1
|
Pemmari A, Moilanen E. Macrophage and chondrocyte phenotypes in inflammation. Basic Clin Pharmacol Toxicol 2024; 135:537-549. [PMID: 39319534 DOI: 10.1111/bcpt.14085] [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: 05/12/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024]
Abstract
Inflammation is a complex biological process protecting the body from diverse external threats. Effectively performing this task requires an intricate, well-regulated interplay of different cells and tissues. Furthermore, several cells participating in inflammation can assume diverse phenotypes. A classic and relatively well-studied example of phenotypic diversity in inflammation is macrophage polarization. Based on the TH1/TH2 phenotypes of T helper cells, this scheme has proinflammatory "classical/M1" activation contrasted with the anti-inflammatory and healing-promoting "alternative/M2" phenotype. Some authors have extended the concept into an M17 phenotype induced by the classic TH17 cytokine IL-17. Phenotypic changes in chondrocytes have also been studied especially in the context of osteoarthritis (OA), and there are indications that these cells can also assume polarized phenotypes at least partly analogous to those of TH cells and macrophages. The therapeutic success of biological agents targeting TH1/TH2/TH17 inductor and/or effector cytokines displays the utility of the concept of polarization. The aim of this focused review is to survey the internal and external factors affecting macrophage and chondrocyte phenotypes (such as inflammatory cytokines, widely used medications and natural products) and to explore the possibility of ameliorating pathological states by modulating these phenotypes.
Collapse
Affiliation(s)
- Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
2
|
Qiu F, Xie D, Chen H, Wang Z, Huang J, Cao C, Liang Y, Yang X, He DY, Fu X, Lu A, Liang C. Generation of cytotoxic aptamers specifically targeting fibroblast-like synoviocytes by CSCT-SELEX for treatment of rheumatoid arthritis. Ann Rheum Dis 2024:ard-2024-225565. [PMID: 39237134 DOI: 10.1136/ard-2024-225565] [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: 01/22/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is an autoimmune disease characterised by aggressive fibroblast-like synoviocytes (FLSs). Very few RA patients-derived FLSs (RA-FLSs)-specific surface signatures have been identified, and there is currently no approved targeted therapy for RA-FLSs. This study aimed to screen therapeutic aptamers with cell-targeting and cytotoxic properties against RA-FLSs and to uncover the molecular targets and mechanism of action of the screened aptamers. METHODS We developed a cell-specific and cytotoxic systematic evolution of ligands by exponential enrichment (CSCT-SELEX) method to screen the therapeutic aptamers without prior knowledge of the surface signatures of RA-FLSs. The molecular targets and mechanisms of action of the screened aptamers were determined by pull-down assays and RNA sequencing. The therapeutic efficacy of the screened aptamers was examined in arthritic mouse models. RESULTS We obtained an aptamer SAPT8 that selectively recognised and killed RA-FLSs. The molecular target of SAPT8 was nucleolin (NCL), a shuttling protein overexpressed on the surface and involved in the tumor-like transformation of RA-FLSs. Mechanistically, SAPT8 interacted with the surface NCL and was internalised to achieve lysosomal degradation of NCL, leading to the upregulation of proapoptotic p53 and downregulation of antiapoptotic B-cell lymphoma 2 (Bcl-2) in RA-FLSs. When administrated systemically to arthritic mice, SAPT8 accumulated in the inflamed FLSs of joints. SAPT8 monotherapy or its combination with tumour necrosis factor (TNF)-targeted biologics was shown to relieve arthritis in mouse models. CONCLUSIONS CSCT-SELEX could be a promising strategy for developing cell-targeting and cytotoxic aptamers. SAPT8 aptamer selectively ablates RA-FLSs via modulating NCL-p53/Bcl-2 signalling, representing a potential alternative or complementary therapy for RA.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | | | - Xu Yang
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Dong-Yi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
3
|
Tian K, He X, Lin X, Chen X, Su Y, Lu Z, Chen Z, Zhang L, Li P, Ma L, Lan Z, Zhao X, Fen G, Hai Q, Xue D, Jin Q. Unveiling the Role of Sik1 in Osteoblast Differentiation: Implications for Osteoarthritis. Mol Cell Biol 2024; 44:411-428. [PMID: 39169784 PMCID: PMC11485870 DOI: 10.1080/10985549.2024.2385633] [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: 03/25/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by subchondral osteosclerosis, mainly due to osteoblast activity. This research investigates the function of Sik1, a member of the AMP-activated protein kinase family, in OA. Proteomic analysis was conducted on clinical samples from 30 OA patients, revealing a negative correlation between Sik1 expression and OA. In vitro experiments utilized BMSCs to examine the effect of Sik1 on osteogenic differentiation. BMSCs were cultured and induced toward osteogenesis with specific media. Sik1 overexpression was achieved through lentiviral transfection, followed by analysis of osteogenesis-associated proteins using Western blotting, RT-qPCR, and alkaline phosphate staining. In vivo experiments involved destabilizing the medial meniscus in mice to establish an OA model, assessing the therapeutic potential of Sik1. The CT scans and histological staining were used to analyze subchondral bone alterations and cartilage damage. The findings show that Sik1 downregulation correlates with advanced OA and heightened osteogenic differentiation in BMSCs. Sik1 overexpression inhibits osteogenesis-related markers in vitro and reduces cartilage damage and subchondral osteosclerosis in vivo. Mechanistically, Sik1 modulates osteogenesis and subchondral bone changes through Runx2 activity regulation. The research emphasizes Sik1 as a promising target for treating OA, suggesting its involvement in controlling bone formation and changes in the subchondral osteosclerosis.
Collapse
Affiliation(s)
- Kuanmin Tian
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaoxin He
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xue Lin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaolei Chen
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yajing Su
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhidong Lu
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhirong Chen
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Liang Zhang
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Peng Li
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Long Ma
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhibin Lan
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xin Zhao
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Gangning Fen
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qinqin Hai
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Di Xue
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qunhua Jin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| |
Collapse
|
4
|
Zheng CQ, Zeng LJ, Liu ZH, Miao CF, Yao LY, Song HT, Hu XM, Zhou X. Insights into the Roles of Natural Killer Cells in Osteoarthritis. Immunol Invest 2024; 53:766-787. [PMID: 38622991 DOI: 10.1080/08820139.2024.2337025] [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: 04/17/2024]
Abstract
Osteoarthritis (OA) is now widely acknowledged as a low-grade inflammatory condition, in which the intrinsic immune system plays a significant role in its pathogenesis. While the involvement of macrophages and T cells in the development of OA has been extensively reviewed, recent research has provided mounting evidence supporting the crucial contribution of NK cells in both the initiation and advancement of OA. Accumulated evidence has emerged in recent years indicating that NK cells play a critical role in OA development and progression. This review will outline the ongoing understanding of the utility of NK cells in the etiology of OA, focusing on how NK cells interact with chondrocytes, synoviocytes, osteoclasts, and other immune cells to influence the course of OA disease.
Collapse
Affiliation(s)
- Chang-Qing Zheng
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, People's Republic of China
| | - Ling-Jun Zeng
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, People's Republic of China
| | - Zhi-Hong Liu
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, People's Republic of China
| | - Chen-Fang Miao
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, People's Republic of China
| | - Ling-Yan Yao
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, People's Republic of China
| | - Hong-Tao Song
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, People's Republic of China
| | - Xiao-Mu Hu
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, People's Republic of China
| | - Xin Zhou
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, People's Republic of China
| |
Collapse
|
5
|
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
|
6
|
Fan Y, Bian X, Meng X, Li L, Fu L, Zhang Y, Wang L, Zhang Y, Gao D, Guo X, Lammi MJ, Peng G, Sun S. Unveiling inflammatory and prehypertrophic cell populations as key contributors to knee cartilage degeneration in osteoarthritis using multi-omics data integration. Ann Rheum Dis 2024; 83:926-944. [PMID: 38325908 PMCID: PMC11187367 DOI: 10.1136/ard-2023-224420] [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: 05/11/2023] [Accepted: 01/23/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVES Single-cell and spatial transcriptomics analysis of human knee articular cartilage tissue to present a comprehensive transcriptome landscape and osteoarthritis (OA)-critical cell populations. METHODS Single-cell RNA sequencing and spatially resolved transcriptomic technology have been applied to characterise the cellular heterogeneity of human knee articular cartilage which were collected from 8 OA donors, and 3 non-OA control donors, and a total of 19 samples. The novel chondrocyte population and marker genes of interest were validated by immunohistochemistry staining, quantitative real-time PCR, etc. The OA-critical cell populations were validated through integrative analyses of publicly available bulk RNA sequencing data and large-scale genome-wide association studies. RESULTS We identified 33 cell population-specific marker genes that define 11 chondrocyte populations, including 9 known populations and 2 new populations, that is, pre-inflammatory chondrocyte population (preInfC) and inflammatory chondrocyte population (InfC). The novel findings that make this an important addition to the literature include: (1) the novel InfC activates the mediator MIF-CD74; (2) the prehypertrophic chondrocyte (preHTC) and hypertrophic chondrocyte (HTC) are potentially OA-critical cell populations; (3) most OA-associated differentially expressed genes reside in the articular surface and superficial zone; (4) the prefibrocartilage chondrocyte (preFC) population is a major contributor to the stratification of patients with OA, resulting in both an inflammatory-related subtype and a non-inflammatory-related subtype. CONCLUSIONS Our results highlight InfC, preHTC, preFC and HTC as potential cell populations to target for therapy. Also, we conclude that profiling of those cell populations in patients might be used to stratify patient populations for defining cohorts for clinical trials and precision medicine.
Collapse
Affiliation(s)
- Yue Fan
- Center for Single-Cell Omics and Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Shaanxi Province; Key Laboratory of Trace Elements and Endemic Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xuzhao Bian
- Center for Single-Cell Omics and Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xiaogao Meng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Lei Li
- Center for Single-Cell Omics and Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Laiyi Fu
- School of Automation Science and Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yanan Zhang
- Center for Single-Cell Omics and Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Long Wang
- Center for Single-Cell Omics and Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yan Zhang
- Center for Single-Cell Omics and Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Department of Orthopaedics, Honghui Hospital, Xi'an, Shaanxi, China
| | - Dalong Gao
- Department of Orthopaedics, The Central Hospital of Xianyang, Xianyang, China
| | - Xiong Guo
- Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Shaanxi Province; Key Laboratory of Trace Elements and Endemic Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mikko Juhani Lammi
- Department of Integrative Medical Biology, University of Umeå, Umeå, Sweden
| | - Guangdun Peng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shiquan Sun
- Center for Single-Cell Omics and Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Shaanxi Province; Key Laboratory of Trace Elements and Endemic Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
7
|
Nguyen M, Battistoni CM, Babiak PM, Liu JC, Panitch A. Chondroitin Sulfate/Hyaluronic Acid-Blended Hydrogels Suppress Chondrocyte Inflammation under Pro-Inflammatory Conditions. ACS Biomater Sci Eng 2024; 10:3242-3254. [PMID: 38632852 PMCID: PMC11094685 DOI: 10.1021/acsbiomaterials.4c00200] [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: 01/29/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
Osteoarthritis is characterized by enzymatic breakdown of the articular cartilage via the disruption of chondrocyte homeostasis, ultimately resulting in the destruction of the articular surface. Decades of research have highlighted the importance of inflammation in osteoarthritis progression, with inflammatory cytokines shifting resident chondrocytes into a pro-catabolic state. Inflammation can result in poor outcomes for cells implanted for cartilage regeneration. Therefore, a method to promote the growth of new cartilage and protect the implanted cells from the pro-inflammatory cytokines found in the joint space is required. In this study, we fabricate two gel types: polymer network hydrogels composed of chondroitin sulfate and hyaluronic acid, glycosaminoglycans (GAGs) known for their anti-inflammatory and prochondrogenic activity, and interpenetrating networks of GAGs and collagen I. Compared to a collagen-only hydrogel, which does not provide an anti-inflammatory stimulus, chondrocytes in GAG hydrogels result in reduced production of pro-inflammatory cytokines and enzymes as well as preservation of collagen II and aggrecan expression. Overall, GAG-based hydrogels have the potential to promote cartilage regeneration under pro-inflammatory conditions. Further, the data have implications for the use of GAGs to generally support tissue engineering in pro-inflammatory environments.
Collapse
Affiliation(s)
- Michael Nguyen
- Department
of Biomedical Engineering, University of
California, Davis, California 95616, United States
| | - Carly M. Battistoni
- Davidson
School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Paulina M. Babiak
- Davidson
School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Julie C. Liu
- Davidson
School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon
School of Biomedical Engineering, Purdue
University, West Lafayette, Indiana 47907, United States
| | - Alyssa Panitch
- Department
of Biomedical Engineering, University of
California, Davis, California 95616, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| |
Collapse
|
8
|
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
|
9
|
Zhou J, Ning E, Lu L, Zhang H, Yang X, Hao Y. Effectiveness of low-intensity pulsed ultrasound on osteoarthritis: molecular mechanism and tissue engineering. Front Med (Lausanne) 2024; 11:1292473. [PMID: 38695024 PMCID: PMC11061361 DOI: 10.3389/fmed.2024.1292473] [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: 12/19/2023] [Accepted: 04/05/2024] [Indexed: 05/04/2024] Open
Abstract
Osteoarthritis (OA) is distinguished by pathological alterations in the synovial membrane, articular cartilage, and subchondral bone, resulting in physical symptoms such as pain, deformity, and impaired mobility. Numerous research studies have validated the effectiveness of low-intensity pulsed ultrasound (LIPUS) in OA treatment. The periodic mechanical waves generated by LIPUS can mitigate cellular ischemia and hypoxia, induce vibration and collision, produce notable thermal and non-thermal effects, alter cellular metabolism, expedite tissue repair, improve nutrient delivery, and accelerate the healing process of damaged tissues. The efficacy and specific mechanism of LIPUS is currently under investigation. This review provides an overview of LIPUS's potential role in the treatment of OA, considering various perspectives such as the synovial membrane, cartilage, subchondral bone, and tissue engineering. It aims to facilitate interdisciplinary scientific research and further exploration of LIPUS as a complementary technique to existing methods or surgery. Ongoing research is focused on determining the optimal dosage, frequency, timing, and treatment strategy of LIPUS for OA. Additional research is required to clarify the precise mechanism of action and potential impacts on cellular, animal, and human systems prior to its integration into therapeutic applications.
Collapse
Affiliation(s)
- Jing Zhou
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Eryu Ning
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Huili Zhang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
10
|
Henning P, Kassem A, Westerlund A, Lundberg P, Engdahl C, Lionikaite V, Wikström P, Wu J, Li L, Lindholm C, de Souza PPC, Movérare-Skrtic S, Lerner UH. Toll-like receptor-2 induced inflammation causes local bone formation and activates canonical Wnt signaling. Front Immunol 2024; 15:1383113. [PMID: 38646530 PMCID: PMC11026618 DOI: 10.3389/fimmu.2024.1383113] [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: 02/06/2024] [Accepted: 03/11/2024] [Indexed: 04/23/2024] Open
Abstract
It is well established that inflammatory processes in the vicinity of bone often induce osteoclast formation and bone resorption. Effects of inflammatory processes on bone formation are less studied. Therefore, we investigated the effect of locally induced inflammation on bone formation. Toll-like receptor (TLR) 2 agonists LPS from Porphyromonas gingivalis and PAM2 were injected once subcutaneously above mouse calvarial bones. After five days, both agonists induced bone formation mainly at endocranial surfaces. The injection resulted in progressively increased calvarial thickness during 21 days. Excessive new bone formation was mainly observed separated from bone resorption cavities. Anti-RANKL did not affect the increase of bone formation. Inflammation caused increased bone formation rate due to increased mineralizing surfaces as assessed by dynamic histomorphometry. In areas close to new bone formation, an abundance of proliferating cells was observed as well as cells robustly stained for Runx2 and alkaline phosphatase. PAM2 increased the mRNA expression of Lrp5, Lrp6 and Wnt7b, and decreased the expression of Sost and Dkk1. In situ hybridization demonstrated decreased Sost mRNA expression in osteocytes present in old bone. An abundance of cells expressed Wnt7b in Runx2-positive osteoblasts and ß-catenin in areas with new bone formation. These data demonstrate that inflammation, not only induces osteoclastogenesis, but also locally activates canonical WNT signaling and stimulates new bone formation independent on bone resorption.
Collapse
Affiliation(s)
- Petra Henning
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ali Kassem
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Anna Westerlund
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Lundberg
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Cecilia Engdahl
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Vikte Lionikaite
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Wikström
- Department of Medical Biosciences, Section of Pathology, Umeå University, Umeå, Sweden
| | - Jianyao Wu
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Lei Li
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Catharina Lindholm
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pedro P. C. de Souza
- Innovation in Biomaterials Laboratory, Federal University of Goiás, Goiania, Brazil
| | - Sofia Movérare-Skrtic
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ulf H. Lerner
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| |
Collapse
|
11
|
Li W, He H, Du M, Gao M, Sun Q, Wang Y, Lu H, Ou S, Xia C, Xu C, Zhao Q, Sun H. Quercetin as a promising intervention for rat osteoarthritis by decreasing M1-polarized macrophages via blocking the TRPV1-mediated P2X7/NLRP3 signaling pathway. Phytother Res 2024; 38:1990-2006. [PMID: 38372204 DOI: 10.1002/ptr.8158] [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: 09/19/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/20/2024]
Abstract
Osteoarthritis (OA) is characterized by an imbalance between M1 and M2 polarized synovial macrophages. Quercetin has shown protective effects against OA by altering M1/M2-polarized macrophages, but the underlying mechanisms remain unclear. In this study, rat chondrocytes were treated with 10 ng/mL of IL-1β. To create M1-polarized macrophages in vitro, rat bone marrow-derived macrophages (rBMDMs) were treated with 100 ng/mL LPS. To mimic OA conditions observed in vivo, a co-culture system of chondrocytes and macrophages was established. ATP release assays, immunofluorescence assays, Fluo-4 AM staining, Transwell assays, ELISA assays, and flow cytometry were performed. Male adult Sprague-Dawley (SD) rats were used to create an OA model. Histological analyses, including H&E, and safranin O-fast green staining were performed. Our data showed a quercetin-mediated suppression of calcium ion influx and ATP release, with concurrent downregulation of TRPV1 and P2X7 in the chondrocytes treated with IL-1β. Activation of TRPV1 abolished the quercetin-mediated effects on calcium ion influx and ATP release in chondrocytes treated with IL-1β. In the co-culture system, overexpression of P2X7 in macrophages attenuated the quercetin-mediated effects on M1 polarization, migration, and inflammation. Either P2X7 or NLRP3 knockdown attenuated IL-1β-induced M1/M2 polarization, migration, and inflammation. Moreover, overexpression of TRPV1 reduced the quercetin-mediated suppressive effects on OA by promoting M1/M2-polarized macrophages in vivo. Collectively, our data showed that quercetin-induced suppression of TRPV1 leads to a delay in OA progression by shifting the macrophage polarization from M1 to M2 subtypes via modulation of the P2X7/NLRP3 pathway.
Collapse
Affiliation(s)
- Wenjun Li
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Hebei He
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Min Du
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Mu Gao
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Qijie Sun
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Yeyang Wang
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Hanyu Lu
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Shuanji Ou
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Changliang Xia
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Changpeng Xu
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Hongtao Sun
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
12
|
Lee KT, Lin CY, Liu SC, He XY, Tsai CH, Ko CY, Tsai YH, Chao CC, Chen PC, Tang CH. IL-17 promotes IL-18 production via the MEK/ERK/miR-4492 axis in osteoarthritis synovial fibroblasts. Aging (Albany NY) 2024; 16:1829-1844. [PMID: 38261743 PMCID: PMC10866453 DOI: 10.18632/aging.205462] [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: 08/19/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024]
Abstract
The concept of osteoarthritis (OA) as a low-grade inflammatory joint disorder has been widely accepted. Many inflammatory mediators are implicated in the pathogenesis of OA. Interleukin (IL)-18 is a pleiotropic cytokine with versatile cellular functions that are pathogenetically important in immune responses, as well as autoimmune, inflammatory, and infectious diseases. IL-17, a proinflammatory cytokine mainly secreted by Th17 cells, is upregulated in OA patients. However, the role of IL-17 in OA progression is unclear. The synovial tissues collected from healthy donors and OA patients were used to detect the expression level of IL-18 by IHC stain. The OA synovial fibroblasts (OASFs) were incubated with recombinant IL-17 and subjected to Western blot, qPCR, and ELISA to examine IL-18 expression level. The chemical inhibitors and siRNAs which targeted signal pathways were used to investigate signal pathways involved in IL-17-induced IL-18 expression. The microRNAs which participated IL-18 expression were surveyed with online databases miRWalk and miRDB, followed by validation with qPCR. This study revealed significantly higher levels of IL-18 expression in synovial tissue from OA patients compared with healthy controls, as well as increased IL-18 expression in OASFs from rats with severe OA. In vitro findings indicated that IL-17 dose-dependently promoted IL-18 production in OASFs. Molecular investigations revealed that the MEK/ERK/miR-4492 axis stimulated IL-18 production when OASFs were treated with IL-17. This study provides novel insights into the role of IL-17 in the pathogenesis of OA, which may help to inform OA treatment in the future.
Collapse
Affiliation(s)
- Kun-Tsan Lee
- Department of Post-Baccalaureate Medicine, National Chung-Hsing University, Taichung, Taiwan
- Department of Orthopedics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Yang Lin
- Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan
| | - Shan-Chi Liu
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Xiu-Yuan He
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Chih-Yuan Ko
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Yuan-Hsin Tsai
- Department of Orthopedics, Show-Chwan Memorial Hospital, Changhua, Taiwan
| | - Chia-Chia Chao
- Department of Respiratory Therapy, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Po-Chun Chen
- Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, Taiwan
| |
Collapse
|
13
|
Xu K, Li J, Wen R, Chang B, Cheng Y, Yi X. Role of SIRT3 in bone homeostasis and its application in preventing and treating bone diseases. Front Pharmacol 2023; 14:1248507. [PMID: 38192409 PMCID: PMC10773770 DOI: 10.3389/fphar.2023.1248507] [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: 06/27/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024] Open
Abstract
Bone homeostasis refers to the balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption and the maintenance of stable bone mass. SIRT3 is a class of mitochondrial protein deacetylase that influences various mitochondrial functions and is involved in the mechanisms underlying resistance to aging; regulation of bone marrow mesenchymal stem cells, osteoblasts, and osteoclasts; and development of osteoporosis, osteoarthritis, and other bone diseases. Moreover, exercise affects bones through SIRT3. Thus, studies on SIRT3 may provide insights for the treatment of bone diseases. Although SIRT3 can exert multiple effects on bone, the specific mechanism by which it regulates bone homeostasis remains unclear. By evaluating the relevant literature, this review discusses the structure and function of SIRT3, reveals the role and associated mechanisms of SIRT3 in regulating bone homeostasis and mediating bone health during exercise, and highlights the potential pharmacological value of SIRT3 in treating bone diseases.
Collapse
Affiliation(s)
- Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, China
| | - Jing Li
- School of Physical Education, Liaoning Normal University, Dalian, China
| | - Ruiming Wen
- School of Sports Health, Shenyang Sport University, Shenyang, China
| | - Bo Chang
- School of Sports Health, Shenyang Sport University, Shenyang, China
| | - Yang Cheng
- School of Sports Health, Shenyang Sport University, Shenyang, China
| | - Xuejie Yi
- School of Sports Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
14
|
Fainor M, Orozco BS, Muir VG, Mahindroo S, Gupta S, Mauck RL, Burdick JA, Smith HE, Gullbrand SE. Mechanical crosstalk between the intervertebral disc, facet joints, and vertebral endplate following acute disc injury in a rabbit model. JOR Spine 2023; 6:e1287. [PMID: 38156057 PMCID: PMC10751980 DOI: 10.1002/jsp2.1287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/21/2023] [Accepted: 09/06/2023] [Indexed: 12/30/2023] Open
Abstract
Background Vertebral endplate sclerosis and facet osteoarthritis have been documented in animals and humans. However, it is unclear how these adjacent pathologies engage in crosstalk with the intervertebral disc. This study sought to elucidate this crosstalk by assessing each compartment individually in response to acute disc injury. Methods Eleven New Zealand White rabbits underwent annular disc puncture using a 16G or 21G needle. At 4 and 10 weeks, individual compartments of the motion segment were analyzed. Discs underwent T 1 relaxation mapping with MRI contrast agent gadodiamide as well T 2 mapping. Both discs and facets underwent mechanical testing via vertebra-disc-vertebra tension-compression creep testing and indentation testing, respectively. Endplate bone density was quantified via μCT. Discs and facets were sectioned and stained for histology scoring. Results Intervertebral discs became more degenerative with increasing needle diameter and time post-puncture. Bone density also increased in endplates adjacent to both 21G and 16G punctured discs leading to reduced gadodiamide transport at 10 weeks. The facet joints, however, did not follow this same trend. Facets adjacent to 16G punctured discs were less degenerative than facets adjacent to 21G punctured discs at 10 weeks. 16G facets were more degenerative at 4 weeks than at 10, suggesting the cartilage had recovered. The formation of severe disc osteophytes in 16G punctured discs between 4 and 10 weeks likely offloaded the facet cartilage, leading to the recovery observed. Conclusions Overall, this study supports that degeneration spans the whole spinal motion segment following disc injury. Vertebral endplate thickening occurred in response to disc injury, which limited the diffusion of small molecules into the disc. This work also suggests that altered disc mechanics can induce facet degeneration, and that extreme bony remodeling adjacent to the disc may promote facet cartilage recovery through offloading of the articular cartilage.
Collapse
Affiliation(s)
- Matthew Fainor
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Brianna S. Orozco
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Victoria G. Muir
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sonal Mahindroo
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
- Department of BiologySt. Bonaventure UniversitySt. BonaventureNew YorkUSA
| | - Sachin Gupta
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Robert L. Mauck
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jason A. Burdick
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- BioFrontiers Institute and Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
| | - Harvey E. Smith
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Sarah E. Gullbrand
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
15
|
Najar M, Alsabri SG, Guedi GG, Merimi M, Lavoie F, Grabs D, Pelletier JP, Martel-Pelletier J, Benderdour M, Fahmi H. Role of epigenetics and the transcription factor Sp1 in the expression of the D prostanoid receptor 1 in human cartilage. Front Cell Dev Biol 2023; 11:1256998. [PMID: 38099292 PMCID: PMC10720455 DOI: 10.3389/fcell.2023.1256998] [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: 07/11/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023] Open
Abstract
D prostanoid receptor 1 (DP1), a prostaglandin D2 receptor, plays a central role in the modulation of inflammation and cartilage metabolism. We have previously shown that activation of DP1 signaling downregulated catabolic responses in cultured chondrocytes and was protective in mouse osteoarthritis (OA). However, the mechanisms underlying its transcriptional regulation in cartilage remained poorly understood. In the present study, we aimed to characterize the human DP1 promoter and the role of DNA methylation in DP1 expression in chondrocytes. In addition, we analyzed the expression level and methylation status of the DP1 gene promoter in normal and OA cartilage. Deletion and site-directed mutagenesis analyses identified a minimal promoter region (-250/-120) containing three binding sites for specificity protein 1 (Sp1). Binding of Sp1 to the DP1 promoter was confirmed using electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assays. Treatment with the Sp1 inhibitor mithramycin A reduced DP1 promoter activity and DP1 mRNA expression. Inhibition of DNA methylation by 5-Aza-2'-deoxycytidine upregulated DP1 expression, and in vitro methylation reduced the DP1 promoter activity. Neither the methylation status of the DP1 promoter nor the DP1 expression level were different between normal and OA cartilage. In conclusion, our results suggest that the transcription factor Sp1 and DNA methylation are important determinants of DP1 transcription regulation. They also suggest that the methylation status and expression level of DP1 are not altered in OA cartilage. These findings will improve our understanding of the regulatory mechanisms of DP1 transcription and may facilitate the development of intervention strategies involving DP1.
Collapse
Affiliation(s)
- Mehdi Najar
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Sami G. Alsabri
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Gadid G. Guedi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Makram Merimi
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Frédéric Lavoie
- Departement of Orthopedic Surgery, University of Montreal Hospital Center (CHUM), Montréal, QC, Canada
| | - Detlev Grabs
- Research Unit in Clinical and Functional Anatomy, Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Mohamed Benderdour
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, QC, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
16
|
Li J, Peng S, Yan Y, Yan S, Cao X, Li Y, Zhu L, Xu J. IL-37 counteracts inflammatory injury in the temporomandibular joint via the intracellular pathway. Front Pharmacol 2023; 14:1250216. [PMID: 38053836 PMCID: PMC10694265 DOI: 10.3389/fphar.2023.1250216] [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: 06/29/2023] [Accepted: 11/02/2023] [Indexed: 12/07/2023] Open
Abstract
Background: The temporomandibular joint is often afflicted by osteoarthritis (TMJOA), causing pain and dysfunction, which is particularly prevalent in the elderly population. IL-37 is effective in avoiding excessive inflammatory damage to the organism. This article investigates the role and mechanism of intracellular IL-37 in TMJOA. Methods: Enzyme-linked immunosorbent assay, quantitative real-time polymerase chain reaction, Western blotting, Senescence-associated β-galactosidase staining, immunofluorescence, and lentivirus were performed to elucidate the underlying mechanism. Results: The results confirmed that IL-37 in synovial cells decreased with aging. Inflammatory stimulus elevated intracellular IL-37 in synoviocytes, while lentiviral knockdown of IL-37 resulted in more inflammatory factor production. Dynamic changes of IL-37 were observed in the nucleus and supernatant. In addition, Caspease-1 inhibitor hindered intracellular IL-37 maturation, and Smad3 inhibitor caused the loss of nuclear translocation of mature IL-37. Transfection of synovial cells with IL-37-expressing lentivirus resulted in relief not only of synovitis but also of the cartilage damage and inflammation caused by synovitis. Conclusion: This study provides new insights into the intracellular anti-inflammatory mechanism of IL-37. It also confirms that IL-37 decreases with cellular senescence and that increasing intracellular IL-37 can effectively treat synovitis and synovitis-induced inflammatory damage to cartilage.
Collapse
Affiliation(s)
- Jun Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- Chongqing Emergency Medical Center, Chongqing, China
- Chongqing University Central Hospital, Chongqing, China
| | - Sisi Peng
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ying Yan
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Shan Yan
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xin Cao
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yong Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Luying Zhu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
17
|
Sugimura N, Aso K, Wada H, Izumi M, Ikeuchi M. Association Between Power Doppler Ultrasound Signals and Chronic Pain After Total Knee Arthroplasty: A Cross-Sectional Explorative Study. J Pain Res 2023; 16:2981-2992. [PMID: 37664487 PMCID: PMC10474857 DOI: 10.2147/jpr.s403641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
Purpose Some patients experience chronic postsurgical pain (CPSP) after total knee arthroplasty (TKA) despite the absence of clinical or radiographic abnormalities. Postoperative synovitis as a cause of CPSP after TKA has received limited research attention. This study aimed to investigate the relationship between synovitis after TKA and CPSP. Patients and Methods A total of 111 knees of 85 patients, with at least 1-year post-TKA follow-up, were assessed retrospectively and cross-sectionally. Power Doppler (PD) ultrasonography was used to detect the synovial hypervascularity associated with synovitis. The knee joint was divided into 15 areas, and PD signals were graded semi-quantitatively (0-3) in each area, the sum of which was defined as the total PD score. Clinical information regarding CPSP, including the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain subscales, was recorded. The relationship between pain and PD ultrasonography findings was accessed. Patients were divided into two groups (CPSP+ and CPSP- groups) based on pain severity. Clinical information, including PD ultrasonography findings and other possible causes, was compared between the groups. Results The WOMAC pain subscale was significantly correlated with the total PD score and maximum PD grade (r=0.3977, p<0.0001; r=0.2797, p=0.0029; respectively). The CPSP+ group had a significantly higher maximum PD grade and total PD score than the CPSP- group (median [interquartile range]: 2 [1, 2] vs 1 [1, 2], p=0.0001; 6 [2, 11] vs 2 [1, 4], p=0.0002; respectively). Multiple and logistic regression analyses showed that the total PD score was an independent factor for the WOMAC pain subscale (β=0.3822, 95% confidence interval [CI]=0.1460, 0.6184, p=0.00176) and CPSP (odds ratio=1.19, 95% CI=1.01, 1.41, p=0.0424). Conclusion This study indicated a possible association between the total PD score and chronic pain after TKA; however, further studies are needed to corroborate these findings.
Collapse
Affiliation(s)
- Natsuki Sugimura
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
- Department of Orthopedic Surgery, Hata Prefectural Hospital, Sukumo, Kochi, 788-0785, Japan
| | - Koji Aso
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Hiroyuki Wada
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Masashi Izumi
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Masahiko Ikeuchi
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
18
|
Chu ZC, Cong T, Zhao JY, Zhang J, Lou ZY, Gao Y, Tang X. The identification of hub-methylated differentially expressed genes in osteoarthritis patients is based on epigenomic and transcriptomic data. Front Med (Lausanne) 2023; 10:1219830. [PMID: 37465641 PMCID: PMC10351907 DOI: 10.3389/fmed.2023.1219830] [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: 05/09/2023] [Accepted: 06/16/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction Osteoarthritis (OA) refers to a commonly seen degenerative joint disorder and a major global public health burden. According to the existing literature, osteoarthritis is related to epigenetic changes, which are important for diagnosing and treating the disease early. Through early targeted treatment, costly treatments and poor prognosis caused by advanced osteoarthritis can be avoided. Methods This study combined gene differential expression analysis and weighted gene co-expression network analysis (WGCNA) of the transcriptome with epigenome microarray data to discover the hub gene of OA. We obtained 2 microarray datasets (GSE114007, GSE73626) in Gene Expression Omnibus (GEO). The R software was utilized for identifying differentially expressed genes (DEGs) and differentially methylated genes (DMGs). By using WGCNA to analyze the relationships between modules and phenotypes, it was discovered that the blue module (MEBlue) has the strongest phenotypic connection with OA (cor = 0.92, p = 4e-16). The hub genes for OA, also known as the hub methylated differentially expressed genes, were identified by matching the MEblue module to differentially methylated differentially expressed genes. Furthermore, this study used Gene set variation analysis (GSVA) to identify specific signal pathways associated with hub genes. qRT-PCR and western blotting assays were used to confirm the expression levels of the hub genes in OA patients and healthy controls. Results Three hub genes were discovered: HTRA1, P2RY6, and RCAN1. GSVA analysis showed that high HTRA1 expression was mainly enriched in epithelial-mesenchymal transition and apical junction; high expression of P2RY6 was mainly enriched in the peroxisome, coagulation, and epithelial-mesenchymal transition; and high expression of RCAN1 was mainly enriched in epithelial-mesenchymal-transition, TGF-β-signaling, and glycolysis. The results of the RT-qPCR and WB assay were consistent with the findings. Discussion The three genes tested may cause articular cartilage degeneration by inducing chondrocyte hypertrophy, regulating extracellular matrix accumulation, and improving macrophage pro-inflammatory response, resulting in the onset and progression of osteoarthritis. They can provide new ideas for targeted treatment of osteoarthritis.
Collapse
Affiliation(s)
- Zhen-Chen Chu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Dalian Medical University, Dalian, Liaoning, China
| | - Ting Cong
- Dalian Medical University, Dalian, Liaoning, China
- Department of Anesthesiology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jian-Yu Zhao
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jian Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Dalian Medical University, Dalian, Liaoning, China
| | - Zhi-Yuan Lou
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yang Gao
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xin Tang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
19
|
Huang H, Dong X, Mao K, Pan W, Nie B, Jiang L. Identification of key candidate genes and pathways in rheumatoid arthritis and osteoarthritis by integrated bioinformatical analysis. Front Genet 2023; 14:1083615. [PMID: 36861127 PMCID: PMC9968929 DOI: 10.3389/fgene.2023.1083615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are the most common joint disorders. Although they have shown analogous clinical manifestations, the pathogenesis of RA and OA are different. In this study, we used the online Gene Expression Omnibus (GEO) microarray expression profiling dataset GSE153015 to identify gene signatures between RA and OA joints. The relevant data on 8 subjects obtained from large joints of RA patients (RA-LJ), 8 subjects obtained from small joints of RA patients (RA-SJ), and 4 subjects with OA were investigated. Differentially expressed genes (DEGs) were screened. Functional enrichment analysis of DEGs including the Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were identified, which were mainly associated with T cell activation or chemokine activity. Besides, protein-protein interaction (PPI) network analysis was performed, and key modules were identified. Hub genes of RA-LJ and OA groups were screened, they were CD8A, GZMB, CCL5, CD2, and CXCL9, whereas CD8A, CD2, IL7R, CD27, and GZMB were hub genes of RA-SJ and OA group. The novel DEGs and functional pathways between RA and OA identified in this study may provide new insight into the underlying molecular mechanisms and therapeutic strategies of RA and OA.
Collapse
Affiliation(s)
- Huijing Huang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyi Dong
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kaimin Mao
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wanwan Pan
- Yankuang New Journey General Hospital, Jingning, Shandong, China
| | - Bin’en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lindi Jiang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China,*Correspondence: Lindi Jiang,
| |
Collapse
|
20
|
Wen P, Liu R, Wang J, Wang Y, Song W, Zhang Y. Bibliometric insights from publications on subchondral bone research in osteoarthritis. Front Physiol 2022; 13:1095868. [PMID: 36620224 PMCID: PMC9814489 DOI: 10.3389/fphys.2022.1095868] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Background: The role of subchondral bone in the pathogenesis of osteoarthritis has received continuous attention worldwide. To date, no comprehensive bibliometric analysis of this topic has been carried out. The purpose of this study was to investigate the knowledge landscape, hot spots, and research trends in subchondral bone research through bibliometrics. Methods: Web of Science Core Collection database was used to collect articles and reviews on subchondral bone in osteoarthritis published between 2003 and 2022. CiteSpace, VOSviewer, Scimago Graphica, and a bibliometric online analysis platform (http://bibliometric.com/) were used to visualize the knowledge network of countries, institutions, authors, references, and keywords in this field. Both curve fitting and statistical plotting were performed using OriginPro, while correlation analysis was done using SPSS. Results: A total of 3,545 articles and reviews were included. The number of publications on subchondral bone showed an exponential growth trend. The US produced the most (980), followed by China (862) and the United Kingdom (364). Scientific output and gross domestic product were significantly correlated (r = .948, p < .001). The University of California System and Professor Pelletier Jean-Pierre were the most prolific institutions and influential authors, respectively. The most active and influential journal for subchondral bone research was Osteoarthritis and Cartilage. The majority of papers were financed by NSFC (474, 13.4%), followed by HHS (445, 12.6%), and NIH (438, 12.4%). In recent years, hot keywords have focused on the research of pathomechanisms (e.g., inflammation, apoptosis, pathogenesis, cartilage degeneration/repair, angiogenesis, TGF beta) and therapeutics (e.g., regeneration, stromal cell, mesenchymal stem cell). Conclusion: Subchondral bone research in osteoarthritis is flourishing. Current topics and next research trends would be centered on the pathomechanisms of cellular and molecular interactions in the subchondral bone microenvironment in the development of osteoarthritis and the exploration of targeted treatment medicines for the altered subchondral bone microenvironment.
Collapse
Affiliation(s)
| | | | | | | | - Wei Song
- *Correspondence: Wei Song, ; Yumin Zhang,
| | | |
Collapse
|
21
|
Liang X, Li SR, Zhang XX, He SH, Li SS, Li TF. Teriparatide prevented synovial inflammation and cartilage destruction in mice with DMM. Connect Tissue Res 2022; 64:274-284. [PMID: 36537662 DOI: 10.1080/03008207.2022.2157723] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIM Emerging data have demonstrated that low-grade inflammation in osteoarthritis, a long-held degenerative disease. The inflamed synovium produces various cytokines that induce cartilage destruction and joint pain. A previous study showed that teriparatide, an FDA approved anti-osteoporotic drug, may enhance cartilage repair. Our study focuses on its role in OA synovitis. MATERIALS AND METHODS Primary mouse articular chondrocytes were used to determine the most potent cytokines involved in OA inflammation and cartilage destruction. A destabilization of the medial meniscus mouse model was established to investigate the effect of teriparatide in OA, particularly, on synovial inflammation and cartilage degradation. RESULTS In vitro experiments showed that TNF-α was the most potent inducer of cartilage matrix-degrading enzymes, and that teriparatide antagonized the TNF-α of effect. Consistently, articular cartilage samples from TNF-α transgenic mice contained more MMP-13 positive chondrocytes than those from wild type mice. In addition, more type II collagen was cleaved in human OA cartilage than in normal cartilage samples. CONCLUSIONS Teriparatide can prevent synovitis and cartilage degradation by suppressing TNF-α mediated MMP-13 overexpression. Together with its chondroregenerative capability, teriparatide may be the first effective disease modifying osteoarthritis drug.
Collapse
Affiliation(s)
| | | | - Xin-Xin Zhang
- Department of Rheumatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Shi-Hao He
- Department of Rheumatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Shan-Shan Li
- Department of Rheumatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Tian-Fang Li
- Department of Rheumatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
22
|
Feng SY, Lei J, Li YX, Shi WG, Wang RR, Yap AU, Wang YX, Fu KY. Increased joint loading induces subchondral bone loss of the temporomandibular joint via the RANTES-CCRs-Akt2 axis. JCI Insight 2022; 7:158874. [PMID: 36173680 PMCID: PMC9675482 DOI: 10.1172/jci.insight.158874] [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: 03/15/2022] [Accepted: 09/21/2022] [Indexed: 12/15/2022] Open
Abstract
Early-stage temporomandibular joint osteoarthritis (TMJOA) is characterized by excessive subchondral bone loss. Emerging evidence suggests that TMJ disc displacement is involved, but the pathogenic mechanism remains unclear. Here, we established a rat model of TMJOA that simulated disc displacement with a capacitance-based force-sensing system to directly measure articular surface pressure in vivo. Micro-CT, histological staining, immunofluorescence staining, IHC staining, and Western blot were used to assess pathological changes and underlying mechanisms of TMJOA in the rat model in vivo as well as in RAW264.7 cells in vitro. We found that disc displacement led to significantly higher pressure on the articular surface, which caused rapid subchondral bone loss via activation of the RANTES-chemokine receptors-Akt2 (RANTES-CCRs-Akt2) axis. Inhibition of RANTES or Akt2 attenuated subchondral bone loss and resulted in improved subchondral bone microstructure. Cytological studies substantiated that RANTES regulated osteoclast formation by binding to its receptor CCRs and activating the Akt2 pathway. The clinical evidence further supported that RANTES was a potential biomarker for predicting subchondral bone loss in early-stage TMJOA. Taken together, this study demonstrates important functions of the RANTES-CCRs-Akt2 axis in the regulation of subchondral bone remodeling and provides further knowledge of how disc displacement causes TMJOA.
Collapse
Affiliation(s)
- Shi-Yang Feng
- Center for Temporomandibular Disorders & Orofacial Pain, and,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China.,National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Jie Lei
- Center for Temporomandibular Disorders & Orofacial Pain, and,National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yu-Xiang Li
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Wen-Ge Shi
- Center for Temporomandibular Disorders & Orofacial Pain, and,National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Ran-Ran Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.,School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Adrian Ujin Yap
- Center for Temporomandibular Disorders & Orofacial Pain, and,Department of Dentistry, Ng Teng Fong General Hospital and Faculty of Dentistry, National University Health System, Singapore, Singapore.,National Dental Research Institute Singapore, National Dental Centre Singapore and Duke-NUS Medical School, Singapore Health Services, Singapore, Singapore
| | - Yi-Xiang Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China.,National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Kai-Yuan Fu
- Center for Temporomandibular Disorders & Orofacial Pain, and,National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
23
|
Analysis of the Anti-Inflammatory and Anti-Osteoarthritic Potential of Flonat Fast®, a Combination of Harpagophytum Procumbens DC. ex Meisn., Boswellia Serrata Roxb., Curcuma longa L., Bromelain and Escin (Aesculus hippocastanum), Evaluated in In Vitro Models of Inflammation Relevant to Osteoarthritis. Pharmaceuticals (Basel) 2022; 15:ph15101263. [PMID: 36297375 PMCID: PMC9609228 DOI: 10.3390/ph15101263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a joint disease characterized by inflammation of the synovium, angiogenesis, cartilage degradation, and osteophyte formation. Harpagophytum Procumbens DC. ex Meisn., Boswellia Serrata Roxb., Curcuma longa L., Bromelain and Escin (Aesculus hippocastanum) are plants which extracts, together to Bromelain and Escin (Aesculus hippocastanum) are traditionally used in OA. However, their mechanistic role remains unclear. We aimed to investigate whether these bioactives alone or in combination (as in Flonat Fast®) can suppress TNF-α-induced inflammation, angiogenesis, and osteophyte formation using two cell models involved in OA: endothelial cells and monocytes. Each plant extract was evaluated for its polyphenol content, antioxidant activity, and toxicity. In endothelial cells and monocytes, expression of genes involved in OA was assessed, functional assays for inflammation and angiogenesis were performed, and impairment of reactive oxygen species production (ROS) was evaluated. Exposure of cells to the bioactives alone and in combination before cytokine stimulation resulted in differential counterregulation of several gene and protein expressions, including those for cyclooxygenases-2, metalloproteinase-9, transforming growth factor β1, and bone morphogenic protein-2. We demonstrated that these bioactives modulated monocyte adhesion to endothelial cells as well as cell migration and endothelial angiogenesis. Consistent with radical scavenging activity in the cell-free system, the bioactives curbed TNF-α-stimulated intracellular ROS production. We confirmed the potential anti-inflammatory and antiangiogenic effects of the combination of Harpagophytum procumbens, Boswellia, Curcuma, Bromelain, and Escin and provided new mechanistic evidence for their use in OA. However, further clinical studies are needed to evaluate the true clinical utility of these bioactives as supportive, preventive, and therapeutic agents.
Collapse
|
24
|
Yang Z, Lin J, Li H, He Z, Wang K, Lei L, Li H, Xing D, Lin J. Bibliometric and visualization analysis of macrophages associated with osteoarthritis from 1991 to 2021. Front Immunol 2022; 13:1013498. [PMID: 36268031 PMCID: PMC9577295 DOI: 10.3389/fimmu.2022.1013498] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundMacrophages significantly contributes to symptomology and structural progression of osteoarthritis (OA) and raise increasing attention in the relative research field. Recent studies have shown that tremendous progress has been made in the research of macrophages associated with osteoarthritis. However, a comprehensive bibliometric analysis is lacking in this research field. This study aimed to introduce the research status as well as hotspots and explore the field of macrophages research in OA from a bibliometric perspective.MethodsThis study collected 1481 records of macrophages associated with osteoarthritis from 1991 to 2021 in the web of science core collection (WoSCC) database. CiteSpace, VOSviewer, and R package “bibliometrix” software were used to analyze regions, institutions, journals, authors, and keywords to predict the latest trends in macrophages associated with osteoarthritis research.ResultsThe number of publications related to macrophages associated with osteoarthritis is increasing annually. China and the USA, contributing more than 44% of publications, were the main drivers for research in this field. League of European Research Universities was the most active institution and contributed the most publications. Arthritis and Rheumatism is the most popular journal in this field with the largest publications, while Osteoarthritis and Cartilage is the most co-cited journal. Koch AE was the most prolific writer, while Bondeson J was the most commonly co-cited author. “Rheumatology”, “Orthopedics”, and “Immunology” were the most widely well-represented research areas of OA associated macrophages. “Rheumatoid arthritis research”, “clinical symptoms”, “regeneration research”, “mechanism research”, “pathological features”, and “surgery research” are the primary keywords clusters in this field.ConclusionThis is the first bibliometric study comprehensively mapped out the knowledge structure and development trends in the research field of macrophages associated with osteoarthritis in recent 30 years. The results comprehensively summarize and identify the research frontiers which will provide a reference for scholars studying macrophages associated with osteoarthritis.
Collapse
Affiliation(s)
- Zhen Yang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Li
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| | - Zihao He
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| | - Kai Wang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| | - Liandi Lei
- Center of Medical and Health Analysis, Peking University, Beijing, China
| | - Hao Li
- School of Medicine, Nankai University, Tianjin, China
- *Correspondence: Hao Li, ; Dan Xing, ; Jianhao Lin,
| | - Dan Xing
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
- *Correspondence: Hao Li, ; Dan Xing, ; Jianhao Lin,
| | - Jianhao Lin
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
- *Correspondence: Hao Li, ; Dan Xing, ; Jianhao Lin,
| |
Collapse
|
25
|
Qiu G, Zhong S, Xie J, Feng H, Sun S, Gao C, Xu X, Kang B, Xu H, Zhao C, Ran L, Xinyu A, Xu B, Meng X, Meng L, Zhang X, Xiao L. Expanded CD1c +CD163 + DC3 Population in Synovial Tissues Is Associated with Disease Progression of Osteoarthritis. J Immunol Res 2022; 2022:9634073. [PMID: 35958878 PMCID: PMC9359855 DOI: 10.1155/2022/9634073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/15/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
The mechanisms underlying osteoarthritis (OA) have recently been hypothesized to involve a dysfunctional immune system. In this study, we collected synovium, synovial fluid (SF), and peripheral blood from 21 patients. Mononuclear cells were characterized using FCM. H&E staining and mIHC histological assessment of synovium were performed. Cytokine levels in the SF were measured using ELISA. We observed similar frequencies of immune cells in the synovium and SF, which were enriched in DCs. Notably, CD1c+CD163+ DC3s were expanded in the synovium and SF. Furthermore, we found that DC3s were primarily located within the ectopic lymphoid-like structure (ELLS) in close proximity to CD8+ T cells. Finally, the level of TNF-α and IL12p70 in the SF correlated with the severity of OA. These data suggest that OA is an immune system-related disease and that DC3s may play an active role in OA progression by promoting ELLS formation and inflammatory responses.
Collapse
Affiliation(s)
- Guowei Qiu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Zhong
- Department of Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional Chinese and Western Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Jun Xie
- Department of Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional Chinese and Western Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Hui Feng
- Department of Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional Chinese and Western Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Songtao Sun
- Department of Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional Chinese and Western Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Chenxin Gao
- Department of Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional Chinese and Western Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Xirui Xu
- Department of Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional Chinese and Western Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| | - Bingxin Kang
- Department of Rehabilitation, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Hui Xu
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Chi Zhao
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Ran
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - A. Xinyu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bo Xu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaohui Meng
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Meng
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences/University of Chinese Academy of Sciences, Shanghai, China
| | - Xiaoming Zhang
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences/University of Chinese Academy of Sciences, Shanghai, China
| | - Lianbo Xiao
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional Chinese and Western Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200052, China
| |
Collapse
|
26
|
Cross-Tissue Analysis Using Machine Learning to Identify Novel Biomarkers for Knee Osteoarthritis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9043300. [PMID: 35785145 PMCID: PMC9246600 DOI: 10.1155/2022/9043300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 04/28/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022]
Abstract
Background Knee osteoarthritis (KOA) is a common degenerative joint disease. In this study, we aimed to identify new biomarkers of KOA to improve the accuracy of diagnosis and treatment. Methods GSE98918 and GSE51588 were downloaded from the Gene Expression Omnibus database as training sets, with a total of 74 samples. Gene differences were analyzed by Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway, and Disease Ontology enrichment analyses for the differentially expressed genes (DEGs), and GSEA enrichment analysis was carried out for the training gene set. Through least absolute shrinkage and selection operator regression analysis, the support vector machine recursive feature elimination algorithm, and gene expression screening, the range of DEGs was further reduced. Immune infiltration analysis was carried out, and the prediction results of the combined biomarker logistic regression model were verified with GSE55457. Results In total, 84 DEGs were identified through differential gene expression analysis. The five biomarkers that were screened further showed significant differences in cartilage, subchondral bone, and synovial tissue. The diagnostic accuracy of the model synthesized using five biomarkers through logistic regression was better than that of a single biomarker and significantly better than that of a single clinical trait. Conclusions CX3CR1, SLC7A5, ARL4C, TLR7, and MTHFD2 might be used as novel biomarkers to improve the accuracy of KOA disease diagnosis, monitor disease progression, and improve the efficacy of clinical treatment.
Collapse
|
27
|
Rothbauer M, Reihs EI, Fischer A, Windhager R, Jenner F, Toegel S. A Progress Report and Roadmap for Microphysiological Systems and Organ-On-A-Chip Technologies to Be More Predictive Models in Human (Knee) Osteoarthritis. Front Bioeng Biotechnol 2022; 10:886360. [PMID: 35782494 PMCID: PMC9240813 DOI: 10.3389/fbioe.2022.886360] [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: 02/28/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Osteoarthritis (OA), a chronic debilitating joint disease affecting hundreds of million people globally, is associated with significant pain and socioeconomic costs. Current treatment modalities are palliative and unable to stop the progressive degeneration of articular cartilage in OA. Scientific attention has shifted from the historical view of OA as a wear-and-tear cartilage disorder to its recognition as a whole-joint disease, highlighting the contribution of other knee joint tissues in OA pathogenesis. Despite much progress in the field of microfluidic systems/organs-on-a-chip in other research fields, current in vitro models in use do not yet accurately reflect the complexity of the OA pathophenotype. In this review, we provide: 1) a detailed overview of the most significant recent developments in the field of microsystems approaches for OA modeling, and 2) an OA-pathophysiology-based bioengineering roadmap for the requirements of the next generation of more predictive and authentic microscale systems fit for the purpose of not only disease modeling but also of drug screening to potentially allow OA animal model reduction and replacement in the near future.
Collapse
Affiliation(s)
- Mario Rothbauer
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Faculty of Technical Chemistry, Vienna University of Technology, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Eva I. Reihs
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Faculty of Technical Chemistry, Vienna University of Technology, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Anita Fischer
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Reinhard Windhager
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Florien Jenner
- Veterinary Tissue Engineering and Regenerative Medicine Vienna (VETERM), Equine Surgery Unit, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Stefan Toegel
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| |
Collapse
|
28
|
Yang X, Liang X, Guo H, Ma L, Jian L, Zhao X, Wang J, Yang L, Meng Z, Jin Q. β2-Adrenergic receptor expression in subchondral bone of patients with varus knee osteoarthritis. Open Med (Wars) 2022; 17:1031-1044. [PMID: 35794997 PMCID: PMC9175016 DOI: 10.1515/med-2022-0498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
An important causative factor in osteoarthritis (OA) is the abnormal mechanical stress-induced bone remodeling of the subchondral bone. β2-adrenergic receptor (Adrb2) plays a major role in mechanical stresses that induce bone remodeling. The medial tibial plateau (MTP) and lateral tibial plateau (LTP) of patients with varus Knee osteoarthritis (KO) bear different mechanical stresses. The present study aimed to investigate the expression of Adrb2 in medial tibial plateau subchondral bone (MTPSB) and lateral tibial plateau subchondral bone (LTPSB) in patients with varus KO. A total of 30 tibial plateau samples from patients undergoing total knee arthroplasty for varus KO and MTPSB and LTPSB were studied. Statistical analysis was performed using paired sample t-tests. Safranin O-Fast Green staining and Micro-computed tomography showed significant differences in the bone structure between MTPSB and LTPSB. Tartrate-resistant acid phosphatase (TRAP)-positive cell density in MTPSB was higher than that in LTPSB. Immunohistochemistry, reverse transcription-quantitative polymerase chain reaction, and Western blot analysis revealed that compared to LTPSB, the levels of Adrb2, tyrosine hydroxylase (TH), and osteocalcin increased significantly in MTPSB. Double-labeling immunofluorescence showed Adrb2 was present in the majority of TRAP-positive multinuclear cells of the MTPSB. The expression of Adrb2 and TH was significantly higher in MTPSB than in LTPSB, confirming the involvement of these molecules in the development of OA.
Collapse
Affiliation(s)
- Xiaochun Yang
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Xuegang Liang
- Department of The General Hospital of Ningxia Medical University, Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Haohui Guo
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Long Ma
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Li Jian
- Department of Pathology, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Xin Zhao
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Jian Wang
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Lvlin Yang
- Department of The General Hospital of Ningxia Medical University, Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Zhiqiang Meng
- Department of The General Hospital of Ningxia Medical University, Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Qunhua Jin
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| |
Collapse
|
29
|
Artificial Intelligence Algorithm-Based Magnetic Resonance Imaging to Evaluate the Effect of Radiation Synovectomy for Hemophilic Arthropathy. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:5694163. [PMID: 35360269 PMCID: PMC8957465 DOI: 10.1155/2022/5694163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022]
Abstract
This study aimed to discuss magnetic resonance imaging (MRI) based on artificial intelligence (AI) algorithm to evaluate the effect of radiation synovectomy for hemophilic arthropathy (HA). MRI based on the Canny algorithm was applied and compared with conventional MRI to evaluate its application effects according to the PSNR and SSIM. Sixty patients diagnosed with HA were selected as the research subjects. According to the detection method, the patients were divided into group A (pathological detection after radiation synovectomy), group B (conventional MRI detection), and group C (MRI detection based on the Canny algorithm). The application value of MRI based on the Canny algorithm was judged by comparing the differences between the two detection methods and pathological results. The results displayed that the reconstruction effect of the Canny algorithm was remarkably better than that of the traditional algorithm regarding the peak signal-to-noise ratio (PSNR) and structural similarity (SSIM), which showed a clearer synovial contour. The results of the IPSG score of joint effusion and hemorrhage showed that there was a difference in the detection rate of joints between conventional MRI and pathological results on the score of 1 and 2 (P < 0.05); and there was no significant difference between the MRI and pathological results based on the Canny algorithm (P > 0.05). The results of the IPSG score of synovial hyperplasia showed that the detection rate of conventional MRI was different from pathological results on the score of 1 and 2 (P < 0.05); and there was no significant difference between the MRI and pathological results based on the Canny algorithm (P > 0.05). The results of the IPSG score of hemosiderin deposition showed that the detection rate of conventional MRI was different from the pathological results on the score of 1 and 2 (P < 0.05); and there was no significant difference between the MRI and pathological results based on the Canny algorithm (P > 0.05). The synovial volume of patients after surgery was reduced compared with that before surgery. One-factor variance was used to analyze the clinical hemorrhage frequency before and after surgery, and the results showed that the differences were statistically significant (P < 0.05). Therefore, MRI on account of AI algorithm made it easier to detect synovial contour, which was helpful to evaluate the efficacy of polygenic risk scores (PRS) surgery in HA patients. MRI based on the Canny algorithm had less differences between the score of hemophilic arthropathy and pathological results, which could replace conventional MRI examination and have clinical application value.
Collapse
|
30
|
Jones GMC, Pitsillides AA, Meeson RL. Moving Beyond the Limits of Detection: The Past, the Present, and the Future of Diagnostic Imaging in Canine Osteoarthritis. Front Vet Sci 2022; 9:789898. [PMID: 35372534 PMCID: PMC8964951 DOI: 10.3389/fvets.2022.789898] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common orthopedic condition in dogs, characterized as the chronic, painful end-point of a synovial joint with limited therapeutic options other than palliative pain control or surgical salvage. Since the 1970s, radiography has been the standard-of-care for the imaging diagnosis of OA, despite its known limitations. As newer technologies have been developed, the limits of detection have lowered, allowing for the identification of earlier stages of OA. Identification of OA at a stage where it is potentially reversible still remains elusive, however, yet there is hope that newer technologies may be able to close this gap. In this article, we review the changes in the imaging of canine OA over the past 50 years and give a speculative view on future innovations which may provide for earlier identification, with the ultimate goal of repositioning the limit of detection to cross the threshold of this potentially reversible disease.
Collapse
Affiliation(s)
- Gareth M. C. Jones
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, United Kingdom
| | - Andrew A. Pitsillides
- Department of Comparative Biological Science, Royal Veterinary College, London, United Kingdom
| | - Richard L. Meeson
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, United Kingdom
| |
Collapse
|
31
|
Hodgkinson T, Amado IN, O'Brien FJ, Kennedy OD. The role of mechanobiology in bone and cartilage model systems in characterizing initiation and progression of osteoarthritis. APL Bioeng 2022. [DOI: 10.1063/5.0068277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Tom Hodgkinson
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Isabel N. Amado
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fergal J. O'Brien
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Oran D. Kennedy
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
32
|
Lin C, Chen Z, Guo D, Zhou L, Lin S, Li C, Li S, Wang X, Lin B, Ding Y. Increased expression of osteopontin in subchondral bone promotes bone turnover and remodeling, and accelerates the progression of OA in a mouse model. Aging (Albany NY) 2022; 14:253-271. [PMID: 34982732 PMCID: PMC8791213 DOI: 10.18632/aging.203707] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/28/2021] [Indexed: 02/05/2023]
Abstract
Osteopontin (OPN) has been proved to be closely related to the pathogenesis of osteoarthritis (OA), but the role of OPN in the pathogenesis of OA has not been fully clarified. Current studies on OPN in OA mostly focus on articular cartilage, synovial membrane and articular fluid, while ignoring its role in OA subchondral bone turnover and remodeling. In this study, we used a destabilization OA mouse model to investigate the role of OPN in OA subchondral bone changes. Our results indicate that increased expression of OPN accelerates the turnover and remodeling of OA subchondral bone, promotes the formation of h-type vessels in subchondral bone, and mediates articular cartilage degeneration induced by subchondral bone metabolism. In addition, our results confirmed that inhibition of PI3K/AKT signaling pathway inhibits OPN-mediated OA subchondral bone remodeling and cartilage degeneration. This study revealed the role and mechanism of OPN in OA subchondral bone, which is of great significance for exploring specific biological indicators for early diagnosis of OA and monitoring disease progression, as well as for developing drugs to regulate the metabolism and turnover of subchondral bone and alleviate the subchondral bone sclerosis of OA.
Collapse
Affiliation(s)
- Chuangxin Lin
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
- Department of Orthopedic Surgery, Shantou Central Hospital, Shantou 515000, P.R. China
| | - Zhong Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Dong Guo
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, P.R China
| | - Laixi Zhou
- Department of Orthopedic Surgery, Shantou Central Hospital, Shantou 515000, P.R. China
| | - Sipeng Lin
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Changchuan Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Shixun Li
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| | - Xinjia Wang
- Department of Orthopedic, Affiliated Cancer Hospital, Shantou University Medical College, Shantou 515041, P.R. China
| | - Bendan Lin
- Department of Orthopedic Surgery, Shantou Central Hospital, Shantou 515000, P.R. China
| | - Yue Ding
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P.R. China
| |
Collapse
|
33
|
Ye X, Liu X. Wnt16 signaling in bone homeostasis and osteoarthristis. Front Endocrinol (Lausanne) 2022; 13:1095711. [PMID: 36619549 PMCID: PMC9815800 DOI: 10.3389/fendo.2022.1095711] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Wnts are secreted cysteine-rich glycoproteins involved in joint development and skeletal homeostasis and have been implicated in the occurrence of osteoarthritis. Over the past decade, Wnt16, a member of the Wnt family, has received widespread attention for its strong association with bone mineral density, cortical bone thickness, bone strength, and osteoporotic fracture risk. In recent years, further studies have shed light on the role of Wnt16 a positive regulator of bone mass and protective regulator of osteoarthritis progression. Transduction mechanisms and crosstalk involving Wnt16 signaling have also been illustrated. More importantly, local Wnt16 treatment has been shown to ease osteoarthritis, inhibit bone resorption, and promote new bone formation in bone defect models. Thus, Wnt16 is now a potential therapeutic target for skeletal diseases and osteoarthritis. This paper reviews our current understanding of the mechanisms by which Wnt16 signaling regulates bone homeostasis and osteoarthritis.
Collapse
|
34
|
Tan Q, Cai Z, Li J, Li J, Xiang H, Li B, Cai G. Imaging Study on Acupuncture Inhibiting Inflammation and Bone Destruction in Knee Osteoarthritis Induced by Monosodium Iodoacetate in Rat Model. J Pain Res 2022; 15:93-103. [PMID: 35046719 PMCID: PMC8760981 DOI: 10.2147/jpr.s346242] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/18/2021] [Indexed: 11/26/2022] Open
Abstract
Objective We aim to explore whether acupuncture inhibits inflammation and bone destruction in rat model monosodium iodoacetate (MIA)-induced knee osteoarthritis (KOA) by 18F-fluorodeoxyglucose (18F-FDG) small-animal positron emission tomography (PET) and micro-computed tomography (CT) imaging. Methods KOA was induced in rats by intra-articular injection MIA (2 mg/50 μL) through the right knee of the rats. Forty male Sprague Dawley rats weighing 280 to 340 g (12 weeks old) were randomly divided into four groups including Control group, KOA group, KOA plus manual acupuncture group (KOA+MA), KOA plus sham acupuncture group (KOA+SA). The acupuncture treatment lasted for three weeks (one-day rest after six days of treatment). Paw withdrawal threshold test and open-field test were used to assess mechanical allodynia and locomotor activity respectively for once a week. Hematoxylin and eosin (H&E) staining was used to assess the damage of the cartilage, synovium and infrapatellar fat pad (IFP). 18F-FDG PET was performed to quantify joint inflammation. The influence on the subchondral bone in these rats was confirmed by micro-CT. Results Mechanical hyperalgesia, joint inflammation, and obvious bone destruction were observed in the KOA group. H&E staining of the knee joint found that manual acupuncture played a protective effect in cartilage, synovium and IFP destruction. However, compared with KOA group, the results in sham acupuncture had no significant difference. After manual acupuncture treatment in KOA rats, inflammation was significantly suppressed shown by 18F-FDG PET imaging. Micro-CT analysis of the knee joint revealed that manual acupuncture protected bone by inhibiting osteophyte development and subchondral bone remodeling. Conclusion The results of 18F-FDG PET and micro-CT showed that manual acupuncture inhibited inflammation and bone destruction, which provides reliable evidence for the effectiveness of acupuncture in hindering development of KOA, and provides reliable evidence for clinical application of acupuncture.
Collapse
Affiliation(s)
- Qian Tan
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhengkun Cai
- Department of Public Administration, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jia Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Jing Li
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Hongchun Xiang
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Bocun Li
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Guowei Cai
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Correspondence: Guowei Cai Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China Email
| |
Collapse
|
35
|
Abstract
The last decade has seen an enormous increase in long non-coding RNA (lncRNA) research within rheumatology. LncRNAs are arbitrarily classed as non-protein encoding RNA transcripts that exceed 200 nucleotides in length. These transcripts have tissue and cell specific patterns of expression and are implicated in a variety of biological processes. Unsurprisingly, numerous lncRNAs are dysregulated in rheumatoid conditions, correlating with disease activity and cited as potential biomarkers and targets for therapeutic intervention. In this chapter, following an introduction into each condition, we discuss the lncRNAs involved in rheumatoid arthritis, osteoarthritis and systemic lupus erythematosus. These inflammatory joint conditions share several inflammatory signalling pathways and therefore not surprisingly many commonly dysregulated lncRNAs are shared across these conditions. In the interest of translational research only those lncRNAs which are strongly conserved have been addressed. The lncRNAs discussed here have diverse roles in regulating inflammation, proliferation, migration, invasion and apoptosis. Understanding the molecular basis of lncRNA function in rheumatology will be crucial in fully determining the inflammatory mechanisms that drive these conditions.
Collapse
|
36
|
Li Y, Zhou Y, Wang Y, Crawford R, Xiao Y. Synovial macrophages in cartilage destruction and regeneration-lessons learnt from osteoarthritis and synovial chondromatosis. Biomed Mater 2021; 17. [PMID: 34823229 DOI: 10.1088/1748-605x/ac3d74] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/25/2021] [Indexed: 01/15/2023]
Abstract
Inflammation is a critical process in disease pathogenesis and the restoration of tissue structure and function, for example, in joints such as the knee and temporomandibular. Within the innate immunity process, the body's first defense response in joints when physical and chemical barriers are breached is the synovial macrophages, the main innate immune effector cells, which are responsible for triggering the initial inflammatory reaction. Macrophage is broadly divided into three phenotypes of resting M0, pro-inflammatory M1-like (referred to below as M1), and anti-inflammatory M2-like (referred to below as M2). The synovial macrophage M1-to-M2 transition can affect the chondrogenic differentiation of mesenchymal stem cells (MSCs) in joints. On the other hand, MSCs can also influence the transition between M1 and M2. Failure of the chondrogenic differentiation of MSCs can result in persistent cartilage destruction leading to osteoarthritis. However, excessive chondrogenic differentiation of MSCs may cause distorted cartilage formation in the synovium, which is evidenced in the case of synovial chondromatosis. This review summarizes the role of macrophage polarization in the process of both cartilage destruction and regeneration, and postulates that the transition of macrophage phenotype in an inflammatory joint environment may play a key role in determining the fate of joint cartilage.
Collapse
Affiliation(s)
- Yingjie Li
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Yinghong Zhou
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Yifan Wang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Ross Crawford
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Yin Xiao
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| |
Collapse
|
37
|
Zhuang C, Wang Z, Chen W, Wang H, Tian B, Lin H. Jintiange Capsules Ameliorate Osteoarthritis by Modulating Subchondral Bone Remodeling and Protecting Cartilage Against Degradation. Front Pharmacol 2021; 12:762543. [PMID: 34858187 PMCID: PMC8631927 DOI: 10.3389/fphar.2021.762543] [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: 08/24/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disease worldwide, making it a major cause of pain and disability. Identified as a chronic and progressive disease, effective treatment at the early stages of OA has become critical to its management. Jintiange (Jtg) capsules are a traditional Chinese medicine produced from multiple organic components of various animal bones and routinely used to treat osteoporosis in China. However, the effect of Jtg on subchondral bone and cartilage degeneration in OA remains unknown. The purpose of the present study was to investigate the biomolecular role and underlying mechanisms of Jtg in OA progression. Herein, we found that Jtg inhibited receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation and it functions through the NF-κB signaling pathway. Jtg also inhibited chondrocyte apoptosis via reducing the reactive oxygen species concentration in these cells. Moreover, in vivo evaluation revealed that Jtg significantly attenuates subchondral bone remodeling and cartilage destruction in anterior cruciate ligament transection (ACLT) mouse models. Taken together, our data demonstrate that Jtg inhibits osteoclast differentiation in subchondral bone and chondrocyte apoptosis in cartilage, supporting its potential therapeutic value for treating OA.
Collapse
Affiliation(s)
- Chenyang Zhuang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zixiang Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weisin Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hanquan Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bo Tian
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hong Lin
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Orthopedics, Shanghai Geriatric Medical Centre, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Li M, Li H, Ran X, Yin H, Luo X, Chen Z. Effects of adenovirus-mediated knockdown of IRAK4 on synovitis in the osteoarthritis rabbit model. Arthritis Res Ther 2021; 23:294. [PMID: 34863246 PMCID: PMC8643028 DOI: 10.1186/s13075-021-02684-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/20/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The use of interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitor as a treatment for the inflammatory joint disease is a promising method. However, its underlying mechanism in osteoarthritis (OA) remains unclear. The purpose of this study is to look into the effects of adenovirus-mediated knockdown of IRAK4 on synovitis in the OA rabbit model. METHODS Ad-shIRAK4 was injected two weeks after anterior cruciate ligament resection. Six weeks later, the rabbits were killed. The expression of IRAK4, TNFR-associated factor 6(TRAF6), TGF-activated kinase 1(TAK1), p-IKB kinase (p-IKK), p-nuclear factor kappa-B (p-NFκB), p38, and p-p38 in the synovial membrane was detected by western blot, qRT-PCR, and immunohistochemistry analysis. Immunohistochemistry was to detect the expression of IRAK4 proteins in articular cartilage. H&E staining was to assess the pathological changes of synovium and cartilage. The levels of interleukin (IL)-1β, tumor necrosis factor-α(TNF-α), and MMP-13 in the synovial fluid were measured by ELISA. X-ray and micro-computerized tomography (μCT) scans were used to assess knee joint conditions and microstructure of subchondral bone. RESULTS IRAK4 expression levels in synovial tissues of the OA model group exhibited a significant upward trend. Ad-shIRAK4 significantly reduced IRAK4 mRNA expression in synovium tissues. Notably, Ad-shIRAK4 suppressed the Toll-like receptor/interleukin-1 receptor (TLR/IL-1R) signaling. In addition, in the Ad-shIRAK4 treatment group, we can see less inflammatory cell infiltration and reduced hyperplasia and angiogenesis. The levels of IL-1β, TNF-α, and MMP-13 in the synovial fluid in the OA model group were significantly higher than that in the control group, which were reduced by Ad-shIRAK4 treatment. Finally, Results of HE stains, immunohistochemistry, and μCT showed that Ad-shIRAK4 treatment has a protective effect on cartilage damage. CONCLUSIONS IRAK4 is significantly upregulated in the synovium from the osteoarthritis rabbit model. In addition, Ad-shIRAK4 reduced the expression of IRAK4 and suppressed TLR/IL-1R signaling in the synovium from the osteoarthritis rabbit model. Ad-shIRAK4 could alleviate synovitis and cartilage degradation in the osteoarthritis rabbit model, and thus alleviate the symptoms of OA and prevent the progression of OA.
Collapse
Affiliation(s)
- Muzhe Li
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Huiyun Li
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Xun Ran
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Han Yin
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xuling Luo
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China.
| | - Zhiwei Chen
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China.
| |
Collapse
|
39
|
Qin Y, Li J, Zhou Y, Yin C, Li Y, Chen M, Du Y, Li T, Yan J. Apolipoprotein D as a Potential Biomarker and Construction of a Transcriptional Regulatory-Immune Network Associated with Osteoarthritis by Weighted Gene Coexpression Network Analysis. Cartilage 2021; 13:1702S-1717S. [PMID: 34719950 PMCID: PMC8808834 DOI: 10.1177/19476035211053824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Synovial inflammation influences the progression of osteoarthritis (OA). Herein, we aimed to identify potential biomarkers and analyze transcriptional regulatory-immune mechanism of synovitis in OA using weighted gene coexpression network analysis (WGCNA). DESIGN A data set of OA synovium samples (GSE55235) was analyzed based on WGCNA. The most significant module with OA was identified and function annotation of the module was performed, following which the hub genes of the module were identified using Pearson correlation and a protein-protein interaction network was constructed. A transcriptional regulatory network of hub genes was constructed using the TRRUST database. The immune cell infiltration of OA samples was evaluated using the single-sample Gene Set Enrichment Analysis (ssGSEA) method. The hub genes coexpressed in multiple tissues were then screened out using data sets of synovium, cartilage, chondrocyte, subchondral bone, and synovial fluid samples. Finally, transcriptional factors and coexpressed hub genes were validated via experiments. RESULTS The turquoise module of GSE55235 was identified via WGCNA. Functional annotation analysis showed that "mineral absorption" and "FoxO signaling pathway" were mostly enriched in the module. JUN, EGR1, FOSB, and KLF4 acted as central nodes in protein-protein interaction network and transcription factors to connect several target genes. "Activated B cell," "activated CD4T cell," "eosinophil," "neutrophil," and "type 17 T helper cell" showed high immune infiltration, while FOSB, KLF6, and MYBL2 showed significant negative correlation with type 17 T helper cell. CONCLUSIONS Our results suggest that the expression level of apolipoprotein D (APOD) was correlated with OA. Furthermore, transcriptional regulatory-immune network was constructed, which may contribute to OA therapy.
Collapse
Affiliation(s)
- Yong Qin
- Department of Orthopedics Surgery, The
Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jia Li
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yonggang Zhou
- Department of Orthopedics Surgery, The
Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chengliang Yin
- Medical Big Data Research Center,
Medical Innovation Research Division of Chinese PLA General Hospital, Beijing,
China,National Engineering Laboratory for
Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing,
China,Faculty of Medicine, Macau University
of Science and Technology, Macau, China
| | - Yi Li
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ming Chen
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yinqiao Du
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Tiejian Li
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jinglong Yan
- Department of Orthopedics Surgery, The
Second Affiliated Hospital of Harbin Medical University, Harbin, China,Jinglong Yan, Department of Orthopedics
Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246
Xuefu Road, Harbin 150086, China.
| |
Collapse
|
40
|
Jiang A, Xu P, Sun S, Zhao Z, Tan Q, Li W, Song C, Leng H. Cellular alterations and crosstalk in the osteochondral joint in osteoarthritis and promising therapeutic strategies. Connect Tissue Res 2021; 62:709-719. [PMID: 33397157 DOI: 10.1080/03008207.2020.1870969] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/28/2020] [Indexed: 02/03/2023]
Abstract
Osteoarthritis (OA) is a joint disorder involving cartilage degeneration and subchondral bone sclerosis. The bone-cartilage interface is implicated in OA pathogenesis due to its susceptibility to mechanical and biological factors. The crosstalk between cartilage and the underlying subchondral bone is elevated in OA due to multiple factors, such as increased vascularization, porosity, microcracks and fissures. Changes in the osteochondral joint are traceable to alterations in chondrocytes and bone cells (osteoblasts, osteocytes and osteoclasts). The phenotypes of these cells can change with the progression of OA. Aberrant intercellular communications among bone cell-bone cell and bone cell-chondrocyte are of great importance and might be the factors promoting OA development. An appreciation of cellular phenotypic changes in OA and the mechanisms by which these cells communicate would be expected to lead to the development of targeted drugs with fewer side effects.
Collapse
Affiliation(s)
- Ai Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Peng Xu
- State Key Laboratory of Systematic and Evolutionary Botany, Institute of Botany, Chinese Academy of Sciences, Beijing, China
| | - Shang Sun
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Zhenda Zhao
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Qizhao Tan
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education Lisbon Portugal
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Lab of Spine Diseases, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
41
|
Wallace KG, Pfeiffer SJ, Pietrosimone LS, Harkey MS, Zong X, Nissman D, Kamath GM, Creighton RA, Spang JT, Blackburn JT, Pietrosimone B. Changes in Infrapatellar Fat Pad Volume 6 to 12 Months After Anterior Cruciate Ligament Reconstruction and Associations With Patient-Reported Knee Function. J Athl Train 2021; 56:1173-1179. [PMID: 33787883 PMCID: PMC8582630 DOI: 10.4085/1062-6050-0458.20] [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] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
CONTEXT Hypertrophy of the infrapatellar fat pad (IFP) in idiopathic knee osteoarthritis has been linked to deleterious synovial changes and joint pain related to mechanical tissue impingement. Yet little is known regarding the IFP's volumetric changes after anterior cruciate ligament reconstruction (ACLR). OBJECTIVES To examine changes in IFP volume between 6 and 12 months after ACLR and determine associations between patient-reported outcomes and IFP volume at each time point as well as the volume change over time. In a subset of individuals, we examined interlimb IFP volume differences 12 months post-ACLR. STUDY DESIGN Prospective cohort study. SETTING Laboratory. PATIENTS OR OTHER PARTICIPANTS We studied 26 participants (13 women, 13 men, age = 21.88 ± 3.58 years, body mass index = 23.82 ± 2.21 kg/m2) for our primary aims and 13 of those participants (8 women, 5 men, age = 21.15 ± 3.85 years, body mass index = 23.01 ± 2.01 kg/m2) for our exploratory aim. MAIN OUTCOME MEASURE(S) Using magnetic resonance imaging, we evaluated the IFP volume change between 6 and 12 months post-ACLR in the ACLR limb and between-limbs differences at 12 months in a subset of participants. International Knee Documentation Committee subjective knee evaluation (IKDC) scores were collected at 6-month and 12-month follow-ups, and associations between IFP volume and patient-reported outcomes were determined. RESULTS The IFP volume in the ACLR limb increased from 6 months (19.67 ± 6.30 cm3) to 12 months (21.26 ± 6.91 cm3) post-ACLR. Greater increases of IFP volume between 6 and 12 months were significantly associated with better 6-month IKDC scores (r = .44, P = .03). The IFP volume was greater in the uninjured limb (22.71 ± 7.87 cm3) than in the ACLR limb (20.75 ± 9.03 cm3) 12 months post-ACLR. CONCLUSIONS The IFP volume increased between 6 and 12 months post-ACLR; however, the IFP volume of the ACLR limb remained smaller than that of the uninjured limb at 12 months. In addition, those with better knee function 6 months post-ACLR demonstrated greater increases in IFP volume between 6 and 12 months post-ACLR. This suggests that greater IFP volumes may play a role in long-term joint health after ACLR.
Collapse
Affiliation(s)
- Kyle G Wallace
- Georgetown University School of Medicine, Washington, DC
- MOTION Science Institute, Department of Exercise and Sport Science, University of North Carolina at Chapel Hill
| | - Steven J Pfeiffer
- MOTION Science Institute, Department of Exercise and Sport Science, University of North Carolina at Chapel Hill
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens
- School of Applied Health Sciences and Wellness, Ohio University, Athens
| | - Laura S Pietrosimone
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
| | - Matthew S Harkey
- Department of Kinesiology, Michigan State University, East Lansing
| | - Xiaopeng Zong
- Department of Radiology, University of North Carolina at Chapel Hill
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill
| | - Daniel Nissman
- Department of Radiology, University of North Carolina at Chapel Hill
| | - Ganesh M Kamath
- Department of Orthopaedics, University of North Carolina at Chapel Hill
| | | | - Jeffrey T Spang
- Department of Orthopaedics, University of North Carolina at Chapel Hill
| | - J Troy Blackburn
- MOTION Science Institute, Department of Exercise and Sport Science, University of North Carolina at Chapel Hill
- Department of Orthopaedics, University of North Carolina at Chapel Hill
- Human Movement Science Curriculum, University of North Carolina at Chapel Hill
| | - Brian Pietrosimone
- MOTION Science Institute, Department of Exercise and Sport Science, University of North Carolina at Chapel Hill
- Department of Orthopaedics, University of North Carolina at Chapel Hill
- Human Movement Science Curriculum, University of North Carolina at Chapel Hill
| |
Collapse
|
42
|
Characterization and miRNA Profiling of Extracellular Vesicles from Human Osteoarthritic Subchondral Bone Multipotential Stromal Cells (MSCs). Stem Cells Int 2021; 2021:7232773. [PMID: 34667479 PMCID: PMC8520657 DOI: 10.1155/2021/7232773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/26/2021] [Accepted: 08/19/2021] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is a heterogeneous disease in which the cross-talk between the cells from different tissues within the joint is affected as the disease progresses. Extracellular vesicles (EVs) are known to have a crucial role in cell-cell communication by means of cargo transfer. Subchondral bone (SB) resident cells and its microenvironment are increasingly recognised to have a major role in OA pathogenesis. The aim of this study was to investigate the EV production from OA SB mesenchymal stromal cells (MSCs) and their possible influence on OA chondrocytes. Small EVs were isolated from OA-MSCs, characterized and cocultured with chondrocytes for viability and gene expression analysis, and compared to small EVs from MSCs of healthy donors (H-EVs). OA-EVs enhanced viability of chondrocytes and the expression of chondrogenesis-related genes, although the effect was marginally lower compared to that of the H-EVs. miRNA profiling followed by unsupervised hierarchical clustering analysis revealed distinct microRNA sets in OA-EVs as compared to their parental MSCs or H-EVs. Pathway analysis of OA-EV miRNAs showed the enrichment of miRNAs implicated in chondrogenesis, stem cells, or other pathways related to cartilage and OA. In conclusion, OA SB MSCs were capable of producing EVs that could support chondrocyte viability and chondrogenic gene expression and contained microRNAs implicated in chondrogenesis support. These EVs could therefore mediate the cross-talk between the SB and cartilage in OA potentially modulating chondrocyte viability and endogenous cartilage regeneration.
Collapse
|
43
|
Chen T, Weng W, Liu Y, Aspera-Werz RH, Nüssler AK, Xu J. Update on Novel Non-Operative Treatment for Osteoarthritis: Current Status and Future Trends. Front Pharmacol 2021; 12:755230. [PMID: 34603064 PMCID: PMC8481638 DOI: 10.3389/fphar.2021.755230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 09/06/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis (OA) is a leading cause of pain and disability which results in a reduced quality of life. Due to the avascular nature of cartilage, damaged cartilage has a finite capacity for healing or regeneration. To date, conservative management, including physical measures and pharmacological therapy are still the principal choices offered for OA patients. Joint arthroplasties or total replacement surgeries are served as the ultimate therapeutic option to rehabilitate the joint function of patients who withstand severe OA. However, these approaches are mainly to relieve the symptoms of OA, instead of decelerating or reversing the progress of cartilage damage. Disease-modifying osteoarthritis drugs (DMOADs) aiming to modify key structures within the OA joints are in development. Tissue engineering is a promising strategy for repairing cartilage, in which cells, genes, and biomaterials are encompassed. Here, we review the current status of preclinical investigations and clinical translations of tissue engineering in the non-operative treatment of OA. Furthermore, this review provides our perspective on the challenges and future directions of tissue engineering in cartilage regeneration.
Collapse
Affiliation(s)
- Tao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Trauma and Reconstructive Surgery, BG Trauma Center Tübingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Weidong Weng
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tübingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Yang Liu
- Department of Clinical Sciences, Orthopedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Romina H Aspera-Werz
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tübingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas K Nüssler
- Department of Trauma and Reconstructive Surgery, BG Trauma Center Tübingen, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Jianzhong Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
44
|
Bedingfield SK, Colazo JM, Di Francesco M, Yu F, Liu DD, Di Francesco V, Himmel LE, Gupta MK, Cho H, Hasty KA, Decuzzi P, Duvall CL. Top-Down Fabricated microPlates for Prolonged, Intra-articular Matrix Metalloproteinase 13 siRNA Nanocarrier Delivery to Reduce Post-traumatic Osteoarthritis. ACS NANO 2021; 15:14475-14491. [PMID: 34409835 PMCID: PMC9074946 DOI: 10.1021/acsnano.1c04005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) associated with joint injury triggers a degenerative cycle of matrix destruction and inflammatory signaling, leading to pain and loss of function. Here, prolonged RNA interference (RNAi) of matrix metalloproteinase 13 (MMP13) is tested as a PTOA disease modifying therapy. MMP13 is upregulated in PTOA and degrades the key cartilage structural protein type II collagen. Short interfering RNA (siRNA) loaded nanoparticles (siNPs) were encapsulated in shape-defined poly(lactic-co-glycolic acid) (PLGA) based microPlates (μPLs) to formulate siNP-μPLs that maintained siNPs in the joint significantly longer than delivery of free siNPs. Treatment with siNP-μPLs against MMP13 (siMMP13-μPLs) in a mechanical load-induced mouse model of PTOA maintained potent (65-75%) MMP13 gene expression knockdown and reduced MMP13 protein production in joint tissues throughout a 28-day study. MMP13 silencing reduced PTOA articular cartilage degradation/fibrillation, meniscal deterioration, synovial hyperplasia, osteophytes, and pro-inflammatory gene expression, supporting the therapeutic potential of long-lasting siMMP13-μPL therapy for PTOA.
Collapse
Affiliation(s)
- Sean K Bedingfield
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States; Vanderbilt University School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States; Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Danielle D. Liu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States; Vanderbilt University School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States; Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Valentina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Lauren E. Himmel
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Hongsik Cho
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee 38104, United States; Research 151, VA Medical Center, Memphis, Tennessee 38104, United States
| | - Karen A. Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center-Campbell Clinic, Memphis, Tennessee 38104, United States; Research 151, VA Medical Center, Memphis, Tennessee 38104, United States
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, Italy
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
45
|
Mao L, Wu W, Wang M, Guo J, Li H, Zhang S, Xu J, Zou J. Targeted treatment for osteoarthritis: drugs and delivery system. Drug Deliv 2021; 28:1861-1876. [PMID: 34515606 PMCID: PMC8439249 DOI: 10.1080/10717544.2021.1971798] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The management of osteoarthritis (OA) is a clinical challenge due to the particular avascular, dense, and occluded tissue structure. Despite numerous clinical reports and animal studies, the pathogenesis and progression of OA are still not fully understood. On the basis of traditional drugs, a large number of new drugs have been continuously developed. Intra-articular (IA) administration for OA hastens the development of targeted drug delivery systems (DDS). OA drugs modification and the synthesis of bioadaptive carriers contribute to a qualitative leap in the efficacy of IA treatment. Nanoparticles (NPs) are demonstrated credible improvement of drug penetration and retention in OA. Targeted nanomaterial delivery systems show the prominent biocompatibility and drug loading-release ability. This article reviews different drugs and nanomaterial delivery systems for IA treatment of OA, in an attempt to resolve the inconsonance between in vitro and in vivo release, and explore more interactions between drugs and nanocarriers, so as to open up new horizons for the treatment of OA.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Wei Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Miao Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Hui Li
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Shihua Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
46
|
Pan L, Ding W, Li J, Gan K, Shen Y, Xu J, Zheng M. Aldehyde dehydrogenase 2 alleviates monosodium iodoacetate-induced oxidative stress, inflammation and apoptosis in chondrocytes via inhibiting aquaporin 4 expression. Biomed Eng Online 2021; 20:80. [PMID: 34362382 PMCID: PMC8349086 DOI: 10.1186/s12938-021-00917-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a common cause of disability among the elderly. We aimed to explore the effects of aldehyde dehydrogenase (ALDH) 2 on the progression of KOA and identifying the potential mechanisms. METHODS First, ALDH2 expression in knee joint effusion of patients with KOA and the levels of oxidative stress-related markers were determined. After ALDH2 overexpression in monosodium iodoacetate (MIA)-treated SW1353 cells, cell viability was tested with CCK-8 assay. Subsequently, oxidative stress and inflammation-associated factors were measured. Meanwhile, cell apoptosis was assessed with TUNEL staining and expression of apoptosis-related proteins was detected by western blotting. To analyze the mechanism of ALDH2 in KOA, aquaporin 4 (AQP4) expression was determined using western blotting following ALDH2-upregulation. Subsequently, AQP4 was overexpressed to evaluate the changing of oxidative stress, inflammation and apoptosis in SW1353 cells exposed to MIA with ALDH2 overexpression. RESULTS Results indicated that knee joint effusion with higher ALDH2 expression displayed lower oxidative stress. In addition, significantly upregulated ALDH2 expression was observed in MIA-treated SW1353 cells. ALDH2 overexpression oxidative stress, inflammation and apoptosis in SW1353 cells exposed to MIA. Moreover, MIA-triggered elevated expression of AQP4, which was reduced by ALDH2 overexpression. By contrast, AQP4-upregulation abrogated the inhibitory effects of ALDH2 on oxidative stress, inflammation and apoptosis in MIA-induced SW1353 cells. CONCLUSIONS ALDH2 inactivates the expression of AQP4, by which mechanism the MIA-induced oxidative stress, inflammation and apoptosis injuries were alleviated, which provides a novel insight for understanding the mechanism of KOA and a promising target for the treatment of this disease.
Collapse
Affiliation(s)
- Lingxiao Pan
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, No. 1111 Jiangnan Road, Ningbo, 315400, Zhejiang, China
| | - Wei Ding
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, No. 1111 Jiangnan Road, Ningbo, 315400, Zhejiang, China
| | - Jie Li
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, No. 1111 Jiangnan Road, Ningbo, 315400, Zhejiang, China
| | - Kaifeng Gan
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, No. 1111 Jiangnan Road, Ningbo, 315400, Zhejiang, China
| | - Yandong Shen
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, No. 1111 Jiangnan Road, Ningbo, 315400, Zhejiang, China
| | - Junxiang Xu
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, No. 1111 Jiangnan Road, Ningbo, 315400, Zhejiang, China
| | - Minzhe Zheng
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, No. 1111 Jiangnan Road, Ningbo, 315400, Zhejiang, China.
| |
Collapse
|
47
|
The expression of Netrin-1 in the MIA-induced osteoarthritic temporomandibular joint in mice. Sci Rep 2021; 11:15695. [PMID: 34344989 PMCID: PMC8333414 DOI: 10.1038/s41598-021-95251-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/14/2021] [Indexed: 11/09/2022] Open
Abstract
Subchondral bone degeneration is the main pathological change during temporomandibular joint (TMJ) osteoarthritis (OA) development. Netrin-1, an axon-guiding factor, might play roles in OA development and pain. The purpose of this study was to investigate the expression of Netrin-1 in TMJ OA and its possible role in the progression of TMJ OA and pain. The synovial fluids of temporomandibular joint disorders (TMDs) patients were collected for Netrin-1 by enzyme linked immunosorbent assay (ELISA). TMJ OA model was built by MIA joint injection, and then the von Frey test, hematoxylin & eosin (H&E) staining, toluidine blue (TB) staining, immunohistochemical (IHC) staining and micro-CT were performed. After induction of osteoclast differentiation of raw264.7 cells, immunofluorescence (IF) was used to detect the Netrin-1 and its receptors on osteoclast membrane. The concentration of Netrin-1 increased in the synovial fluid of TMJ OA patients. After MIA injection to TMJ, the head withdrawal threshold (HWT) was significantly decreased. Microscopically, the structural disorder of subchondral bone was the most obvious at the 2nd week after MIA injection. In addition, Netrin-1 expression increased in the subchondral bone at the 2nd week after MIA injection. In vitro, the expressions of Netrin-1 and its receptor Unc5B were upregulated on the osteoclast membrane. Netrin-1 might be an important regulator during bone degeneration and pain in the process of TMJ OA.
Collapse
|
48
|
Hu X, Feng G, Meng Z, Ma L, Jin Q. The protective mechanism of SIRT1 on cartilage through regulation of LEF-1. BMC Musculoskelet Disord 2021; 22:642. [PMID: 34315467 PMCID: PMC8317295 DOI: 10.1186/s12891-021-04516-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 07/07/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic degenerative disease that suppresses middle-aged and older people worldwide. Silent information regulator 1(SIRT-1) is associated with several age-related diseases, such as cardiovascular diseases, neurodegenerative diseases and tumors, etc. The protective role of SIRT-1 in bone and joint diseases has become increasingly well known. OBJECTIVE To explore the relationship between SIRT-1 and its related factors in OA. METHODS Fresh tibial plateau specimens were collected from 30 patients with knee OA who underwent total knee arthroplasty. According to the results of Safranin O Fast Green Staining, hematoxylin-eosin staining and the OARSI grade developed by the International Association for the Study of Osteoarthropathy, the specimens were divided into the mild group, moderate group and severe group, and the damage of cartilage was evaluated. SIRT-1 protein levels in cartilage samples were analyzed by immunohistochemistry. Then, take 60 8-week-old female C57BL/6 J mice and apply the Destabilization of the medial meniscus (DMM) to induce OA. Mice were randomly divided into normal group (sham), model group (model), and post-modeling drug administration group (srt), and each group was further divided into 2 weeks after modeling (2 W) and 8 weeks after modeling (8 W) according to the time after surgery. The degenerative degree of a knee joint in mouse knee cartilage samples was evaluated using Safranin O Fast Green Staining and OARSI grade. Immunohistochemical techniques assessed the protein levels of SIRT-1, β-catenin, LEF-1, MMP-13 and Collagen II in cartilage samples. The protein levels of β-catenin, LEF-1 and MMP-13 in the samples were assessed by the immunohistofluorescence technique. The mRNA expression of SIRT-1 and LEF-1 in mouse cartilage samples was evaluated by real-time quantitative polymerase chain reaction (qPCR). RESULTS In the human cartilage samples, according to the results of Safranin O Fast Green Staining, compared with the mild group, the moderate group and the severe group showed damage cartilage layer structure, the number of chondrocytes decreased, the cell hypertrophic, the cartilage surface discontinuous, and the OARSI grade increased. The severe group had severe cartilage injury and the highest OARSI grade. In the mice cartilage samples, according to immunohistochemical analysis, the protein levels of β-catenin, LEF-1 and MMP-13 in cartilage specimens of model 2 W and model 8 W groups were significantly increased than the sham 2 W and sham 8 W groups. The protein levels of SIRT-1 and Collagen II were significantly decreased (P < 0.05), the results of srt 2 W and srt 8 W groups were between the sham group and the model group. According to immunofluorescence analysis, the protein levels of β-catenin, LEF-1 and MMP-13 in model 2 W and model 8 W groups were significantly increased than sham 2 W and sham 8 W groups. The results of srt 2w and srt 8w groups were between the sham group and the model group. According to the real-time qPCR results: Compared with sham 2 W and sham 8 W groups, the mRNA expression of SIRT-1 in model 2 W and model 8 W groups was significantly decreased, while the mRNA expression of LEF-1 was significantly increased. In contrast, the results of srt 2 W and srt 8 W groups were between the sham group and the model group. CONCLUSION SRT-1720, as a specific activator of SIRT-1, does increase the protein level of SIRT-1. SIRT-1 may play a protective role in cartilage by regulating the expression of LEF-1 and related inflammatory factors in OA.
Collapse
Affiliation(s)
- Xueyu Hu
- Ningxia Medical University, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, P.R. China
| | - Gangning Feng
- Ningxia Medical University, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, P.R. China
| | - Zhiqiang Meng
- Ningxia Medical University, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, P.R. China
| | - Long Ma
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia, P.R. China
| | - Qunhua Jin
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia, P.R. China.
| |
Collapse
|
49
|
Oo WM, Little C, Duong V, Hunter DJ. The Development of Disease-Modifying Therapies for Osteoarthritis (DMOADs): The Evidence to Date. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2921-2945. [PMID: 34262259 PMCID: PMC8273751 DOI: 10.2147/dddt.s295224] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/16/2021] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) is a complex heterogeneous articular disease with multiple joint tissue involvement of varying severity and no regulatory-agency-approved disease-modifying drugs (DMOADs). In this review, we discuss the reasons necessitating the development of DMOADs for OA management, the classifications of clinical phenotypes or molecular/mechanistic endotypes from the viewpoint of targeted drug discovery, and then summarize the efficacy and safety profile of a range of targeted drugs in Phase 2 and 3 clinical trials directed to cartilage-driven, bone-driven, and inflammation-driven endotypes. Finally, we briefly put forward the reasons for failures in OA clinical trials and possible steps to overcome these barriers.
Collapse
Affiliation(s)
- Win Min Oo
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar
| | - Christopher Little
- Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Vicky Duong
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David J Hunter
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
50
|
Jin H, Jiang S, Wang R, Zhang Y, Dong J, Li Y. Mechanistic Insight Into the Roles of Integrins in Osteoarthritis. Front Cell Dev Biol 2021; 9:693484. [PMID: 34222261 PMCID: PMC8250141 DOI: 10.3389/fcell.2021.693484] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/19/2021] [Indexed: 01/13/2023] Open
Abstract
Osteoarthritis (OA), one of the most common degenerative diseases, is characterized by progressive degeneration of the articular cartilage and subchondral bone, as well as the synovium. Integrins, comprising a family of heterodimeric transmembrane proteins containing α subunit and β subunit, play essential roles in various physiological functions of cells, such as cell attachment, movement, growth, differentiation, and mechanical signal conduction. Previous studies have shown that integrin dysfunction is involved in OA pathogenesis. This review article focuses on the roles of integrins in OA, especially in OA cartilage, subchondral bone and the synovium. A clear understanding of these roles may influence the future development of treatments for OA.
Collapse
Affiliation(s)
- Hongfu Jin
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shigang Jiang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ruomei Wang
- Department of Endocrinology and Metabolic Diseases, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jiangtao Dong
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|