mediated aberrant subchondral bone loss and subsequent cartilage deterioration. Therefore, GW4064 demonstrates the potential as an alternative therapeutic option against OA for further drug development.
Collapse
Affiliation(s)
- Wenhui Hu
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chenhui Cai
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuheng Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fei Kang
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tongwei Chu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
24
|
Karila T, Tervahartiala T, Cohen B, Sorsa T. The collagenases: are they tractable targets for preventing cartilage destruction in osteoarthritis? Expert Opin Ther Targets 2022; 26:93-105. [PMID: 35081858 DOI: 10.1080/14728222.2022.2035362] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The etiology and pathogenesis of osteoarthritis (OA) have been intensely investigated; however, the disease course and progression are not completely understood. A prominent role for interstitial collagenases is recognized in this degenerative process, hence strategies to target them are of major interest. AREAS COVERED The pathogenesis of OA, the role of interstitial collagenases (MMP-1, -8 and -13) and collagenase modifying drugs are examined and discussed. We reviewed relevant papers from PubMed and Google Scholar. EXPERT OPINION There is strong evidence for the therapeutic potential of MMP inhibitors in OA; however, they are not expected to impact the inflammatory process. Therefore, there is a need for a relative inhibitor of MMP-13 collagenase which possesses anti-inflammatory properties. The identification of novel broad-spectrum relative multiple peptidase inhibitors could provide desirable tools for the prophylaxis, cure, or treatment of diseases involving articular cartilage (AC) degradation, in particular OA.
Collapse
Affiliation(s)
- Tuomo Karila
- Hospital Orton, Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Taina Tervahartiala
- Department of Oral and Maxillofacial Diseases, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
| | | | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland.,Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
25
|
Liu CC, Lee HC, Peng YS, Tseng AH, Wu JL, Tsai WY, Wong CS, Su LJ. Transcriptome Analysis Reveals Novel Genes Associated with Cartilage Degeneration in Posttraumatic Osteoarthritis Progression. Cartilage 2021; 13:1249S-1262S. [PMID: 31104480 PMCID: PMC8804845 DOI: 10.1177/1947603519847744] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The current therapeutic strategy for posttraumatic osteoarthritis (PTOA) focuses on early intervention to attenuate disease progression, preserve joint function, and defer joint replacement timing. Sequential transcriptomic changes of articular cartilage in a rat model were investigated to explore the molecular mechanism in early PTOA progression. DESIGN Anterior cruciate ligament transection and medial meniscectomy (ACLT + MMx)-induced PTOA model was applied on male Wistar rats. Articular cartilages were harvested at time 0 (naïve), 2 week, and 4 weeks after surgery. Affymetrix Rat genome 230 2.0 array was utilized to analyze the gene expression changes of articular cartilages. RESULTS We identified 849 differentially expressed genes (DEGs) at 2 weeks and 223 DEGs at 4 weeks post-ACLT + MMx surgery compared with time 0 (naïve group). Gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to gain further insights from these DEGs. 22 novel genes and 1 novel KEGG pathway (axon guidance) in cartilage degeneration of osteoarthritis were identified. Axon guidance molecules-Gnai1, Sema4d, Plxnb1, and Srgap2 commonly dysregulated in PTOA progression. Gnai1 gene showed a concordant change in protein expression by immunohistochemistry staining. CONCLUSIONS Our study identified 22 novel dysregulated genes and axon guidance pathway associated with articular cartilage degeneration in PTOA progression. These findings provide the potential candidates of biomarkers and therapeutic targets for further investigation.
Collapse
Affiliation(s)
- Chih-Chung Liu
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei, Taiwan,Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hoong-Chien Lee
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan,Department of Physics, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Yi-Shian Peng
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | | | - Jia-Lin Wu
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan
| | - Wei-Yuan Tsai
- Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwna
| | - Chih-Shung Wong
- Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwna,Graduate Institute of Medical Sciences, National Defence Medical Center, Taipei, Taiwan,Chih-Shung Wong, Department of Anesthesiology, Cathay General Hospital, No. 280, Renai Road, Sec. 4, Daan District, Taipei 10630, Taiwan.
| | - Li-Jen Su
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| |
Collapse
|
26
|
Cho Y, Jeong S, Kim H, Kang D, Lee J, Kang SB, Kim JH. Disease-modifying therapeutic strategies in osteoarthritis: current status and future directions. Exp Mol Med 2021; 53:1689-1696. [PMID: 34848838 PMCID: PMC8640059 DOI: 10.1038/s12276-021-00710-y] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 08/18/2021] [Accepted: 09/22/2021] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis. It is characterized by progressive destruction of articular cartilage and the development of chronic pain and constitutes a considerable socioeconomic burden. Currently, pharmacological treatments mostly aim to relieve the OA symptoms associated with inflammation and pain. However, with increasing understanding of OA pathology, several potential therapeutic targets have been identified, enabling the development of disease-modifying OA drugs (DMOADs). By targeting inflammatory cytokines, matrix-degrading enzymes, the Wnt pathway, and OA-associated pain, DMOADs successfully modulate the degenerative changes in osteoarthritic cartilage. Moreover, regenerative approaches aim to counterbalance the loss of cartilage matrix by stimulating chondrogenesis in endogenous stem cells and matrix anabolism in chondrocytes. Emerging strategies include the development of senolytic drugs or RNA therapeutics to eliminate the cellular or molecular sources of factors driving OA. This review describes the current developmental status of DMOADs and the corresponding results from preclinical and clinical trials and discusses the potential of emerging therapeutic approaches to treat OA.
Collapse
Affiliation(s)
- Yongsik Cho
- grid.31501.360000 0004 0470 5905Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826 South Korea ,grid.410720.00000 0004 1784 4496Center for RNA Research, Institute for Basic Science, Seoul, 08826 South Korea
| | - Sumin Jeong
- grid.31501.360000 0004 0470 5905Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826 South Korea ,grid.31501.360000 0004 0470 5905Department of Business Administration, Business School, Seoul National University, Seoul, 08826 South Korea
| | - Hyeonkyeong Kim
- grid.31501.360000 0004 0470 5905Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826 South Korea ,grid.410720.00000 0004 1784 4496Center for RNA Research, Institute for Basic Science, Seoul, 08826 South Korea
| | - Donghyun Kang
- grid.31501.360000 0004 0470 5905Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826 South Korea ,grid.410720.00000 0004 1784 4496Center for RNA Research, Institute for Basic Science, Seoul, 08826 South Korea
| | - Jeeyeon Lee
- grid.31501.360000 0004 0470 5905Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826 South Korea ,grid.410720.00000 0004 1784 4496Center for RNA Research, Institute for Basic Science, Seoul, 08826 South Korea
| | - Seung-Baik Kang
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Boramae Hospital, Seoul, 07061, South Korea.
| | - Jin-Hong Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea. .,Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
27
|
Zhang L, Kirkwood CL, Sohn J, Lau A, Bayers-Thering M, Bali SK, Rachala S, Marzo JM, Anders MJ, Beier F, Kirkwood KL. Expansion of myeloid-derived suppressor cells contributes to metabolic osteoarthritis through subchondral bone remodeling. Arthritis Res Ther 2021; 23:287. [PMID: 34784965 PMCID: PMC8594239 DOI: 10.1186/s13075-021-02663-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) subsequent to acute joint injury accounts for a significant proportion of all arthropathies. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of myeloid progenitor cells classically known for potent immune-suppressive activity; however, MDSCs can also differentiate into osteoclasts. In addition, this population is known to be expanded during metabolic disease. The objective of this study was to determine the role of MDSCs in the context of OA pathophysiology. METHODS In this study, we examined the differentiation and functional capacity of MDSCs to become osteoclasts in vitro and in vivo using mouse models of OA and in MDSC quantitation in humans with OA pathology relative to obesity status. RESULTS We observed that MDSCs are expanded in mice and humans during obesity. MDSCs were expanded in peripheral blood of OA subjects relative to body mass index and in mice fed a high-fat diet (HFD) compared to mice fed a low-fat diet (LFD). In mice, monocytic MDSC (M-MDSC) was expanded in diet-induced obesity (DIO) with a further expansion after destabilization of the medial meniscus (DMM) surgery to induce post-traumatic OA (PTOA) (compared to sham-operated controls). M-MDSCs from DIO mice had a greater capacity to form osteoclasts in culture with increased subchondral bone osteoclast number. In humans, we observed an expansion of M-MDSCs in peripheral blood and synovial fluid of obese subjects compared to lean subjects with OA. CONCLUSION These data suggest that MDSCs are reprogrammed in metabolic disease, with the potential to contribute towards OA progression and severity.
Collapse
Affiliation(s)
- Lixia Zhang
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, 645 Biomedical Research Building, 3435 Main St, Buffalo, NY, 14214-8006, USA
| | - Cameron L Kirkwood
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, 645 Biomedical Research Building, 3435 Main St, Buffalo, NY, 14214-8006, USA
| | - Jiho Sohn
- Department of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Ashley Lau
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, 645 Biomedical Research Building, 3435 Main St, Buffalo, NY, 14214-8006, USA
| | | | - Supinder Kour Bali
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Western Bone and Joint Institute, University of Western Ontario, London, Ontario, Canada
| | - Sridhar Rachala
- Department of Orthopaedics, University at Buffalo, Buffalo, NY, USA
| | - John M Marzo
- Department of Orthopaedics, University at Buffalo, Buffalo, NY, USA
| | - Mark J Anders
- Department of Orthopaedics, University at Buffalo, Buffalo, NY, USA
| | - Frank Beier
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Western Bone and Joint Institute, University of Western Ontario, London, Ontario, Canada
| | - Keith L Kirkwood
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, 645 Biomedical Research Building, 3435 Main St, Buffalo, NY, 14214-8006, USA. .,Department of Head and Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
28
|
Yajun W, Jin C, Zhengrong G, Chao F, Yan H, Weizong W, Xiaoqun L, Qirong Z, Huiwen C, Hao Z, Jiawei G, Xinchen Z, Shihao S, Sicheng W, Xiao C, Jiacan S. Betaine Attenuates Osteoarthritis by Inhibiting Osteoclastogenesis and Angiogenesis in Subchondral Bone. Front Pharmacol 2021; 12:723988. [PMID: 34658862 PMCID: PMC8511433 DOI: 10.3389/fphar.2021.723988] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/13/2021] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis with no effective therapy. Subchondral bone and overlying articular cartilage are closely associated and function as “osteo-chondral unit” in the joint. Abnormal mechanical load leads to activated osteoclast activity and increased bone resorption in the subchondral bone, which is implicated in the onset of OA pathogenesis. Thus, inhibiting subchondral bone osteoclast activation could prevent OA onset. Betaine, isolated from the Lycii Radicis Cortex (LRC), has been demonstrated to exert anti-inflammatory, antifibrotic and antiangiogenic properties. Here, we evaluated the effects of betaine on anterior cruciate ligament transection (ACLT)-induced OA mice. We observed that betaine decreased the number of matrix metalloproteinase 13 (MMP-13)-positive and collagen X (Col X)-positive cells, prevented articular cartilage proteoglycan loss and lowered the OARSI score. Betaine decreased the thickness of calcified cartilage and increased the expression level of lubricin. Moreover, betaine normalized uncoupled subchondral bone remodeling as defined by lowered trabecular pattern factor (Tb.pf) and increased subchondral bone plate thickness (SBP). Additionally, aberrant angiogenesis in subchondral bone was blunted by betaine treatment. Mechanistically, we demonstrated that betaine suppressed osteoclastogenesis in vitro by inhibiting reactive oxygen species (ROS) production and subsequent mitogen-activated protein kinase (MAPK) signaling. These data demonstrated that betaine attenuated OA progression by inhibiting hyperactivated osteoclastogenesis and maintaining microarchitecture in subchondral bone.
Collapse
Affiliation(s)
- Wang Yajun
- Graduate Management Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Cui Jin
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Gu Zhengrong
- Department of Orthopedics, Luodian Hospital, Shanghai, China
| | - Fang Chao
- Graduate Management Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hu Yan
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China.,Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Weng Weizong
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Li Xiaoqun
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhou Qirong
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chen Huiwen
- Graduate Management Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhang Hao
- Graduate Management Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guo Jiawei
- Graduate Management Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhuang Xinchen
- Graduate Management Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Sheng Shihao
- Graduate Management Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wang Sicheng
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,Department of Orthopedics, Zhongye Hospital, Shanghai, China
| | - Chen Xiao
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Su Jiacan
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China.,Institute of Translational Medicine, Shanghai University, Shanghai, China.,Shanghai Clinical Research Center for Aging and Medicine, Shanghai, China
| |
Collapse
|
29
|
Chen T, Weng W, Liu Y, Aspera-Werz RH, Nüssler AK, Xu J. Update on Novel Non-Operative Treatment for Osteoarthritis: Current Status and Future Trends. Front Pharmacol 2021; 12:755230. [PMID: 34603064 PMCID: PMC8481638 DOI: 10.3389/fphar.2021.755230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 09/06/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis (OA) is a leading cause of pain and disability which results in a reduced quality of life. Due to the avascular nature of cartilage, damaged cartilage has a finite capacity for healing or regeneration. To date, conservative management, including physical measures and pharmacological therapy are still the principal choices offered for OA patients. Joint arthroplasties or total replacement surgeries are served as the ultimate therapeutic option to rehabilitate the joint function of patients who withstand severe OA. However, these approaches are mainly to relieve the symptoms of OA, instead of decelerating or reversing the progress of cartilage damage. Disease-modifying osteoarthritis drugs (DMOADs) aiming to modify key structures within the OA joints are in development. Tissue engineering is a promising strategy for repairing cartilage, in which cells, genes, and biomaterials are encompassed. Here, we review the current status of preclinical investigations and clinical translations of tissue engineering in the non-operative treatment of OA. Furthermore, this review provides our perspective on the challenges and future directions of tissue engineering in cartilage regeneration.
Collapse
Affiliation(s)
- Tao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Trauma and Reconstructive Surgery, BG Trauma Center Tübingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Weidong Weng
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tübingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Yang Liu
- Department of Clinical Sciences, Orthopedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Romina H Aspera-Werz
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tübingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas K Nüssler
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tübingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Jianzhong Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Oo WM, Little C, Duong V, Hunter DJ. The Development of Disease-Modifying Therapies for Osteoarthritis (DMOADs): The Evidence to Date. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2921-2945. [PMID: 34262259 PMCID: PMC8273751 DOI: 10.2147/dddt.s295224] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/16/2021] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) is a complex heterogeneous articular disease with multiple joint tissue involvement of varying severity and no regulatory-agency-approved disease-modifying drugs (DMOADs). In this review, we discuss the reasons necessitating the development of DMOADs for OA management, the classifications of clinical phenotypes or molecular/mechanistic endotypes from the viewpoint of targeted drug discovery, and then summarize the efficacy and safety profile of a range of targeted drugs in Phase 2 and 3 clinical trials directed to cartilage-driven, bone-driven, and inflammation-driven endotypes. Finally, we briefly put forward the reasons for failures in OA clinical trials and possible steps to overcome these barriers.
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, Australia.,Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar
| | - Christopher Little
- Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Vicky Duong
- 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, Australia
| | - 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, Australia
| |
Collapse
|
31
|
Wang T, Guo Y, Shi XW, Gao Y, Zhang JY, Wang CJ, Yang X, Shu Q, Chen XL, Fu XY, Xie WS, Zhang Y, Li B, Guo CQ. Acupotomy Contributes to Suppressing Subchondral Bone Resorption in KOA Rabbits by Regulating the OPG/RANKL Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:8168657. [PMID: 34335838 PMCID: PMC8298142 DOI: 10.1155/2021/8168657] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/11/2020] [Accepted: 04/11/2021] [Indexed: 01/19/2023]
Abstract
Subchondral bone lesions, as the crucial inducement for accelerating cartilage degeneration, have been considered as the initiating factor and the potential therapeutic target of knee osteoarthritis (KOA). Acupotomy, the biomechanical therapy guided by traditional Chinese meridians theory, alleviates cartilage deterioration by correcting abnormal mechanics. Whether this mechanical effect of acupotomy inhibits KOA subchondral bone lesions is indistinct. This study aimed to investigate the effects of acupotomy on inhibiting subchondral bone resorption and to define the possible mechanism in immobilization-induced KOA rabbits. After KOA modeling, 8 groups of rabbits (4w/6w acupotomy, 4w/6w electroacupuncture, 4w/6w model, and 4w/6w control groups) received the indicated intervention for 3 weeks. Histological and bone histomorphometry analyses revealed that acupotomy prevented both cartilage surface erosion and subchondral bone loss. Further, acupotomy suppressed osteoclast activity and enhanced osteoblast activity in KOA subchondral bone, showing a significantly decreased expression of tartrate-resistant acid phosphatase (TRAP), matrix metalloproteinases-9 (MMP-9), and cathepsin K (Ctsk) and a significantly increased expression of osteocalcin (OCN); this regulation may be mediated by blocking the decrease in osteoprotegerin (OPG) and the increase in NF-κB receptor activated protein ligand (RANKL). These findings indicated that acupotomy inhibited osteoclast activity and promoted osteoblast activity to ameliorate hyperactive subchondral bone resorption and cartilage degeneration in immobilization-induced KOA rabbits, which may be mediated by the OPG/RANKL signaling pathway. Taken together, our results indicate that acupotomy may have therapeutic potential in KOA by restoring the balance between bone formation and bone resorption to attenuate subchondral bone lesions.
Collapse
Affiliation(s)
- Tong Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan Guo
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated with Capital Medical University, Beijing 100010, China
| | - Xiao-Wei Shi
- Massage Department, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Gao
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jia-Yi Zhang
- Traditional Chinese Medicine Department, Beijing Nankou Hospital, Beijing 102200, China
| | - Chun-Jiu Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xue Yang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qi Shu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xi-Lin Chen
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xin-Yi Fu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wen-Shan Xie
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yi Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Bin Li
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine affiliated with Capital Medical University, Beijing 100010, China
| | - Chang-Qing Guo
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
32
|
From Pathogenesis to Therapy in Knee Osteoarthritis: Bench-to-Bedside. Int J Mol Sci 2021; 22:ijms22052697. [PMID: 33800057 PMCID: PMC7962130 DOI: 10.3390/ijms22052697] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 03/04/2021] [Indexed: 12/22/2022] Open
Abstract
Osteoarthritis (OA) is currently the most widespread musculoskeletal condition and primarily affects weight-bearing joints such as the knees and hips. Importantly, knee OA remains a multifactorial whole-joint disease, the appearance and progression of which involves the alteration of articular cartilage as well as the synovium, subchondral bone, ligaments, and muscles through intricate pathomechanisms. Whereas it was initially depicted as a predominantly aging-related and mechanically driven condition given its clear association with old age, high body mass index (BMI), and joint malalignment, more recent research identified and described a plethora of further factors contributing to knee OA pathogenesis. However, the pathogenic intricacies between the molecular pathways involved in OA prompted the study of certain drugs for more than one therapeutic target (amelioration of cartilage and bone changes, and synovial inflammation). Most clinical studies regarding knee OA focus mainly on improvement in pain and joint function and thus do not provide sufficient evidence on the possible disease-modifying properties of the tested drugs. Currently, there is an unmet need for further research regarding OA pathogenesis as well as the introduction and exhaustive testing of potential disease-modifying pharmacotherapies in order to structure an effective treatment plan for these patients.
Collapse
|
33
|
Fernández-Martín S, López-Peña M, Muñoz F, Permuy M, González-Cantalapiedra A. Bisphosphonates as disease-modifying drugs in osteoarthritis preclinical studies: a systematic review from 2000 to 2020. Arthritis Res Ther 2021; 23:60. [PMID: 33618776 PMCID: PMC7898436 DOI: 10.1186/s13075-021-02446-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/11/2021] [Indexed: 01/26/2023] Open
Abstract
Bisphosphonates have been proposed as possible disease-modifying drugs in osteoarthritis. However, the evidence of their efficacy is poor and their outcomes presented a great heterogeneity. Therefore, the aim of this study is to systematically review the main effects of bisphosphonate use on synovial joint tissues and biochemical markers in preclinical studies over the past two decades (2000-2020). Three databases (Pubmed, Scopus, and Web of Science) were searched, and after screening, twenty-six studies with five different types of bisphosphonates were included in the review. The animal model selected, the type of bisphosphonate used, the therapy duration, and the main effects of individual drugs on synovial tissues were evaluated. Additionally, the quality and risk of bias assessments were performed using the Animals in Research Reporting In Vivo Experiments guidelines and the Systematic Review Centre for Laboratory animal Experimentation tool. Studies showed high variability in experimental designs. Consequently, the comparison of the findings in order to draw specific conclusions about the effectiveness of the drugs is complicated. However, the results of this systematic review suggested that bisphosphonates seemed to reduce the osteoarthritic changes in a dose-dependent manner showing better chondroprotective effects at high doses. Besides, a time-dependent efficacy was also detected in terms of cartilage status. One can conclude that the disease stage of the time-point of treatment initiation may constitute a key factor in the antiresorptive drug efficacy. Generally, we noted that bisphosphonate administration seemed to show positive subchondral bone conservation and fewer biomarker alterations. However, they did not appear to suppress the osteophyte development and their chondroprotective effect is highly variable among the studies. Bisphosphonates appeared to show a positive anti-inflammatory effect on the synovial membrane. However, only a few included publications were focused on their investigation. Regarding the therapy duration, there is a significant lack of evidence on evaluating their effectiveness in preclinical long-term studies and further experimental studies may be needed to examine the pharmacological response in these circumstances. This systematic review might help to clarify the efficacy of bisphosphonates and their function as disease-modifying treatments in osteoarthritis.
Collapse
Affiliation(s)
- Silvia Fernández-Martín
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002, Lugo, Spain.
| | - Mónica López-Peña
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002, Lugo, Spain
| | - Fernando Muñoz
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002, Lugo, Spain
| | - María Permuy
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002, Lugo, Spain
| | - Antonio González-Cantalapiedra
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002, Lugo, Spain
| |
Collapse
|
34
|
He Z, Li H, Han X, Zhou F, Du J, Yang Y, Xu Q, Zhang S, Zhang S, Zhao N, Yan M, Yu Z. Irisin inhibits osteocyte apoptosis by activating the Erk signaling pathway in vitro and attenuates ALCT-induced osteoarthritis in mice. Bone 2020; 141:115573. [PMID: 32768686 DOI: 10.1016/j.bone.2020.115573] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/20/2020] [Accepted: 07/26/2020] [Indexed: 01/29/2023]
Abstract
Moderate exercise can alleviate symptoms of osteoarthritis (OA) such as pain, stiffness, and joint deformities that are associated with progressive cartilaginous degeneration, osteophyte formation, subchondral bone changes, and synovial inflammation. Irisin is an exercise-related myokine that reportedly plays a crucial role in bone remodeling. However, its role in OA remains unknown. This study aimed to determine whether irisin can attenuate OA progression and the mechanism of its therapeutic effect. Three-month-old male C57BL/6J mice were randomized to groups that underwent sham operation, and anterior cruciate ligament transection (ACLT) intraperitoneally injected with vehicle or irisin in vivo. Apoptosis was induced by stretching murine osteocyte-like MLO-Y4 cells in vitro. Irisin reduced wear, maintained the proportion of hyaline cartilage, a more complete cartilage structure, and lower Osteoarthritis Research Society International (OARSI) scores at 4 weeks after ACLT. Irisin reduced the expression of matrix metalloproteinase (MMP)-13 in cartilage and caspase 3 in the subchondral bone. Irisin exerted rescue effects in microstructural parameters of subchondral trabecular bone including bone volume fraction (BV/TV), trabecular number (Tb.N), connection density (Conn. D), and the structure model index (SMI) compared with ACLT-vehicle group. Bone histomorphometry showed that irisin increased subchondral bone remodeling. The decreasing ratio (%) of the eroded surface (ES/BS) was reversed by irisin in the ACLT+vehicle group. Staining with tartrate-resistant acid phosphatase showed a decreased number of osteoclasts. Irisin significantly increased the proliferation of osteocytes, protected them from apoptosis, and maintained cellular activity by regulating the expression of Bax, Bcl-2, and osteoprotegerin/receptor activator of nuclear factor (NF)-kB-ligand (OPG/Rankl). Irisin activated serine/threonine-selective protein kinases (Erk) and p38 signaling, and its anti-apoptosis function depended on the Erk signaling pathway. Irisin attenuated OA progression by decreasing osteocyte apoptosis and improving the microarchitecture of subchondral bone. Activation of the Erk pathway by irisin plays an important role in reducing osteocyte apoptosis in vitro.
Collapse
Affiliation(s)
- Zihao He
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hanjun Li
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xuequan Han
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Feng Zhou
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jingke Du
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yiqi Yang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qi Xu
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shuhong Zhang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shuangyan Zhang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Ning Zhao
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mengning Yan
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
35
|
Blaker CL, Ashton DM, Doran N, Little CB, Clarke EC. Sex- and injury-based differences in knee biomechanics in mouse models of post-traumatic osteoarthritis. J Biomech 2020; 114:110152. [PMID: 33285491 DOI: 10.1016/j.jbiomech.2020.110152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 01/14/2023]
Abstract
Sex and joint injury are risk factors implicated in the onset and progression of osteoarthritis (OA). In mouse models of post-traumatic OA (ptOA), the pathogenesis of disease is notably impacted by sex (often worse in males) and injury model (e.g. meniscal versus ligament injury). Increasing ptOA progression and severity is often associated with greater relative instability of the joint but few studies have directly quantified changes in joint mechanics after injury and compared outcomes across multiple models in both male and female mice. Passive anterior-posterior knee biomechanics were evaluated in 10-week-old, male and female C57BL/6J mice. PtOA injury models included destabilisation of the medial meniscus (DMM), anterior cruciate ligament transection (ACLT) or mechanical rupture (ACLR), and combined DMM and ACLT (DMM + ACLT). Sham operated and non-operated controls (NOC) were included for baseline comparisons. The test apparatus loaded hindlimbs at 60° flexion between ± 1 N at 0.5 mm/s (build specifications available for download: https://doi.org/10.17632/z754455x3c.1). Measures of joint laxity (range of motion, neutral zone) and stiffness were calculated. Joint laxity was comparable between male and female mice while joint stiffness was greater in females (P ≤ 0.002, correcting for body-mass and injury-model). Anterior-posterior joint mechanics were minimally altered by DMM but significantly affected by loss of the ACL (P < 0.001), with equivalent changes between ACL-injury models despite different injury mechanisms and adjacent meniscal damage. These findings suggest that despite the important role of joint injury; sex- and model-specific differences in ptOA progression and severity are not primarily driven by altered anterior-posterior knee biomechanics.
Collapse
Affiliation(s)
- Carina L Blaker
- Murray Maxwell Biomechanics Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, University of Sydney, St. Leonards, New South Wales, Australia; Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, University of Sydney, St. Leonards, New South Wales, Australia
| | - Dylan M Ashton
- Murray Maxwell Biomechanics Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, University of Sydney, St. Leonards, New South Wales, Australia
| | - Nathan Doran
- Murray Maxwell Biomechanics Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, University of Sydney, St. Leonards, New South Wales, Australia; School of Biomedical Engineering, University of New South Wales, Kensington, New South Wales, Australia
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, University of Sydney, St. Leonards, New South Wales, Australia
| | - Elizabeth C Clarke
- Murray Maxwell Biomechanics Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, University of Sydney, St. Leonards, New South Wales, Australia.
| |
Collapse
|
36
|
Induced Pluripotent Stem Cell-Differentiated Chondrocytes Repair Cartilage Defect in a Rabbit Osteoarthritis Model. Stem Cells Int 2020; 2020:8867349. [PMID: 33224204 PMCID: PMC7671807 DOI: 10.1155/2020/8867349] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
The aim of this study was to explore the therapeutic effect of iPSC-mesenchymal stem cell (MSC)-derived chondrocytes in a rabbit osteoarthritis (OA) model. The iPSCs were characterized by gene expressions, immunostaining of pluripotent markers, and in vivo teratoma formation. iPSC-differentiated MSCs were characterized by flow cytometry and trilineage differentiation. A rabbit OA model was established by the transection of the anterior cruciate ligament. The therapeutic effect of transplanted iPSC-MSC-chondrocytes on the OA was evaluated by the histology, immunostaining, and qPCR of defective cartilage. The results showed iPSC could express pluripotency markers such as OCT4, SOX2, and NANOG and form an embryoid body and a teratoma. After differentiation of iPSCs for 30 days, MSCs were established. The iPSC-MSC could express typical MSC markers such as CD29, CD44, CD90, CD105, and HLA-ABC. They could differentiate into adipocytes, osteocytes, and chondrocytes. In this model, iPSC-MSC-chondrocytes significantly improved the histology and ICRS (International Cartilage Repair Society) scores. The transplanted cartilage expressed less IL-1β, TNF-α, and MMP13 than control cartilage. In conclusion, the iPSCs we derived might represent an emerging source for differentiated MSC-chondrocyte and might rescue cartilage defects through its anti-inflammatory and anti-catabolic effects.
Collapse
|
37
|
Abstract
The prevalence of osteoarthritis (OA) and the burden associated with the disease are steadily increasing worldwide, representing a major public health challenge for the coming decades. The lack of specific treatments for OA has led to it being recognized as a serious disease that has an unmet medical need. Advances in the understanding of OA pathophysiology have enabled the identification of a variety of potential therapeutic targets involved in the structural progression of OA, some of which are promising and under clinical investigation in randomized controlled trials. Emerging therapies include those targeting matrix-degrading proteases or senescent chondrocytes, promoting cartilage repair or limiting bone remodelling, local low-grade inflammation or Wnt signalling. In addition to these potentially disease-modifying OA drugs (DMOADs), several targets are being explored for the treatment of OA-related pain, such as nerve growth factor inhibitors. The results of these studies are expected to considerably reshape the landscape of OA management over the next few years. This Review describes the pathophysiological processes targeted by emerging therapies for OA, along with relevant clinical data and discussion of the main challenges for the further development of these therapies, to provide context for the latest advances in the field of pharmaceutical therapies for OA.
Collapse
|
38
|
Migliore A, Paoletta M, Moretti A, Liguori S, Iolascon G. The perspectives of intra-articular therapy in the management of osteoarthritis. Expert Opin Drug Deliv 2020; 17:1213-1226. [PMID: 32543240 DOI: 10.1080/17425247.2020.1783234] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Management of Osteoarthritis (OA) still is a challenge for clinicians. Taking into account a multidisciplinary approach including pharmacological and non-pharmacological treatments, intra-articular (IA) injection could be considered as an effective local therapy. Areas covered This review provides a new perspective of IA treatment going beyond current available IA agents. We describe novel biological targets for developing new IA agents and innovative modalities of delivery systems. Additional topics include predictors of response for a better choice of IA agents for each patient, diagnostic and prognostic role of biomarkers, accuracy of IA injection, and cost-effectiveness of IA injection. Expert opinion IA treatments seem to be very promising for the management of OA. Identifying clinical and biochemical predictive factors could drive clinician to the appropriate therapeutic approach. To date, there is a gap regarding the benefit of IA treatments in the 'real practice' once they have been adopted. However, considering these promising effects of IA approach, several open questions remain not clarified.
Collapse
Affiliation(s)
| | - Marco Paoletta
- Department of Medical and Surgical Specialties and Dentistry, University of Campania "Luigi Vanvitelli" , Naples, Italy
| | - Antimo Moretti
- Department of Medical and Surgical Specialties and Dentistry, University of Campania "Luigi Vanvitelli" , Naples, Italy
| | - Sara Liguori
- Department of Medical and Surgical Specialties and Dentistry, University of Campania "Luigi Vanvitelli" , Naples, Italy
| | - Giovanni Iolascon
- Department of Medical and Surgical Specialties and Dentistry, University of Campania "Luigi Vanvitelli" , Naples, Italy
| |
Collapse
|
39
|
Trivedi S, Fang W, Ayyalasomayajula I, Vangsness CT. Pharmacotherapeutic considerations and options for the management of osteoarthritis in women. Expert Opin Pharmacother 2020; 21:557-566. [DOI: 10.1080/14656566.2020.1718649] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sunny Trivedi
- University of Florida College of Medicine, Gainesville, FL, USA
| | - William Fang
- Department of Orthopaedic Surgery, Keck School of Medicine USC, Los Angeles, CA, USA
| | | | - C. Thomas Vangsness
- Department of Orthopaedic Surgery, Keck School of Medicine USC, Los Angeles, CA, USA
| |
Collapse
|
40
|
Zhou F, Mei J, Yang S, Han X, Li H, Yu Z, Qiao H, Tang T. Modified ZIF-8 Nanoparticles Attenuate Osteoarthritis by Reprogramming the Metabolic Pathway of Synovial Macrophages. ACS APPLIED MATERIALS & INTERFACES 2020; 12:2009-2022. [PMID: 31849213 DOI: 10.1021/acsami.9b16327] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Accumulating evidence suggests that activation of proinflammatory M1-type macrophages in the synovium plays a vital role in the progression of osteoarthritis (OA). Redundant nitric oxide (NO) and hydrogen peroxide (H2O2) are key factors that drive macrophages to polarize to the M1 type. Herein, modified zeolitic imidazolate framework-8 (ZIF-8) nanoparticles (NPs) have been synthesized. By regulating intracellular gases and reprogramming the metabolism phenotype, modified NPs transformed macrophage polarization from proinflammatory M1 to anti-inflammatory M2 phenotype. Specifically, S-methylisothiourea hemisulfate salt was loaded into ZIF-8 NPs to inhibit inducible nitric oxide synthase, hence reducing NO production. Catalase was encapsulated to catalyze the production of oxygen (O2) from H2O2. Results demonstrated that modified NPs were capable of catalyzing H2O2 to produce O2 and eliminate NO, hence inhibiting hypoxia-inducible factor 1α, further rescuing mitochondrial function. Moreover, anti-CD16/32 antibody modification could prolong the retention time of NPs in knee joints of OA mice with anterior cruciate ligament transection. More significantly, modified NPs suppressed M1 macrophages and up-regulated M2 macrophage infiltration in the synovium, further inhibiting cartilage degeneration. This ZIF-8 NP-based gas regulation and metabolic reprogramming strategy may pave a new avenue for OA treatment.
Collapse
Affiliation(s)
- Feng Zhou
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , China
| | - Jingtian Mei
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , China
| | - Xiuguo Han
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , China
| | - Hanjun Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , China
| | - Han Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200011 , China
| |
Collapse
|
41
|
Hyaluronan suppresses enhanced cathepsin K expression via activation of NF-κB with mechanical stress loading in a human chondrocytic HCS-2/8 cells. Sci Rep 2020; 10:216. [PMID: 31937805 PMCID: PMC6959248 DOI: 10.1038/s41598-019-57073-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022] Open
Abstract
Cathepsin K is a protease known to be involved in not only bone remodeling and resorption, but also articular cartilage degradation that leads to osteoarthritis (OA). Hyaluronan (HA) plays a pivotal role in maintaining homeostasis within articular chondrocytes. Intra-articular supplementation of high molecular weight hyaluronan (HMW-HA) has been widely used in OA treatment. However, its prospective mechanism of action is still unclear. In this study, we examined the suppressive effect of HA on enhanced cathepsin K expression induced by mechanical stress loading. A human chondrocytic HCS-2/8 cells were cultured in silicon chambers and subjected to cyclic tensile stress (CTS) loading. CTS loading significantly increased messenger ribonucleic acid and protein expression of cathepsin K, which appeared to be suppressed by pre-treatment with HMW-HA. Activation of nuclear factor-kappa B (NF-κB) was induced by CTS loading, and suppressed by pre-treatment with HMW-HA. Helenalin, a chemical inhibitor of NF-κB, clearly suppressed the enhanced expression of cathepsin K, as well as NF-κB activation induced by CTS loading. The suppressive effect of HMW-HA on enhanced cathepsin K expression via NF-κB inhibition impacts the effectiveness of HMW-HA in OA treatment. Our findings provide new evidence supporting the biological effectiveness of intra-articular HMW-HA injections for treatment of OA.
Collapse
|
42
|
Dai R, Wu Z, Chu HY, Lu J, Lyu A, Liu J, Zhang G. Cathepsin K: The Action in and Beyond Bone. Front Cell Dev Biol 2020; 8:433. [PMID: 32582709 PMCID: PMC7287012 DOI: 10.3389/fcell.2020.00433] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/08/2020] [Indexed: 02/05/2023] Open
Abstract
Cathepsin K (CatK) is one of the most potent proteases in lysosomal cysteine proteases family, of which main function is to mediate bone resorption. Currently, CatK is among the most attractive targets for anti-osteoporosis drug development. Although many pharmaceutical companies are working on the development of selective inhibitors for CatK, there is no FDA approved drug till now. Odanacatib (ODN) developed by Merck & Co. is the only CatK inhibitor candidate which demonstrated high therapeutic efficacy in patients with postmenopausal osteoporosis in Phase III clinical trials. Unfortunately, the development of ODN was finally terminated due to the cardio-cerebrovascular adverse effects. Therefore, it arouses concerns on the undesirable CatK inhibition in non-bone sites. It is known that CatK has far-reaching actions throughout various organs besides bone. Many studies have also demonstrated the involvement of CatK in various diseases beyond the musculoskeletal system. This review not only summarized the functional roles of CatK in bone and beyond bone, but also discussed the potential relevance of the CatK action beyond bone to the adverse effects of inhibiting CatK in non-bone sites.
Collapse
Affiliation(s)
- Rongchen Dai
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Zeting Wu
- International Medical Service Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Hang Yin Chu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Jun Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
- *Correspondence: Jin Liu,
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
- Ge Zhang,
| |
Collapse
|
43
|
Atobe M. Activation of Transient Receptor Potential Vanilloid (TRPV) 4 as a Therapeutic Strategy in Osteoarthritis. Curr Top Med Chem 2019; 19:2254-2267. [DOI: 10.2174/1568026619666191010162850] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/21/2019] [Accepted: 09/13/2019] [Indexed: 01/29/2023]
Abstract
Transient receptor potential vanilloid (TRPV) 4 belongs to the TRPV subfamily of TRP ion
channels. TRPV4 channels play a critical role in chondrocytes and thus TRPV4 is an attractive target of
Disease-Modifying Osteoarthritis Drugs (DMOADs). Initial investigations of small molecules by Glaxo
Smith Klein (GSK) as both agonists and antagonists via oral/intravenous administration have led to the
use of existing agonists as lead compounds for biological studies. Our recent results suggest that local
injection of a TRPV4 agonist is a potential treatment for osteoarthritis (OA). This review briefly summarizes
updates regarding TRPV4 agonists based on recent advances in drug discovery, and particularly
the local administration of TRPV4 agonists.
Collapse
Affiliation(s)
- Masakazu Atobe
- Laboratory for Medicinal Chemistry, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| |
Collapse
|
44
|
Soliman SA, Kamal BM, Abd-Elhafeez HH. Cellular Invasion and Matrix Degradation, a Different Type of Matrix-Degrading Cells in the Cartilage of Catfish ( Clarias gariepinus) and Japanese Quail Embryos ( Coturnix coturnix japonica). MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2019; 25:1283-1292. [PMID: 31583991 DOI: 10.1017/s1431927619014892] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We previously studied the phenomena of the mesenchymal cell-dependent mode of cartilage growth in quail and catfish. Thus, we selected the two cartilage models in which mesenchymal cells participate in their growth. In such models, cartilage degradation occurred to facilitate cellular invasion. The studies do not explain the nature of the cartilage degrading cells. The current study aims to explore the nature of the cartilage-degrading cells using transmission electron microscopy (TEM) and immunohistochemistry. Samples of cartilage have been isolated from the air-breathing organ of catfish and the cartilage of the prospective occipital bone of quail embryos. Samples have been processed for TEM and immunohistochemistry. We found that two different cell types are involved in cartilage degradation; the macrophage in the cartilage of catfish and mesenchymal cells in the cartilage of the quail. Areas of cellular invasion in both catfish cartilage and quail embryo cartilage had an immunological affinity for MMP-9. In catfish, cartilage-degrading cells had identical morphological features of macrophages, whereas in quail embryos, cartilage-degrading cells were mesenchymal-like cells which had cell processes rich in vesicles and expressed CD117. Further study should consider the role of macrophage and mesenchymal cells during cartilage degradation. This could be valuable to be applied to remove the defective cartilage matrix formed in osteoarthritic patients to improve cartilage repair strategies.
Collapse
Affiliation(s)
- Soha A Soliman
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Basma Mohamed Kamal
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Sadat-City University, Sadat City, Egypt
| | - Hanan H Abd-Elhafeez
- Department of Anatomy, Embryology and Histology, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
45
|
Kinsenoside attenuates osteoarthritis by repolarizing macrophages through inactivating NF- κB/MAPK signaling and protecting chondrocytes. Acta Pharm Sin B 2019; 9:973-985. [PMID: 31649847 PMCID: PMC6804452 DOI: 10.1016/j.apsb.2019.01.015] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/14/2018] [Accepted: 01/28/2019] [Indexed: 12/13/2022] Open
Abstract
The objective was to investigate the effect of kinsenoside (Kin) treatments on macrophage polarity and evaluate the resulting protection of chondrocytes to attenuate osteoarthritis (OA) progression. RAW264.7 macrophages were polarized to M1/M2 subtypes then administered with different concentrations of Kin. The polarization transitions were evaluated with quantitative real-time polymerase chain reaction (qRT-PCR), confocal observation and flow cytometry analysis. The mechanism of Kin repolarizing M1 macrophages was evaluated by Western blot. Further, macrophage conditioned medium (CM) and IL-1β were administered to chondrocytes. Micro-CT scanning and histological observations were conducted in vivo on anterior cruciate ligament transection (ACLT) mice with or without Kin treatment. We found that Kin repolarized M1 macrophages to the M2 phenotype. Mechanistically, Kin inhibited the phosphorylation of IκBα, which further reduced the downstream phosphorylation of P65 in nuclear factor-κB (NF-κB) signaling. Moreover, Kin inhibited mitogen-activated protein kinases (MAPK) signaling molecules p-JNK, p-ERK and p-P38. Additionally, Kin attenuated macrophage CM and IL-1β-induced chondrocyte damage. In vivo, Kin reduced the infiltration of M1 macrophages, promoted M2 macrophages in the synovium, inhibited subchondral bone destruction and reduced articular cartilage damage induced by ACLT. All the results indicated that Kin is an effective therapeutic candidate for OA treatment.
Collapse
Key Words
- AP-1, activator protein-1
- Arg-1, arginase-1
- BV, bone volume
- BV/TV, bone volume/total tissue volume
- C/EBP β, CCAAT/enhancer-binding protein β
- CM, conditioned medium
- Chondrocytes
- DMEM, Dulbecco׳s minimum essential medium
- GA, gouty arthritis
- H&E, hematoxylin & eosin
- HUVECs, human umbilical vein endothelial cells
- IFN-γ, interferon-γ
- IRF4, interferon regulatory factor 4
- Kin, kinsenoside
- Kinsenoside
- LPS, lipopolysaccharides
- MAPK, mitogen-activated protein kinases
- MSU, monosodium urate
- Macrophages
- NF-κB, nuclear factor-κB
- NSAIDs, non-steroidal anti-inflammatory drugs
- OA, osteoarthritis
- OARSI, Osteoarthritis Research Society International
- Osteoarthritis
- PPARγ, peroxisome proliferator-activated receptor γ
- Polarization
- RA, rheumatoid arthritis
- ROS, reactive oxygen species
- S&F, safranin O-fast green
- TLRs, toll-like receptors
- TNF-α, tumor necrosis factor-α
- Tb.N, trabecular number
- Tb.Sp, trabecular separation
- Tb.Th, trabecular thickness
- iNOS, inducible nitric oxide synthase
Collapse
|
46
|
Zhou F, Mei J, Yuan K, Han X, Qiao H, Tang T. Isorhamnetin attenuates osteoarthritis by inhibiting osteoclastogenesis and protecting chondrocytes through modulating reactive oxygen species homeostasis. J Cell Mol Med 2019; 23:4395-4407. [PMID: 30983153 PMCID: PMC6533508 DOI: 10.1111/jcmm.14333] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/22/2019] [Accepted: 03/19/2019] [Indexed: 12/20/2022] Open
Abstract
Increasing evidence indicates that osteoarthritis (OA) is a musculoskeletal disease affecting the whole joint, including both cartilage and subchondral bone. Reactive oxygen species (ROS) have been demonstrated to be one of the important destructive factors during early‐stage OA development. The objective of this study was to investigate isorhamnetin (Iso) treatment on osteoclast formation and chondrocyte protection to attenuate OA by modulating ROS. Receptor activator of nuclear factor‐kappa B ligand (RANKL) was used to establish the osteoclast differentiation model in bone marrow macrophages (BMMs) in vivo. H2O2 was used to induce ROS, which could further cause chondrocyte apoptosis. We demonstrated that Iso suppressed RANKL‐induced ROS generation, which could mediate osteoclastogenesis. Moreover, we found that Iso inhibited osteoclast formation and function by suppressing the expression of osteoclastogenesis‐related genes and proteins. We proved that Iso inhibited RANKL‐induced activation of mitogen‐activated protein kinase activation of mitogen‐activated protein kinase (MAPK), nuclear factor‐kappa B (NF‐κB) and AKT signalling pathways in BMMs. In addition, Iso inhibited ROS‐induced chondrocyte apoptosis by regulating apoptosis‐related proteins. Moreover, Iso was administered to an anterior cruciate ligament transection (ACLT)‐induced OA mouse model. The results indicated that Iso exerted beneficial effects on inhibiting excessive osteoclast activity and chondrocyte apoptosis, which further remedied cartilage damage. Overall, our data showed that Iso is an effective candidate for treating OA.
Collapse
Affiliation(s)
- Feng Zhou
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. of China
| | - Jingtian Mei
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. of China
| | - Kai Yuan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. of China
| | - Xiuguo Han
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. of China
| | - Han Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. of China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. of China
| |
Collapse
|
47
|
Design, synthesis and biological evaluation of inhibitors of cathepsin K on dedifferentiated chondrocytes. Bioorg Med Chem 2019; 27:1034-1042. [DOI: 10.1016/j.bmc.2019.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/27/2019] [Accepted: 02/01/2019] [Indexed: 01/18/2023]
|
48
|
Ma B, Wesolowski G, Luo B, Lifsted T, Wessner K, Adamson G, Glantschnig H, Lubbers LS. Suppression of cathepsin K biomarker in synovial fluid as a free-drug-driven process. J Circ Biomark 2019; 8:1849454418821819. [PMID: 30671145 PMCID: PMC6327326 DOI: 10.1177/1849454418821819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 12/01/2018] [Indexed: 11/17/2022] Open
Abstract
Cathepsin K (CatK) inhibitors exhibited chondroprotective and pain-reducing effects in animal models, however, improvements were relatively modest at dose levels achieving maximal suppression of CatK biomarkers in urine. In this report, a previously characterized CatK inhibitor (MK-1256) is utilized to explore the potential of reduced target engagement and/or suboptimal exposure (free drug) as limiting factors to the pharmacological potential of CatK inhibitors in the knee joint. Following oral administration of MK-1256 at a dose level achieving maximal inhibition of urinary biomarker (helical peptide) in dogs, full suppression of the biomarker in synovial fluid was observed. Subsequent tissue distribution studies conducted in dogs and rabbits revealed that MK-1256 levels in synovial fluid and cartilage were consistent with the free-drug hypothesis. Reasonable projection (within twofold) of drug levels in these tissues can be made based on plasma drug concentration with adjustments for binding factors. These results indicate that the previously observed efficacies in the animal models were not limited by compound distribution or target engagement in the knee tissues.
Collapse
Affiliation(s)
- Bennett Ma
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., West Point, PA, USA
| | | | - Bin Luo
- Department of Pharmacology, Merck & Co., West Point, PA, USA
| | - Traci Lifsted
- Department of Pharmacology, Merck & Co., West Point, PA, USA
| | - Keith Wessner
- Department of Pharmacology, Merck & Co., West Point, PA, USA
| | - Gary Adamson
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., West Point, PA, USA
| | | | - Laura S Lubbers
- Department of Pharmacology, Merck & Co., West Point, PA, USA
| |
Collapse
|
49
|
Nwosu LN, Gowler PR, Burston JJ, Rizoska B, Tunblad K, Lindström E, Grabowska U, Li L, McWilliams DF, Walsh DA, Chapman V. Analgesic effects of the cathepsin K inhibitor L-006235 in the monosodium iodoacetate model of osteoarthritis pain. Pain Rep 2018; 3:e685. [PMID: 30706033 PMCID: PMC6344135 DOI: 10.1097/pr9.0000000000000685] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/03/2018] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION The mounting evidence that osteoclasts play an important role in osteoarthritis (OA) pain lead us to investigate the effects of L-006235, a potent and selective inhibitor of cathepsin K, on pain behaviour and joint pathology in a model of OA pain. METHODS Effects of preventative (30 and 100 mg/kg) and therapeutic (100 mg/kg) oral dosing with L-006235 on weight-bearing asymmetry, hind paw withdrawal thresholds, cartilage and bone pathology, synovial inflammation, and drug exposure were studied in the monosodium iodoacetate rat model of OA pain. RESULTS Preventative L-006235 inhibited weight-bearing asymmetry from day 14, with this measure nearly abolished by the higher dose. In the same treatment setting, L-006235 prevented lowering of hind paw withdrawal thresholds from day 7. Exposure to L-006235 in plasma was higher for the 100 mg/kg dose, compared with 30 mg/kg. Therapeutic dosing with L-006235 from day 14 significantly inhibited weight-bearing asymmetry, compared with monosodium iodoacetate vehicle rats. Regression analysis revealed a significant interaction coefficient of the effects of L-006235 on weight-bearing asymmetry and synovitis score, but not for cartilage damage nor osteophyte scores. CONCLUSION Our novel finding that cathepsin K inhibition is analgesic in a clinically relevant model of OA pain provides new evidence for the therapeutic potential of this target.
Collapse
Affiliation(s)
- Lilian N. Nwosu
- Arthritis Research UK Pain Centre, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Peter R.W. Gowler
- Arthritis Research UK Pain Centre, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - James J. Burston
- Arthritis Research UK Pain Centre, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | | | - Li Li
- Arthritis Research UK Pain Centre, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Dan F. McWilliams
- Arthritis Research UK Pain Centre, Academic Rheumatology, City Hospital, University of Nottingham, Nottingham, United Kingdom
| | - David A. Walsh
- Arthritis Research UK Pain Centre, Academic Rheumatology, City Hospital, University of Nottingham, Nottingham, United Kingdom
| | - Victoria Chapman
- Arthritis Research UK Pain Centre, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
50
|
Zhu Z, Li J, Ruan G, Wang G, Huang C, Ding C. Investigational drugs for the treatment of osteoarthritis, an update on recent developments. Expert Opin Investig Drugs 2018; 27:881-900. [PMID: 30345826 DOI: 10.1080/13543784.2018.1539075] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Osteoarthritis (OA) is the leading cause of pain, loss of function, and disability among elderly, with the knee the most affected joint. It is a heterogeneous condition characterized by complex and multifactorial etiologies which contribute to the broad variation in symptoms presentation and treatment responses that OA patients present. This poses a challenge for the development of effective treatment on OA. AREAS COVERED This review will discuss recent development of agents for the treatment of OA, updating our previous narrative review published in 2015. They include drugs for controlling local and systemic inflammation, regulating articular cartilage, targeting subchondral bone, and relieving pain. EXPERT OPINION Although new OA drugs such as monoclonal antibodies have shown marked effects and favorable tolerance, current treatment options for OA remain limited. The authors believe there is no miracle drug that can be used for all OA patients'; treatment and disease stage is crucial for the effectiveness of drugs. Therefore, early diagnosis, phenotyping OA patients and precise therapy would expedite the development of investigational drugs targeting at symptoms and disease progression of OA.
Collapse
Affiliation(s)
- Zhaohua Zhu
- a Clinical Research Centre, Zhujiang Hospital, Southern Medical University , Guangzhou , China
| | - Jia Li
- a Clinical Research Centre, Zhujiang Hospital, Southern Medical University , Guangzhou , China
| | - Guangfeng Ruan
- a Clinical Research Centre, Zhujiang Hospital, Southern Medical University , Guangzhou , China.,b Department of Rheumatology and Immunology , Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University , Hefei , China
| | - Guoliang Wang
- c Menzies Institute for Medical Research, University of Tasmania , Hobart , Australia
| | - Cibo Huang
- d Department of Rheumatology & Immunology , Beijing Hospital , Beijing , China
| | - Changhai Ding
- a Clinical Research Centre, Zhujiang Hospital, Southern Medical University , Guangzhou , China.,b Department of Rheumatology and Immunology , Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University , Hefei , China.,c Menzies Institute for Medical Research, University of Tasmania , Hobart , Australia
| |
Collapse
|