1
|
Konteles V, Papathanasiou I, Tzetis M, Kriebardis A, Tsezou A. Synovial Fibroblast Extracellular Vesicles Induce Inflammation via Delivering miR-21-5p in Osteoarthritis. Cells 2025; 14:519. [PMID: 40214473 PMCID: PMC11989074 DOI: 10.3390/cells14070519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
Small extracellular vesicles (sEVs) derived from different osteoarthritic (OA) tissues regulate OA-related biological processes through transporting their content (proteins, miRNAs, etc.) to recipient cells. This study aimed to characterize the miRNA profile of synovial fibroblasts-derived small EVs (FS_OA_sEVs) and investigate their role in inflammation in chondrocytes. Chondrocytes were isolated from macroscopically preserved and lesioned OA cartilage (C_OAmin and C_OAmax) and synovial fibroblasts from OA synovium. Synovial fibroblasts-derived small EVs (FS_OA_sEVs) were characterized according to ISEV guidelines and used for miRNA profiling and bioinformatics analysis. miR-21-5p was identified as one of the most abundant, and its target genes, such as KLF6, were enriched in OA-related processes including inflammation. Treatment of C_OAmin chondrocytes with FS_OA_sEVs resulted in decreased expression of COL2A1 and ACAN and an increase in catabolic markers MMP-3 and MMP-13. Moreover, C-OAmin receiving FS_OA_sEVs exhibited increased levels of inflammatory markers and miR-21-5p expression, resembling chondrocytes' phenotype from lesioned OA cartilage, whereas miR-21-5p inhibition reversed their expression of inflammatory markers and miR-21-5p. Compared to C_OA min, C_OAmax chondrocytes exhibited increased miR-21-5p and inflammatory markers expression and decreased KLF6 expression. miR-21-5p inhibition in C_OAmax led to KLF6 upregulation and suppression of inflammatory mediators, whereas co-treatment with siRNA against KLF6 negated this effect, confirming a potential direct regulatory relationship between miR-21-5p and KLF6. Our results provide novel insights into the FS_OA_sEV-mediated inflammation axis, highlighting FS_OA_sEV-derived miR-21-5p as a driver of OA progression via regulating inflammation in chondrocytes.
Collapse
Affiliation(s)
- Vasileios Konteles
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, 41222 Larissa, Greece or (V.K.); (I.P.)
| | - Ioanna Papathanasiou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, 41222 Larissa, Greece or (V.K.); (I.P.)
| | - Maria Tzetis
- Choremion Research Laboratory, Department of Medical Genetics, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Anastasios Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece;
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, 41222 Larissa, Greece or (V.K.); (I.P.)
| |
Collapse
|
2
|
Jiang Y, Wang S, Zhu W, Liu X, Yang Y, Huo L, Ye J, Ma Y, Zhou Y, Yang Z, Mao J, Wang X. Lysyl Oxidase-Like 1 (LOXL1) Up-Regulation in Chondrocytes Promotes M1 Macrophage Activation in Osteoarthritis via NF-κB and STAT3 Signaling. Immunotargets Ther 2025; 14:259-278. [PMID: 40161479 PMCID: PMC11951931 DOI: 10.2147/itt.s512768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/16/2025] [Indexed: 04/02/2025] Open
Abstract
Purpose Osteoarthritis (OA) constitutes a widespread degenerative joint disease predominantly affecting the elderly, leading to disability. There is still a lack of biomarkers for OA, so it cannot be intervened in time. Methods OA biomarkers were identified from human cartilage datasets using LASSO and SVM-RFE, followed by ROC analysis. LOXL1 was prioritized for further research due to its high expression in OA cartilage and robust predictive performance. Anterior cruciate ligament transection (ACLT) surgery-induced OA rats were used to explore the correlation between LOXL1 and inflammatory factors and macrophages. Macrophage markers and cytokine secretion were detected from macrophages treated with LOXL1, or co-cultured with chondrocytes after LOXL1 siRNA silencing. Results Five hub biomarkers with OA-specific expression were identified. Elevated LOXL1 correlated with IL-6 and IL-8 in patients and increased M1 macrophages in OA rats. LOXL1-stimulated macrophages upregulated CD86 and inflammatory cytokines. Silencing LOXL1 in chondrocytes reduced CD86, inflammatory cytokines, and NF-κB p65 and p-STAT3 expression in co-cultured macrophages, mitigating MMP13 and chondrocyte apoptosis. STAT3 and NF-κB signal inhibition reduces p-STAT3, p-p65, CD86, IL-6 and IL-1β expression in LOXL1-stimulated macrophages. Conclusion This study underscores the pivotal role of LOXL1 in activating M1 macrophages through NF-κB and STAT3 signaling, thereby promoting pro-inflammatory cytokine secretion and contributing to OA pathogenesis. LOXL1 holds promise as a potential marker for early diagnosis of OA inflammation and as a novel therapeutic target.
Collapse
Affiliation(s)
- Yuyun Jiang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Shang Wang
- Tzu Chi International College of Traditional Chinese Medicine, Vancouver, BC, Canada
| | - Wei Zhu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
- Department of Sports Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Xi Liu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Yanwei Yang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Liyue Huo
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jixian Ye
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Yongbin Ma
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
- Department of Central Laboratory, Jintan Hospital, Jiangsu University, Jintan, 213200, People’s Republic of China
| | - Yuepeng Zhou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Zhe Yang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jiahui Mao
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
- Department of Nuclear Medicine, Institute of Digestive Diseases, and Institute of Endocrinology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| |
Collapse
|
3
|
Brizuela L, Buchet R, Bougault C, Mebarek S. Cathepsin K Inhibitors as Potential Drugs for the Treatment of Osteoarthritis. Int J Mol Sci 2025; 26:2896. [PMID: 40243480 PMCID: PMC11988852 DOI: 10.3390/ijms26072896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Links between cathepsin K and the pathophysiology of osteoarthritis (OA) can be established, not least because of the overabundance of cathepsin K in the serum of OA patients and the upregulation of cathepsin K in degraded cartilage in animal models of OA. Chondrocytes, chondroclasts, or osteoclasts contribute to the accumulated cathepsin K at the diseased osteochondral junction. After a general presentation of OA and cartilage physiology, as well as its degradation processes, we describe the function of cathepsin K and its effect on cartilage degradation via type II collagen cleavage. An overview of the most promising cathepsin K inhibitors is then presented, together with their in vitro effects. Although intensive research on cathepsin K inhibitors initially focused on bone resorption, there is growing interest in the potential of these drugs to prevent cartilage degradation. In this review, we summarize the pre-clinical and clinical trials that support the use of cathepsin K inhibitors in the treatment of OA. To date, no molecules of this type are commercially available, although a few have undergone clinical trials, but we believe that the development of cathepsin K inhibitors could broaden the therapeutic arsenal for the treatment of OA.
Collapse
Affiliation(s)
| | | | | | - Saida Mebarek
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, Université de Lyon, Université Lyon 1, UMR CNRS 5246, 69 622 Villeurbanne Cedex, France
| |
Collapse
|
4
|
Huang C, Zeng B, Zhou B, Chen G, Zhang Q, Hou W, Xiao G, Duan L, Hong N, Jin W. Single-cell transcriptomic analysis of chondrocytes in cartilage and pathogenesis of osteoarthritis. Genes Dis 2025; 12:101241. [PMID: 39759119 PMCID: PMC11697194 DOI: 10.1016/j.gendis.2024.101241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/26/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2025] Open
Abstract
Chondrocyte is considered the only cell type in cartilage. However, the cell heterogeneity of chondrocytes in human articular cartilage is still not well defined, which hinders our understanding of the pathogenesis of osteoarthritis (OA). Here, we constructed a single-cell transcriptomic atlas of chondrocytes in healthy cartilage and identified nine chondrocyte subsets including homeostatic chondrocytes, proliferate fibrochondrocytes, and hypertrophic chondrocytes (HTC). Interestingly, we identified two distinct HTC subpopulations, among which HTC-1 specifically expressed genes associated with apoptosis and programmed cell death. We identified two main trajectories of chondrocytes, one of which differentiates into fibrochondrocytes, while the other terminates in apoptosis. Comparison of chondrocyte subsets between healthy and OA cartilage showed that proliferate fibrochondrocytes and HTC-1 expanded in OA patients, whereas homeostatic chondrocytes decreased. Interestingly, we discovered an OA-specific proliferate fibrochondrocyte subset that may contribute to the development of OA via inflammation. In summary, this study significantly enhanced our understanding of cell heterogeneity of chondrocytes in articular cartilage and provides insight into the pathogenesis of OA.
Collapse
Affiliation(s)
- Changyuan Huang
- Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Bin Zeng
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University, Shenzhen, Guangdong 518035, China
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi 53020, China
| | - Bo Zhou
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Guanming Chen
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Qi Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Wenhong Hou
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523710, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, Guangdong 518055, China
| | - Li Duan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University, Shenzhen, Guangdong 518035, China
| | - Ni Hong
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Wenfei Jin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
5
|
Xu Z, Liu J, Hu H, Ma J, Yang H, Chen J, Xu H, Hu H, Luo H, Chen G. Recent applications of stimulus-responsive smart hydrogels for osteoarthritis therapy. Front Bioeng Biotechnol 2025; 13:1539566. [PMID: 40035023 PMCID: PMC11872905 DOI: 10.3389/fbioe.2025.1539566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Osteoarthritis is one of the most common degenerative joint diseases, which seriously affects the life of middle-aged and elderly people. Traditional treatments such as surgical treatment and systemic medication, often do not achieve the expected or optimal results, which leads to severe trauma and a variety of side effects. Therefore, there is an urgent need to develop novel therapeutic options to overcome these problems. Hydrogels are widely used in biomedical tissue repairing as a platform for loading drugs, proteins and stem cells. In recent years, smart-responsive hydrogels have achieved excellent results as novel drug delivery systems in the treatment of osteoarthritis. This review focuses on the recent advances of endogenous stimuli (including enzymes, pH, reactive oxygen species and temperature, etc.) responsive hydrogels and exogenous stimuli (including light, shear, ultrasound and magnetism, etc.) responsive hydrogels in osteoarthritis treatment. Finally, the current limitations of application and future prospects of smart responsive hydrogels are summarized.
Collapse
Affiliation(s)
- Zhuoming Xu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jintao Liu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hanyin Hu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jun Ma
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Haiyang Yang
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jiayi Chen
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hongwei Xu
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Haodong Hu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Huanhuan Luo
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Gang Chen
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
6
|
Lu P, Li Y, Yang S, Yao H, Tu B, Ning R. B Cell Activation, Differentiation, and Their Potential Molecular Mechanisms in Osteoarthritic Synovial Tissue. J Inflamm Res 2025; 18:2137-2151. [PMID: 39959649 PMCID: PMC11829641 DOI: 10.2147/jir.s503597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Objective The objective of this study was to characterize the activation and differentiation of B cells in the synovium of osteoarthritis (OA) and to explore the underlying molecular mechanisms. Methods Peripheral blood and synovial samples from OA patients at different stages were collected, and flow cytometry was employed to analyze the activation and differentiation of B cells. Immunofluorescence staining of joint synovium from OA mice at different stages was conducted to assess mice joint synovium B cell activation and differentiation. Co-culture experiments of synovial fibroblasts with B cells were performed to investigate the influence of synovial cells on B cell activation and differentiation. Finally, transcriptome analysis was utilized to identify potential key molecules and pathways. Results In OA patients, the infiltration, activation, and differentiation of B cells in synovium and peripheral blood exhibited distinct characteristics. Specifically, the proportion of activated CD86+ B cells and the differentiation marker HLA-DR+ increased with disease severity, whereas the proportion of the differentiation marker IgM decreased. The proportion of CD38+ B cells also decreased with increasing severity, although this change lacked statistical significance. Immunofluorescence staining of CD19+ and CD86+ cells in mice indicated increased expression with greater OA severity. Co-culture experiments demonstrated that OA synovial fibroblasts promoted B cell activation and differentiation, as evidenced by higher expression levels of CD86+ and HLA-DR+ in the OA group compared to controls. Additionally, the proportion of naive B cells decreased as disease severity progressed. Conclusion Synovial fibroblasts in OA have been shown to promote the differentiation and activation of B cells, indicating that B cells play a significant role in the pathogenesis of synovium inflammation in OA.
Collapse
Affiliation(s)
- Peizhi Lu
- Graduate School, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Department of Orthopedics, the Third Affiliated Hospital of Anhui Medical University, the First People’s Hospital of Hefei, Hefei, Anhui, People’s Republic of China
| | - Ya Li
- Graduate School, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Department of Orthopedics, the Third Affiliated Hospital of Anhui Medical University, the First People’s Hospital of Hefei, Hefei, Anhui, People’s Republic of China
| | - Shuo Yang
- Department of Orthopedics, the Third Affiliated Hospital of Anhui Medical University, the First People’s Hospital of Hefei, Hefei, Anhui, People’s Republic of China
| | - Haoyu Yao
- Department of Orthopedics, the Third Affiliated Hospital of Anhui Medical University, the First People’s Hospital of Hefei, Hefei, Anhui, People’s Republic of China
| | - Bizhi Tu
- Department of Orthopedics, the Third Affiliated Hospital of Anhui Medical University, the First People’s Hospital of Hefei, Hefei, Anhui, People’s Republic of China
| | - Rende Ning
- Graduate School, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Department of Orthopedics, the Third Affiliated Hospital of Anhui Medical University, the First People’s Hospital of Hefei, Hefei, Anhui, People’s Republic of China
| |
Collapse
|
7
|
Lin CP, Chung CH, Lu CH, Su SC, Kuo FC, Liu JS, Li PF, Huang CL, Ho LJ, Chen KC, Chang CY, Lin MS, Liu YC, Cheng AC, Lin HH, Kuo SW, Lee CH, Hsieh CH, Hung YJ, Liu HY, Guo LY, Chien WC. Glucagon-like peptide-1 receptor agonists therapy to attenuate the risk of knee osteoarthritis and total knee replacement in type 2 diabetes mellitus: A nation-wide population-based cohort study. Medicine (Baltimore) 2025; 104:e41243. [PMID: 39928811 PMCID: PMC11813052 DOI: 10.1097/md.0000000000041243] [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: 01/17/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 02/12/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an independent risk factor of knee osteoarthritis (KOA). This study was mainly based on data from the Taiwan National Health Insurance Database. Using big data analysis, we showed that glucagon-like peptide-1 receptor agonist (GLP-1RA) treatment is helpful for patients with T2DM who have a lower risk of KOA or total knee replacement (TKR). A total of 35,762 patients with T2DM were included in this study. We divided these patients into 988 patients with T2DM without KOA and 372 patients with T2DM with KOA who received GLP-1RA treatment and those who did not receive GLP-1RA treatment. The patients were matched for sex, age, and inclusion date by 1:1 propensity score, which was included in the control group. Cox proportional hazards analyses were performed to compare KOA risk and TKR rate during a maximum follow-up period of 5 years. There were 1976/744 patients with T2DM without/with KOA who received and did not receive GLP-1RA treatment, including 1052/322 men (53.24/43.28%) and 924/422 women (46.76/56.72%). At the end of follow-up, there were 46/39 (4.66/10.48%) patients with T2DM without/with KOA who received GLP-1RA treatment and underwent KOA/TKR were lower than those without GLP-1RA treatment 87/70 (8.81/18.82%). Cox proportional hazard regression analysis showed a lower rate of KOA/TKR among patients with GLP-1RA treatment (adjusted hazard ratio [HR] = .852; 95% confidence interval [CI] = .784-.930, P < .001/ adjusted HR = .913; 95% CI = .885-.977, P = .015, respectively). Kaplan-Meier analysis showed that the cumulative risk of KOA/TKR in patient with/without GLP-1RA was significantly different (log-rank test, P < .001/P < .001, respectively). This study aimed to provide clinicians with the option of GLP-1RA as a treatment for patients with T2DM with or without KOA to reduce the risk of KOA or TKR among such patients.
Collapse
Affiliation(s)
- Chih-Ping Lin
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan, ROC
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chi-Hsiang Chung
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan, ROC
| | - Chieh-Hua Lu
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Sheng-Chiang Su
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Feng-Chih Kuo
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Jhih-Syuan Liu
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Peng-Fei Li
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chia-Luen Huang
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Li-Ju Ho
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Kuan-Chan Chen
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chun-Yung Chang
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan, ROC
| | - Ming-Shiun Lin
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Chen Liu
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - An-Che Cheng
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hong-Han Lin
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Shi-Wen Kuo
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Taipei Tzu Chi Hospital, Taipei, Taiwan, ROC
| | - Chien-Hsing Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chang-Hsun Hsieh
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Jen Hung
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hsin-Ya Liu
- BeYoung Research Institute, Taipei, Taiwan, ROC
| | - Lan-Yuen Guo
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Wu-Chien Chien
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
8
|
Yan B, Chen H, Yan L, Yuan Q, Guo L. Cryopreserved Umbilical Cord Mesenchymal Stem Cells Show Comparable Effects to Un-Cryopreserved Cells in Treating Osteoarthritis. Cell Transplant 2025; 34:9636897241297631. [PMID: 39874109 PMCID: PMC11776000 DOI: 10.1177/09636897241297631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 01/30/2025] Open
Abstract
Non-cryo and hypothermic preservations are two available options for short-term storage of living cells. For long-term cell storage, cryopreservation is an essential procedure as it prolongs the storage time, allowing for the transport and testing of cells, as well as the establishment of cell banks. But it is unclear whether cryopreservation reduces the therapeutic effects of human umbilical cord mesenchymal stem cells (hucMSCs) on osteoarthritis (OA). To investigate this, we compared the basic biological characteristics and the anti-OA efficacy of un-cryopreserved hucMSCs (UC-MSCs) and cryopreserved hucMSCs (C-MSCs). A mono-iodoacetate-induced rat OA model was established to evaluate the anti-OA properties of UC-MSCs and C-MSCs. And the conditioned medium of UC-MSCs (UC-CM) and cell freezing medium of C-MSCs (C-CFM) were collected for the mechanism study. No significant differences were found between UC-MSCs and C-MSCs in cell viability, immunophenotype, and trilineage differentiation capacity. In vivo, UC-MSCs and C-MSCs exhibited similar cartilage-repairing effects by attenuating pain and alleviating pathological changes in OA rat joints. In vitro, C-CFM and UC-CM promoted the proliferation of chondrocytes, improved the expression of anabolism-related molecules (Col2, COL2, and SOX9), and decreased the expression of catabolism-related molecules (Adamts5, Mmp13, Il6, COL10, and MMP13). These results indicated that UC-MSCs and C-MSCs had comparable anti-OA effects, and cryopreservation did not alter the anti-OA capability of hucMSCs, which provides further support for clinical use of C-MSCs in treating OA.
Collapse
Affiliation(s)
- Bo Yan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China
| | - Huixin Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China
| | - Qiang Yuan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Le Guo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China
| |
Collapse
|
9
|
Jiang P, Song Y, Li P, Yang Y, Zhang J. LncRNA-HHCP5 Regulates KLF5 in ceRNA and m6A Pathways to Inhibit the Progression of Osteoarthritis. Int J Rheum Dis 2025; 28:e70035. [PMID: 39835513 DOI: 10.1111/1756-185x.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Osteoarthritis (OA) is one of the most common bone disorders and has a serious impact on the quality of life of patients. LncRNA-HCP5 (HCP5) is downregulated in OA tissues. However, the latent function and regulatory mechanisms of HCP5 in OA are unclear. METHODS In the current study, IL-1β-induced C28/I2 cells were used to establish an in vitro model of OA. The expression of HCP5 in OA cartilage tissue and in the in vitro model of OA was detected by RT-qPCR. Cell viability and apoptosis were assessed by CCK-8 and Annexin V-PI double staining. Western blotting was employed to detect the protein expression of MMP-13 and aggrecan. RESULTS The results showed that the findings suggested that HCP5 was downregulated in OA cartilage tissue and IL-1β-induced C28/I2 cells. HCP5 overexpression greatly enhanced IL-1β-induced proliferation of C28/I2 cells, as well as prevented cell apoptosis and degradation of extracellular matrix (ECM). Besides, we have shown that HCP5 is a ceRNA that regulates KLF5 by sponging miR-375. Furthermore, KLF5 is also regulated by m6A regulation induced by HCP5. Finally, overexpression of miR-375, the m6A modification inhibitor, as well as KLF5 inhibition reversed the impact of HCP5 on IL-1β-induced C28/I2 cells. CONCLUSION In summary, the present study demonstrated that the HCP5/KLF5 axis inhibited the progression of osteoarthritis.
Collapse
Affiliation(s)
- Peng Jiang
- Department of Orthopaedics, Shaanxi Rehbilitation Hospital, Xi'an, Shaanxi, China
| | - Yuxuan Song
- Department of Orthopaedics, Shaanxi Rehbilitation Hospital, Xi'an, Shaanxi, China
| | - Pengfei Li
- Department of Orthopaedics, Shaanxi Rehbilitation Hospital, Xi'an, Shaanxi, China
| | - Yanhui Yang
- Department of Occupational Therapy, Shaanxi Rehbilitation Hospital, Xi'an, Shaanxi, China
| | - Jiyang Zhang
- Department of Orthopaedics, Shaanxi Rehbilitation Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Pattison LA, Rickman RH, Hilton H, Dannawi M, Wijesinghe SN, Ladds G, Yang LV, Jones SW, Smith ESJ. Activation of the proton-sensing GPCR, GPR65 on fibroblast-like synoviocytes contributes to inflammatory joint pain. Proc Natl Acad Sci U S A 2024; 121:e2410653121. [PMID: 39661058 PMCID: PMC11665855 DOI: 10.1073/pnas.2410653121] [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: 05/28/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Inflammation is associated with localized acidosis, however, attributing physiological and pathological roles to proton-sensitive receptors is challenging due to their diversity and widespread expression. Here, agonists of the proton-sensing GPCR, GPR65, were systematically characterized. The synthetic agonist BTB09089 (BTB) recapitulated many proton-induced signaling events and demonstrated selectivity for GPR65. BTB was used to show that GPR65 activation on fibroblast-like synoviocytes (FLS), cells that line synovial joints, results in the secretion of proinflammatory mediators capable of recruiting immune cells and sensitizing sensory neurons. Intra-articular injection of BTB resulted in GPR65-dependent sensitization of knee-innervating neurons and nocifensive behaviors in mice. Stimulation of GPR65 on human FLS also triggered the release of inflammatory mediators and synovial fluid samples from human osteoarthritis patients were shown to activate GPR65. These results suggest a role of GPR65 in mediating cell-cell interactions that drive inflammatory joint pain in both mice and humans.
Collapse
Affiliation(s)
- Luke A. Pattison
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Rebecca H. Rickman
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Maya Dannawi
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Susanne N. Wijesinghe
- Institute of Inflammation and Ageing, University of Birmingham, BirminghamB15 2TT, United Kingdom
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Li V. Yang
- Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC27834
| | - Simon W. Jones
- Institute of Inflammation and Ageing, University of Birmingham, BirminghamB15 2TT, United Kingdom
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| |
Collapse
|
11
|
Jia X, Zhang G, Yu D. Application of extracellular vesicles in diabetic osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1466775. [PMID: 39720256 PMCID: PMC11666354 DOI: 10.3389/fendo.2024.1466775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/15/2024] [Indexed: 12/26/2024] Open
Abstract
As the population ages, the occurrence of osteoporosis is becoming more common. Diabetes mellitus is one of the factors in the development of osteoporosis. Compared with the general population, the incidence of osteoporosis is significantly higher in diabetic patients. Diabetic osteoporosis (DOP) is a metabolic bone disease characterized by abnormal bone tissue structure due to hyperglycemia and insulin resistance, reduced bone strength and increased risk of fractures. This is a complex mechanism that occurs at the cellular level due to factors such as blood vessels, inflammation, and hyperglycemia and insulin resistance. Although the application of some drugs in clinical practice can reduce the occurrence of DOP, the incidence of fractures caused by DOP is still very high. Extracellular vesicles (EVs) are a new communication mode between cells, which can transfer miRNAs and proteins from mother cells to target cells through membrane fusion, thereby regulating the function of target cells. In recent years, the role of EVs in the pathogenesis of DOP has been widely demonstrated. In this article, we first describe the changes in the bone microenvironment of osteoporosis. Second, we describe the pathogenesis of DOP. Finally, we summarize the research progress and challenges of EVs in DOP.
Collapse
Affiliation(s)
- Xiaopeng Jia
- Trauma Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Gongzi Zhang
- Department of Rehabilitation Medicine, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Deshui Yu
- Trauma Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
12
|
Pohl PC, Villas-Boas IM, Pidde G, Tambourgi DV. Pararamosis, a Neglected Tropical Disease Induced by Premolis semirufa Caterpillar Toxins: Investigating Their Effects on Synovial Cell Inflammation. Int J Mol Sci 2024; 25:13149. [PMID: 39684859 DOI: 10.3390/ijms252313149] [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: 10/12/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Pararamosis, also known as Pararama-associated phalangeal periarthritis, is a neglected tropical disease primarily affecting rubber tappers in the Amazon region. It is caused by contact with the urticating hairs of the Premolis semirufa moth caterpillar, which resides in rubber plantations. The condition is marked by the thickening of the articular synovial membrane and cartilage impairment, features associated with chronic synovitis. Given the significance of synovial inflammation in osteoarticular diseases, in this study, the role of synoviocytes and their interactions with macrophages and chondrocytes are examined when stimulated by Pararama toxins. Synoviocytes and macrophages treated with Pararama hair extract showed an increased production of cytokines IL-6, IL-1β, and TNF-α, indicating a direct effect on these cells. In cocultures, there was a significant rise in inflammation, with levels of IL-1β, IL-6, and chemokines CCL2, CCL5, and CXCL8 increasing up to seven times compared to monocultures. Additionally, matrix-degrading enzymes MMP-1 and MMP-3 were significantly elevated in cocultures. Chondrocytes exposed to the extract also produced IL-6, CCL2, and CCL5, and in cocultures with synoviocytes, there was a notable increase in IL-6, CCL5, and CXCL8, as well as a doubling of MMP-1 and MMP-3 levels. These findings underscore the critical role of cell crosstalk in the inflammatory and catabolic processes associated with pararamosis and demonstrate how Pararama hair extract can influence factors affecting cartilage health, providing valuable insights into this condition.
Collapse
Affiliation(s)
- Paula C Pohl
- Immunochemistry Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil
| | | | - Giselle Pidde
- Immunochemistry Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Denise V Tambourgi
- Immunochemistry Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil
| |
Collapse
|
13
|
Du C, Liu J, Liu S, Xiao P, Chen Z, Chen H, Huang W, Lei Y. Bone and Joint-on-Chip Platforms: Construction Strategies and Applications. SMALL METHODS 2024; 8:e2400436. [PMID: 38763918 DOI: 10.1002/smtd.202400436] [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: 03/25/2024] [Revised: 04/28/2024] [Indexed: 05/21/2024]
Abstract
Organ-on-a-chip, also known as "tissue chip," is an advanced platform based on microfluidic systems for constructing miniature organ models in vitro. They can replicate the complex physiological and pathological responses of human organs. In recent years, the development of bone and joint-on-chip platforms aims to simulate the complex physiological and pathological processes occurring in human bones and joints, including cell-cell interactions, the interplay of various biochemical factors, the effects of mechanical stimuli, and the intricate connections between multiple organs. In the future, bone and joint-on-chip platforms will integrate the advantages of multiple disciplines, bringing more possibilities for exploring disease mechanisms, drug screening, and personalized medicine. This review explores the construction and application of Organ-on-a-chip technology in bone and joint disease research, proposes a modular construction concept, and discusses the new opportunities and future challenges in the construction and application of bone and joint-on-chip platforms.
Collapse
Affiliation(s)
- Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Xiao
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
14
|
Jin P, Liu H, Chen X, Liu W, Jiang T. From Bench to Bedside: The Role of Extracellular Vesicles in Cartilage Injury Treatment. Biomater Res 2024; 28:0110. [PMID: 39583872 PMCID: PMC11582190 DOI: 10.34133/bmr.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/20/2024] [Accepted: 10/26/2024] [Indexed: 11/26/2024] Open
Abstract
Cartilage repair is the key to the treatment of joint-related injury. However, because cartilage lacks vessels and nerves, its self-repair ability is extremely low. Extracellular vesicles (EVs) are bilayer nanovesicles with membranes mainly composed of ceramides, cholesterol, phosphoglycerides, and long-chain free fatty acids, containing DNA, RNA, and proteins (such as integrins and enzymes). For mediating intercellular communication and regulating mechanisms, EVs have been shown by multiple studies to be effective treatment options for cartilage repair. This review summarizes recent findings of different sources (mammals, plants, and bacteria) and uses of EVs in cartilage repair, mechanisms of EVs captured by injured chondrocytes, and quantification and storage of EVs, which may provide scientific guidance for promoting the development of EVs in the field of cartilage injury treatment.
Collapse
Affiliation(s)
- Pan Jin
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Huan Liu
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Xichi Chen
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Wei Liu
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Tongmeng Jiang
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University,
Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma; Hainan Provincial Stem Cell Research Institute; Hainan Academy of Medical Sciences,
Hainan Medical University, Haikou 571199, China
| |
Collapse
|
15
|
D'Arrigo D, Salerno M, De Marziani L, Boffa A, Filardo G. A call for standardization for secretome and extracellular vesicles in osteoarthritis: results show disease-modifying potential, but protocols are too heterogeneous-a systematic review. Hum Cell 2024; 37:1243-1275. [PMID: 38909330 DOI: 10.1007/s13577-024-01084-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/22/2024] [Indexed: 06/24/2024]
Abstract
The currently available osteoarthritis (OA) treatments offer symptoms' relief without disease-modifying effects. Increasing evidence supports the role of human mesenchymal stem cells (MSCs) to drive beneficial effects provided by their secretome and extracellular vesicles (EVs), which includes trophic and biologically active factors. Aim of this study was to evaluate the in vitro literature to understand the potential of human secretome and EVs for OA treatment and identify trends, gaps, and potential translational challenges. A systematic review was performed on PubMed, Embase, and Web-of-Science, identifying 58 studies. The effects of secretome and EVs were analysed on osteoarthritic cells regarding anabolic, anti-apoptotic/anti-inflammatory and catabolic/pro-inflammatory/degenerative activity, chondroinduction, and immunomodulation. The results showed that MSC-derived EVs elicit an increase in proliferation and migration, reduction of cell death and inflammation, downregulation of catabolic pathways, regulation of immunomodulation, and promotion of anabolic processes in arthritic cells. However, a high heterogeneity in several technical or more applicative aspects emerged. In conclusion, the use of human secretome and EVs as strategy to address OA processes has overall positive effects and disease-modifying potential. However, it is crucial to reduce protocol variability and strive toward a higher standardization, which will be essential for the translation of this promising OA treatment from the in vitro research setting to the clinical practice.
Collapse
Affiliation(s)
- Daniele D'Arrigo
- Regenerative Medicine Technologies Laboratory, EOC, Via Francesco Chiesa 5, 6500, Bellinzona, Switzerland
- Laboratoire Matière et Systèmes Complexes, Université Paris Cité, 45 Rue des Saints Pères, 75006, Paris, France
- Abbelight, Cachan, 191 Av. Aristide Briand, 94230, Cachan, France
| | - Manuela Salerno
- Applied and Translational Research center, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136, Bologna, Italy.
| | - Luca De Marziani
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136, Bologna, Italy
| | - Angelo Boffa
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136, Bologna, Italy
| | - Giuseppe Filardo
- Applied and Translational Research center, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136, Bologna, Italy
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Via Tesserete 46, 6900, Lugano, Switzerland
- Università Della Svizzera Italiana, Faculty of Biomedical Sciences, Via Buffi 13, 6900, Lugano, Switzerland
| |
Collapse
|
16
|
Zhang Z, Li B, Wu S, Yang Y, Wu B, Lai Q, Lai F, Mo F, Zhong Y, Wang S, Guo R, Zhang B. Bergenin protects against osteoarthritis by inhibiting STAT3, NF-κB and Jun pathways and suppressing osteoclastogenesis. Sci Rep 2024; 14:20292. [PMID: 39217193 PMCID: PMC11366014 DOI: 10.1038/s41598-024-71259-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by articular cartilage destruction and subchondral bone reconstruction in the early stages. Bergenin (Ber) is a cytoprotective polyphenol found in many medicinal plants. It has been proven to have anti-inflammatory, antioxidant, and other biological activities, which may reveal its potential role in the treatment of OA. This study aimed to determine the potential efficacy of Ber in treating OA and explore the possible underlying mechanism through network pharmacology and validation experiments. The potential co-targets and processes of Ber and OA were predicted by using network pharmacology, including a Venn diagram for intersection targets, a protein‒protein interaction (PPI) network to obtain key potential targets, and GO and KEGG pathway enrichment to reveal the probable mechanism of action of Ber on OA. Subsequently, validation experiments were carried out to investigate the effects and mechanisms of Ber in treating OA in vitro and vivo. Ber suppressed IL-1β-induced chondrocyte apoptosis and extracellular matrix catabolism by inhibiting the STAT3, NF-κB and Jun signalling pathway in vitro. Furthermore, Ber suppressed the expression of osteoclast marker genes and RANKL-induced osteoclastogenesis. Ber alleviated the progression of OA in DMM-induced OA mice model. These results demonstrated the protective efficacy and potential mechanisms of Ber against OA, which suggested that Ber could be adopted as a potential therapeutic agent for treating OA.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Bo Li
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Shuqin Wu
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Yuxin Yang
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Binkang Wu
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Qi Lai
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Fuchong Lai
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Fengbo Mo
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Yufei Zhong
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Song Wang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| | - Runsheng Guo
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| | - Bin Zhang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
17
|
Li K, Leng Y, Lei D, Zhang H, Ding M, Lo WLA. Causal link between metabolic related factors and osteoarthritis: a Mendelian randomization investigation. Front Nutr 2024; 11:1424286. [PMID: 39206315 PMCID: PMC11349640 DOI: 10.3389/fnut.2024.1424286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Metabolic syndrome (MetS) is significantly associated with osteoarthritis (OA), especially in MetS patients with blood glucose abnormalities, such as elevated fasting blood glucose (FG), which may increase OA risk. Dietary modifications, especially the intake of polyunsaturated fatty acids (PUFAs), are regarded as a potential means of preventing MetS and its complications. However, regarding the effects of FG, Omega-3s, and Omega-6s on OA, the research conclusions are conflicting, which is attributed to the complexity of the pathogenesis of OA. Therefore, it is imperative to thoroughly evaluate multiple factors to fully understand their role in OA, which needs further exploration and clarification. Methods Two-sample univariable Mendelian randomization (UVMR) and multivariable Mendelian randomization (MVMR) were employed to examine the causal effect of metabolic related factors on hip OA (HOA) or knee OA (KOA). The exposure and outcome datasets were obtained from Open GWAS IEU. All cases were independent European ancestry data. Three MR methods were performed to estimate the causal effect: inverse-variance weighting (IVW), weighted median method (WMM), and MR-Egger regression. Additionally, the intercept analysis in MR-Egger regression is used to estimate pleiotropy, and the IVW method and MR-Egger regression are used to test the heterogeneity. Results The UVMR analysis revealed a causal relationship between FG and HOA. By MVMR analysis, the study discovered a significant link between FG (OR = 0.79, 95%CI: 0.64∼0.99, p = 0.036) and KOA after accounting for body mass index (BMI), age, and sex hormone-binding globulin (SHBG). However, no causal effects of FG on HOA were seen. Omega-3s and Omega-6s did not have a causal influence on HOA or KOA. No significant evidence of pleiotropy was identified. Discussion The MR investigation showed a protective effect of FG on KOA development but no causal relationship between FG and HOA. No causal effect of Omega-3s and Omega-6s on HOA and KOA was observed. Shared genetic overlaps might also exist between BMI and age, SHBG and PUFAs for OA development. This finding offers a novel insight into the treatment and prevention of KOA from glucose metabolism perspective. The FG cutoff value should be explored in the future, and consideration should be given to demonstrating the study in populations other than Europeans.
Collapse
Affiliation(s)
- Kai Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Leng
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Lei
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haojie Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minghui Ding
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wai Leung Ambrose Lo
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering and Technology Research Centre for Rehabilitation Medicine and Translation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Sengprasert P, Waitayangkoon P, Kamenkit O, Sawatpanich A, Chaichana T, Wongphoom J, Ngarmukos S, Taweevisit M, Lotinun S, Tumwasorn S, Tanavalee A, Reantragoon R. Catabolic mediators from TLR2-mediated proteoglycan aggrecan peptide-stimulated chondrocytes are reduced by Lactobacillus-conditioned media. Sci Rep 2024; 14:18043. [PMID: 39103466 PMCID: PMC11300663 DOI: 10.1038/s41598-024-68404-9] [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: 09/20/2023] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
In osteoarthritis (OA), extracellular matrix (ECM) digestion by cartilage-degrading enzymes drives cartilage destruction and generates ECM fragments, such as proteoglycan aggrecan (PG) peptides. PG peptides have been shown to induce immunological functions of chondrocytes. However, the role of PG peptides in stimulating catabolic mediators from chondrocytes has not been investigated. Therefore, we aim to determine the effects and its mechanism by which PG peptides induce chondrocytes to produce catabolic mediators in OA. Human chondrocytes were stimulated with IFNγ and various PG peptides either (i) with or (ii) without TLR2 blockade or (iii) with Lactobacillus species-conditioned medium (LCM), a genus of bacteria with anti-inflammatory properties. Transcriptomic analysis, cartilage-degrading enzyme production and TLR2-intracellular signaling activation were investigated. Chondrocytes treated with PG peptides p16-31 and p263-280 increased expression levels of genes associated with chondrocyte hypertrophy, cartilage degradation and proteolytic enzyme production. TLR2 downstream signaling proteins (STAT3, IkBα and MAPK9) were significantly phosphorylated in p263-280 peptide-stimulated chondrocytes. MMP-1 and ADAMTS-4 were significantly reduced in p263-280 peptides-treated condition with TLR2 blockade or LCM treatment. Phosphorylation levels of IkBa, ERK1/2 and MAPK9 were significantly decreased with TLR2 blockade, but only phosphorylation levels of MAPK9 was significantly decreased with LCM treatment. Our study showed that PG peptide stimulation via TLR2 induced cartilage-degrading enzyme production via activation of MAPK, NFκB and STAT3 pathways.
Collapse
Affiliation(s)
- Panjana Sengprasert
- Immunology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Bangkok, 10330, Thailand
| | - Palapun Waitayangkoon
- Immunology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Bangkok, 10330, Thailand
| | - Ousakorn Kamenkit
- Medical Microbiology Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Ajcharaporn Sawatpanich
- Bacteriology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thiamjit Chaichana
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jutamas Wongphoom
- Department of Pathology, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Srihatach Ngarmukos
- Department of Orthopedics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Biologics for Knee Osteoarthritis Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Mana Taweevisit
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sutada Lotinun
- Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Somying Tumwasorn
- Bacteriology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Aree Tanavalee
- Department of Orthopedics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Biologics for Knee Osteoarthritis Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Rangsima Reantragoon
- Immunology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Bangkok, 10330, Thailand.
- Center of Excellence in Immunology and Immune-Mediated Diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
19
|
Zhang Y, Li Z, Chen C, Wei W, Li Z, Zhou H, He W, Xia J, Li B, Yang Y. SRGN promotes macrophage recruitment through CCL3 in osteoarthritis. Connect Tissue Res 2024; 65:330-342. [PMID: 39067006 DOI: 10.1080/03008207.2024.2380313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/18/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative disease that affects synovial joints and leads to significant pain and disability, particularly in older adults. Infiltration of macrophages plays a key role in the progression of OA. However, the mechanisms underlying macrophage recruitment in OA are not fully understood. METHODS The Serglycin (SRGN) expression pattern was analyzed, along with its association with macrophage infiltration in OA, using bioinformatic methods. SRGN expression in chondrocytes was altered by small interfering RNA (siRNA) and plasmids. Conditioned media (CM) was obtained from transfected chondrocytes to establish a co-culture model of chondrocytes and THP-1 derived macrophages. The impact of SRGN on macrophage recruitment was evaluated using a transwell assay. Furthermore, the regulatory effect of SRGN on CCL3 was validated through qPCR, WB, and ELISA experiments. RESULTS In OA patients, the upregulation of SRGN positively correlated with K-L grade and macrophage infiltration. It was found that SRGN expression and secretion were up-regulated in OA and that it can promote macrophage migration in vitro. Further investigation showed that SRGN affects macrophage migration by regulating the expression of CCL3. CONCLUSION SRGN in chondrocytes plays a role in promoting the recruitment of THP-1 derived macrophages in vitro by regulating production of CCL3.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihua Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cheng Chen
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Wei
- The First Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhendong Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haichao Zhou
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenbao He
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiang Xia
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bing Li
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunfeng Yang
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Cai R, Jiang Q, Chen D, Feng Q, Liang X, Ouyang Z, Liao W, Zhang R, Fang H. Identification of osteoblastic autophagy-related genes for predicting diagnostic markers in osteoarthritis. iScience 2024; 27:110130. [PMID: 38952687 PMCID: PMC11215306 DOI: 10.1016/j.isci.2024.110130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/15/2024] [Accepted: 05/24/2024] [Indexed: 07/03/2024] Open
Abstract
The development of osteoarthritis (OA) involves subchondral bone lesions, but the role of osteoblastic autophagy-related genes (ARGs) in osteoarthritis is unclear. Through integrated analysis of single-cell dataset, Bulk RNA dataset, and 367 ARGs extracted from GeneCards, 40 ARGs were found. By employing multiple machine learning algorithms and PPI networks, three key genes (DDIT3, JUN, and VEGFA) were identified. Then the RF model constructed from these genes indicated great potential as a diagnostic tool. Furthermore, the model's effectiveness in predicting OA has been confirmed through external validation datasets. Moreover, the expression of ARGs was examined in osteoblasts subject to excessive mechanical stress, human and mouse tissues. Finally, the role of ARGs in OA was confirmed through co-culturing explants and osteoblasts. Thus, osteoblastic ARGs could be crucial in OA development, providing potential diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Rulong Cai
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- Academy of Orthopedics · Guangdong Province, Guangzhou, 510630, China
- Orthopedic Hospital of Guangdong Province, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qijun Jiang
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
- Department of Urology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
| | - Dongli Chen
- Department of Ultrasound, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Qi Feng
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- Academy of Orthopedics · Guangdong Province, Guangzhou, 510630, China
- Orthopedic Hospital of Guangdong Province, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xinzhi Liang
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- Academy of Orthopedics · Guangdong Province, Guangzhou, 510630, China
- Orthopedic Hospital of Guangdong Province, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhaoming Ouyang
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- Academy of Orthopedics · Guangdong Province, Guangzhou, 510630, China
- Orthopedic Hospital of Guangdong Province, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Weijian Liao
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- Academy of Orthopedics · Guangdong Province, Guangzhou, 510630, China
- Orthopedic Hospital of Guangdong Province, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Rongkai Zhang
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- Academy of Orthopedics · Guangdong Province, Guangzhou, 510630, China
- Orthopedic Hospital of Guangdong Province, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Hang Fang
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, China
- Academy of Orthopedics · Guangdong Province, Guangzhou, 510630, China
- Orthopedic Hospital of Guangdong Province, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
21
|
Faeed M, Ghiasvand M, Fareghzadeh B, Taghiyar L. Osteochondral organoids: current advances, applications, and upcoming challenges. Stem Cell Res Ther 2024; 15:183. [PMID: 38902814 PMCID: PMC11191177 DOI: 10.1186/s13287-024-03790-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024] Open
Abstract
In the realm of studying joint-related diseases, there is a continuous quest for more accurate and representative models. Recently, regenerative medicine and tissue engineering have seen a growing interest in utilizing organoids as powerful tools for studying complex biological systems in vitro. Organoids, three-dimensional structures replicating the architecture and function of organs, provide a unique platform for investigating disease mechanisms, drug responses, and tissue regeneration. The surge in organoid research is fueled by the need for physiologically relevant models to bridge the gap between traditional cell cultures and in vivo studies. Osteochondral organoids have emerged as a promising avenue in this pursuit, offering a better platform to mimic the intricate biological interactions within bone and cartilage. This review explores the significance of osteochondral organoids and the need for their development in advancing our understanding and treatment of bone and cartilage-related diseases. It summarizes osteochondral organoids' insights and research progress, focusing on their composition, materials, cell sources, and cultivation methods, as well as the concept of organoids on chips and application scenarios. Additionally, we address the limitations and challenges these organoids face, emphasizing the necessity for further research to overcome these obstacles and facilitate orthopedic regeneration.
Collapse
Affiliation(s)
- Maryam Faeed
- Cell and Molecular School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mahsa Ghiasvand
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahar Fareghzadeh
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Leila Taghiyar
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
22
|
Chen B, Sun Y, Xu G, Jiang J, Zhang W, Wu C, Xue P, Cui Z. Role of crosstalk between synovial cells and chondrocytes in osteoarthritis (Review). Exp Ther Med 2024; 27:201. [PMID: 38590580 PMCID: PMC11000048 DOI: 10.3892/etm.2024.12490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 04/10/2024] Open
Abstract
Osteoarthritis (OA) is a low-grade, nonspecific inflammatory disease that affects the entire joint. This condition is characterized by synovitis, cartilage erosion, subchondral bone defects, and subpatellar fat pad damage. There is mounting evidence demonstrating the significance of crosstalk between synovitis and cartilage destruction in the development of OA. To comprehensively explore the phenotypic alterations of synovitis and cartilage destruction, it is important to elucidate the crosstalk mechanisms between chondrocytes and synovial cells. Furthermore, the updated iteration of single-cell sequencing technology reveals the interaction between chondrocyte and synovial cells. In the present review, the histological and pathological alterations between cartilage and synovium during OA progression are described, and the mode of interaction and molecular mechanisms between synovial cells and chondrocytes in OA, both of which affect the OA process mainly by altering the inflammatory environment and cellular state, are elucidated. Finally, the current OA therapeutic approaches are summarized and emerging therapeutic targets are reviewed in an attempt to provide potential insights into OA treatment.
Collapse
Affiliation(s)
- Baisen Chen
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yuyu Sun
- Department of Orthopedics, Nantong Third People's Hospital, Nantong, Jiangsu 226003, P.R. China
| | - Guanhua Xu
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jiawei Jiang
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wenhao Zhang
- Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Chunshuai Wu
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Pengfei Xue
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhiming Cui
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
23
|
Ding X, Huang J, Zhou R, Che X, Pang Y, Liang D, Lu C, Zhuo Y, Cao F, Wu G, Li W, Li P, Zhao L, Rong X, Li P, Wang C. Bibliometric study and visualization of cellular senescence associated with osteoarthritis from 2009 to 2023. Medicine (Baltimore) 2024; 103:e37611. [PMID: 38669405 PMCID: PMC11049721 DOI: 10.1097/md.0000000000037611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/23/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Osteoarthritis is a common degenerative joint disease that is highly prevalent in the elderly population. Along with the occurrence of sports injuries, osteoarthritis is gradually showing a younger trend. Osteoarthritis has many causative factors, and its pathogenesis is currently unknown. Cellular senescence is a stable form of cell cycle arrest exhibited by cells in response to external stimuli and plays a role in a variety of diseases. And it is only in the last decade or so that cellular senescence has gradually become cross-linked with osteoarthritis. However, there is no comprehensive bibliometric analysis in this field. The aim of this study is to present the current status and research hotspots of cellular senescence in the field of osteoarthritis, and to predict the future trends of cellular senescence in osteoarthritis research from a bibliometric perspective. METHODS This study included 298 records of cellular senescence associated with osteoarthritis from 2009 to 2023, with data from the Web of Science Core Collection database. CiteSpace, Scimago Graphica software, VOSviewer, and the R package "bibliometrix" software were used to analyze regions, institutions, journals, authors, and keywords to predict recent trends in cellular senescence related to osteoarthritis research. RESULTS The number of publications related to cellular senescence associated with osteoarthritis is increasing year by year. China and the United States contribute more than 70% of the publications and are the mainstay of research in this field. Central South University is the most active institution with the largest number of publications. International Journal of Molecular Sciences is the most popular journal in the field with the largest number of publications, while Osteoarthritis and Cartilage is the most cited journal. Loeser, Richard F. is not only the most prolific author, but also the most frequently cited author, contributing greatly to the field. CONCLUSION In the last decade or so, this is the first bibliometric study that systematically describes the current status and development trend of research on cellular senescence associated with osteoarthritis. The study comprehensively and systematically summarizes and concludes the research hotspots and development trends, providing valuable references for researchers in this field.
Collapse
Affiliation(s)
- Xueting Ding
- Department of Embryology, School of Basic Medical Sciences, Shanxi Medical University, Shanxi, China
- Animal Experiment Center, Shanxi Medical University, Shanxi, China
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Jingrui Huang
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Raorao Zhou
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Xianda Che
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Yiming Pang
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Dan Liang
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Chengyang Lu
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Yuhao Zhuo
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Fuyang Cao
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Gaige Wu
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Wenjin Li
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Penghua Li
- Laboratory department, Fenyang Hospital of Shanxi Province, Shanxi, China
| | - Litao Zhao
- Pain Department, The Third People's Hospital of Hainan Province, Hainan, China
| | - XueQin Rong
- Pain Department, The Third People's Hospital of Hainan Province, Hainan, China
| | - Pengcui Li
- Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, Shanxi, China
| | - Chunfang Wang
- Department of Embryology, School of Basic Medical Sciences, Shanxi Medical University, Shanxi, China
- Animal Experiment Center, Shanxi Medical University, Shanxi, China
| |
Collapse
|
24
|
Chen W, Wang Q, Tao H, Lu L, Zhou J, Wang Q, Huang W, Yang X. Subchondral osteoclasts and osteoarthritis: new insights and potential therapeutic avenues. Acta Biochim Biophys Sin (Shanghai) 2024; 56:499-512. [PMID: 38439665 DOI: 10.3724/abbs.2024017] [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: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, and good therapeutic results are often difficult to obtain due to its complex pathogenesis and diverse causative factors. After decades of research and exploration of OA, it has been progressively found that subchondral bone is essential for its pathogenesis, and pathological changes in subchondral bone can be observed even before cartilage lesions develop. Osteoclasts, the main cells regulating bone resorption, play a crucial role in the pathogenesis of subchondral bone. Subchondral osteoclasts regulate the homeostasis of subchondral bone through the secretion of degradative enzymes, immunomodulation, and cell signaling pathways. In OA, osteoclasts are overactivated by autophagy, ncRNAs, and Rankl/Rank/OPG signaling pathways. Excessive bone resorption disrupts the balance of bone remodeling, leading to increased subchondral bone loss, decreased bone mineral density and consequent structural damage to articular cartilage and joint pain. With increased understanding of bone biology and targeted therapies, researchers have found that the activity and function of subchondral osteoclasts are affected by multiple pathways. In this review, we summarize the roles and mechanisms of subchondral osteoclasts in OA, enumerate the latest advances in subchondral osteoclast-targeted therapy for OA, and look forward to the future trends of subchondral osteoclast-targeted therapies in clinical applications to fill the gaps in the current knowledge of OA treatment and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Wenlong Chen
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiufei Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Huaqiang Tao
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiang Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Wei Huang
- Department of Orthopaedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| |
Collapse
|
25
|
Pan J, Cai Y, Zhang C, Xu S. Intra-articular delivery of geraniol encapsulated by pH/redox-responsive nanogel ameliorates osteoarthritis by regulating oxidative stress and inflammation. J Mol Histol 2023; 54:579-591. [PMID: 37848748 PMCID: PMC10635995 DOI: 10.1007/s10735-023-10163-4] [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: 07/05/2022] [Accepted: 09/30/2023] [Indexed: 10/19/2023]
Abstract
Osteoarthritis (OA) remains a challenging condition due to limited drug bioavailability within the avascular and dense cartilage matrix. This study introduces a pH/redox-responsive nanogel for enhanced delivery of geraniol in OA therapy. We investigated geraniol's role in preventing chondrocyte matrix degradation and designed a pH/redox-responsive nanogel as a delivery platform. Our methods included Western blot, histological staining, and immunohistochemistry. Geraniol treatment reduced Keap1 expression while elevating Nrf2 and HO-1 levels, effectively inhibiting cartilage matrix degradation. The pH/redox-responsive nanogel further enhanced geraniol's therapeutic impact. Our study demonstrates that geraniol encapsulated within a pH/redox-responsive nanogel mitigates OA by regulating oxidative stress and inflammation. This innovative approach holds potential as an effective OA therapeutic strategy.
Collapse
Affiliation(s)
- Jun Pan
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Youzhi Cai
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chi Zhang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Sanzhong Xu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
26
|
Li Z, Bi R, Zhu S. The Dual Role of Small Extracellular Vesicles in Joint Osteoarthritis: Their Global and Non-Coding Regulatory RNA Molecule-Based Pathogenic and Therapeutic Effects. Biomolecules 2023; 13:1606. [PMID: 38002288 PMCID: PMC10669328 DOI: 10.3390/biom13111606] [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: 09/13/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
OA is the most common joint disease that affects approximately 7% of the global population. Current treatment methods mainly relieve its symptoms with limited repairing effect on joint destructions, which ultimately contributes to the high morbidity rate of OA. Stem cell treatment is a potential regenerative medical therapy for joint repair in OA, but the uncertainty in differentiation direction and immunogenicity limits its clinical usage. Small extracellular vesicles (sEVs), the by-products secreted by stem cells, show similar efficacy levels but have safer regenerative repair effect without potential adverse outcomes, and have recently drawn attention from the broader research community. A series of research works and reviews have been performed in the last decade, providing references for the application of various exogenous therapeutic sEVs for treating OA. However, the clinical potential of target intervention involving endogenous pathogenic sEVs in the treatment of OA is still under-explored and under-discussed. In this review, and for the first time, we emphasize the dual role of sEVs in OA and explain the effects of sEVs on various joint tissues from both the pathogenic and therapeutic aspects. Our aim is to provide a reference for future research in the field.
Collapse
Affiliation(s)
- Zhi Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
| | - Ruiye Bi
- Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Songsong Zhu
- Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
27
|
Huang L, Dong G, Peng J, Li T, Zou M, Hu K, Shu Y, Cheng T, Hao L. The role of exosomes and their enhancement strategies in the treatment of osteoarthritis. Hum Cell 2023; 36:1887-1900. [PMID: 37603220 DOI: 10.1007/s13577-023-00970-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/12/2023] [Indexed: 08/22/2023]
Abstract
With the increasingly prominent problem of population aging, osteoarthritis (OA), which is closely related to aging, has become a serious illness affecting the lives and health of elderly individuals. However, effective treatments are still lacking. OA is typically considered a low-grade inflammatory state. The inflammatory infiltration of macrophages, neutrophils, T cells, and other cells is common in diseased joints. These cells create the inflammatory environment of OA and are involved in the onset and progression of the disease. Exosomes, a type of complex vesicle containing abundant RNA molecules and proteins, play a crucial role in the physiological and pathological processes of an organism. In comparison to other therapeutic methods such as stem cells, exosomes have distinct advantages of precise targeting and low immunogenicity. Moreover, research and techniques related to exosomes are more mature, indicating a promising future in disease treatment. Many studies have shown that the impact of exosomes on the inflammatory microenvironment directly or indirectly leads to the occurrence of various diseases. Furthermore, exosomes can be helpful in the management of illnesses. This article provides a comprehensive review and update on the research of exosomes, a type of extracellular vesicle, in the treatment of OA by modulating the inflammatory microenvironment. It also combines innovative studies on the modification of exosomes. In general, the application of exosomes in the treatment of OA has been validated, and the introduction of modified exosome technology holds potential for enhancing its therapeutic efficacy.
Collapse
Affiliation(s)
- Linzhen Huang
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Ge Dong
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Jie Peng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ting Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Mi Zou
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Kaibo Hu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Yuan Shu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Tao Cheng
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai, China
| | - Liang Hao
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
28
|
Xiao J, Zhang P, Cai FL, Luo CG, Pu T, Pan XL, Tian M. IL-17 in osteoarthritis: A narrative review. Open Life Sci 2023; 18:20220747. [PMID: 37854319 PMCID: PMC10579884 DOI: 10.1515/biol-2022-0747] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/18/2023] [Accepted: 09/07/2023] [Indexed: 10/20/2023] Open
Abstract
Osteoarthritis (OA) is a painful joint disease that is common among the middle-aged and elderly populations, with an increasing prevalence. Therapeutic options for OA are limited, and the pathogenic mechanism of OA remains unclear. The roles of cytokines and signaling pathways in the development of OA is a current research hot spot. Interleukin (IL)-17 is a pleiotropic inflammatory cytokine produced mainly by T helper 17 cells that has established roles in host defense, tissue repair, lymphoid tissue metabolism, tumor progression, and pathological processes of immune diseases, and studies in recent years have identified an important role for IL-17 in the progression of OA. This narrative review focuses on the mechanisms by which IL-17 contributes to articular cartilage degeneration and synovial inflammation in OA and discusses how IL-17 and the IL-17 signaling pathway affect the pathological process of OA. Additionally, therapeutic targets that have been proposed in recent years based on IL-17 and its pathway in OA are summarized as well as recent advances in the study of IL-17 pathway inhibitors and the potential challenges of their use for OA treatment.
Collapse
Affiliation(s)
- Juan Xiao
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, Zunyi563000, China
| | - Ping Zhang
- The First School of Clinical Medicine, Zunyi Medical University, Zunyi563000, China
| | - Fang-Lan Cai
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi563000, China
| | - Cheng-Gen Luo
- The First School of Clinical Medicine, Zunyi Medical University, Zunyi563000, China
| | - Tao Pu
- Department of Nephrology and Rheumatology, Moutai Hospital, Renhuai 564500Guizhou, China
| | - Xiao-Li Pan
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, Zunyi563000, China
| | - Mei Tian
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, Zunyi563000, China
| |
Collapse
|
29
|
Kong R, Ji L, Pang Y, Zhao D, Gao J. Exosomes from osteoarthritic fibroblast-like synoviocytes promote cartilage ferroptosis and damage via delivering microRNA-19b-3p to target SLC7A11 in osteoarthritis. Front Immunol 2023; 14:1181156. [PMID: 37691947 PMCID: PMC10484587 DOI: 10.3389/fimmu.2023.1181156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/10/2023] [Indexed: 09/12/2023] Open
Abstract
Objective Our previous studies revealed that normal synovial exosomes promoted chondrogenesis, and microRNA (miR)-19b-3p independently related to osteoarthritis (OA) risk. Subsequently, this study intended to further explore the effect of OA fibroblast-like synoviocyte (OA-FLS) exosomal miR-19b-3p on OA ferroptosis and its potential mechanisms. Methods Interleukin (IL)-1β-stimulated chondrocytes and medial meniscus surgery were used to construct the OA cellular model and the OA rat model, respectively. OA-FLS exosomes with/without miR-19b-3p modification were added to the IL-1β-stimulated chondrocytes and OA rat models, followed by direct miR-19b-3p mimic/inhibitor transfection with/without SLC7A11 overexpression plasmids. miR-19b-3p, ferroptosis-related markers (malondialdehyde (MDA), glutathione (GSH)/oxidized glutathione (GSSG), ferrous ion (Fe2+), glutathione peroxidase 4 (GPX4), solute carrier family 7 member 11 (SLC7A11), and acyl-CoA synthetase long-chain family member 4 (ACSL4)), mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) levels were detected. Results Enhanced ferroptosis reflected by dysregulated ferroptosis-related markers, a reduced MMP, and an increased ROS was observed in cartilage tissues from OA patients vs. controls, IL-1β-stimulated chondrocytes vs. normal ones, and OA rat models vs. sham, so did miR-19b-3p. OA-FLS exosomes promoted MDA, Fe2+, ACSL4, and ROS but reduced cell viability, GSH/GSSG, GPX4, SLC7A11, and MMP in IL-1β-stimulated chondrocytes, whose effect was enhanced by miR-19b-3p mimics and attenuated by miR-19b-3p inhibitors. miR-19b-3p negatively regulated SLC7A11 and directly bound to SLC7A11 via luciferase reporter gene assay. Furthermore, SLC7A11 overexpression weakened miR-19b-3p mimics' effect on ferroptosis-related markers, MMP, or ROS in IL-1β-stimulated chondrocytes. OA-FLS exosomes also induced cartilage damage and ferroptosis in OA rats whose influence was tempered by miR-19b-3p inhibitors. Conclusion OA-FLS exosomal miR-19b-3p enhances cartilage ferroptosis and damage by sponging SLC7A11 in OA, indicating a potential linkage among synovium, cartilage, and ferroptosis during the OA process.
Collapse
Affiliation(s)
- Ruina Kong
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lianmei Ji
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yafei Pang
- Department of Rheumatology, Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Dongbao Zhao
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Gao
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
30
|
Tang Y, Zhao C, Zhuang Y, Zhong A, Wang M, Zhang W, Zhu L. Mechanosensitive Piezo1 protein as a novel regulator in macrophages and macrophage-mediated inflammatory diseases. Front Immunol 2023; 14:1149336. [PMID: 37334369 PMCID: PMC10275567 DOI: 10.3389/fimmu.2023.1149336] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Macrophages are the most important innate immune cells in humans. They are almost ubiquitous in peripheral tissues with a large variety of different mechanical milieus. Therefore, it is not inconceivable that mechanical stimuli have effects on macrophages. Emerging as key molecular detectors of mechanical stress, the function of Piezo channels in macrophages is becoming attractive. In this review, we addressed the architecture, activation mechanisms, biological functions, and pharmacological regulation of the Piezo1 channel and review the research advancements in functions of Piezo1 channels in macrophages and macrophage-mediated inflammatory diseases as well as the potential mechanisms involved.
Collapse
Affiliation(s)
- Yu Tang
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chuanxiang Zhao
- Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai’an, Jiangsu, China
| | - Ying Zhuang
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Anjing Zhong
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ming Wang
- Department of Medical Imaging, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Liqun Zhu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
31
|
Kehayova YS, Wilkinson JM, Rice SJ, Loughlin J. Osteoarthritis genetic risk acting on the galactosyltransferase gene COLGALT2 has opposing functional effects in articulating joint tissues. Arthritis Res Ther 2023; 25:83. [PMID: 37208701 DOI: 10.1186/s13075-023-03066-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/10/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Investigation of cartilage and chondrocytes has revealed that the osteoarthritis risk marked by the independent DNA variants rs11583641 and rs1046934 mediate their effects by decreasing the methylation status of CpG dinucleotides in enhancers and increasing the expression of shared target gene COLGALT2. We set out to investigate if these functional effects operate in a non-cartilaginous joint tissue. METHODS Nucleic acids were extracted from the synovium of osteoarthritis patients. Samples were genotyped, and DNA methylation was quantified by pyrosequencing at CpGs within the COLGALT2 enhancers. CpGs were tested for enhancer effects using a synovial cell line and a reporter gene assay. DNA methylation was altered using epigenetic editing, with the impact on gene expression determined using quantitative polymerase chain reaction. In silico analysis complemented laboratory experiments. RESULTS The rs1046934 genotype did not associate with DNA methylation or COLGALT2 expression in the synovium, whereas the rs11583641 genotype did. Surprisingly, the effects for rs11583641 were opposite to those previously observed in cartilage. Epigenetic editing in synovial cells revealed that enhancer methylation is causally linked to COLGALT2 expression. CONCLUSIONS This is the first direct demonstration for osteoarthritis genetic risk of a functional link between DNA methylation and gene expression operating in opposite directions between articular joint tissues. It highlights pleiotropy in the action of osteoarthritis risk and provides a cautionary note in the application of future genetically based osteoarthritis therapies: an intervention that decreases the detrimental effect of a risk allele in one joint tissue may inadvertently increase its detrimental effect in another joint tissue.
Collapse
Affiliation(s)
- Yulia S Kehayova
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle Upon Tyne, NE1 3BZ, UK
| | - J Mark Wilkinson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Sarah J Rice
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle Upon Tyne, NE1 3BZ, UK.
| | - John Loughlin
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle Upon Tyne, NE1 3BZ, UK.
| |
Collapse
|
32
|
Yoshimoto M, Sadamori K, Tokumura K, Tanaka Y, Fukasawa K, Hinoi E. Bioinformatic analysis reveals potential relationship between chondrocyte senescence and protein glycosylation in osteoarthritis pathogenesis. Front Endocrinol (Lausanne) 2023; 14:1153689. [PMID: 37265706 PMCID: PMC10229820 DOI: 10.3389/fendo.2023.1153689] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/04/2023] [Indexed: 06/03/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative and progressive joint disease. Cellular senescence is an irreversible cell cycle arrest progressive with age, while protein glycosylation is the most abundant post-translational modification, regulating various cellular and biological pathways. The implication of either chondrocyte senescence or protein glycosylation in the OA pathogenesis has been extensively and individually studied. In this study, we aimed to investigate the possible relationship between chondrocyte senescence and protein glycosylation on the pathogenesis of OA using single-cell RNA sequencing datasets of clinical OA specimens deposited in the Gene Expression Omnibus database with a different cohort. We demonstrated that both cellular senescence signal and protein glycosylation pathways in chondrocytes are validly associated with OA pathogenesis. In addition, the cellular senescence signal is well-connected to the O-linked glycosylation pathway in OA chondrocyte and vice-versa. The expression levels of the polypeptide N-acetylgalactosaminyltransferase (GALNT) family, which is essential for the biosynthesis of O-Glycans at the early stage, are highly upregulated in OA chondrocytes. Moreover, the expression levels of the GALNT family are prominently associated with chondrocyte senescence as well as pathological features of OA. Collectively, these findings uncover a crucial relationship between chondrocyte senescence and O-linked glycosylation on the OA pathophysiology, thereby revealing a potential target for OA.
Collapse
Affiliation(s)
- Makoto Yoshimoto
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Koki Sadamori
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Kazuya Tokumura
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuki Tanaka
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Kazuya Fukasawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Eiichi Hinoi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
- Center for One Medicine Innovative Translational Research, Division of Innovative Modality Development, Gifu University, Gifu, Japan
| |
Collapse
|
33
|
Conditioned Medium - Is it an Undervalued Lab Waste with the Potential for Osteoarthritis Management? Stem Cell Rev Rep 2023:10.1007/s12015-023-10517-1. [PMID: 36790694 PMCID: PMC10366316 DOI: 10.1007/s12015-023-10517-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND The approaches currently used in osteoarthritis (OA) are mainly short-term solutions with unsatisfactory outcomes. Cell-based therapies are still controversial (in terms of the sources of cells and the results) and require strict culture protocol, quality control, and may have side-effects. A distinct population of stromal cells has an interesting secretome composition that is underrated and commonly ends up as biological waste. Their unique properties could be used to improve the existing techniques due to protective and anti-ageing properties. SCOPE OF REVIEW In this review, we seek to outline the advantages of the use of conditioned media (CM) and exosomes, which render them superior to other cell-based methods, and to summarise current information on the composition of CM and their effect on chondrocytes. MAJOR CONCLUSIONS CM are obtainable from a variety of mesenchymal stromal cell (MSC) sources, such as adipose tissue, bone marrow and umbilical cord, which is significant to their composition. The components present in CMs include proteins, cytokines, growth factors, chemokines, lipids and ncRNA with a variety of functions. In most in vitro and in vivo studies CM from MSCs had a beneficial effect in enhance processes associated with chondrocyte OA pathomechanism. GENERAL SIGNIFICANCE This review summarises the information available in the literature on the function of components most commonly detected in MSC-conditioned media, as well as the effect of CM on OA chondrocytes in in vitro culture. It also highlights the need to standardise protocols for obtaining CM, and to conduct clinical trials to transfer the effects obtained in vitro to human subjects.
Collapse
|
34
|
Chang B, Hu Z, Chen L, Jin Z, Yang Y. Development and validation of cuproptosis-related genes in synovitis during osteoarthritis progress. Front Immunol 2023; 14:1090596. [PMID: 36817415 PMCID: PMC9932029 DOI: 10.3389/fimmu.2023.1090596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common refractory degenerative joint diseases worldwide. Synovitis is believed to drive joint cartilage destruction during OA pathogenesis. Cuproptosis is a novel form of copper-induced cell death. However, few studies have examined the correlations between cuproptosis-related genes (CRGs), immune infiltration, and synovitis. Therefore, we analyzed CRGs in synovitis during OA. Microarray datasets (GSE55235, GSE55457, GSE12021, GSE82107 and GSE176308) were downloaded from the Gene Expression Omnibus database. Next, we conducted differential and subtype analyses of CRGs across synovitis. Immune infiltration and correlation analyses were performed to explore the association between CRGs and immune cell abundance in synovitis. Finally, single-cell RNA-seq profiling was performed using the GSE176308 dataset to investigate the expression of CRGs in the various cell clusters. We found that the expression of five CRGs (FDX1, LIPT1, PDHA1, PDHB, and CDKN2A) was significantly increased in the OA synovium. Moreover, abundant and various types of immune cells infiltrated the synovium during OA, which was correlated with the expression of CRGs. Additionally, single-cell RNA-seq profiling revealed that the cellular composition of the synovium was complex and that their proportions varied greatly as OA progressed. The expression of CRGs differed across various cell types in the OA synovium. The current study predicted that cuproptosis may be involved in the pathogenesis of synovitis. The five screened CRGs (FDX1, LIPT1, PDHA1, PDHB, and CDKN2A) could be explored as candidate biomarkers or therapeutic targets for OA synovitis.
Collapse
Affiliation(s)
- Bohan Chang
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhehan Hu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liang Chen
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhuangzhuang Jin
- Department of Emergence Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Yang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
35
|
Kwapisz A, Bowman M, Walters J, Cosh H, Burnikel B, Tokish J, Ye T, Mercuri J. Human Adipose- and Amnion-Derived Mesenchymal Stromal Cells Similarly Mitigate Osteoarthritis Progression in the Dunkin Hartley Guinea Pig. Am J Sports Med 2022; 50:3963-3973. [PMID: 36300544 DOI: 10.1177/03635465221126683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Clinical trials are currently underway to investigate the efficacy of intra-articular administration of mesenchymal stromal cells (MSCs) to mitigate osteoarthritis (OA) progression in the knee. Although multiple MSC sources exist, studies have yet to determine whether differences in therapeutic efficacy exist between them. PURPOSE To compare the ability of intra-articularly injected adipose-derived MSCs (AD-MSCs) and amnion-derived MSCs (AM-MSCs) to mitigate the progression of knee OA in a small animal model of spontaneous OA, as well as to compare the therapeutic potential of MSCs in hyaluronic acid (HA) and in HA only with saline (OA) controls. STUDY DESIGN Controlled laboratory study. METHODS Injections of AD-MSCs or AM-MSCs suspended in HA or HA only were performed in the rear stifle joints of 3-month-old Dunkin Hartley guinea pigs (DHGPs). Repeat injections occurred at 2 and 4 months after the initial injection in each animal. Contralateral limbs received saline injections and served as untreated controls. Subsequently, joints were analyzed for osteoarthritic changes of the cartilage and subchondral bone via histologic and biochemical analyses. To evaluate MSC retention time in the joint space, DHGPs received a single intra-articular injection of fluorescently labeled AD-MSCs or AM-MSCs, and the fluorescence intensity was longitudinally tracked via an in vivo imaging system. RESULTS No statistically significant differences in outcomes were found when comparing the ability of AD-MSCs and AM-MSCs to mitigate OA. However, the injection of AD-MSCs, AM-MSCs, and HA-only treatments more effectively mitigated cartilage damage compared with that of saline controls by demonstrating higher amounts of cartilage glycosaminoglycan content and improved histological proteoglycan scoring while reducing the percentage of osteophytes present. CONCLUSION Intra-articular injection of AD-MSCs, AM-MSCs, or HA only was able to similarly mitigate the progression of cartilage damage and reduce the percentage of osteophytes compared with that of saline controls in the DHGP. However, this study was unable to establish the superiority of AD-MSCs versus AM-MSCs as a treatment to mitigate spontaneous OA. CLINICAL RELEVANCE MSCs demonstrate the ability to mitigate the progression of knee OA and thus may be used in a prophylactic approach to delay the need for end-stage treatment strategies.
Collapse
Affiliation(s)
- Adam Kwapisz
- Clinic of Orthopedics and Pediatric Orthopedics, Medical University of Lodz, Lodz, Poland
- Steadman Hawkins Clinic of the Carolinas, Department of Orthopaedic Surgery, Prisma Health, Greenville, South Carolina, USA
| | - Mackenzie Bowman
- Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Joshua Walters
- Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Heather Cosh
- Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Brian Burnikel
- Steadman Hawkins Clinic of the Carolinas, Department of Orthopaedic Surgery, Prisma Health, Greenville, South Carolina, USA
| | - John Tokish
- Mayo Clinic, Department of Orthopedic Surgery, Phoenix, Arizona, USA
| | - Tong Ye
- Nano and Functional Imaging Lab, Department of Bioengineering, Clemson University, Charleston, South Carolina, USA
| | - Jeremy Mercuri
- Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
- Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, South Carolina, USA
| |
Collapse
|
36
|
Xin Y, Wang W, Mao E, Yang H, Li S. Targeting NLRP3 Inflammasome Alleviates Synovitis by Reducing Pyroptosis in Rats with Experimental Temporomandibular Joint Osteoarthritis. Mediators Inflamm 2022; 2022:2581151. [PMID: 36466156 PMCID: PMC9712023 DOI: 10.1155/2022/2581151] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/29/2022] [Accepted: 10/29/2022] [Indexed: 11/14/2023] Open
Abstract
The mechanism of temporomandibular joint osteoarthritis (TMJOA), which leads to the final erosion of cartilage and subchondral bone, has been widely demonstrated, but still not clearly elucidated. Many studies have pointed that NLRP3-mediated inflammation played a vital role in degenerative diseases. However, its interaction with synovitis of TMJOA has remained poorly investigated. In our study, we explored the role of NLRP3 inflammasome in TMJOA synovitis and the therapeutic potential of caspase-1 and NLRP3 inhibitors. By establishing a rat TMJOA model, we found that NLRP3 was upregulated in synovial tissue of TMJOA. It was involved in the progress of a programmed cell death called pyroptosis, which was caspase-1 dependent and ultimately triggered inflammatory mediator interleukin IL-1β release. Treatment with Ac-YVAD-cmk and MCC950, inhibitors targeting caspase-1 and NLRP3, respectively, significantly suppressed pyroptosis in TMJOA synovial tissue. Then, a macrophage- and fibroblast-like synoviocyte (FLS) cocultured model further verified the above results. Macrophage somehow promoted FLS pyroptosis in this study. Our results suggested that the NLRP3 inflammasome-mediated pyroptosis participated in synovial inflammation of TMJOA. Interfering with the progress could be a potential option for controlling TMJOA development.
Collapse
Affiliation(s)
- Yinzi Xin
- Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Wei Wang
- Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Enyu Mao
- Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Hefeng Yang
- Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Song Li
- Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| |
Collapse
|
37
|
Shesternya PA, Savchenko AA, Gritsenko OD, Vasileva AO, Kudryavtsev IV, Masterova AA, Isakov DV, Borisov AG. Features of Peripheral Blood Th-Cell Subset Composition and Serum Cytokine Level in Patients with Activity-Driven Ankylosing Spondylitis. Pharmaceuticals (Basel) 2022; 15:ph15111370. [PMID: 36355542 PMCID: PMC9695783 DOI: 10.3390/ph15111370] [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: 10/09/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Th cells may exhibit pathological activity depending on the regulatory and functional signals sensed under a wide range of immunopathological conditions, including ankylosing spondylitis (AS). The relationship between Th cells and cytokines is important for diagnoses and for determining treatment. Accordingly, the aim of this study was to investigate the relationship between Th-cell subset composition and serum cytokine profile for patients with activity-driven AS. In our study, patients were divided into two groups according to disease activity: low-activity AS (ASDAS-CRP < 2.1) and high-activity AS (ASDAS-CRP > 2.1). The peripheral blood Th cell subset composition was studied by flow cytometry. Using multiplex analysis, serum cytokine levels were quantified and investigated. It was found that only patients with high-activity AS had reduced central memory (CM) Th1 cells (p = 0.035) but elevated numbers of CM (p = 0.014) and effector memory (EM) Th2 cells (p < 0.001). However, no activity-driven change in the Th17 cell subset composition was observed in AS patients. Moreover, low-AS activity patients had increased numbers of Tfh17 EM cells (p < 0.001), whereas high-AS activity was associated with elevated Tfh2 EM level (p = 0.031). The serum cytokine profiles in AS patients demonstrated that cues stimulating cellular immunity were increased, but patients with high-AS activity reveled increased IL-5 level (p = 0.017). Analyzing the data obtained from AS patients allowed us to conclude that Th cell subset differentiation was mainly affected during the CM stage and characterized the IL-23/IL-17 regulatory axis, whereas increased humoral immunity was observed in the high-AS activity group.
Collapse
Affiliation(s)
- Pavel A. Shesternya
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Healthcare, 660022 Krasnoyarsk, Russia
- Correspondence:
| | - Andrei A. Savchenko
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Healthcare, 660022 Krasnoyarsk, Russia
- Federal Research Center “Krasnoyarsk Science Center”, Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Olga D. Gritsenko
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Healthcare, 660022 Krasnoyarsk, Russia
| | - Alexandra O. Vasileva
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Healthcare, 660022 Krasnoyarsk, Russia
| | | | - Alena A. Masterova
- Federal Research Center “Krasnoyarsk Science Center”, Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Dmitry V. Isakov
- Academician I.P. Pavlov First St. Petersburg State Medical University, Ministry of Healthcare, 197022 St. Peterburg, Russia
| | - Alexandr G. Borisov
- Federal Research Center “Krasnoyarsk Science Center”, Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| |
Collapse
|
38
|
Xia Y, Yang R, Hou Y, Wang H, Li Y, Zhu J, Fu C. Application of mesenchymal stem cell-derived exosomes from different sources in intervertebral disc degeneration. Front Bioeng Biotechnol 2022; 10:1019437. [PMID: 36277386 PMCID: PMC9585200 DOI: 10.3389/fbioe.2022.1019437] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 12/12/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is a main cause of lower back pain, leading to psychological and economic burdens to patients. Physical therapy only delays pain in patients but cannot eliminate the cause of IVDD. Surgery is required when the patient cannot tolerate pain or has severe neurological symptoms. Although surgical resection of IVD or decompression of the laminae eliminates the diseased segment, it damages adjacent normal IVD. There is also a risk of re-protrusion after IVD removal. Cell therapy has played a crucial role in the development of regenerative medicine. Cell transplantation promotes regeneration of degenerative tissue. However, owing to the lack of vascular structure in IVD, sufficient nutrients cannot be provided for transplanted mesenchymal stem cells (MSCs). In addition, dead cells release harmful substances that aggravate IVDD. Extracellular vesicles (EVs) have been extensively studied as an emerging therapeutic approach. EVs generated by paracrine MSCs retain the potential of MSCs and serve as carriers to deliver their contents to target cells to regulate target cell activity. Owing to their double-layered membrane structure, EVs have a low immunogenicity and no immune rejection. Therefore, EVs are considered an emerging therapeutic modality in IVDD. However, they are limited by mass production and low loading rates. In this review, the structure of IVD and advantages of EVs are introduced, and the application of MSC-EVs in IVDD is discussed. The current limitations of EVs and future applications are described.
Collapse
Affiliation(s)
- Yuanliang Xia
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ruohan Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yulin Hou
- Department of Cardiology, Guangyuan Central Hospital, Guangyuan, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuehong Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Changfeng Fu,
| |
Collapse
|
39
|
Anderson JR, Jacobsen S, Walters M, Bundgaard L, Diendorfer A, Hackl M, Clarke EJ, James V, Peffers MJ. Small non-coding RNA landscape of extracellular vesicles from a post-traumatic model of equine osteoarthritis. Front Vet Sci 2022; 9:901269. [PMID: 36003409 PMCID: PMC9393553 DOI: 10.3389/fvets.2022.901269] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/18/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles comprise an as yet inadequately investigated intercellular communication pathway in the field of early osteoarthritis. We hypothesised that the small non-coding RNA expression pattern in synovial fluid and plasma would change during progression of experimental osteoarthritis. In this study, we conducted small RNA sequencing to provide a comprehensive overview of the temporal expression profiles of small non-coding transcripts carried by extracellular vesicles derived from plasma and synovial fluid for the first time in a posttraumatic model of equine osteoarthritis. Additionally, we characterised synovial fluid and plasma-derived extracellular vesicles with respect to quantity, size, and surface markers. The different temporal expressions of seven microRNAs in plasma and synovial fluid-derived extracellular vesicles, eca-miR-451, eca-miR-25, eca-miR-215, eca-miR-92a, eca-miR-let-7c, eca-miR-486-5p, and eca-miR-23a, and four snoRNAs, U3, snord15, snord46, and snord58, represent potential biomarkers for early osteoarthritis. Bioinformatics analysis of the differentially expressed microRNAs in synovial fluid highlighted that in early osteoarthritis these related to the inhibition of cell cycle, cell cycle progression, DNA damage and cell proliferation as well as increased cell viability and differentiation of stem cells. Plasma and synovial fluid-derived extracellular vesicle small non-coding signatures have been established for the first time in a temporal model of osteoarthritis. These could serve as novel biomarkers for evaluation of osteoarthritis progression or act as potential therapeutic targets.
Collapse
Affiliation(s)
- James R. Anderson
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Marie Walters
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Louise Bundgaard
- Department of Veterinary Clinical Sciences, University of Copenhagen, Taastrup, Denmark
| | | | | | - Emily J. Clarke
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Mandy J. Peffers
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
40
|
Wu Y, Li J, Zeng Y, Pu W, Mu X, Sun K, Peng Y, Shen B. Exosomes rewire the cartilage microenvironment in osteoarthritis: from intercellular communication to therapeutic strategies. Int J Oral Sci 2022; 14:40. [PMID: 35927232 PMCID: PMC9352673 DOI: 10.1038/s41368-022-00187-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage loss and accounts for a major source of pain and disability worldwide. However, effective strategies for cartilage repair are lacking, and patients with advanced OA usually need joint replacement. Better comprehending OA pathogenesis may lead to transformative therapeutics. Recently studies have reported that exosomes act as a new means of cell-to-cell communication by delivering multiple bioactive molecules to create a particular microenvironment that tunes cartilage behavior. Specifically, exosome cargos, such as noncoding RNAs (ncRNAs) and proteins, play a crucial role in OA progression by regulating the proliferation, apoptosis, autophagy, and inflammatory response of joint cells, rendering them promising candidates for OA monitoring and treatment. This review systematically summarizes the current insight regarding the biogenesis and function of exosomes and their potential as therapeutic tools targeting cell-to-cell communication in OA, suggesting new realms to improve OA management.
Collapse
Affiliation(s)
- Yuangang Wu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Li
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zeng
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenchen Pu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Mu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kaibo Sun
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Bin Shen
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
41
|
Sun X, Li KX, Figueiredo ML, Lin CC, Li BY, Yokota H. Generation of the Chondroprotective Proteomes by Activating PI3K and TNFα Signaling. Cancers (Basel) 2022; 14:cancers14133039. [PMID: 35804814 PMCID: PMC9264838 DOI: 10.3390/cancers14133039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/13/2022] [Accepted: 06/18/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Chondrosarcoma and inflammatory arthritis are two joint-damaging diseases. Here, we examined whether a counterintuitive approach of activating tumorigenic and inflammatory signaling may generate joint-protective proteomes in mesenchymal stem cells and chondrocytes for the treatment of chondrosarcoma and inflammatory arthritis. While activating PI3K signaling and the administration of TNFα to chondrosarcoma cells and chondrocytes promoted tumor progression and inflammatory responses, those cells paradoxically generated a chondroprotective conditioned medium. Notably, the chondroprotective conditioned medium was enriched with Hsp90ab1 that interacted with GAPDH. Extracellular GAPDH interacted with L1CAM, an oncogenic transmembrane protein, and inhibited tumorigenic behaviors, whereas intracellular GAPDH downregulated p38 in chondrocytes and exerted anti-inflammatory effects. The result supports the unconventional approach of generating chondroprotective proteomes. Abstract Purpose: To develop a novel treatment option for Chondrosarcoma (CS) and inflammatory arthritis, we evaluated a counterintuitive approach of activating tumorigenic and inflammatory signaling for generating joint-protective proteomes. Methods: We employed mesenchymal stem cells and chondrocytes to generate chondroprotective proteomes by activating PI3K signaling and the administration of TNFα. The efficacy of the proteomes was examined using human and mouse cell lines as well as a mouse model of CS. The regulatory mechanism was analyzed using mass spectrometry-based whole-genome proteomics. Results: While tumor progression and inflammatory responses were promoted by activating PI3K signaling and the administration of TNFα to CS cells and chondrocytes, those cells paradoxically generated a chondroprotective conditioned medium (CM). The application of CM downregulated tumorigenic genes in CS cells and TNFα and MMP13 in chondrocytes. Mechanistically, Hsp90ab1 was enriched in the chondroprotective CM, and it immunoprecipitated GAPDH. Extracellular GAPDH interacted with L1CAM and inhibited tumorigenic behaviors, whereas intracellular GAPDH downregulated p38 and exerted anti-inflammatory effects. Conclusions: We demonstrated that the unconventional approach of activating oncogenic and inflammatory signaling can generate chondroprotective proteomes. The role of Hsp90ab1 and GAPDH differed in their locations and they acted as the uncommon protectors of the joint tissue from tumor and inflammatory responses.
Collapse
Affiliation(s)
- Xun Sun
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China; (X.S.); (K.-X.L.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Ke-Xin Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China; (X.S.); (K.-X.L.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Marxa L. Figueiredo
- Department of Basic Medical Sciences and Interdisciplinary Biomedical Sciences Program, Purdue University, West Lafayette, IN 47907, USA;
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China; (X.S.); (K.-X.L.)
- Correspondence: (B.-Y.L.); (H.Y.); Tel.: +86-451-8667-1354 (B.-Y.L.); +1-317-278-5177 (H.Y.); Fax: +86-451-8667-1354 (B.-Y.L.); +1-317-278-2455 (H.Y.)
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: (B.-Y.L.); (H.Y.); Tel.: +86-451-8667-1354 (B.-Y.L.); +1-317-278-5177 (H.Y.); Fax: +86-451-8667-1354 (B.-Y.L.); +1-317-278-2455 (H.Y.)
| |
Collapse
|
42
|
Sanchez-Lopez E, Coras R, Torres A, Lane NE, Guma M. Synovial inflammation in osteoarthritis progression. Nat Rev Rheumatol 2022; 18:258-275. [PMID: 35165404 PMCID: PMC9050956 DOI: 10.1038/s41584-022-00749-9] [Citation(s) in RCA: 434] [Impact Index Per Article: 144.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2022] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a progressive degenerative disease resulting in joint deterioration. Synovial inflammation is present in the OA joint and has been associated with radiographic and pain progression. Several OA risk factors, including ageing, obesity, trauma and mechanical loading, play a role in OA pathogenesis, likely by modifying synovial biology. In addition, other factors, such as mitochondrial dysfunction, damage-associated molecular patterns, cytokines, metabolites and crystals in the synovium, activate synovial cells and mediate synovial inflammation. An understanding of the activated pathways that are involved in OA-related synovial inflammation could form the basis for the stratification of patients and the development of novel therapeutics. This Review focuses on the biology of the OA synovium, how the cells residing in or recruited to the synovium interact with each other, how they become activated, how they contribute to OA progression and their interplay with other joint structures.
Collapse
Affiliation(s)
- Elsa Sanchez-Lopez
- Department of Orthopaedic Surgery, University of California San Diego, San Diego, CA, USA
| | - Roxana Coras
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, San Diego, CA, USA
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Alyssa Torres
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Nancy E Lane
- Division of Rheumatology, Department of Medicine, University of California Davis, Davis, CA, USA
| | - Monica Guma
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, San Diego, CA, USA.
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain.
- San Diego VA Healthcare Service, San Diego, CA, USA.
| |
Collapse
|
43
|
Connection between Mesenchymal Stem Cells Therapy and Osteoclasts in Osteoarthritis. Int J Mol Sci 2022; 23:ijms23094693. [PMID: 35563083 PMCID: PMC9102843 DOI: 10.3390/ijms23094693] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
The use of mesenchymal stem cells constitutes a promising therapeutic approach, as it has shown beneficial effects in different pathologies. Numerous in vitro, pre-clinical, and, to a lesser extent, clinical trials have been published for osteoarthritis. Osteoarthritis is a type of arthritis that affects diarthritic joints in which the most common and studied effect is cartilage degradation. Nowadays, it is known that osteoarthritis is a disease with a very powerful inflammatory component that affects the subchondral bone and the rest of the tissues that make up the joint. This inflammatory component may induce the differentiation of osteoclasts, the bone-resorbing cells. Subchondral bone degradation has been suggested as a key process in the pathogenesis of osteoarthritis. However, very few published studies directly focus on the activity of mesenchymal stem cells on osteoclasts, contrary to what happens with other cell types of the joint, such as chondrocytes, synoviocytes, and osteoblasts. In this review, we try to gather the published bibliography in relation to the effects of mesenchymal stem cells on osteoclastogenesis. Although we find promising results, we point out the need for further studies that can support mesenchymal stem cells as a therapeutic tool for osteoclasts and their consequences on the osteoarthritic joint.
Collapse
|
44
|
von Mentzer U, Corciulo C, Stubelius A. Biomaterial Integration in the Joint: Pathological Considerations, Immunomodulation, and the Extracellular Matrix. Macromol Biosci 2022; 22:e2200037. [PMID: 35420256 DOI: 10.1002/mabi.202200037] [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: 02/27/2022] [Revised: 03/30/2022] [Indexed: 11/08/2022]
Abstract
Defects of articular joints are becoming an increasing societal burden due to a persistent increase in obesity and aging. For some patients suffering from cartilage erosion, joint replacement is the final option to regain proper motion and limit pain. Extensive research has been undertaken to identify novel strategies enabling earlier intervention to promote regeneration and cartilage healing. With the introduction of decellularized extracellular matrix (dECM), researchers have tapped into the potential for increased tissue regeneration by designing biomaterials with inherent biochemical and immunomodulatory signals. Compared to conventional and synthetic materials, dECM-based materials invoke a reduced foreign body response. It is therefore highly beneficial to understand the interplay of how these native tissue-based materials initiate a favorable remodeling process by the immune system. Yet, such an understanding also demands increasing considerations of the pathological environment and remodeling processes, especially for materials designed for early disease intervention. This knowledge would avoid rejection and help predict complications in conditions with inflammatory components such as arthritides. This review outlines general issues facing biomaterial integration and emphasizes the importance of tissue-derived macromolecular components in regulating essential homeostatic, immunological, and pathological processes to increase biomaterial integration for patients suffering from joint degenerative diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ula von Mentzer
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| | - Carmen Corciulo
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation, Sahlgrenska Academy at the University of Gothenburg, Guldhedsgatan 10A, Gothenburg, 41296, Sweden
| | - Alexandra Stubelius
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| |
Collapse
|
45
|
Minguzzi M, Panichi V, D’Adamo S, Cetrullo S, Cattini L, Flamigni F, Mariani E, Borzì RM. Pleiotropic Roles of NOTCH1 Signaling in the Loss of Maturational Arrest of Human Osteoarthritic Chondrocytes. Int J Mol Sci 2021; 22:ijms222112012. [PMID: 34769441 PMCID: PMC8585104 DOI: 10.3390/ijms222112012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 02/07/2023] Open
Abstract
Notch signaling has been identified as a critical regulator of cartilage development and homeostasis. Its pivotal role was established by both several joint specific Notch signaling loss of function mouse models and transient or sustained overexpression. NOTCH1 is the most abundantly expressed NOTCH receptors in normal cartilage and its expression increases in osteoarthritis (OA), when chondrocytes exit from their healthy “maturation arrested state” and resume their natural route of proliferation, hypertrophy, and terminal differentiation. The latter are hallmarks of OA that are easily evaluated in vitro in 2-D or 3-D culture models. The aim of our study was to investigate the effect of NOTCH1 knockdown on proliferation (cell count and Picogreen mediated DNA quantification), cell cycle (flow cytometry), hypertrophy (gene and protein expression of key markers such as RUNX2 and MMP-13), and terminal differentiation (viability measured in 3-D cultures by luminescence assay) of human OA chondrocytes. NOTCH1 silencing of OA chondrocytes yielded a healthier phenotype in both 2-D (reduced proliferation) and 3-D with evidence of decreased hypertrophy (reduced expression of RUNX2 and MMP-13) and terminal differentiation (increased viability). This demonstrates that NOTCH1 is a convenient therapeutic target to attenuate OA progression.
Collapse
Affiliation(s)
- Manuela Minguzzi
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy; (M.M.); (S.D.); (E.M.)
| | - Veronica Panichi
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40138 Bologna, Italy; (V.P.); (S.C.); (F.F.)
| | - Stefania D’Adamo
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy; (M.M.); (S.D.); (E.M.)
| | - Silvia Cetrullo
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40138 Bologna, Italy; (V.P.); (S.C.); (F.F.)
| | - Luca Cattini
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Flavio Flamigni
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40138 Bologna, Italy; (V.P.); (S.C.); (F.F.)
| | - Erminia Mariani
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy; (M.M.); (S.D.); (E.M.)
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Rosa Maria Borzì
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
- Correspondence:
| |
Collapse
|