1
|
Canalis E, Guzzo R, Schilling L, Denker E. NOTCH2 disrupts the synovial fibroblast identity and the inflammatory response of epiphyseal chondrocytes. J Biol Chem 2025:110206. [PMID: 40345585 DOI: 10.1016/j.jbc.2025.110206] [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: 03/20/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025] Open
Abstract
Notch signaling plays a fundamental role in the inflammatory response and has been linked to the pathogenesis of osteoarthritis in murine models of the disease and in humans. To address how Notch signaling modifies transcriptomes and cell populations, we examined the effects of NOTCH2 in chondrocytes from mice harboring a NOTCH2 gain-of-function mutation (Notch2tm1.1Ecan) and a conditional NOTCH2 gain-of-function model expressing the NOTCH2 intracellular domain (NICD2) from the Rosa26 locus (R26-NICD2 mice). Bulk RNA-Sequencing (RNA-Seq) of primary epiphyseal cells from both gain-of-function models established increased expression of pathways associated with the phagosome, genes linked to osteoclast activity in rheumatoid arthritis signaling and pulmonary fibrosis signaling. Expression of genes linked to collagen degradation was enhanced in Notch2tm1.1Ecan cells, while genes related to osteoarthritis pathways were increased in NICD2-expressing cells. Single cell (sc)RNA-Seq of cultured Notch2tm1.1Ecan cells revealed clusters of cells related to limb mesenchyme, chondrogenic cells and fibroblasts including articular synovial fibroblasts. Pseudotime trajectory revealed close associations among clusters in control cultures, but the cluster of articular/synovial fibroblasts was disrupted in cells from Notch2tm1.1Ecan mice. ScRNA-Seq showed similarities in the cluster distributions and pseudotime trajectories of NICD2-expressing and control cells, except for altered progression in a cluster of NICD2-expressing cells. In conclusion, NOTCH2 enhances the activity of pathways associated with inflammation in epiphyseal chondrocytes and disrupts the transcriptome profile of articular/synovial fibroblasts.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030; Department of Medicine, UConn Health, Farmington, CT 06030; UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030.
| | - Rosa Guzzo
- Department of Neuroscience, UConn Health, Farmington, CT 06030
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030
| | - Emily Denker
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030
| |
Collapse
|
2
|
Meng P, Liu L, Jiang N, Hao S, Yao C, Liu H, Zhang Y, Zhang F, Jia Y, Guo X. Selenium deficiency exacerbates cartilage degradation caused by HT-2 toxin via notch signaling pathway activation. Eur J Nutr 2025; 64:129. [PMID: 40105990 DOI: 10.1007/s00394-025-03621-0] [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: 03/31/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE This study aims to explore the interaction of Selenium (Se) deficiency and HT-2 toxin on cartilage homeostasis and the effect of Notch signaling pathway in this process. METHODS Male C57BL/6 mice were randomly assigned to different dietary groups and subjected to either a Se-deficiency diet or a control diet for 4 weeks, followed by exposure to varying doses of HT-2 toxin for 4 weeks. Primary mouse chondrocytes were extracted and treated with DAPT (N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester), a γ-secretase inhibitor for the Notch signaling pathway, before combined intervention. Histological evaluation and transmission electron microscopy (TEM) were applied to assess cartilage damage, while immunohistochemical (IHC) analysis and Quantitative real-time polymerase chain reaction (qRT-PCR) were performed to detect extracellular matrix (ECM) metabolism and Notch signaling. RESULTS HT-2 toxin, alone or in combination with Se deficiency, led to significant cartilage injury characterized by chondrocyte necrosis and ultrastructural abnormalities. IHC revealed increased expression of Adamts5 and decreased expression of Col2a1 and Acan in cartilage following exposure to HT-2 toxin, indicative of ECM degradation, which could be aggravated under Se deficiency. Additionally, activation of the Notch signaling pathway was observed in response to HT-2 toxin and Se deficiency, with upregulation of Notch pathway-related components. In vitro experiments further confirmed the role of the Notch pathway in ECM metabolism regulation, with partial protection against ECM depletion caused by HT-2 toxin and Se deficiency observed upon inhibition of the Notch pathway using DAPT. CONCLUSION This study demonstrate that Se deficiency exacerbates HT-2 toxin-induced cartilage degradation via Notch signaling activation, highlighting the interplay of environmental mycotoxins and nutritional deficits in KBD etiology and identify Notch signaling as a therapeutic target to mitigate disease progression.
Collapse
Affiliation(s)
- Peilin Meng
- Northwest Women's and Children's Hospital, Xi'an, 710061, People's Republic of China
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
- Key Laboratory of Environment and Endemic Diseases, National Health Commission of China, and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region in Shaanxi, Xi'an, 710061, People's Republic of China
| | - Li Liu
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
- Key Laboratory of Environment and Endemic Diseases, National Health Commission of China, and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region in Shaanxi, Xi'an, 710061, People's Republic of China
| | - Ning Jiang
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
- Key Laboratory of Environment and Endemic Diseases, National Health Commission of China, and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region in Shaanxi, Xi'an, 710061, People's Republic of China
| | - Shuichu Hao
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Cong Yao
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
- Nursing Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Huan Liu
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
- Key Laboratory of Environment and Endemic Diseases, National Health Commission of China, and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region in Shaanxi, Xi'an, 710061, People's Republic of China
| | - Yingang Zhang
- Department of Orthopaedics of the First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Feng Zhang
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
- Key Laboratory of Environment and Endemic Diseases, National Health Commission of China, and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region in Shaanxi, Xi'an, 710061, People's Republic of China
| | - Yumeng Jia
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
- Key Laboratory of Environment and Endemic Diseases, National Health Commission of China, and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region in Shaanxi, Xi'an, 710061, People's Republic of China.
| | - Xiong Guo
- Institute of Endemic Diseases, School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
- Key Laboratory of Environment and Endemic Diseases, National Health Commission of China, and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region in Shaanxi, Xi'an, 710061, People's Republic of China.
| |
Collapse
|
3
|
Zhao F, Jia Z, Zhang L, Liu G, Li J, Zhao J, Xie Y, Chen L, Jiang H, He W, Wang A, Peng J, Zheng Y. A MnO 2 nanosheets doping double crosslinked hydrogel for cartilage defect repair through alleviating inflammation and guiding chondrogenic differentiation. Biomaterials 2025; 314:122875. [PMID: 39454507 DOI: 10.1016/j.biomaterials.2024.122875] [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: 06/07/2024] [Revised: 08/29/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024]
Abstract
The inflammatory microenvironment and inferior chondrogenesis are major symptoms after cartilage defect. Although various modifications strategies associated with hydrogels exhibit remarkable capacity of pro-cartilage regeneration, the adverse effect by prolonging inflammation is still formidable to hamper potential biomedical applications of different hydrogel implants. Herein, inspired by the repair microenvironment of articular cartilage defects, an injectable, immunomodulatory, and chondrogenic L-MNS-CMDA hydrogel is prepared through grafting vinyl and catechol groups to chitosan macromolecules using amide reaction, then further loading MnO2 nanosheets (MNS). The double crosslinking of photopolymerization and catechol oxidative polymerization endows L-MNS-CMDA hydrogel with preferable mechanical property, affording a suitable mechanical support for cartilage defect repair. Additionally, the robust tissue adhesion capability stemming from catechol groups guarantees the long-term retention of the hydrogel in the defect site. Meanwhile, L-MNS-CMDA hydrogel decomposes exogenous and intracellular H2O2 into O2 and H2O, to effectively alleviate cellular oxidative stress caused by long-term hypoxia. Under the synergies of catechol groups and MNS, L-MNS-CMDA hydrogel not only inhibits macrophages polarizing into M1 phenotype, but encourages them turn into M2 phenotype, thereby, reconstructing an immunization friendly microenvironment to ultimately enhance cartilage regeneration. Predictably, the hydrogel markedly induces rat bone marrow mesenchymal stem cells differentiating into chondrocytes by expressing abundant glycosaminoglycan and type II collagen. A cartilage defect model of rat knee joint indicates that L-MNS-CMDA hydrogel visually regulate the early inflammatory response of post-implantation, and facilitate cartilage regeneration and recovery of joint function after 12 weeks of post-implantation. All in all, this multifunctional L-MNS-CMDA hydrogel exhibits superior immunomodulatory and chondrogenic properties, holding immense clinical potential in the treatment of cartilage defects.
Collapse
Affiliation(s)
- Feilong Zhao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Zhibo Jia
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Liyang Zhang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Guodong Liu
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Junfei Li
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Jianming Zhao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Yajie Xie
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Lu Chen
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Hongyu Jiang
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei He
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Aiyuan Wang
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yudong Zheng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| |
Collapse
|
4
|
Dilawar M, Yu X, Jin Y, Yang J, Lin S, Liao J, Dai Q, Zhang X, Nisar MF, Chen G. Notch signaling pathway in osteogenesis, bone development, metabolism, and diseases. FASEB J 2025; 39:e70417. [PMID: 39985304 DOI: 10.1096/fj.202402545r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/18/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
The skeletal system provides vital importance to support organ development and functions. The Notch signaling pathway possesses well-established functions in organ development and cellular homeostasis. The Notch signaling pathway comprises five typical ligands (JAG1, JAG2, DLL1, DLL3, and DLL4), four receptors (Notch1-4), and four intracellular domains (NICD1-4). Each component of the Notch signaling pathway has been demonstrated to be fundamental in osteoblast differentiation and bone formation. The dysregulation in the Notch signaling pathway is highly linked with skeletal disorders or diseases at the developmental and postnatal stages. Recent studies have highlighted the importance of the elements of the Notch signaling pathway in the skeletal system, as well as its interaction with signaling, such as Wnt/β-catenin, BMP, TGF-β, FGF, autophagy, and hedgehog (Hh) to construct a potential gene regulatory network to orchestrate osteogenesis and ossification. Our review has provided a comprehensive summary of the Notch signaling pathway in the skeletal system, as well as the insights targeting Notch signaling for innovative potential drug discovery targets or therapeutic interventions to treat bone disorders, such as osteoporosis and osteoarthritis. An in-depth molecular mechanistic strategy to modulate the Notch signaling pathway and its associated signaling pathway will be encouraged for consideration to trigger enhanced therapeutic approaches for bone disorders by defining Notch-regulating drugs for clinical use.
Collapse
Affiliation(s)
- Muhammad Dilawar
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xuan Yu
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuanyuan Jin
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jing Yang
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Sisi Lin
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Junguang Liao
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qi Dai
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Muhammad Farrukh Nisar
- Department of Physiology & Biochemistry, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
- Ministry of Education and Jiangxi Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Jiangxi Agricultural University, Nanchang, China
| | - Guiqian Chen
- Department of Biopharmaceutics, Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
5
|
Canalis E, Schilling L, Denker E. TNFα has differential effects on the transcriptome profile of selected populations in murine cartilage. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100528. [PMID: 39494399 PMCID: PMC11530803 DOI: 10.1016/j.ocarto.2024.100528] [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: 07/30/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
Objective To further our understanding of the role of tumor necrosis factor (TNF)α on the inflammatory response in chondrocytes. Design We explored the effects of TNFα on the transcriptome of epiphyseal chondrocytes from newborn C57BL/6 mice at the total and single cell (sc) resolution. Results Gene set enrichment analysis of total RNA-Seq from TNFα-treated chondrocytes revealed enhanced response to biotic stimulus, defense and immune response and cytokine signaling and suppressed cartilage and skeletal morphogenesis and development. scRNA-Seq analyzed 14,239 cells and 24,320 genes and distinguished 16 cell clusters. The more prevalent ones were constituted by limb bud and chondrogenic cells and fibroblasts comprising ∼73 % of the cell population. Genes expressed by joint fibroblasts were detected in 5 clusters comprising ∼45 % of the cells isolated. Pseudotime trajectory finding revealed an association between fibroblast and chondrogenic clusters which was not modified by TNFα. TNFα decreased the total cells recovered by 18.5 % and the chondrogenic, limb bud and mesenchymal clusters by 32 %, 27 % and 7 %, respectively. TNFα had profound effects on the insulin-like growth factor (IGF) axis decreasing Igf1, Igf2 and Igfbp4 and inducing Igfbp3 and Igfbp5, explaining an inhibition of collagen biosynthesis, cartilage and skeletal morphogenesis. Ingenuity Pathway Analysis of scRNA-Seq data revealed that TNFα enhanced the osteoarthritis, rheumatoid arthritis, pathogen induced cytokine storm and interleukin 6 signaling pathways and suppressed fibroblast growth factor signaling. Conclusions Epiphyseal chondrocytes are constituted by diverse cell populations distinctly regulated by TNFα to promote inflammation and suppression of matrix biosynthesis and growth.
Collapse
Affiliation(s)
- Ernesto Canalis
- Departments of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA
- Departments of Medicine, UConn Health, Farmington, CT 06030, USA
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, USA
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, USA
| | - Emily Denker
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
6
|
Meng P, Liu H, Liu L, Wen Y, Zhang F, Zhang Y, Jia Y, Zhang Y, Zhang F, Guo X. Activation of Notch Signaling Pathway is involved in Extracellular Matrix Degradation in human induced pluripotent stem cells chondrocytes induced by HT-2 toxin. Food Chem Toxicol 2024; 189:114724. [PMID: 38734200 DOI: 10.1016/j.fct.2024.114724] [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: 03/27/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Notch signaling regulates cartilage formation and homeostasis. Kashin-Beck Disease (KBD), an endemic osteochondropathy, is characterized by severe cartilage degradation. The etiology of KBD is related to the exposure of HT-2 toxin, a mycotoxin and primary metabolite of T-2 toxin. This study aims to explore the role of HT-2 toxin in the Notch signaling regulation and extracellular matrix (ECM) metabolism of hiPSCs-Chondrocytes. Immunohistochemistry and qRT-PCR were employed to investigate the expression of Notch pathway molecules in KBD articular cartilage and primary chondrocytes. hiPSCs-Chondrocytes, derived from hiPSCs, were treated with 100 ng/mL HT-2 toxin and the γ-secretase inhibitor (DAPT) for 48h, respectively. The markers related to the Notch signaling pathway and ECM were assessed using qRT-PCR and Western blot. Notch pathway dysregulation was prominent in KBD cartilage. HT-2 toxin exposure caused cytotoxicity in hiPSCs-Chondrocytes, and activated Notch signaling by increasing the mRNA and protein levels of NOTCH1 and HES1. HT-2 toxin also upregulated ECM catabolic enzymes and downregulated ECM components (COL2A1 and ACAN), indicating ECM degradation. DAPT-mediated Notch signaling inhibition suppressed the mRNA and protein level of ADAMTS5 expression while enhancing ECM component expression in hiPSCs-Chondrocytes. This study suggests that HT-2 toxin may induce ECM degradation in hiPSCs-Chondrocytes through activating Notch signaling.
Collapse
Affiliation(s)
- Peilin Meng
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China
| | - Huan Liu
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China
| | - Li Liu
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China
| | - Yan Wen
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China
| | - Feng'e Zhang
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China
| | - Yanan Zhang
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China; School of Nursing, Lanzhou University, Lanzhou, 730000, PR China
| | - Yumeng Jia
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China
| | - Yingang Zhang
- Department of Orthopaedics of the First Affiliated Hospital, Medical School, Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Feng Zhang
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China.
| | - Xiong Guo
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, 710061, PR China; Clinical Research Center for Endemic Disease of Shaanxi Province, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, PR China.
| |
Collapse
|
7
|
Canalis E, Yu J, Singh V, Mocarska M, Schilling L. NOTCH2 sensitizes the chondrocyte to the inflammatory response of tumor necrosis factor α. J Biol Chem 2023; 299:105372. [PMID: 37865314 PMCID: PMC10692730 DOI: 10.1016/j.jbc.2023.105372] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/06/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023] Open
Abstract
Notch regulates the immune and inflammatory response and has been associated with the pathogenesis of osteoarthritis in humans and preclinical models of the disease. Notch2tm1.1Ecan mice harbor a NOTCH2 gain-of-function and are sensitized to osteoarthritis, but the mechanisms have not been explored. We examined the effects of tumor necrosis factor α (TNFα) in chondrocytes from Notch2tm1.1Ecan mice and found that NOTCH2 enhanced the effect of TNFα on Il6 and Il1b expression. Similar results were obtained in cells from a conditional model of NOTCH2 gain-of-function, Notch22.1Ecan mice, and following the expression of the NOTCH2 intracellular domain in vitro. Recombination signal-binding protein for immunoglobulin Kappa J region partners with the NOTCH2 intracellular domain to activate transcription; in the absence of Notch signaling it inhibits transcription, and Rbpj inactivation in chondrocytes resulted in Il6 induction. Although TNFα induced IL6 to a greater extent in the context of NOTCH2 activation, there was a concomitant inhibition of Notch target genes Hes1, Hey1, Hey2, and Heyl. Electrophoretic mobility shift assay demonstrated displacement of recombination signal-binding protein for immunoglobulin Kappa J region from DNA binding sites by TNFα explaining the increased Il6 expression and the concomitant decrease in Notch target genes. NOTCH2 enhanced the effect of TNFα on NF-κB signaling, and RNA-Seq revealed increased expression of pathways associated with inflammation and the phagosome in NOTCH2 overexpressing cells in the absence and presence of TNFα. Collectively, NOTCH2 has important interactions with TNFα resulting in the enhanced expression of Il6 and inflammatory pathways in chondrocytes.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA; Department of Medicine, UConn Health, Farmington, Connecticut, USA; UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA.
| | - Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA; UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA
| | - Vijender Singh
- Computational Biology Core, Institute for System Genomics, UConn, Storrs, Connecticut, USA
| | - Magda Mocarska
- UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
8
|
Chen N, Wu RW, Lam Y, Chan WC, Chan D. Hypertrophic chondrocytes at the junction of musculoskeletal structures. Bone Rep 2023; 19:101698. [PMID: 37485234 PMCID: PMC10359737 DOI: 10.1016/j.bonr.2023.101698] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/12/2023] [Accepted: 07/01/2023] [Indexed: 07/25/2023] Open
Abstract
Hypertrophic chondrocytes are found at unique locations at the junction of skeletal tissues, cartilage growth plate, articular cartilage, enthesis and intervertebral discs. Their role in the skeleton is best understood in the process of endochondral ossification in development and bone fracture healing. Chondrocyte hypertrophy occurs in degenerative conditions such as osteoarthritis. Thus, the role of hypertrophic chondrocytes in skeletal biology and pathology is context dependent. This review will focus on hypertrophic chondrocytes in endochondral ossification, in which they exist in a transient state, but acting as a central regulator of differentiation, mineralization, vascularization and conversion to bone. The amazing journey of a chondrocyte from being entrapped in the extracellular matrix environment to becoming proliferative then hypertrophic will be discussed. Recent studies on the dynamic changes and plasticity of hypertrophic chondrocytes have provided new insights into how we view these cells, not as terminally differentiated but as cells that can dedifferentiate to more progenitor-like cells in a transition to osteoblasts and adipocytes, as well as a source of skeletal stem and progenitor cells residing in the bone marrow. This will provide a foundation for studies of hypertrophic chondrocytes at other skeletal sites in development, tissue maintenance, pathology and therapy.
Collapse
Affiliation(s)
- Ning Chen
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Robin W.H. Wu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Yan Lam
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Wilson C.W. Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen 518053, China
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
9
|
Wolfe PN, Stoker AM, Crist BD, Leary E, Bozynski CC, Cook JL. Evaluation of mechanistic serum and urine biomarkers for secondary osteoarthritis associated with developmental dysplasia of the hip. J Orthop 2023; 42:24-29. [PMID: 37601815 PMCID: PMC10435905 DOI: 10.1016/j.jor.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 08/22/2023] Open
Abstract
Objective Determine measurable differences for mechanistic urine and serum biomarkers in patients with developmental dysplasia of the hip (DDH) prior to, and following, secondary hip osteoarthritis (OA) when compared to controls. Design Urine and serum were collected from individuals with developmental dysplasia of the hip (n = 39), prior to (Pre-OA DDH, n = 32) and following diagnosis of secondary hip OA (Post-OA DDH, n = 7), age-matched Pre-OA controls (n = 35), and age-matched Post-OA controls (n = 12). Samples were analyzed for protein biomarkers with potential for differentiation of hip status through a Mann-Whitney U test with a Benjamini-Hochberg correction. Results Several interleukin and degradation related proteins were found to be differentially expressed when comparing DDH-related hip status prior to and following diagnosis of hip OA. In addition, MCP-1 and TIMP-1 were significantly different between younger and older patients in the control cohorts. Conclusion These results provide initial evidence for serum and urine protein biomarkers that define clinically relevant stages of symptomatic DDH and its progression to secondary hip osteoarthritis categorized by known mechanisms of disease. Level of evidence III.
Collapse
Affiliation(s)
- Preston N. Wolfe
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
- Arthrex, Inc, Naples, FL, United States
| | - Aaron M. Stoker
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| | - Brett D. Crist
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| | - Emily Leary
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| | - Chantelle C. Bozynski
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| | - James L. Cook
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| |
Collapse
|
10
|
Yigit S, Tekcan A, Inanir A, Nursal AF, Akkanat S, Tural E. Effect of IL-6 -174G/C and -572G/C variants on susceptibility to osteoarthritis in Turkish population. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 42:65-76. [PMID: 35924736 DOI: 10.1080/15257770.2022.2107219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Osteoarthritis (OA) is a complex disorder characterized by degenerative articular cartilage in which inflammatory mechanisms play a major role in the pathogenesis. Interleukin-6 (IL6), a multifunctional cytokine, can trigger osteoclast differentiation and bone resorption. Our purpose in this study was to evaluate the association of IL-6 -174 G/C (rs1800795) and -572 G/C (rs1800796) variants with the susceptibility to OA. One hundred fifty OA patients and 150 healthy individuals were enrolled in the study. Polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) was used for genotyping the IL-6 gene variants. The results of analyses were evaluated for statistical significance. The pain intensity was assessed using the Visual Analogue Scale (VAS). There was a statistically significant difference in the genotype and allele frequencies of the IL-6 -174 G/C variant between patients with OA and control groups (p = 0.001, p = 0.002, respectively). IL-6 -174 G/C GG genotype and G allele were more prevalent in patients with OA. We found that the IL-6 -572 G/C variant was not different between patients and controls in either genotype distribution and allele frequency. IL-6 174 G/C and -572 G/C loci GG-GG combined genotype was significantly higher in OA patients (p = 0.00). Our study suggests that there was a strong association between the IL-6 -174 G/C variant and OA in the Turkish population. Further studies on populations of different ethnic background are necessary to prove the association of IL-6 variants with OA.
Collapse
Affiliation(s)
- Serbulent Yigit
- Faculty of Veterinary, Department of Genetics, Ondokuz Mayıs University, Samsun, Turkey.,Faculty of Medicine, Department of Medical Biology, Gaziosmanpasa University, Tokat, Turkey
| | - Akin Tekcan
- Faculty of Medicine, Departments of Medical Biology, Amasya University, Amasya, Turkey
| | - Ahmet Inanir
- Dr. Ahmet Inanir Clinic, Kecioren, Ankara, Turkey
| | - Ayse Feyda Nursal
- Faculty of Medicine, Departments of Medical Genetics, Hitit University, Corum, Turkey
| | - Songul Akkanat
- Faculty of Medicine, Department of Medical Biology, Gaziosmanpasa University, Tokat, Turkey
| | - Ercan Tural
- Faculty of Health Sciences, Department of Physiotherapy and Rehabilitation, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
11
|
Li M, Yin H, Yan Z, Li H, Wu J, Wang Y, Wei F, Tian G, Ning C, Li H, Gao C, Fu L, Jiang S, Chen M, Sui X, Liu S, Chen Z, Guo Q. The immune microenvironment in cartilage injury and repair. Acta Biomater 2022; 140:23-42. [PMID: 34896634 DOI: 10.1016/j.actbio.2021.12.006] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023]
Abstract
The ability of articular cartilage to repair itself is limited because it lacks blood vessels, nerves, and lymph tissue. Once damaged, it can lead to joint swelling and pain, accelerating the progression of osteoarthritis. To date, complete regeneration of hyaline cartilage exhibiting mechanical properties remains an elusive goal, despite the many available technologies. The inflammatory milieu created by cartilage damage is critical for chondrocyte death and hypertrophy, extracellular matrix breakdown, ectopic bone formation, and progression of cartilage injury to osteoarthritis. In the inflammatory microenvironment, mesenchymal stem cells (MSCs) undergo aberrant differentiation, and chondrocytes begin to convert or dedifferentiate into cells with a fibroblast phenotype, thereby resulting in fibrocartilage with poor mechanical qualities. All these factors suggest that inflammatory problems may be a major stumbling block to cartilage repair. To produce a milieu conducive to cartilage repair, multi-dimensional management of the joint inflammatory microenvironment in place and time is required. Therefore, this calls for elucidation of the immune microenvironment of cartilage repair after injury. This review provides a brief overview of: (1) the pathogenesis of cartilage injury; (2) immune cells in cartilage injury and repair; (3) effects of inflammatory cytokines on cartilage repair; (4) clinical strategies for treating cartilage defects; and (5) strategies for targeted immunoregulation in cartilage repair. STATEMENT OF SIGNIFICANCE: Immune response is increasingly considered the key factor affecting cartilage repair. It has both negative and positive regulatory effects on the process of regeneration and repair. Proinflammatory factors are secreted in large numbers, and necrotic cartilage is removed. During the repair period, immune cells can secrete anti-inflammatory factors and chondrogenic cytokines, which can inhibit inflammation and promote cartilage repair. However, inflammatory factors persist, which accelerate the degradation of the cartilage matrix. Furthermore, in an inflammatory microenvironment, MSCs undergo abnormal differentiation, and chondrocytes begin to transform or dedifferentiate into fibroblast-like cells, forming fibrocartilage with poor mechanical properties. Consequently, cartilage regeneration requires multi-dimensional regulation of the joint inflammatory microenvironment in space and time to make it conducive to cartilage regeneration.
Collapse
|
12
|
Minguzzi M, Panichi V, D’Adamo S, Cetrullo S, Cattini L, Flamigni F, Mariani E, Borzì RM. Pleiotropic Roles of NOTCH1 Signaling in the Loss of Maturational Arrest of Human Osteoarthritic Chondrocytes. Int J Mol Sci 2021; 22:ijms222112012. [PMID: 34769441 PMCID: PMC8585104 DOI: 10.3390/ijms222112012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 02/07/2023] Open
Abstract
Notch signaling has been identified as a critical regulator of cartilage development and homeostasis. Its pivotal role was established by both several joint specific Notch signaling loss of function mouse models and transient or sustained overexpression. NOTCH1 is the most abundantly expressed NOTCH receptors in normal cartilage and its expression increases in osteoarthritis (OA), when chondrocytes exit from their healthy “maturation arrested state” and resume their natural route of proliferation, hypertrophy, and terminal differentiation. The latter are hallmarks of OA that are easily evaluated in vitro in 2-D or 3-D culture models. The aim of our study was to investigate the effect of NOTCH1 knockdown on proliferation (cell count and Picogreen mediated DNA quantification), cell cycle (flow cytometry), hypertrophy (gene and protein expression of key markers such as RUNX2 and MMP-13), and terminal differentiation (viability measured in 3-D cultures by luminescence assay) of human OA chondrocytes. NOTCH1 silencing of OA chondrocytes yielded a healthier phenotype in both 2-D (reduced proliferation) and 3-D with evidence of decreased hypertrophy (reduced expression of RUNX2 and MMP-13) and terminal differentiation (increased viability). This demonstrates that NOTCH1 is a convenient therapeutic target to attenuate OA progression.
Collapse
Affiliation(s)
- Manuela Minguzzi
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy; (M.M.); (S.D.); (E.M.)
| | - Veronica Panichi
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40138 Bologna, Italy; (V.P.); (S.C.); (F.F.)
| | - Stefania D’Adamo
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy; (M.M.); (S.D.); (E.M.)
| | - Silvia Cetrullo
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40138 Bologna, Italy; (V.P.); (S.C.); (F.F.)
| | - Luca Cattini
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Flavio Flamigni
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40138 Bologna, Italy; (V.P.); (S.C.); (F.F.)
| | - Erminia Mariani
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy; (M.M.); (S.D.); (E.M.)
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Rosa Maria Borzì
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
- Correspondence:
| |
Collapse
|
13
|
Liu WB, Feng QJ, Li GS, Shen P, Li YN, Zhang FJ. Long non-coding RNA HOTAIRM1-1 silencing in cartilage tissue induces osteoarthritis through microRNA-125b. Exp Ther Med 2021; 22:933. [PMID: 34306202 PMCID: PMC8280714 DOI: 10.3892/etm.2021.10365] [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: 09/04/2020] [Accepted: 03/11/2021] [Indexed: 11/29/2022] Open
Abstract
Aberrations in long noncoding RNA (lncRNA) expression have been recognized in numerous human diseases. In the present study, the of role the long noncoding RNA HOX antisense intergenic RNA myeloid 1 variant (HOTAIRM1-1) in regulating the pathological progression of osteoarthritis (OA) was investigated. The aberrant expression of HOTAIRM1-1 in OA was demonstrated, but the molecular mechanisms require further analysis. The aim of the present study was to explore the function of miR-125b in modulating chondrocyte viability and apoptosis, and to address the functional association between HOTAIRM1-1 and miR-125b as potential targets. A miR-125b inhibitor was used, which laid the foundation for the following investigation. The study confirmed that HOTAIRM1-1 and miR-125b are inversely expressed in chondrocytes. The expression of HOTAIRM1-1 was downregulated and the expression of miR-125b was upregulated in tissues from patients with OA. HOTAIRM1-1 directly interacted with miR-125b in chondrocytes. HOTAIRM1-1 knockdown was associated with chondrocyte proliferation and extracellular matrix degradation. Furthermore, miR-125b reversed the effect of HOTAIRM1-1 on cell proliferation and apoptosis. In conclusion, the present study indicates that the loss of HOTAIRM1-1 function leads to aberrant increases in the proliferation and apoptosis of chondrocytes. miR-125b may be a potential downstream mechanism that regulates the function of HOTAIRM1-1, and this finding provides a therapeutic strategy for OA.
Collapse
Affiliation(s)
- Wen-Bin Liu
- Department of Joint Surgery, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Qi-Jin Feng
- Department of Orthopedics, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300150, P.R. China
| | - Gui-Shi Li
- Department of Joint Surgery, Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Peng Shen
- Department of Rheumatology and Immunology, Tianjin First Center Hospital, Tianjin 300192, P.R. China
| | - Ya-Nan Li
- Department of Orthopedics, Tianjin Dongli Hospital, Tianjin 300300, P.R. China
| | - Fu-Jiang Zhang
- Department of Joint Surgery, Tianjin Hospital, Tianjin 300211, P.R. China
| |
Collapse
|
14
|
Wu YT, Tang MX, Wang YJ, Li J, Wang YX, Deng A, Guo CF, Zhang HQ. Lower androgen levels promote abnormal cartilage development in female patients with adolescent idiopathic scoliosis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:784. [PMID: 34268397 PMCID: PMC8246169 DOI: 10.21037/atm-20-3171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 01/25/2021] [Indexed: 11/14/2022]
Abstract
Background Adolescent idiopathic scoliosis (AIS) is a disease characterized by changes in the three-dimensional structure of the spine. Studies have shown that the development of AIS might be associated with genetic, biomechanics, endocrine factors and abnormal bone or cartilage development. Methods Blood samples collected from 301 female patients (161 females with AIS and 140 females without AIS) were used for genotyping. Forty-eight serum samples from 161 females with AIS and 40 serum samples from 140 females without AIS were subjected to enzyme-linked immunosorbent assays (ELISAs). We also evaluated 32 facet joints (18 females with AIS and 14 females without AIS from the 301 female patients) using immunohistochemistry, Western blotting, and isolation of human primary chondrocytes, among other methods. We treated the AIS primary chondrocytes with dihydrotestosterone (DHT) to verify the relationship among androgen, the androgen receptor (AR), and its downstream pathway proteins. Results The serum androgen level in the AIS group was significantly decreased (1.94±0.09 vs. 2.284±0.103) compared with that in the non-AIS (control) group. The single nucleotide polymorphism genotyping results showed that the mutation rates of rs6259 between the AIS and control groups were significantly different (G/G genotype: 48.4% vs. 42.1%, G/A genotype: 40.4% vs. 35.7%, P<0.05). The levels of interleukin (IL)-6 and metalloproteinase (MMP)-13 were increased in the cartilage of AIS patients, and these patients also exhibited decreased AR levels. The cell experiment results showed that androgen reduced the degree of abnormal cartilage development in female AIS patients through the AR/IL-6/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Conclusions Our study provides a new perspective on the pathogenesis of AIS and indicates that decreased androgen levels in female AIS patients play a potential role in the development of AIS via the AR/IL-6/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yuan-Tao Wu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Spine Surgery, Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ming-Xing Tang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yun-Jia Wang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jiong Li
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Xiang Wang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ang Deng
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chao-Feng Guo
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong-Qi Zhang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Wiegertjes R, van de Loo FAJ, Blaney Davidson EN. A roadmap to target interleukin-6 in osteoarthritis. Rheumatology (Oxford) 2021; 59:2681-2694. [PMID: 32691066 PMCID: PMC7516110 DOI: 10.1093/rheumatology/keaa248] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/26/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023] Open
Abstract
Joint inflammation is present in the majority of OA patients and pro-inflammatory mediators, such as IL-6, are actively involved in disease progression. Increased levels of IL-6 in serum or synovial fluid from OA patients correlate with disease incidence and severity, with IL-6 playing a pivotal role in the development of cartilage pathology, e.g. via induction of matrix-degrading enzymes. However, IL-6 also increases expression of anti-catabolic factors, suggesting a protective role. Until now, this dual role of IL-6 is incompletely understood and may be caused by differential effects of IL-6 classic vs trans-signalling. Here, we review current evidence regarding the role of IL-6 classic- and trans-signalling in local joint pathology of cartilage, synovium and bone. Furthermore, we discuss targeting of IL-6 in experimental OA models and provide future perspective for OA treatment by evaluating currently available IL-6 targeting strategies.
Collapse
Affiliation(s)
- Renske Wiegertjes
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fons A J van de Loo
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esmeralda N Blaney Davidson
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Lo Monaco M, Gervois P, Beaumont J, Clegg P, Bronckaers A, Vandeweerd JM, Lambrichts I. Therapeutic Potential of Dental Pulp Stem Cells and Leukocyte- and Platelet-Rich Fibrin for Osteoarthritis. Cells 2020; 9:cells9040980. [PMID: 32326610 PMCID: PMC7227024 DOI: 10.3390/cells9040980] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/07/2020] [Accepted: 04/13/2020] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative and inflammatory joint disorder with cartilage loss. Dental pulp stem cells (DPSCs) can undergo chondrogenic differentiation and secrete growth factors associated with tissue repair and immunomodulation. Leukocyte- and platelet-rich fibrin (L-PRF) emerges in regenerative medicine because of its growth factor content and fibrin matrix. This study evaluates the therapeutic application of DPSCs and L-PRF in OA via immunomodulation and cartilage regeneration. Chondrogenic differentiation of DPSCs, with or without L-PRF exudate (ex) and conditioned medium (CM), and of bone marrow-mesenchymal stem cells was compared. These cells showed differential chondrogenesis. L-PRF was unable to increase cartilage-associated components. Immature murine articular chondrocytes (iMACs) were cultured with L-PRF ex, L-PRF CM, or DPSC CM. L-PRF CM had pro-survival and proliferative effects on unstimulated and cytokine-stimulated iMACs. L-PRF CM stimulated the release of IL-6 and PGE2, and increased MMP-13, TIMP-1 and IL-6 mRNA levels in cytokine-stimulated iMACs. DPSC CM increased the survival and proliferation of unstimulated iMACs. In cytokine-stimulated iMACs, DPSC CM increased TIMP-1 gene expression, whereas it inhibited nitrite release in 3D culture. We showed promising effects of DPSCs in an in vitro OA model, as they undergo chondrogenesis in vitro, stimulate the survival of chondrocytes and have immunomodulatory effects.
Collapse
Affiliation(s)
- Melissa Lo Monaco
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
- Department of Veterinary Medicine, Integrated Veterinary Research Unit (IVRU) - Namur Research Institute for Life Science (NARILIS), University of Namur, 5000 Namur, Belgium;
- Correspondence: ; Tel.: +32-(0)-26-92-09
| | - Pascal Gervois
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
| | - Joel Beaumont
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
- Maastricht Radiation Oncology (MaastRO) Lab, GROW—School for Oncology and Developmental Biology, Maastricht University, 6229ER Maastricht, The Netherlands
| | - Peter Clegg
- Department of Musculoskeletal and Ageing Sciences, Institute of Lifecourse and Medical Sciences, University of Liverpool, L7 8TX Liverpool, UK;
| | - Annelies Bronckaers
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
| | - Jean-Michel Vandeweerd
- Department of Veterinary Medicine, Integrated Veterinary Research Unit (IVRU) - Namur Research Institute for Life Science (NARILIS), University of Namur, 5000 Namur, Belgium;
| | - Ivo Lambrichts
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
| |
Collapse
|
17
|
Abstract
PURPOSE OF THE REVIEW Osteoarthritis (OA) is an aging-associated and injury-induced joint disease characterized by cartilage degradation, bone sclerosis, and persistent low-grade inflammation in the joint. Aging and injury are triggers of joint pathological changes mediated by pro-inflammatory factors, some of which are secreted by white adipose tissue. Adipokines including adiponectin, leptin, resistin, chemerin, IL-6, and TNF-α are major players not only during inflammation but also in metabolic regulation of joint cells including chondrocytes, osteoblasts, osteoclasts as well as mesenchymal stem cells. The purpose of this review is to summarize the signal transduction pathways of adipokines in the articular joint to provide new information on potential targets for intervention of OA. RECENT FINDINGS The risk of knee osteoarthritis is associated with adipokine gene polymorphism. While the infrapatellar fat pad is a major source of adipokines in knee synovial fluid, adipocytes also accumulate in the bone marrow during aging and obesity. Adipokines can act as SASPs (senescence associated secretory phenotype factors) that participate in cellular senescence of chondrocytes, but they also regulate energy metabolism impacting bone remodeling. Thus, adipokines are closely related to the metabolic syndrome and degenerative pathological changes in cartilage and bone during OA. Modulating the effects of adipokines on different cell types in the intra-articular joint will be a promising new option for OA intervention.
Collapse
Affiliation(s)
- Chenxi Xie
- Bone and Joint Research Center, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qian Chen
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, 02903, USA.
| |
Collapse
|
18
|
Charlier E, Deroyer C, Ciregia F, Malaise O, Neuville S, Plener Z, Malaise M, de Seny D. Chondrocyte dedifferentiation and osteoarthritis (OA). Biochem Pharmacol 2019; 165:49-65. [PMID: 30853397 DOI: 10.1016/j.bcp.2019.02.036] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
|
19
|
Zhang T, Yao Y. Effects of inflammatory cytokines on bone/cartilage repair. J Cell Biochem 2019; 120:6841-6850. [PMID: 30335899 DOI: 10.1002/jcb.27953] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/02/2018] [Indexed: 01/24/2023]
Abstract
Many inflammatory factors can affect cell behaviors and work as a form of inter-regulatory networks through the inflammatory pathway. Inflammatory cytokines are critical for triggering bone regeneration after fracture or bone injury. Also, inflammatory cytokines play an important role in cartilage repair. The synergistic or antagonistic effects of both proinflammatory and anti-inflammatory cytokines have a great influence on fracture healing. This review discusses key inflammatory cytokines and signaling pathways involved in bone or cartilage repair.
Collapse
Affiliation(s)
- Tingshuai Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
20
|
Zanotti S, Yu J, Bridgewater D, Wolf JM, Canalis E. Mice harboring a Hajdu Cheney Syndrome mutation are sensitized to osteoarthritis. Bone 2018; 114:198-205. [PMID: 29940267 PMCID: PMC6083868 DOI: 10.1016/j.bone.2018.06.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/01/2018] [Accepted: 06/20/2018] [Indexed: 11/27/2022]
Abstract
Osteoarthritis is a joint disease characterized by cartilage degradation, altered gene expression and inflammation. NOTCH1 and NOTCH2 receptors and the JAGGED1 ligand regulate chondrocyte biology; however, the contribution of Notch signaling to osteoarthritis is controversial. Hajdu Cheney Syndrome (HCS) is a rare genetic disorder affecting the skeleton and associated with NOTCH2 mutations that lead to NOTCH2 gain-of-function. A murine model of the disease (Notch2tm1.1Ecan) was used to test whether the HCS mutation increases the susceptibility to osteoarthritis. The knee of three-month-old Notch2tm1.1Ecan male mice and control sex-matched littermates was destabilized by resection of the medial meniscotibial ligament, and changes in the joint analyzed two months thereafter. Expression of Notch target genes was increased in the femoral heads of Notch2tm1.1Ecan mice, documenting Notch signal activation. Periarticular bone and cartilage structures were unaffected in Notch2tm1.1Ecan mutants subjected to sham surgery, indicating that NOTCH2 gain-of-function had no discernible impact on joint structure under basal conditions. However, destabilization of the medial meniscus increased osteophyte volume and thickened subchondral bone in Notch2tm1.1Ecan mice compared to wild type littermates. Moreover, destabilized Notch2tm1.1Ecan mutants exhibited histological signs of moderate to severe cartilage degeneration, demonstrating joint sensitization to the development of osteoarthritis. Chondrocyte cultures from Notch2tm1.1Ecan mutants expressed increased Il6 mRNA levels following exposure to JAGGED1, possibly explaining the susceptibility of Notch2tm1.1Ecan mice to osteoarthritis. In conclusion, Notch2tm1.1Ecan mutants are sensitized to the development of osteoarthritis in destabilized joints and NOTCH2 activation may play a role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- S Zanotti
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, United States of America; Department of Medicine, UConn Health, Farmington, CT 06030, United States of America; UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, United States of America
| | - J Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, United States of America; UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, United States of America
| | - D Bridgewater
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, United States of America
| | - J M Wolf
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, United States of America; UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, United States of America
| | - E Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, United States of America; Department of Medicine, UConn Health, Farmington, CT 06030, United States of America; UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, United States of America.
| |
Collapse
|
21
|
Meo Burt P, Xiao L, Hurley MM. FGF23 Regulates Wnt/β-Catenin Signaling-Mediated Osteoarthritis in Mice Overexpressing High-Molecular-Weight FGF2. Endocrinology 2018; 159:2386-2396. [PMID: 29718273 PMCID: PMC6457004 DOI: 10.1210/en.2018-00184] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/21/2018] [Indexed: 12/23/2022]
Abstract
Although humans with X-linked hypophosphatemia (XLH) and the Hyp mouse, a murine homolog of XLH, are known to develop degenerative joint disease, the exact mechanism that drives the osteoarthritis (OA) phenotype remains unclear. Mice that overexpress high-molecular-weight fibroblast growth factor (FGF) 2 isoforms (HMWTg mice) phenocopy both XLH and Hyp, including OA with increased FGF23 production in bone and serum. Because HMWTg cartilage also has increased FGF23 and there is cross-talk between FGF23-Wnt/β-catenin signaling, the purpose of this study was to determine if OA observed in HMWTg mice is due to FGF23-mediated canonical Wnt signaling in chondrocytes, given that both pathways are implicated in OA pathogenesis. HMWTg OA joints had decreased Dkk1, Sost, and Lrp6 expression with increased Wnt5a, Wnt7b, Lrp5, Axin2, phospho-GSK3β, Lef1, and nuclear β-catenin, as indicated by immunohistochemistry or quantitative PCR analysis. Chondrocytes from HMWTg mice had enhanced alcian blue and alkaline phosphatase staining as well as increased FGF23, Adamts5, Il-1β, Wnt7b, Wnt16, and Wisp1 gene expression and phospho-GSK3β protein expression as indicated by Western blot, compared with chondrocytes of vector control and chondrocytes from mice overexpressing the low-molecular-weight isoform, which were protected from OA. Canonical Wnt inhibitor treatment rescued some of those parameters in HMWTg chondrocytes, seemingly delaying the initially accelerated chondrogenic differentiation. FGF23 neutralizing antibody treatment was able to partly ameliorate OA abnormalities in subchondral bone and reduce degradative/hypertrophic chondrogenic marker expression in HMWTg joints in vivo. These results demonstrate that osteoarthropathy of HMWTg is at least partially due to FGF23-modulated Wnt/β-catenin signaling in chondrocytes.
Collapse
Affiliation(s)
- Patience Meo Burt
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, UConn Health, Farmington, Connecticut
| | - Liping Xiao
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, UConn Health, Farmington, Connecticut
| | - Marja M Hurley
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine, UConn Health, Farmington, Connecticut
- Correspondence: Marja M. Hurley, MD, Department of Medicine MC-3023, UConn Health, 263 Farmington Avenue, Farmington, Connecticut 06030. E-mail:
| |
Collapse
|
22
|
Wyss JC, Kumar R, Mikulic J, Schneider M, Aebi JD, Juillerat-Jeanneret L, Golshayan D. Targeted γ-secretase inhibition of Notch signaling activation in acute renal injury. Am J Physiol Renal Physiol 2018; 314:F736-F746. [DOI: 10.1152/ajprenal.00414.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Notch pathway has been reported to control tissue damage in acute kidney diseases. To investigate potential beneficial nephroprotective effects of targeting Notch, we developed chemically functionalized γ-secretase inhibitors (GSIs) targeting γ-glutamyltranspeptidase (γ-GT) and/or γ-glutamylcyclotransferase (γ-GCT), two enzymes overexpressed in the injured kidney, and evaluated them in in vivo murine models of acute tubular and glomerular damage. Exposure of the animals to disease-inducing drugs together with the functionalized GSIs improved proteinuria and, to some extent, kidney dysfunction. The expression of genes involved in the Notch pathway, acute inflammatory stress responses, and the renin-angiotensin system was enhanced in injured kidneys, which could be downregulated upon administration of functionalized GSIs. Immunohistochemistry staining and Western blots demonstrated enhanced activation of Notch1 as detected by its cleaved active intracellular domain during acute kidney injury, and this was downregulated by concomitant treatment with the functionalized GSIs. Thus targeted γ-secretase-based prodrugs developed as substrates for γ-GT/γ-GCT have the potential to selectively control Notch activation in kidney diseases with subsequent regulation of the inflammatory stress response and the renin-angiotensin pathways.
Collapse
Affiliation(s)
- Jean-Christophe Wyss
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Rajesh Kumar
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Josip Mikulic
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Manfred Schneider
- Medicinal Chemistry, Roche Pharma Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche, Basel, Switzerland
| | - Johannes D. Aebi
- Medicinal Chemistry, Roche Pharma Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche, Basel, Switzerland
| | - Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- University Institute of Pathology, CHUV and UNIL, Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
23
|
Abstract
BACKGROUND The pathogenesis of osteoarthritis (OA) is not clear; leptin may be related to its pathogenesis. METHODS We reviewed articles on leptin in OA, chondrocytes, and in vitro experiments. It is concluded that leptin may lead to OA via some signaling pathways. At the same time, the concentration of leptin in vitro experiments and OA/rheumatoid arthritis (RA) patients was summarized. RESULTS Leptin levels in serum and synovial fluid of OA/RA patients were higher than normal person. In the condition of infection and immunity, serum leptin levels in the peripheral blood significantly increase. Because of the close relationship between obesity, leptin, and OA, it is crucial to study the effects of weight loss and exercise intervention on serum leptin levels to improve the symptoms of OA patients. CONCLUSION Treatment for leptin-increased obesity may be a treatment for OA. The role of leptin in OA cannot be ignored and needs to be further studied.
Collapse
Affiliation(s)
- Moqi Yan
- Department of Orthopedics, The First Affiliated Hospital of Soochow University
- Orthopedic Institute, Soochow University, Suzhou, China
| | - Junxin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University
- Orthopedic Institute, Soochow University, Suzhou, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University
- Orthopedic Institute, Soochow University, Suzhou, China
| | - Ye Sun
- Department of Orthopedics, The First Affiliated Hospital of Soochow University
- Orthopedic Institute, Soochow University, Suzhou, China
| |
Collapse
|
24
|
Zheng Y, Liu C, Ni L, Liu Z, Mirando AJ, Lin J, Saijilafu, Chen D, Hilton MJ, Li B, Chen J. Cell type-specific effects of Notch signaling activation on intervertebral discs: Implications for intervertebral disc degeneration. J Cell Physiol 2018; 233:5431-5440. [PMID: 30025440 DOI: 10.1002/jcp.26385] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/29/2017] [Indexed: 01/03/2023]
Abstract
Intervertebral disc (IVD) degeneration is the major cause of back pain. Notch signaling is activated in annulus fibrosus (AF) and nucleus pulposus (NP) tissues of degenerated IVDs, and induced by IL1-β and TNF-α in NP cells. However, the role of Notch activatin in the pathogenesis of IVD degeneration is largely unknown. In this study, we overexpressed the Notch1 intracellular domain (NICD1) in AF, NP, and chondrogenic ATDC5 cells via adenoviruses. Overexpression of NICD1 activated transcription of Notch signaling target genes in AF, NP, and ATDC5 cells, and caused cell type-specific effects on expression of matrix anabolic and catabolic genes. Activation of Notch signaling promoted expression of matrix catabolic genes and inhibited expression of matrix anabolic genes in both AF and ATDC5 cells, whereas its activation suppressed expression of matrix catabolic genes (including Mmp3, Mmp13, Adamts4, and Adamts5) and attenuated TNF-α and inflammatory macrophage-induced Mmp13 expression in NP cells. Consistently, sustained activation of Notch1 signaling in postnatal IVDs in mice severely disrupted growth plate and endplate cartilage tissues, but did not overly affect NP tissues. Together, these data indicated that activation of Notch signaling exerted differential and cell type-specific effects in intervertebral discs, and specific Notch signaling regulation may be considered during the treatment of IVD degeneration.
Collapse
Affiliation(s)
- Yixin Zheng
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Cunchang Liu
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Li Ni
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Zhaoyang Liu
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Anthony J Mirando
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina
| | - Jun Lin
- Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Saijilafu
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois
| | - Matthew J Hilton
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, North Carolina
| | - Bin Li
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Jianquan Chen
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.,Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
25
|
Young IC, Chuang ST, Gefen A, Kuo WT, Yang CT, Hsu CH, Lin FH. A novel compressive stress-based osteoarthritis-like chondrocyte system. Exp Biol Med (Maywood) 2017; 242:1062-1071. [PMID: 28492349 DOI: 10.1177/1535370217699534] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mechanical stress damage and insufficient self-repair can contribute to osteoarthritis (OA) in the affected joint. As the effects of stress on chondrocyte metabolism can regulate cartilage homeostasis, the specific stress-response condition is therefore a key to the generation of an OA disease model. We aimed to produce a specific stress- and cell-based OA model after evaluating the metabolic responses of chondrocytes in response to a series of static and cyclic compression stressors. A static load exceeding 40 psi initiated extracellular matrix (ECM) degradation through a decrease in the sulphated-glycosaminoglycan (GAG) content, upregulation of catabolic matrix metalloproteinase (MMP)-13 encoding gene expression, and downregulation of the ECM-related aggrecan and type II collagen encoding genes within 24 h. Indicators of pro-inflammatory events and oxidative stress were found to correlate with increased IL-6 expression and reactive oxygen species (ROS) production, respectively. However, chondrocytes stimulated by moderate cyclic loading (30-40 psi) exhibited increased ECM-related gene expression without significant changes in catabolic and pro-inflammatory gene expression. BMP-7 expression increased at cyclic loading levels above 30-60 psi. These results demonstrated that static compression exceeding 60 psi is sufficient to produce OA-like chondrocytes that exhibit signs of ECM degradation and inflammation. These OA-like chondrocytes could therefore be used as a novel cell-based drug screening system. Impact statement The lack of an effective treatment for osteoarthritis (OA) reflects the great need for alternative therapies and drug discovery. Disease models can be used for early-stage compound screening and disease studies. Chondrocytes are solely responsible for the maintenance of the articular cartilage extracellular matrix. Our strategy involved the generation of a cell-based model of OA, a more readily studied disease. Instead of using animal cartilage explants, we incorporated isolated porcine chondrocytes with hydrogel to form three-dimensional assemblies. We could identify the specific magnitude-dependent metabolic responses of chondrocytes by applying a series of static and cyclic compression, and therefore successfully generated a novel OA-like cell-based model for drug screening.
Collapse
Affiliation(s)
- In-Chi Young
- 1 Institute of Biomedical Engineering, National Taiwan University, Taipei 10672, Taiwan
| | - Sung-Ting Chuang
- 2 Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Amit Gefen
- 3 Department of Biomedical Engineering, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Wei-Ting Kuo
- 1 Institute of Biomedical Engineering, National Taiwan University, Taipei 10672, Taiwan
| | - Chun-Ting Yang
- 1 Institute of Biomedical Engineering, National Taiwan University, Taipei 10672, Taiwan
| | - Chia-Hsien Hsu
- 4 Institute of Biomedical Engineering and Nanomedicine, National Health Research Institute, Miaoli 35053, Taiwan
| | - Feng-Huei Lin
- 1 Institute of Biomedical Engineering, National Taiwan University, Taipei 10672, Taiwan
| |
Collapse
|
26
|
Young IC, Chuang ST, Hsu CH, Sun YJ, Liu HC, Chen YS, Lin FH. Protective effects of aucubin on osteoarthritic chondrocyte model induced by hydrogen peroxide and mechanical stimulus. Altern Ther Health Med 2017; 17:91. [PMID: 28153003 PMCID: PMC5288878 DOI: 10.1186/s12906-017-1581-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 01/13/2017] [Indexed: 01/06/2023]
Abstract
Background During the onset of osteoarthritis (OA), certain biochemical events have been shown to accelerate cartilage degradation, including the dysregulation of cartilage ECM anabolism, abnormal generation of reactive oxygen species (ROS) and overproduction of proteolytic enzymes and inflammatory cytokines. The potency of aucubin in protecting cellular components against oxidative stress, inflammation and apoptosis effects are well documented, which makes it a potential candidate for OA treatment. In this study, we aimed to evaluate the protective benefits of aucubin against OA using H2O2 and compression induced OA-like chondrocyte models. Methods The effects of aucubin were studied in porcine chondrocytes after 1 mM H2O2 stimulation for 30 min or sustained compression for 24 h. Effects of aucubin on cell proliferation and cytotoxicity of chondrocytes were measured with WST-1 and LDH assays. ROS production was evaluated by the Total ROS/Superoxide Detection Kit. Caspase-3 activity was evaluated by the CaspACE assay system. The levels of apoptosis were evaluated by the Annexin V-FITC apoptosis detection kit. OA-related gene expression was measured by reverse transcription quantitative polymerase chain reaction (RT-qPCR). Total DNA quantification was evaluated by the DNeasy Blood and Tissue kit. Sulfated-glycosaminoglycans (sGAGs) production and content were evaluated by DMMB assay and Alcian blue staining. Results The results showed that the ROS scavenge effects of aucubin appeared after 1 h of pretreatment. Aucubin could reduce the caspase-3 activity induced by H2O2, and reduced the apoptosis cell population in flowcytometry. In RT-qPCR results, aucubin could maintain ACAN and COL2A1 gene expressions, and prevent IL6 and MMP13 gene up-regulation induced by H2O2 and compression stimulations. In the DMMB assay and Alcian blue staining, aucubin could maintain the sGAG content and protect chondrocytes against compressive stress, but not oxidative stress from H2O2. Conclusions These results indicated that aucubin has protective effects in an osteoarthritic chondrocyte model induced by H2O2 and mechanical stimulus.
Collapse
|
27
|
Young IC, Chuang ST, Hsu CH, Sun YJ, Lin FH. C-phycocyanin alleviates osteoarthritic injury in chondrocytes stimulated with H 2 O 2 and compressive stress. Int J Biol Macromol 2016; 93:852-859. [DOI: 10.1016/j.ijbiomac.2016.09.051] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 01/08/2023]
|
28
|
Abstract
Notch 1 to 4 receptors are important determinants of cell fate and function, and Notch signaling plays an important role in skeletal development and bone remodeling. After direct interactions with ligands of the Jagged and Delta-like families, a series of cleavages release the Notch intracellular domain (NICD), which translocates to the nucleus where it induces transcription of Notch target genes. Classic gene targets of Notch are hairy and enhancer of split (Hes) and Hes-related with YRPW motif (Hey). In cells of the osteoblastic lineage, Notch activation inhibits cell differentiation and causes cancellous bone osteopenia because of impaired bone formation. In osteocytes, Notch1 has distinct effects that result in an inhibition of bone resorption secondary to an induction of osteoprotegerin and suppression of sclerostin with a consequent enhancement of Wnt signaling. Notch1 inhibits, whereas Notch2 enhances, osteoclastogenesis and bone resorption. Congenital disorders of loss- and gain-of-Notch function present with severe clinical manifestations, often affecting the skeleton. Enhanced Notch signaling is associated with osteosarcoma, and Notch can influence the invasive potential of carcinoma of the breast and prostate. Notch signaling can be controlled by the use of inhibitors of Notch activation, small peptides that interfere with the formation of a transcriptional complex, or antibodies to the extracellular domain of specific Notch receptors or to Notch ligands. In conclusion, Notch plays a critical role in skeletal development and homeostasis, and serious skeletal disorders can be attributed to alterations in Notch signaling.
Collapse
Affiliation(s)
- Stefano Zanotti
- Departments of Orthopaedic Surgery and Medicine and the UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030
| | - Ernesto Canalis
- Departments of Orthopaedic Surgery and Medicine and the UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030
| |
Collapse
|
29
|
Potential Application of Biological Products for the Treatment of Ocular Surface Inflammation. Cornea 2015; 34 Suppl 11:S153-7. [DOI: 10.1097/ico.0000000000000585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
30
|
Lu H, Zeng C, Chen M, Lian L, Dai Y, Zhao H. Lentiviral vector-mediated over-expression of Sox9 protected chondrocytes from IL-1β induced degeneration and apoptosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:10038-10049. [PMID: 26617711 PMCID: PMC4637526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/21/2015] [Indexed: 06/05/2023]
Abstract
To explore whether the over-expression of Sry-related HMG box (Sox9) in degenerative chondrocytes is able to improve cell regeneration and protects cells from inflammation induced apoptosis, we generated a Sox9 over-expressing vector delivery system in which the Sox9 gene was inserted into a lentiviral vector. After infecting mouse chondrocytes with the Sox9-encoding vector, we observed a high level of gene transduction efficiency and achieved a high level of Sox9 expression in the infected chondrocytes. To explore whether over-expression of Sox9 is able to induce cell regeneration and improve cell survival, we induced Sox9 over-expression by lentiviral vector infection 48 hours before IL-1β treatment. The cells were infected with the reporter gene GFP-encoded lentiviral vector as a negative control or left uninfected. 48-hours after IL-1β treatment, the chrondrocytes treated with IL-1β alone, underwent a degenerative process, with elevated expression of MMP-3, MMP-13, ADAMTS-5 and ALP, but the cell specific anabolic proteins collagen II and aggrecan were significantly suppressed. The cells infected with the GFP reporter vector had no increased regeneration after IL-1β treatment. The results indicated that Sox9 is an important chondrocyte transcription factor, promoting chondrocyte regeneration and cell survival, which were mediated through affecting multiple cell differentiation as well as anti-apoptotic signaling pathways.
Collapse
Affiliation(s)
- Huading Lu
- Department of Orthopedics, Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, P. R. China
| | - Chun Zeng
- Department of Joint Surgery, Third Affiliated Hospital of Southern Medical UniversityGuangzhou 510630, China
| | - Mingwei Chen
- Department of Orthopedics, Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, P. R. China
| | - Liyi Lian
- Department of Orthopedics, Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, P. R. China
| | - Yuhu Dai
- Department of Orthopedics, Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, P. R. China
| | - Huiqing Zhao
- Department of Orthopedics, Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, P. R. China
| |
Collapse
|
31
|
Liu Z, Chen J, Mirando AJ, Wang C, Zuscik MJ, O'Keefe RJ, Hilton MJ. A dual role for NOTCH signaling in joint cartilage maintenance and osteoarthritis. Sci Signal 2015. [PMID: 26198357 DOI: 10.1126/scisignal.aaa3792] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Loss of NOTCH signaling in postnatal murine joints results in osteoarthritis, indicating a requirement for NOTCH during maintenance of joint cartilage. However, NOTCH signaling components are substantially increased in abundance in posttraumatic osteoarthritis in humans and mice, suggesting either a reparative or a pathological role for NOTCH activation in osteoarthritis. We investigated a potential dual role for NOTCH in joint maintenance and osteoarthritis by generating two mouse models overexpressing the NOTCH1 intracellular domain (NICD) within postnatal joint cartilage. The first mouse model exhibited sustained NOTCH activation to resemble pathological NOTCH signaling, whereas the second model had transient NOTCH activation, which more closely reflected physiological NOTCH signaling. Sustained NOTCH signaling in joint cartilage led to an early and progressive osteoarthritic-like pathology, whereas transient NOTCH activation enhanced the synthesis of cartilage matrix and promoted joint maintenance under normal physiological conditions. Through RNA-sequencing, immunohistochemical, and biochemical approaches, we identified several targets that could be responsible for NOTCH-mediated cartilage degradation, fibrosis, and osteoarthritis progression. These targets included components of the interleukin-6 (IL-6)-signal transducer and activator of transcription 3 (STAT3) and mitogen-activated protein kinase signaling pathways, which may also contribute to the posttraumatic development of osteoarthritis. Together, these data suggest a dual role for the NOTCH pathway in joint cartilage, and they identify downstream effectors of NOTCH signaling as potential targets for disease-modifying osteoarthritis drugs.
Collapse
Affiliation(s)
- Zhaoyang Liu
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA. Department of Biology, University of Rochester, Rochester, NY 14642, USA
| | - Jianquan Chen
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anthony J Mirando
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA. Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Cuicui Wang
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA. Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Michael J Zuscik
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Regis J O'Keefe
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA. Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J Hilton
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA. Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
32
|
Ratneswaran A, Beier F. A top-notch dilemma: The complex role of NOTCH signaling in osteoarthritis. Sci Signal 2015. [PMID: 26198356 DOI: 10.1126/scisignal.aac7862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A study by Liu et al. in the current issue of Science Signaling explores the complex dual role of NOTCH in the etiology of osteoarthritis by comparing gain-of-function mouse models representing aberrant pathological signaling and transient physiological signaling.
Collapse
Affiliation(s)
- Anusha Ratneswaran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| |
Collapse
|
33
|
|
34
|
Silencing of miR-101 Prevents Cartilage Degradation by Regulating Extracellular Matrix-related Genes in a Rat Model of Osteoarthritis. Mol Ther 2015; 23:1331-1340. [PMID: 25921548 PMCID: PMC4817865 DOI: 10.1038/mt.2015.61] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/24/2015] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a common, degenerative joint disease characterized by articular cartilage degradation. Currently, clinical trials based on microRNA therapy have been performed to treat various diseases. However, no treatment has been found for arthritis. This study investigated the functions of miR-101 in cartilage degradation in vivo and evaluated the feasibility of using miR-101 as a therapeutic agent for OA. Mono-iodoacetate-induced arthritis (MIA) rats were used as an animal model of OA. miR-101 mimic or miR-101 inhibitor was injected into the rats' knees to evaluate its effects on cartilage degradation. Cartilage degradation aggravated at 14 days after the injection of miR-101 mimic. By contrast, miR-101 silencing reduced cartilage degradation. Moreover, the administration of miR-101 mimic is sufficient to cause cartilage degradation in the normal cartilage of rats. By contrast, miR-101 inhibitor could prevent this change. Microarray and qPCR were used to investigate the different expressed genes after injecting miR-101 mimic and miR-101 inhibitor in the rats' articular cartilage. Several cartilage degradation-related genes were selected and validated to function in cartilage degradation with miR-101. Our results demonstrated the therapeutic effect of miR-101 inhibition on cartilage degradation in MIA rats by regulating several cartilage degradation-related genes.
Collapse
|
35
|
Duan L, Ma B, Liang Y, Chen J, Zhu W, Li M, Wang D. Cytokine networking of chondrocyte dedifferentiation in vitro and its implications for cell-based cartilage therapy. Am J Transl Res 2015; 7:194-208. [PMID: 25901191 PMCID: PMC4399086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/25/2014] [Indexed: 06/04/2023]
Abstract
Autologous chondrocyte implantation (ACI) is a golden treatment for large defects of the knee joint without osteoarthritis or other complications. Despite notable progresses, generation of a stable chondrocyte phenotype using progenitor cells remains a main obstacle for chondrocyte-based cartilage treatment. Monolayer chondrocyte expansion in vitro is accompanied by chondrocyte dedifferentiation, which produces a non-specific mechanically inferior extracellular matrix (ECM) unsuitable for ACI. In-depth understanding of the molecular events during chondrocyte dedifferentiation is required to maintain the capacity of in vitro expanded chondrocytes to produce hyaline cartilage-specific ECM. This review discusses key cytokines and signaling pathways involved in chondrocyte dedifferentiation from the standpoint of catabolism and anabolism. Some potential therapeutic strategies are also presented to counteract chondrocyte dedifferentiation for cell-based cartilage therapy.
Collapse
Affiliation(s)
- Li Duan
- School of Medicine, Sun Yat-sen UniversityGuangzhou 510182, Guangdong Province, China
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| | - Bin Ma
- Division of Immunology, University Children’s Hospital ZurichZurich 8032, Switzerland
| | - Yujie Liang
- School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate SchoolShenzhen 518055, Guangdong Province, China
| | - Jielin Chen
- School of Medicine, Sun Yat-sen UniversityGuangzhou 510182, Guangdong Province, China
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| | - Weimin Zhu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| | - Mingtao Li
- School of Medicine, Sun Yat-sen UniversityGuangzhou 510182, Guangdong Province, China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| |
Collapse
|