1
|
Yu L, Tian D, Su Z, Zhang L, Jie L, Guo S, Zhu W, Zhang N, Wang P. Mechanical stress overload promotes NF-κB/NLRP3-mediated osteoarthritis synovitis and fibrosis through Piezo1. Cell Signal 2025; 132:111786. [PMID: 40221068 DOI: 10.1016/j.cellsig.2025.111786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Mechanical stress is a pivotal factor in the development of knee osteoarthritis (KOA). Piezo1, an innovative mechanosensitive ion channel, plays a key role in detecting variations in mechanical stress and transforming them into electrical signals. This research focuses on examining how Piezo1 influences synovial inflammation and fibrosis induced by mechanical stress in KOA, as well as delving into the potential underlying mechanisms. In vivo, pathological changes and immunohistochemical staining were conducted on both normal and overexercise rat synovial tissues to analyze the expression of Piezo1 and the NF-κB/NLRP3 pathways. In vitro utilized a cell stretcher to replicate the mechanical conditions seen in KOA. Levels of pro-inflammatory cytokines and fibrosis-related markers were assessed to investigate the impact of Piezo1 on mechanical stress in fibroblast-like synoviocytes (FLS). Subsequently, following cell stretching interventions, the effects on synovial inflammation and fibrosis were observed with the use of the Piezo1 inhibitor GsMTx4 or the NLRP3 inhibitor MCC950. Mechanical stress significantly promoted the activation of Piezo1, increased the phosphorylation ratio of p65, and elevated the levels of NLRP3, caspase-1, ASC, GSDMD, IL-1β, IL-18, IL-6, and TNF-α. Both in vitro and in vivo, mechanical stress also promoted the occurrence and development of synovial fibrosis, with significant increases in the expression levels of fibrosis-related markers. Under mechanical stress overload, upregulation of Piezo1 can promote the secretion of pro-inflammatory cytokines and the fibrotic process in synovium through the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Likai Yu
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Di Tian
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Zishan Su
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Li Zhang
- Orthopedics of traditional Chinese Medicine, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu, China
| | - Lishi Jie
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Shaobo Guo
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Wenhui Zhu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Nongshan Zhang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China.
| | - Peimin Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
2
|
Philpott HT, Birmingham TB, Blackler G, Klapak JD, Knights AJ, Farrell EC, Fiset B, Walsh LA, Giffin JR, Vasarhelyi EM, MacDonald SJ, Lanting BA, Maerz T, Appleton CT, the WOREO Knee Study Group. Association of Synovial Innate Immune Exhaustion With Worse Pain in Knee Osteoarthritis. Arthritis Rheumatol 2025; 77:664-676. [PMID: 39690716 PMCID: PMC12123257 DOI: 10.1002/art.43089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/23/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVE Uncontrolled pain remains a major clinical challenge in the management of knee osteoarthritis (OA), the most common disabling joint disease. Worse pain is associated with synovial innate immune cell infiltration (synovitis), but the role of innate immune-regulatory cells in pain is unknown. Our objective was to identify synovial innate immune cell subsets and pathophysiologic mechanisms associated with worse pain in patients with knee OA. METHODS Synovial tissue biopsies from 122 patients with mild-to-severe knee OA pain (Knee Injury and OA Outcome Score [KOOS]) were analyzed to identify associations between synovial histopathology and worse pain. We then used spatial transcriptomics and proteomics of synovial tissue microenvironments (n = 32), followed by single-cell RNA sequencing (n = 8), to identify synovial cell composition and cell-cell communication networks in patients with more severe OA pain. RESULTS Histopathological signs of synovial microvascular dysfunction and perivascular edema were associated with worse KOOS pain (-10.76; 95% confidence interval [CI] -18.90 to -2.61). Patients with worse pain had fewer immune-regulatory macrophages, expanded fibroblast subsets, and enrichment in neurovascular remodeling pathways. Synovial macrophages from patients with worse pain expressed markers of immune exhaustion and decreased phagocytic function (-19.42%; 95% CI -35.96 to -2.89) and their conditioned media increased neuronal cell stress in dorsal root ganglia. CONCLUSION Although synovitis increases during OA, our findings suggest that exhaustion, dysfunction, and loss of immune-regulatory macrophages is associated with worse pain and may be an important therapeutic target.
Collapse
Affiliation(s)
- Holly T. Philpott
- University of Western Ontario and London Health Sciences Centre‐University HospitalLondonOntarioCanada
| | - Trevor B. Birmingham
- University of Western Ontario and London Health Sciences Centre‐University HospitalLondonOntarioCanada
| | | | | | | | | | | | | | - J. Robert Giffin
- University of Western Ontario and London Health Sciences Centre‐University HospitalLondonOntarioCanada
| | - Edward M. Vasarhelyi
- University of Western Ontario and London Health Sciences Centre‐University HospitalLondonOntarioCanada
| | - Steven J. MacDonald
- University of Western Ontario and London Health Sciences Centre‐University HospitalLondonOntarioCanada
| | - Brent A. Lanting
- University of Western Ontario and London Health Sciences Centre‐University HospitalLondonOntarioCanada
| | | | - C. Thomas Appleton
- University of Western Ontario and London Health Sciences Centre‐University HospitalLondonOntarioCanada
| | | |
Collapse
|
3
|
Raut R, Chakraborty A, Neogi T, Albro M, Snyder B, Schaer T, Zhang C, Grinstaff M, Bais M. Constructing a cross-tissue human knee single-cell atlas identified osteoarthritis reduces regenerative tissue stem cells while increasing inflammatory pain macrophages. RESEARCH SQUARE 2025:rs.3.rs-6247502. [PMID: 40386432 PMCID: PMC12083644 DOI: 10.21203/rs.3.rs-6247502/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Osteoarthritis (OA) affects the entire knee joint; however, cross-tissue molecular mechanisms are poorly understood due to a lack of comprehensive, integrated analysis. We constructed the first comprehensive single-cell RNA sequencing knee OA atlas from articular cartilage, meniscus, synovium, and subchondral bone which showed active communication between them. Healthy synovium and meniscus contain the largest populations of tissue stem cells (TSCs) and immune cells that are altered in OA. The regenerative TSCs expressing SDF1, SOX9, CD146, PDGFRB, and CD105 decrease during OA, whereas osteogenic TSCs expressing osteogenic differentiation-related factor NT5E (CD73) are increased. In OA, the balance between regenerative and osteogenic TSCs shifts in the OA state with an increased number of osteogenic TSCs. We also report an increased level of quadruple-positive inflammatory (IL1B-IL6-NOS2-TNF) and pain marker (P2RX7) specific macrophages in OA. Fibroblasts are enriched in OA-synovium and may contribute to fibrosis. Importantly, OA cartilage contains unique MMP13-producing detrimental chondrocytes along with RUNX2-producing chondrocytes that worsen OA pathophysiology. This atlas provides a novel avenue for potential therapeutic applications in human knee OA and other musculoskeletal diseases and injuries, targeting synovium and meniscus to intervene in OA-specific molecular and cellular alterations.
Collapse
Affiliation(s)
| | | | | | | | - Brian Snyder
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center and Harvard Medical School
| | - Thomas Schaer
- University of Pennsylvania School of Veterinary Medicine
| | - Chao Zhang
- Department of Medicine Section of Computational Biomedicine, Boston University Chobanian and Avedisian School of Medicine, Boston MA 02118
| | | | | |
Collapse
|
4
|
Liu Y, Molchanov V, Brass D, Yang T. Recent advances in omics and the integration of multi-omics in osteoarthritis research. Arthritis Res Ther 2025; 27:100. [PMID: 40319309 PMCID: PMC12049056 DOI: 10.1186/s13075-025-03563-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/20/2025] [Indexed: 05/07/2025] Open
Abstract
Osteoarthritis (OA) is a complex disorder driven by the combination of environmental and genetic factors. Given its high global prevalence and heterogeneity, developing effective and personalized treatment methods is crucial. This requires identifying new disease mechanisms, drug targets, and biomarkers. Various omics approaches have been applied to identify OA-related genes, pathways, and biomarkers, including genomics, epigenomics, transcriptomics, proteomics, and metabolomics. These omics studies have generated vast datasets that are shaping the field of OA research. The emergence of high-resolution methodologies, such as single-cell and spatial omics techniques, further enhances our ability to dissect molecular complexities within the OA microenvironment. By integrating these multi-layered datasets, researchers can uncover central signaling hubs and disease mechanisms, ultimately facilitating the development of targeted therapies and precision medicine approaches for OA treatment.
Collapse
Affiliation(s)
- Ye Liu
- Department of Cell Biology, Van Andel Research Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
| | - Vladimir Molchanov
- Department of Cell Biology, Van Andel Research Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
| | - David Brass
- Department of Cell Biology, Van Andel Research Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
| | - Tao Yang
- Department of Cell Biology, Van Andel Research Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
5
|
Qiu F, Xie D, Chen H, Wang Z, Huang J, Cao C, Liang Y, Yang X, He DY, Fu X, Lu A, Liang C. Generation of cytotoxic aptamers specifically targeting fibroblast-like synoviocytes by CSCT-SELEX for treatment of rheumatoid arthritis. Ann Rheum Dis 2025; 84:726-745. [PMID: 39237134 DOI: 10.1136/ard-2024-225565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is an autoimmune disease characterised by aggressive fibroblast-like synoviocytes (FLSs). Very few RA patients-derived FLSs (RA-FLSs)-specific surface signatures have been identified, and there is currently no approved targeted therapy for RA-FLSs. This study aimed to screen therapeutic aptamers with cell-targeting and cytotoxic properties against RA-FLSs and to uncover the molecular targets and mechanism of action of the screened aptamers. METHODS We developed a cell-specific and cytotoxic systematic evolution of ligands by exponential enrichment (CSCT-SELEX) method to screen the therapeutic aptamers without prior knowledge of the surface signatures of RA-FLSs. The molecular targets and mechanisms of action of the screened aptamers were determined by pull-down assays and RNA sequencing. The therapeutic efficacy of the screened aptamers was examined in arthritic mouse models. RESULTS We obtained an aptamer SAPT8 that selectively recognised and killed RA-FLSs. The molecular target of SAPT8 was nucleolin (NCL), a shuttling protein overexpressed on the surface and involved in the tumor-like transformation of RA-FLSs. Mechanistically, SAPT8 interacted with the surface NCL and was internalised to achieve lysosomal degradation of NCL, leading to the upregulation of proapoptotic p53 and downregulation of antiapoptotic B-cell lymphoma 2 (Bcl-2) in RA-FLSs. When administrated systemically to arthritic mice, SAPT8 accumulated in the inflamed FLSs of joints. SAPT8 monotherapy or its combination with tumour necrosis factor (TNF)-targeted biologics was shown to relieve arthritis in mouse models. CONCLUSIONS CSCT-SELEX could be a promising strategy for developing cell-targeting and cytotoxic aptamers. SAPT8 aptamer selectively ablates RA-FLSs via modulating NCL-p53/Bcl-2 signalling, representing a potential alternative or complementary therapy for RA.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | | | - Xu Yang
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Dong-Yi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China; Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| |
Collapse
|
6
|
Araya-Sapag MJ, Lara-Barba E, García-Guerrero C, Herrera-Luna Y, Flores-Elías Y, Bustamante-Barrientos FA, Albornoz GG, Contreras-Fuentes C, Yantén-Fuentes L, Luque-Campos N, Vega-Letter AM, Toledo J, Luz-Crawford P. New mesenchymal stem/stromal cell-based strategies for osteoarthritis treatment: targeting macrophage-mediated inflammation to restore joint homeostasis. J Mol Med (Berl) 2025:10.1007/s00109-025-02547-8. [PMID: 40272537 DOI: 10.1007/s00109-025-02547-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Macrophages are pivotal in osteoarthritis (OA) pathogenesis, as their dysregulated polarization can contribute to chronic inflammatory processes. This review explores the molecular and metabolic mechanisms that influence macrophage polarization and identifies potential strategies for OA treatment. Currently, non-surgical treatments for OA focus only on symptom management, and their efficacy is limited; thus, mesenchymal stem/stromal cells (MSCs) have gained attention for their anti-inflammatory and immunomodulatory capabilities. Emerging evidence suggests that small extracellular vesicles (sEVs) derived from MSCs can modulate macrophage function, thus offering potential therapeutic benefits in OA. Additionally, the transfer of mitochondria from MSCs to macrophages has shown promise in enhancing mitochondrial functionality and steering macrophages toward an anti-inflammatory M2-like phenotype. While further research is needed to confirm these findings, MSC-based strategies, including the use of sEVs and mitochondrial transfer, hold great promise for the treatment of OA and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- María Jesús Araya-Sapag
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliana Lara-Barba
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García-Guerrero
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yesenia Flores-Elías
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Guillermo G Albornoz
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Consuelo Contreras-Fuentes
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Liliana Yantén-Fuentes
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Red de Equipamiento Científico Avanzado (REDECA), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ana María Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado (REDECA), Facultad de Medicina, Universidad de Chile, Santiago, Chile.
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico Universidad de Chile, Santiago, Chile.
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| |
Collapse
|
7
|
Gao K, Huang Z, Liao Z, Wang Y, Chen D. Machine learning analysis of FOSL2 and RHoBTB1 as central immunological regulators in knee osteoarthritis synovium. J Int Med Res 2025; 53:3000605251333646. [PMID: 40287984 PMCID: PMC12035077 DOI: 10.1177/03000605251333646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 02/25/2025] [Indexed: 04/29/2025] Open
Abstract
BackgroundKnee osteoarthritis is a debilitating disease with a complex pathogenesis. Synovitis, which refers to inflammation of the synovial membrane surrounding the joint, is believed to play an important role in the development and progression of knee osteoarthritis. To better understand the molecular mechanisms underlying knee osteoarthritis, we conducted a comprehensive analysis of gene expression in knee osteoarthritis synovium using machine learning.MethodsDifferentially expressed genes between knee osteoarthritis and control synovial tissues were analyzed using the GSE55235 dataset. We employed several machine learning algorithms, including least absolute shrinkage and selection operator and support vector machine-recursive feature elimination, to screen for key genes. Then, we validated the key genes using an external dataset (GSE51588) and an in vitro knee osteoarthritis animal model. CIBERSORT was used to compare immune cell infiltration levels between knee osteoarthritis and control synovial tissues and determine their relationship with the key genes. Finally, we performed a Connectivity Map analysis to screen for potential small-molecule compounds. Moreover, we conducted single-cell RNA sequencing analysis using knee joint tissues to annotate different subtypes of cells.ResultsA total of 930 differentially expressed genes were identified. Least absolute shrinkage and selection operator regression and support vector machine-recursive feature elimination identified FOSL2 and RHoBTB1 as key genes. The expression levels of both genes were further validated in the GSE51588 dataset as well as verified through an in vitro experiment involving a knee osteoarthritis mouse model. Multiple significant correlation pairs were found between the immune cell infiltration levels. We unveiled the genetic basis of knee osteoarthritis using genome-wide association study and specific signaling pathways through gene set enrichment analysis. The GeneCards database was used to obtain 3032 pathogenic genes associated with knee osteoarthritis, and we found that RHoBTB1 expression was significantly negatively correlated and FOSL2 expression was significantly positively correlated with interleukin-1β expression. We predicted several small-molecule compounds based on Connectivity Map analysis. Finally, single-cell RNA sequencing analysis revealed the expression levels of the two key genes in chondrocytes and tissue stem cells.ConclusionFOSL2 and RHoBTB1 may play key roles in the pathogenesis of knee osteoarthritis, exhibiting correlations with immune cell infiltration levels. These findings indicate that these genes have potential as therapeutic targets. However, further research and validation are necessary to confirm their exact roles and therapeutic potential in knee osteoarthritis.
Collapse
Affiliation(s)
- Kun Gao
- Department of Orthopedics, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhenyu Huang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhouwei Liao
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yanfei Wang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Dayu Chen
- Department of Orthopedics, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
8
|
Henry ÓC, O'Neill LAJ. Metabolic Reprogramming in Stromal and Immune Cells in Rheumatoid Arthritis and Osteoarthritis: Therapeutic Possibilities. Eur J Immunol 2025; 55:e202451381. [PMID: 40170391 PMCID: PMC11962241 DOI: 10.1002/eji.202451381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/03/2025]
Abstract
Metabolic reprogramming of stromal cells, including fibroblast-like synoviocytes (FLS) and chondrocytes, as well as osteoclasts (OCs), are involved in the inflammatory and degenerative processes underlying rheumatoid arthritis (RA) and osteoarthritis (OA). In RA, FLS exhibit mTOR activation, enhanced glycolysis and reduced oxidative phosphorylation, fuelling inflammation, angiogenesis, and cartilage degradation. In OA, chondrocytes undergo metabolic rewiring, characterised by mTOR and NF-κB activation, mitochondrial dysfunction, and increased glycolysis, which promotes matrix metalloproteinase production, extracellular matrix (ECM) degradation, and angiogenesis. Macrophage-derived immunometabolites, including succinate and itaconate further modulate stromal cell function, acting as signalling molecules that modulate inflammatory and catabolic processes. Succinate promotes inflammation whilst itaconate is anti-inflammatory, suppressing inflammatory joint disease in models. Itaconate deficiency also correlates inversely with disease severity in RA in humans. Emerging evidence highlights the potential of targeting metabolic processes as promising therapeutic strategies for connective tissue disorders.
Collapse
Affiliation(s)
- Órlaith C. Henry
- Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | | |
Collapse
|
9
|
Karsdal M, Cox TR, Parker AL, Willumsen N, Sand JMB, Jenkins G, Hansen HH, Oldenburger A, Geillinger-Kaestle KE, Larsen AT, Black D, Genovese F, Eckersley A, Heinz A, Nyström A, Holm Nielsen S, Bennink L, Johannsson L, Bay-Jensen AC, Orange DE, Friedman S, Røpke M, Fiore V, Schuppan D, Rieder F, Simona B, Borthwick L, Skarsfeldt M, Wennbo H, Thakker P, Stoffel R, Clarke GW, Kalluri R, Ruane D, Zannad F, Mortensen JH, Sinkeviciute D, Sundberg F, Coseno M, Thudium C, Croft AP, Khanna D, Cooreman M, Broermann A, Leeming DJ, Mobasheri A, Ricard-Blum S. Advances in Extracellular Matrix-Associated Diagnostics and Therapeutics. J Clin Med 2025; 14:1856. [PMID: 40142664 PMCID: PMC11943371 DOI: 10.3390/jcm14061856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 03/28/2025] Open
Abstract
The extracellular matrix (ECM) is the common denominator of more than 50 chronic diseases. Some of these chronic pathologies lead to enhanced tissue formation and deposition, whereas others are associated with increased tissue degradation, and some exhibit a combination of both, leading to severe tissue alterations. To develop effective therapies for diseases affecting the lung, liver, kidney, skin, intestine, musculoskeletal system, heart, and solid tumors, we need to modulate the ECM's composition to restore its organization and function. Across diverse organ diseases, there are common denominators and distinguishing factors in this fibroinflammatory axis, which may be used to foster new insights into drug development across disease indications. The 2nd Extracellular Matrix Pharmacology Congress took place in Copenhagen, Denmark, from 17 to 19 June 2024 and was hosted by the International Society of Extracellular Matrix Pharmacology. The event was attended by 450 participants from 35 countries, among whom were prominent scientists who brought together state-of-the-art research on organ diseases and asked important questions to facilitate drug development. We highlight key aspects of the ECM in the liver, kidney, skin, intestine, musculoskeletal system, lungs, and solid tumors to advance our understanding of the ECM and its central targets in drug development. We also highlight key advances in the tools and technology that enable this drug development, thereby supporting the ECM.
Collapse
Affiliation(s)
- Morten Karsdal
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Thomas R. Cox
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Amelia L. Parker
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Nicholas Willumsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Jannie Marie Bülow Sand
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, NIHR Imperial Biomedical Research Centre, Imperial College London, London SW7 2AZ, UK;
| | | | | | - Kerstin E. Geillinger-Kaestle
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Anna Thorsø Larsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | - Federica Genovese
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Alexander Eckersley
- Wellcome Centre for Cell Matrix Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK;
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center—University of Freiburg, 79106 Breisgau, Germany;
| | - Signe Holm Nielsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | | | - Anne-Christine Bay-Jensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dana E. Orange
- Hospital for Special Surgery, The Rockefeller University, New York, NY 10065, USA;
| | - Scott Friedman
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA;
| | | | - Vincent Fiore
- Boehringer Ingelheim, 55218 Ingelheim am Rhein, Germany;
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Florian Rieder
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | | | - Lee Borthwick
- FibroFind Ltd., FibroFind Laboratories, Medical School, Newcastle upon Tyne NE2 4HH, UK;
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Mark Skarsfeldt
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Haakan Wennbo
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Paresh Thakker
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Ruedi Stoffel
- Roche Diagnostics International Ltd., 6343 Rotkreuz, Switzerland;
| | - Graham W. Clarke
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden;
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College, London E1 9RT, UK
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Darren Ruane
- Janssen Immunology, Translational Sciences and Medicine, La Jolla, CA 92037, USA;
| | - Faiez Zannad
- Division of Heart Failure and Hypertension, and of the Inserm CIC, University of Lorraine, 54000 Metz, France;
| | - Joachim Høg Mortensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dovile Sinkeviciute
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Fred Sundberg
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Molly Coseno
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Christian Thudium
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Adam P. Croft
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, University of Birmingham, Birmingham B15 2TT, UK;
- Institute of Inflammation and Ageing, Queen Elizabeth Hospital, University of Birmingham, Birmingham B15 2TT, UK
| | - Dinesh Khanna
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | | | - Andre Broermann
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Diana Julie Leeming
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Ali Mobasheri
- Faculty of Medicine, University of Oulu, 90570 Oulu, Finland;
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
- Faculté de Médecine, Université de Liège, 4000 Liège, Belgium
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Sylvie Ricard-Blum
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, ICBMS, University Lyon 1, 69622 Villeurbanne Cedex, France;
| |
Collapse
|
10
|
Tsukada A, Uekusa Y, Ohta E, Hattori A, Mukai M, Iwase D, Aikawa J, Ohashi Y, Inoue G, Takaso M, Uchida K. Association Between Synovial NTN4 Expression and Pain Scores, and Its Effects on Fibroblasts and Sensory Neurons in End-Stage Knee Osteoarthritis. Cells 2025; 14:395. [PMID: 40136644 PMCID: PMC11941210 DOI: 10.3390/cells14060395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease marked by synovial inflammation, cartilage degradation, and persistent pain. Although Netrin-4 (NTN4) has been implicated in pain modulation in rheumatoid arthritis (RA), its role in OA pain remains less understood. Previous research has documented that NTN4 promotes axonal growth in rodent-derived neurons; however, its effects on human sensory neurons are yet to be fully explored. NTN4 also plays a multifactorial role in various non-neuronal cells, such as endothelial cells, tumor cells, and stromal cells. Nevertheless, its specific impact on synovial fibroblasts, which are key components of the synovium and have been linked to OA pain, is still unclear. This study examined the correlation between NTN4 expression levels and pain severity in OA, specifically investigating its effects on human iPSC-derived sensory neurons (iPSC-SNs) and synovial fibroblasts from OA patients. Our findings indicate a positive correlation between synovial NTN4 expression and pain severity. Recombinant human Netrin-4 (rh-NTN4) was also shown to enhance neurite outgrowth in human iPSC-SNs, suggesting a potential role in neuronal sensitization. Additionally, rh-NTN4 stimulated the production of pro-inflammatory cytokines (IL-6, IL-8) and chemokines (CXCL1, CXCL6, CXCL8) in synovium-derived fibroblastic cells, implicating it in synovial inflammation. Collectively, these results suggest that NTN4 may contribute to KOA pathology by promoting synovial inflammation and potentially sensitizing sensory neurons, thereby influencing the mechanisms of underlying pain.
Collapse
Affiliation(s)
- Ayumi Tsukada
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
| | - Yui Uekusa
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
| | - Etsuro Ohta
- Division of Blood Transfusion and Transplantation, Kitasato University School of Health Sciences, Minamiuonuma 949-7241, Nigata, Japan; (E.O.); (A.H.)
- Program in Cellular Immunology, Graduate School of Medical Science, Kitasato, Sagamihara City 252-0375, Kanagawa, Japan
- Center for Cell Design, Institute for Regenerative Medicine and Cell Design, Kitasato University School of Allied Health Sciences, Sagamihara City 252-0374, Kanagawa, Japan
| | - Akito Hattori
- Division of Blood Transfusion and Transplantation, Kitasato University School of Health Sciences, Minamiuonuma 949-7241, Nigata, Japan; (E.O.); (A.H.)
| | - Manabu Mukai
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
| | - Dai Iwase
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
| | - Jun Aikawa
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
| | - Yoshihisa Ohashi
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (Y.U.); (M.M.); (D.I.); (J.A.); (Y.O.); (G.I.); (M.T.)
- Medical Sciences Research Institute, Shonan University, Chigasaki City 253-0083, Kanagawa, Japan
| |
Collapse
|
11
|
Ragni E, Papait A, Taiana MM, De Luca P, Grieco G, Vertua E, Romele P, Colombo C, Silini AR, Parolini O, de Girolamo L. Cell culture expansion media choice affects secretory, protective and immuno-modulatory features of adipose mesenchymal stromal cell-derived secretomes for orthopaedic applications. Regen Ther 2025; 28:481-497. [PMID: 39980717 PMCID: PMC11840939 DOI: 10.1016/j.reth.2025.01.016] [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: 11/26/2024] [Revised: 01/08/2025] [Accepted: 01/19/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Mesenchymal stromal cells (MSCs) gained attention for their anti-inflammatory and trophic properties, with musculoskeletal diseases and osteoarthritis (OA) being among the most studied conditions. Alongside cells, their released factors and extracellular vesicles (EVs), overall termed "secretome", are actively sifted being envisioned as the main therapeutic actors. In addition to standard supplementation given by foetal bovine serum (FBS) or human platelet lysate (hPL), new good manufacturing practice (GMP)-compliant serum/xeno (S/X)-free media formulations have been proposed, although their influence on MSCs phenotype and potential is scarcely described. The aim of this study is therefore to evaluate, in the OA context, the differences in secretome composition and potential after adipose-MSCs (ASCs) cultivation in both standard (FBS and hPL) and two next generation (S/X) GMP-ready supplements. Methods Immunophenotype and secretory ability at soluble protein and EV-related levels, including embedded miRNAs, were analysed in the secretomes by means of flow cytometry, nanoparticle tracking analysis, high throughput ELISA and qRT-PCR arrays. Secretomes effect was tested in in vitro models of chondrocytes, lymphocytes and monocytes to mimic the OA microenvironment. Results Within a conserved molecular signature, a divergent fingerprint emerged for ASCs' secretomes collected after expansion in standard FBS/hPL or next-generation S/X formulations. Regarding soluble factors, a less protective feature for those in the secretome collected after ASCs were cultured in S/X media emerged. Moreover, the overall message for EV-miRNAs was characterized by a preponderance of protective signals in FBS and hPL conditions in a context of general safeguard given by ASCs released molecules. This dichotomy was reflected on secretomes' potential in vitro, with expansion in hPL resulting in the most effective secretome for chondrocytes and in FBS for immune cells. Conclusions These data open the question about the implications from using new media for MSCs expansion for clinical application. Although the undeniable advantages for GMP compliant processes, this study results suggest that new media formulations would deserve a deep characterization to drive the choice of the most effective one tailored to each specific application.
Collapse
Affiliation(s)
- Enrico Ragni
- IRCCS Ospedale Galeazzi - Sant’Ambrogio, Laboratorio di Biotecnologie Applicate all’Ortopedia, Via Cristina Belgioioso 173, 20157 Milano, Italy
| | - Andrea Papait
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Roma, Italy
| | - Michela Maria Taiana
- IRCCS Ospedale Galeazzi - Sant’Ambrogio, Laboratorio di Biotecnologie Applicate all’Ortopedia, Via Cristina Belgioioso 173, 20157 Milano, Italy
| | - Paola De Luca
- IRCCS Ospedale Galeazzi - Sant’Ambrogio, Laboratorio di Biotecnologie Applicate all’Ortopedia, Via Cristina Belgioioso 173, 20157 Milano, Italy
| | - Giulio Grieco
- IRCCS Ospedale Galeazzi - Sant’Ambrogio, Laboratorio di Biotecnologie Applicate all’Ortopedia, Via Cristina Belgioioso 173, 20157 Milano, Italy
| | - Elsa Vertua
- Centro di Ricerca "E. Menni", Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy
| | - Pietro Romele
- Centro di Ricerca "E. Menni", Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy
| | - Cecilia Colombo
- IRCCS Ospedale Galeazzi - Sant’Ambrogio, Laboratorio di Biotecnologie Applicate all’Ortopedia, Via Cristina Belgioioso 173, 20157 Milano, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca "E. Menni", Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy
| | - Ornella Parolini
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Roma, Italy
| | - Laura de Girolamo
- IRCCS Ospedale Galeazzi - Sant’Ambrogio, Laboratorio di Biotecnologie Applicate all’Ortopedia, Via Cristina Belgioioso 173, 20157 Milano, Italy
| |
Collapse
|
12
|
Tang S, Zhang C, Oo WM, Fu K, Risberg MA, Bierma-Zeinstra SM, Neogi T, Atukorala I, Malfait AM, Ding C, Hunter DJ. Osteoarthritis. Nat Rev Dis Primers 2025; 11:10. [PMID: 39948092 DOI: 10.1038/s41572-025-00594-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2025] [Indexed: 05/09/2025]
Abstract
Osteoarthritis is a heterogeneous whole-joint disease that can cause pain and is a leading cause of disability and premature work loss. The predominant disease risk factors - obesity and joint injury - are well recognized and modifiable. A greater understanding of the complex mechanisms, including inflammatory, metabolic and post-traumatic processes, that can lead to disease and of the pathophysiology of pain is helping to delineate mechanistic targets. Currently, management is primarily focused on alleviating the main symptoms of pain and obstructed function through lifestyle interventions such as self-management programmes, education, physical activity, exercise and weight management. However, lack of adherence to known effective osteoarthritis therapeutic strategies also contributes to the high global disease burden. For those who have persistent symptoms that are compromising quality of life and have not responded adequately to core treatments, joint replacement is an option to consider. The burden imparted by the disease causes a substantial impact on individuals affected in terms of quality of life. For society, this disease is a substantial driver of increased health-care costs and underemployment. This Primer highlights advances and controversies in osteoarthritis, drawing key insights from the current evidence base.
Collapse
Affiliation(s)
- Su'an Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Institute of Exercise and Rehabilitation Science, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Win Min Oo
- Department of Rheumatology, Royal North Shore Hospital and Sydney Musculoskeletal Health, Faculty of Medicine and Health Science, Kolling Institute, University of Sydney, Sydney, Australia
- Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Myanmar
| | - Kai Fu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - May Arna Risberg
- Department of Sport Medicine, Norwegian School Sport Sciences, Oslo, Norway
| | - Sita M Bierma-Zeinstra
- Department of General Practice, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tuhina Neogi
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Inoshi Atukorala
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- Institute of Exercise and Rehabilitation Science, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia.
| | - David J Hunter
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Rheumatology, Royal North Shore Hospital and Sydney Musculoskeletal Health, Faculty of Medicine and Health Science, Kolling Institute, University of Sydney, Sydney, Australia.
| |
Collapse
|
13
|
Shi J, Gong T, Zhou Y. Pioglitazone Regulates Chondrocyte Metabolism and Attenuates Osteoarthritis by Activating Peroxisome Proliferator-Activated Receptor Gamma. J Cell Mol Med 2025; 29:e70456. [PMID: 40008494 PMCID: PMC11862886 DOI: 10.1111/jcmm.70456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/28/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Osteoarthritis presents a significant clinical challenge due to its high prevalence and the resultant impairment of patients' motor function. Osteoarthritic chondrocytes are characterised by inflammation and metabolic disturbances. Pioglitazone, an agonist of peroxisome proliferator-activated receptor γ (PPAR-γ), has been demonstrated to exert anti-inflammatory effects across various diseases. This study aims to investigate the potential protective effects of Pioglitazone on osteoarthritic chondrocytes. An in vitro chondrocyte inflammation model was established utilising IL-1β. The impact of Pioglitazone on chondrocyte inflammation and extracellular matrix synthesis was evaluated through enzyme-linked immunosorbent assay, immunofluorescence staining and Alcian blue staining. The affinity of Pioglitazone for PPAR-γ was investigated using molecular docking techniques. Alterations in chondrocyte glycolysis and oxidative phosphorylation were examined using the Seahorse XF Analyser, and the influence of Pioglitazone on glucose uptake and the mitochondrial electron transport chain was further analysed. Pioglitazone was gavaged in a mouse OA model established by anterior cruciate ligament transection to evaluate the therapeutic efficacy of Pioglitazone. Our findings indicate that Pioglitazone mitigates chondrocyte inflammation and osteoarthritis in murine models by inhibiting the expression of inflammatory mediators such as TNF-α, IL-6 and PGE2, and by preventing the degradation of aggrecan and collagen II. Furthermore, Pioglitazone significantly upregulated the expression of glucose transporter 1 and stabilised the mitochondrial proton delivery chain in a PPAR-γ-dependent manner, thereby enhancing chondrocyte glucose uptake, glycolysis, and oxidative phosphorylation. These effects were partially reversed by the PPAR-γ antagonist GW9662. Pioglitazone can confer chondroprotective benefits in osteoarthritis by activating PPAR-γ.
Collapse
Affiliation(s)
- Jiaqi Shi
- Department of Orthopedics, Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangHubeiPeople's Republic of China
| | - Tianlun Gong
- Department of Orthopedics, Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangHubeiPeople's Republic of China
| | - Yi Zhou
- Department of Orthopedics, Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangHubeiPeople's Republic of China
| |
Collapse
|
14
|
Wang L. Complement 3 and 4 impact in osteoarthritis. Biomark Med 2025; 19:81-90. [PMID: 39893562 PMCID: PMC11792862 DOI: 10.1080/17520363.2024.2409062] [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: 10/22/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Aim: To investigate the association between serum complement 3 and 4 (C3 and C4) levels and clinical characteristics in osteoarthritis (OA) patients.Methods: This retrospective study included 361 OA patients divided into groups based on complement levels. Clinical data and laboratory test results, including C3, C4, ESR and CRP levels, were analyzed using non-parametric tests, Spearman correlation and multivariate regression.Results: The AC-OA group had lower PA, C3, C4, ESR, CRP and IgA levels compared with controls. C3 levels were positively correlated with ESR (r = 0.260, p < 0.001) and CRP (r = 0.243, p < 0.001). C4 levels also correlated with ESR (r = 0.175, p = 0.001) and CRP (r = 0.263, p < 0.001) and were significantly associated with the number of affected joints.Conclusion: Serum C4 levels are effective indicators of disease activity in OA, particularly in terms of joint involvement and CRP levels, while C3 levels showed no significant association. These findings suggest a potential role for C4 in predicting OA activity.
Collapse
Affiliation(s)
- Lifang Wang
- Department of Rheumatology & Immunology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| |
Collapse
|
15
|
Miyahara J, Omata Y, Chijimatsu R, Okada H, Ishikura H, Higuchi J, Tachibana N, Nagata K, Tani S, Kono K, Kawaguchi K, Yamagami R, Inui H, Taketomi S, Iwanaga Y, Terashima A, Yano F, Seki M, Suzuki Y, Baron R, Tanaka S, Saito T. CD34hi subset of synovial fibroblasts contributes to fibrotic phenotype of human knee osteoarthritis. JCI Insight 2025; 10:e183690. [PMID: 39846253 PMCID: PMC11790023 DOI: 10.1172/jci.insight.183690] [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] [Indexed: 01/24/2025] Open
Abstract
Osteoarthritis (OA) shows various clinical manifestations depending on the status of its joint components. We aimed to identify the synovial cell subsets responsible for OA pathophysiology by comprehensive analyses of human synovium samples in single-cell resolution. Two distinct OA synovial tissue groups were classified by gene expression profiles in RNA-Seq: inflammatory and fibrotic. The inflammatory group exhibited high expression of inflammatory cytokines, histologically inflammatory infiltrate, and a more severe pain score. The fibrotic group showed higher expression of fibroblast growth factor (FGFs) and bone morphogenetic proteins (BMPs), showed histologically perivascular fibrosis, and showed a lower pain score. In single-cell RNA-Seq (scRNA-Seq) of synovial cells, MERTKloCD206lo macrophages and CD34hi fibroblasts were associated with the inflammatory and fibrotic groups, respectively. Among the 3 fibroblast subsets, CD34loTHY1lo and CD34loTHY1hi fibroblasts were influenced by synovial immune cells, whereas CD34hi fibroblasts were influenced by mural and endothelial cells. Particularly, in CD34hi fibroblast subsets, CD34hiCD70hi fibroblasts promoted proliferation of Tregs, potentially suppressing synovitis and protecting articular cartilage. Elucidation of the mechanisms underlying the regulation of these synovial cell subsets may lead to novel strategies for OA therapeutics.
Collapse
Affiliation(s)
| | - Yasunori Omata
- Sensory & Motor System Medicine
- Bone and Cartilage Regenerative Medicine
| | | | - Hiroyuki Okada
- Sensory & Motor System Medicine
- Center for Disease Biology and Integrative Medicine, and
| | | | | | | | | | - Shoichiro Tani
- Sensory & Motor System Medicine
- Center for Disease Biology and Integrative Medicine, and
| | | | | | | | | | | | - Yasuhide Iwanaga
- Sensory & Motor System Medicine
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | | | | | - Masahide Seki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, Japan
| | - Yutaka Suzuki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, Japan
| | - Roland Baron
- Department of Medicine, Harvard Medical School and Endocrine Unit, MGH, Boston, Massachusetts, USA
| | | | | |
Collapse
|
16
|
Carstens MF, Alder KD, Dilger OB, Bothun CE, Payne AN, Thaler R, Morrey ME, Sanchez-Sotelo J, Berry DJ, Dudakovic A, Abdel MP. Effect of Antiseptic Irrigation Solutions on Primary Human Knee Fibroblasts Cultured in Human Platelet Lysate. J Arthroplasty 2025:S0883-5403(25)00036-1. [PMID: 39837392 DOI: 10.1016/j.arth.2025.01.021] [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: 09/09/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Native and periprosthetic joint infections are devastating conditions fraught with patient morbidity and mortality. Aseptic and septic joints are often debrided and irrigated to decrease bacterial loads when preventing or treating infection. However, the effect of clinically used irrigation solutions on the native cellular components of the synovial joint is unknown. METHODS Patients consented, and their suprapatellar knee tissue was surgically excised for fibroblast isolation. Cultured knee fibroblasts were treated with normal saline for three minutes or one of the following experimental solutions: acetic acid, chlorhexidine-gluconate, Dakin's solution, hydrogen peroxide, or povidone-iodine. The exposure time for the antiseptic solutions was one and three minutes. At 24 hours after irrigation treatment, metabolic activity was measured via MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] activity assay, and deoxyribonucleic acid content was assessed by Hoechst staining as a surrogate for cell number. Phase-contrast imaging elucidated proliferation potential, progressive cell loss, and cell morphology over a 5-day period. All experiments were repeated in triplicate. RESULTS All experimental antiseptic irrigation solutions, regardless of application time, caused a significant reduction in metabolic activity and DNA content, indicating extensive cell death. Phase-contrast imaging showed halted cellular proliferation, progressive cell loss, and distinct changes in cellular morphology, indicating decreased cellular viability and progressive cell death. CONCLUSIONS All antiseptic irrigation solutions investigated in this study were severely cytotoxic to human knee fibroblasts regardless of their chemical composition. The concentrations of these solutions are commonly used in orthopaedic surgery. Although these solutions have high bactericidal properties, it may be beneficial to use them in combination at lower doses to retain their effect on bacteria while remaining benign to native synovial cells.
Collapse
Affiliation(s)
- Mason F Carstens
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Kareme D Alder
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Oliver B Dilger
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Cole E Bothun
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Ashley N Payne
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Mark E Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | | | - Daniel J Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
17
|
De Marziani L, Zanasi L, Roveda G, Boffa A, Andriolo L, Di Martino A, Zaffagnini S, Filardo G. Symptoms and joint degeneration correlate with the temperature of osteoarthritic knees: an infrared thermography analysis. INTERNATIONAL ORTHOPAEDICS 2025; 49:101-108. [PMID: 39589460 PMCID: PMC11703876 DOI: 10.1007/s00264-024-06376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE This study aim was to analyze the joint temperature of patients affected by bilateral knee osteoarthritis (OA) using infrared thermography to investigate whether thermographic imaging patterns are influenced by the severity of symptoms and joint degeneration. METHODS Sixty-sixpatients ranging from 43 to 78 years old (63.3 ± 8.8 years) with bilateral knee OA and one symptomatic knee were enrolled. Thermograms of the two knees were captured using a thermographic camera FLIR T1020 and analyzed with the ResearchIR software to calculate the temperature of the overall knee and the four regions of interest (ROIs): patella, suprapatellar, medial, and lateral areas. RESULTS The temperature of knees affected by OA was influenced by joint degeneration level and symptoms: patients with higher OA grade in the symptomatic knees presented higher total knee temperatures compared to the asymptomatic ones (p = 0.002), as well as in the patellar (p = 0.005), lateral (p = 0.002), and medial (p = 0.001) areas. On the other hand, patients with the same OA level in the two knees presented a higher temperature in the symptomatic knee only in the medial area (p = 0.037). Symptomatic knees demonstrated a different pattern compared to asymptomatic knees, with the medial area presenting the highest temperature changes (p = 0.020). Patients reporting prevalent pain in the lateral knee area presented higher differences in total knee temperature (0.7 ± 0.7 °C) than patients with pain in the medial area (0.1 ± 0.5 °C) (p = 0.023). CONCLUSION The temperature of knees affected by OA is influenced by the degree of joint degeneration and by the presence of symptoms, with higher temperatures found in symptomatic joints, especially with prevalent lateral knee pain, and in more severe OA. Symptomatic knees demonstrated a different pattern compared to asymptomatic knees, with the medial area presenting the highest temperature changes.
Collapse
Affiliation(s)
- Luca De Marziani
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli, 1, Bologna, 40136, Italy
| | - Lorenzo Zanasi
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli, 1, Bologna, 40136, Italy
| | - Giacomo Roveda
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli, 1, Bologna, 40136, Italy
| | - Angelo Boffa
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli, 1, Bologna, 40136, Italy.
| | - Luca Andriolo
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli, 1, Bologna, 40136, Italy
| | - Alessandro Di Martino
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli, 1, Bologna, 40136, Italy
| | - Stefano Zaffagnini
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli, 1, Bologna, 40136, Italy
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
18
|
Pattison LA, Rickman RH, Hilton H, Dannawi M, Wijesinghe SN, Ladds G, Yang LV, Jones SW, Smith ESJ. Activation of the proton-sensing GPCR, GPR65 on fibroblast-like synoviocytes contributes to inflammatory joint pain. Proc Natl Acad Sci U S A 2024; 121:e2410653121. [PMID: 39661058 PMCID: PMC11665855 DOI: 10.1073/pnas.2410653121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Inflammation is associated with localized acidosis, however, attributing physiological and pathological roles to proton-sensitive receptors is challenging due to their diversity and widespread expression. Here, agonists of the proton-sensing GPCR, GPR65, were systematically characterized. The synthetic agonist BTB09089 (BTB) recapitulated many proton-induced signaling events and demonstrated selectivity for GPR65. BTB was used to show that GPR65 activation on fibroblast-like synoviocytes (FLS), cells that line synovial joints, results in the secretion of proinflammatory mediators capable of recruiting immune cells and sensitizing sensory neurons. Intra-articular injection of BTB resulted in GPR65-dependent sensitization of knee-innervating neurons and nocifensive behaviors in mice. Stimulation of GPR65 on human FLS also triggered the release of inflammatory mediators and synovial fluid samples from human osteoarthritis patients were shown to activate GPR65. These results suggest a role of GPR65 in mediating cell-cell interactions that drive inflammatory joint pain in both mice and humans.
Collapse
Affiliation(s)
- Luke A. Pattison
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Rebecca H. Rickman
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Maya Dannawi
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Susanne N. Wijesinghe
- Institute of Inflammation and Ageing, University of Birmingham, BirminghamB15 2TT, United Kingdom
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| | - Li V. Yang
- Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC27834
| | - Simon W. Jones
- Institute of Inflammation and Ageing, University of Birmingham, BirminghamB15 2TT, United Kingdom
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, CambridgeCB2 1PD, United Kingdom
| |
Collapse
|
19
|
Liu Z, Sun Y, Pan J, Guo K, Tang Z, Wang X. Single-cell profiling uncovers synovial fibroblast subpopulations associated with chondrocyte injury in osteoarthritis. Front Endocrinol (Lausanne) 2024; 15:1479909. [PMID: 39720254 PMCID: PMC11666364 DOI: 10.3389/fendo.2024.1479909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Background Chondrocytes and synovial cells participate in the pathogenesis of osteoarthritis (OA). Nonetheless, the interactions and correlations between OA synovial cells and chondrocytes remain unclear. This study aims to elucidate the interactions and correlations between OA synovial cells and chondrocytes, so as to deepen understanding of OA pathogenesis. Methods Single-cell sequencing analysis was employed to analyze clusters of synovial and chondrocyte cells within the OA dataset. Through cell interaction analysis, the potential interactions between these two cell types were further explored. Differential gene expression analysis was used to examine the differences among synovial-related cell clusters. Results The study identified specific characteristics of synovial fibroblasts through single-cell sequencing analysis. Subsequent cell interaction analysis revealed interactions and correlations between synovial fibroblast clusters and cell clusters in both damaged and non-damaged cartilages. CILP+ fibroblasts showed significant interactions with non-damaged chondrocytes, while POSTN+ fibroblasts exhibited significant interactions with damaged chondrocytes. Furthermore, differential gene expression analysis revealed that genes such as PRELP, CLU, COMP, TNFRSF12A, INHBA, CILP, and SERPINE2, were significantly upregulated in CILP+ fibroblasts. These genes are involved in promoting cell proliferation, inhibiting inflammatory pathways, and stabilizing cell structure, thereby exerting reparative and protective effects on chondrocytes. In contrast, COL6A3, COL6A1, COL1A2, COL1A1, COL3A1, TGF-β1, MMP2, AEBP1, SPARC, FNDC1, and POSTN were upregulated in POSTN+ fibroblasts. These genes may contribute to chondrocyte damage and further degeneration by promoting chondrocyte catabolism, driving inflammation, activating inflammatory pathways, and facilitating chondrocyte apoptosis and destruction. Conclusion Our study elucidated the interactions and correlations between OA synovial cells and chondrocytes. CILP+ synovial fibroblasts may exert reparative and protective effects on chondrocytes of patients with OA by promoting cell proliferation, inhibiting inflammation, and stabilizing cellular structures, thereby potentially mitigating the progression of cartilage lesions in affected patients. In contrast, POSTN+ synovial fibroblasts may exacerbate chondrocyte deterioration in patients with OA by enhancing degradation, inflammation, and apoptosis, thereby exacerbating cartilage lesions. Investigating the underlying molecular mechanisms between OA synovial cells and chondrocytes refines the understanding of OA pathogenesis and provides valuable insights for the clinical diagnosis and treatment of OA.
Collapse
Affiliation(s)
- Zezhong Liu
- Spinal Surgery, Zhejiang Chinese Medical University Affiliated Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Bonesetting Center, Xiangtan Chinese Medicine Hospital, Xiangtan, Hunan, China
| | - Yongqi Sun
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiaoyi Pan
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kechun Guo
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhi Tang
- Bonesetting Center, Xiangtan Chinese Medicine Hospital, Xiangtan, Hunan, China
| | - Xiaofeng Wang
- Spinal Surgery, Zhejiang Chinese Medical University Affiliated Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
20
|
Henriques J, Berenbaum F, Mobasheri A. Obesity-induced fibrosis in osteoarthritis: Pathogenesis, consequences and novel therapeutic opportunities. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100511. [PMID: 39483440 PMCID: PMC11525450 DOI: 10.1016/j.ocarto.2024.100511] [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: 02/06/2024] [Revised: 07/26/2024] [Accepted: 08/12/2024] [Indexed: 11/03/2024] Open
Abstract
Osteoarthritis (OA) is a significant global burden, affecting more than half a billion people across the world. It is characterized by degeneration and loss of articular cartilage, synovial inflammation, and subchondral bone sclerosis, leading to pain and functional impairment. After age, obesity is a major modifiable risk factor for OA, and it has recently been identified as a chronic disease by the World Health Organization (WHO). Obesity is associated with high morbidity and mortality, imposing a significant cost on individuals and society. Obesity increases the risk of knee OA through increased joint loading, altered body composition, and elevated pro-inflammatory adipokines in the systemic circulation. Moreover, obesity triggers fibrotic processes in different organs and tissues, including those involved in OA. Fibrosis in OA refers to the abnormal accumulation of fibrous tissue within and around the joints. It can be driven by increased adiposity, low-grade inflammation, oxidative stress, and metabolic alterations. However, the clinical outcomes of fibrosis in OA are unclear. This review focuses on the link between obesity and OA, explores the mechanism of obesity-driven fibrosis, and examines potential therapeutic opportunities for targeting fibrotic processes in OA.
Collapse
Affiliation(s)
- João Henriques
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Francis Berenbaum
- Sorbonne University, Paris, France
- Department of Rheumatology, Saint-Antoine Hospital, Assistance Publique-Hopitaux de Paris, Paris, France
- INSERM CRSA, Paris, France
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| |
Collapse
|
21
|
Morgan M, Nazemian V, Thai J, Lin I, Northfield S, Ivanusic JJ. BDNF sensitizes bone and joint afferent neurons at different stages of MIA-induced osteoarthritis. Bone 2024; 189:117260. [PMID: 39299629 DOI: 10.1016/j.bone.2024.117260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
There is emerging evidence that Brain Derived Neurotrophic Factor (BDNF), and one of its receptors TrkB, play important roles in the pathogenesis of osteoarthritis (OA) pain. Whilst these studies clearly highlight the potential for targeting BDNF/TrkB signaling to treat OA pain, the mechanism for how BDNF/TrkB signaling contributes to OA pain remains unclear. In this study, we used an animal model of mono-iodoacetate (MIA)-induced OA, in combination with electrophysiology, behavioral testing, Western blot analysis, and retrograde tracing and immunohistochemistry, to identify roles for BDNF/TrkB signaling in the pathogenesis of OA pain. We found that: 1) TrkB is expressed in myelinated medium diameter neurons that innervate the knee joint and bone in naïve animals; 2) peripheral application of BDNF increases the sensitivity of Aδ, but not C knee joint and bone afferent neurons, in response to mechanical stimulation, in naïve animals; 3) BDNF expression increases in synovial tissue in early MIA-induced OA, when pathology is confined to the joint, and in the subchondral bone in late MIA-induced OA, when there is additional damage to the surrounding bone; and 4) TrkB inhibition reverses MIA-induced changes in the sensitivity of Aδ but not C knee joint afferent neurons early in MIA-induced OA, and Aδ but not C bone afferent neurons late in MIA-induced OA. Our findings suggest that BDNF/TrkB signaling may have a role to play in the pathogenesis of OA pain, through effects on knee joint afferent neurons early in disease when there is inflammation confined to the joint, and bone afferent neurons late in disease when there is involvement of damage to subchondral bone. Targeted manipulation of BDNF/TrkB signaling may provide therapeutic benefit for the management of OA pain.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Vida Nazemian
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Irene Lin
- Department of Biochemistry and Pharmacology, University of Melbourne, Victoria, Australia
| | - Susan Northfield
- Department of Biochemistry and Pharmacology, University of Melbourne, Victoria, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
22
|
Wijesinghe SN, Ditchfield C, Flynn S, Agrawal J, Davis ET, Dajas-Bailador F, Chapman V, Jones SW. Immunomodulation and fibroblast dynamics driving nociceptive joint pain within inflammatory synovium: Unravelling mechanisms for therapeutic advancements in osteoarthritis. Osteoarthritis Cartilage 2024; 32:1358-1370. [PMID: 38960140 DOI: 10.1016/j.joca.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/21/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVE Synovitis is a widely accepted sign of osteoarthritis (OA), characterised by tissue hyperplasia, where increased infiltration of immune cells and proliferation of resident fibroblasts adopt a pro-inflammatory phenotype, and increased the production of pro-inflammatory mediators that are capable of sensitising and activating sensory nociceptors, which innervate the joint tissues. As such, it is important to understand the cellular composition of synovium and their involvement in pain sensitisation to better inform the development of effective analgesics. METHODS Studies investigating pain sensitisation in OA with a focus on immune cells and fibroblasts were identified using PubMed, Web of Science and SCOPUS. RESULTS In this review, we comprehensively assess the evidence that cellular crosstalk between resident immune cells or synovial fibroblasts with joint nociceptors in inflamed OA synovium contributes to peripheral pain sensitisation. Moreover, we explore whether the elucidation of common mechanisms identified in similar joint conditions may inform the development of more effective analgesics specifically targeting OA joint pain. CONCLUSION The concept of local environment and cellular crosstalk within the inflammatory synovium as a driver of nociceptive joint pain presents a compelling opportunity for future research and therapeutic advancements.
Collapse
Affiliation(s)
- Susanne N Wijesinghe
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Caitlin Ditchfield
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Sariah Flynn
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| | - Jyoti Agrawal
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK.
| | | | | | - Victoria Chapman
- Pain Centre Versus Arthritis, NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
23
|
Rocha PS, Silva AA, Queiroz-Junior CM, Braga AD, Moreira TP, Teixeira MM, Amaral FA. Trained immunity of synovial macrophages is associated with exacerbated joint inflammation and damage after Staphylococcus aureus infection. Inflamm Res 2024; 73:1995-2008. [PMID: 39340660 DOI: 10.1007/s00011-024-01946-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVES Investigate whether and which synoviocytes would acquire trained immunity characteristics that could exacerbate joint inflammation following a secondary Staphylococcus aureus infection. METHODS Lipopolysaccharide (LPS) and S. aureus were separately or double injected (21 days of interval) into the tibiofemoral joint cavity of male C57BL/6 mice. At different time points after these stimulations, mechanical nociception was analyzed followed by the analysis of signs of inflammation and damage in the affected joints. The trained immunity markers, including the glycolytic and mTOR pathway, were analyzed in whole tissue or isolated synoviocytes. A group of mice was treated with Rapamycin, an mTOR inhibitor before LPS or S. aureus stimulation. RESULTS The double LPS - S. aureus hit promoted intense joint inflammation and damage compared to single joint stimulation, including markers in synoviocyte activation, production of proinflammatory cytokines, persistent nociception, and bone damage, despite not reducing the bacterial clearance. The double LPS - S. aureus hit joints increased the synovial macrophage population expressing CX3CR1 alongside triggering established epigenetic modifications associated with trained immunity events in these cells, such as the upregulation of the mTOR signaling pathway (p-mTOR and HIF1α) and the trimethylation of histone H3. Mice treated with Rapamycin presented reduced CX3CR1+ macrophage activation, joint inflammation, and bone damage. CONCLUSIONS There is a trained immunity phenotype in CX3CR1+ synovial macrophages that contributes to the exacerbation of joint inflammation and damage during septic arthritis caused by S. aureus.
Collapse
Affiliation(s)
- Peter Silva Rocha
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Adryan Aparecido Silva
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Amanda Dias Braga
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Thaiane Pinto Moreira
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Flávio Almeida Amaral
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil.
| |
Collapse
|
24
|
Bergman RF, Lammlin L, Junginger L, Farrell E, Goldman S, Darcy R, Rasner C, Obeidat AM, Malfait AM, Miller RE, Maerz T. Sexual dimorphism of the synovial transcriptome underpins greater PTOA disease severity in male mice following joint injury. Osteoarthritis Cartilage 2024; 32:1060-1073. [PMID: 37716404 DOI: 10.1016/j.joca.2023.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a disease with sex-dependent prevalence and severity in both human and animal models. We sought to elucidate sex differences in synovitis, mechanical sensitization, structural damage, bone remodeling, and the synovial transcriptome in the anterior cruciate ligament rupture (ACLR) mouse model of post-traumatic OA (PTOA). DESIGN Male and female 12-week-old C57/BL6J mice were randomized to Sham or noninvasive ACLR with harvests at 7d or 28d post-ACLR (n = 9 per sex in each group - Sham, 7d ACLR, 28d ACLR). Knee hyperalgesia, mechanical allodynia, and intra-articular matrix metalloproteinase (MMP) activity (via intravital imaging) were measured longitudinally. Trabecular and subchondral bone (SCB) remodeling and osteophyte formation were assessed by µCT. Histological scoring of PTOA, synovitis, and anti-MMP13 immunostaining were performed. NaV1.8-Cre;tdTomato mice were used to document localization and sprouting of nociceptors. Bulk RNA-seq of synovium in Sham, 7d, and 28d post-ACLR, and contralateral joints (n = 6 per group per sex) assessed injury-induced and sex-dependent gene expression. RESULTS Male mice exhibited more severe joint damage at 7d and 28d and more severe synovitis at 28d, accompanied by 19% greater MMP activity, 8% lower knee hyperalgesia threshold, and 43% lower hindpaw withdrawal threshold in injured limbs compared to female injured limbs. Females had injury-induced catabolic responses in trabecular and SCB, whereas males exhibited 133% greater normalized osteophyte volume relative to females and sclerotic remodeling of trabecular and SCB. NaV1.8+ nociceptor sprouting in SCB and medial synovium was induced by injury and comparable between sexes. RNA-seq of synovium demonstrated similar injury-induced transcriptomic programs between the sexes at 7d, but only female mice exhibited a transcriptomic signature indicative of synovial inflammatory resolution by 28d, whereas males had persistent pro-inflammatory, pro-fibrotic, pro-neurogenic, and pro-angiogenic gene expression. CONCLUSION Male mice exhibited more severe overall joint damage and pain behavior after ACLR, which was associated with persistent activation of synovial inflammatory, fibrotic, and neuroangiogenic processes, implicating persistent synovitis in driving sex differences in murine PTOA.
Collapse
Affiliation(s)
- Rachel F Bergman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Lindsey Lammlin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Lucas Junginger
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Easton Farrell
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Sam Goldman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Rose Darcy
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Cody Rasner
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Tristan Maerz
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
25
|
Zhao L, Lai Y, Jiao H, Li J, Lu K, Huang J. CRISPR-mediated Sox9 activation and RelA inhibition enhance cell therapy for osteoarthritis. Mol Ther 2024; 32:2549-2562. [PMID: 38879753 PMCID: PMC11405173 DOI: 10.1016/j.ymthe.2024.06.016] [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: 10/25/2023] [Revised: 03/10/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
Osteoarthritis (OA) is a painful and debilitating disease affecting over 500 million people worldwide. Intraarticular injection of mesenchymal stromal cells (MSCs) shows promise for the clinical treatment of OA, but the lack of consistency in MSC preparation and application makes it difficult to further optimize MSC therapy and to properly evaluate the clinical outcomes. In this study, we used Sox9 activation and RelA inhibition, both mediated by the CRISPR-dCas9 technology simultaneously, to engineer MSCs with enhanced chondrogenic potential and downregulated inflammatory responses. We found that both Sox9 and RelA could be fine-tuned to the desired levels, which enhances the chondrogenic and immunomodulatory potentials of the cells. Intraarticular injection of modified cells significantly attenuated cartilage degradation and palliated OA pain compared with the injection of cell culture medium or unmodified cells. Mechanistically, the modified cells promoted the expression of factors beneficial to cartilage integrity, inhibited the production of catabolic enzymes in osteoarthritic joints, and suppressed immune cells. Interestingly, a substantial number of modified cells could survive in the cartilaginous tissues including articular cartilage and meniscus. Together, our results suggest that CRISPR-dCas9-based gene regulation is useful for optimizing MSC therapy for OA.
Collapse
Affiliation(s)
- Lan Zhao
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA.
| | - Yumei Lai
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Hongli Jiao
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Jun Li
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Ke Lu
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Jian Huang
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
26
|
Arruda AL, Katsoula G, Chen S, Reimann E, Kreitmaier P, Zeggini E. The Genetics and Functional Genomics of Osteoarthritis. Annu Rev Genomics Hum Genet 2024; 25:239-257. [PMID: 39190913 DOI: 10.1146/annurev-genom-010423-095636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Osteoarthritis is the most prevalent whole-joint degenerative disorder, and is characterized by the degradation of articular cartilage and the underlying bone structures. Almost 600 million people are affected by osteoarthritis worldwide. No curative treatments are available, and management strategies focus mostly on pain relief. Here, we provide a comprehensive overview of the available human genetic and functional genomics studies for osteoarthritis to date and delineate how these studies have helped shed light on disease etiopathology. We highlight genetic discoveries from genome-wide association studies and provide a detailed overview of molecular-level investigations in osteoarthritis tissues, including methylation-, transcriptomics-, and proteomics-level analyses. We review how functional genomics data from different molecular levels have helped to prioritize effector genes that can be used as drug targets or drug-repurposing opportunities. Finally, we discuss future directions with the potential to drive a step change in osteoarthritis research.
Collapse
Affiliation(s)
- Ana Luiza Arruda
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- Munich School for Data Science, Helmholtz Munich, Neuherberg, Germany
| | - Georgia Katsoula
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Shibo Chen
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
| | - Ene Reimann
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Peter Kreitmaier
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| |
Collapse
|
27
|
Peng Y, Yang Z, Li J, Liu S. Research progress on nanotechnology of traditional Chinese medicine to enhance the therapeutic effect of osteoarthritis. Drug Deliv Transl Res 2024; 14:1517-1534. [PMID: 38225521 DOI: 10.1007/s13346-024-01517-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2023] [Indexed: 01/17/2024]
Abstract
Osteoarthritis (OA) is a prevalent chronic condition that primarily impacts the articular cartilage and surrounding bone tissue, resulting in joint inflammation and structural deterioration. The etiology of OA is multifaceted and intricately linked to the oxidative stress response of joint tissue. Oxidative stress (OS) in OA leads to the creation of reactive oxygen species (ROS) and other oxidizing agents, resulting in detrimental effects on chondrocytes. This oxidative damage diminishes the flexibility and robustness of cartilage, thereby expediting the progression of joint deterioration. Therefore, the antioxidant effect is crucial in the treatment of OA. Currently, a considerable number of components found in traditional Chinese medicine (TCM) have been scientifically demonstrated to exhibit remarkable antioxidant and anti-inflammatory properties. Nevertheless, the utilization of this program is considerably constrained as a result of intrinsic deficiencies, notably stability concerns. The successful amalgamation of TCM components with nanotechnology has properly tackled these concerns and enhanced the efficacy of therapeutic results. The objective of this study is to delineate the antioxidant characteristics of nano-TCM and assess the current inventory of literature pertaining to the application of nano-TCM in the treatment of OA. In conclusion, this paper will now turn to the constraints and potential avenues for the advancement of nano-TCM within the realm of OA therapy.
Collapse
Affiliation(s)
- Yue Peng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, 530021, Nanning, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, 530021, Nanning, Guangxi, People's Republic of China
| | - Zhengshuang Yang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, 530021, Nanning, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, 530021, Nanning, Guangxi, People's Republic of China
| | - Jinling Li
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, 530021, Nanning, Guangxi, People's Republic of China.
- Laboratory of Basic Medicine Center, Guangxi Medical University, Shuangyong Road, 530021, Nanning, Guangxi, People's Republic of China.
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, 530021, Nanning, Guangxi, People's Republic of China.
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, 530021, Nanning, Guangxi, People's Republic of China.
| |
Collapse
|
28
|
Xu S, Jiemy WF, Brouwer E, Burgess JK, Heeringa P, van der Geest KSM, Alba-Rovira R, Corbera-Bellalta M, Boots AH, Cid MC, Sandovici M. Current evidence on the role of fibroblasts in large-vessel vasculitides: From pathogenesis to therapeutics. Autoimmun Rev 2024; 23:103574. [PMID: 38782083 DOI: 10.1016/j.autrev.2024.103574] [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: 03/11/2024] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Large-vessel vasculitides (LVV) comprise a group of chronic inflammatory diseases of the aorta and its major branches. The most common forms of LVV are giant cell arteritis (GCA) and Takayasu arteritis (TAK). Both GCA and TAK are characterized by granulomatous inflammation of the vessel wall accompanied by a maladaptive immune and vascular response that promotes vascular damage and remodeling. The inflammatory process in LVV starts in the adventitia where fibroblasts constitute the dominant cell population. Fibroblasts are traditionally recognized for synthesizing and renewing the extracellular matrix thereby being major players in maintenance of normal tissue architecture and in tissue repair. More recently, fibroblasts have emerged as a highly plastic cell population exerting various functions, including the regulation of local immune processes and organization of immune cells at the site of inflammation through production of cytokines, chemokines and growth factors as well as cell-cell interaction. In this review, we summarize and discuss the current knowledge on fibroblasts in LVV. Furthermore, we identify key questions that need to be addressed to fully understand the role of fibroblasts in the pathogenesis of LVV.
Collapse
Affiliation(s)
- Shuang Xu
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - William F Jiemy
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Elisabeth Brouwer
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, the Netherlands
| | - Peter Heeringa
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, the Netherlands
| | - Kornelis S M van der Geest
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Roser Alba-Rovira
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marc Corbera-Bellalta
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Annemieke H Boots
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Maria C Cid
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Sandovici
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands.
| |
Collapse
|
29
|
Lin P, Gan YB, He J, Lin SE, Xu JK, Chang L, Zhao LM, Zhu J, Zhang L, Huang S, Hu O, Wang YB, Jin HJ, Li YY, Yan PL, Chen L, Jiang JX, Liu P. Advancing skeletal health and disease research with single-cell RNA sequencing. Mil Med Res 2024; 11:33. [PMID: 38816888 PMCID: PMC11138034 DOI: 10.1186/s40779-024-00538-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
Orthopedic conditions have emerged as global health concerns, impacting approximately 1.7 billion individuals worldwide. However, the limited understanding of the underlying pathological processes at the cellular and molecular level has hindered the development of comprehensive treatment options for these disorders. The advent of single-cell RNA sequencing (scRNA-seq) technology has revolutionized biomedical research by enabling detailed examination of cellular and molecular diversity. Nevertheless, investigating mechanisms at the single-cell level in highly mineralized skeletal tissue poses technical challenges. In this comprehensive review, we present a streamlined approach to obtaining high-quality single cells from skeletal tissue and provide an overview of existing scRNA-seq technologies employed in skeletal studies along with practical bioinformatic analysis pipelines. By utilizing these methodologies, crucial insights into the developmental dynamics, maintenance of homeostasis, and pathological processes involved in spine, joint, bone, muscle, and tendon disorders have been uncovered. Specifically focusing on the joint diseases of degenerative disc disease, osteoarthritis, and rheumatoid arthritis using scRNA-seq has provided novel insights and a more nuanced comprehension. These findings have paved the way for discovering novel therapeutic targets that offer potential benefits to patients suffering from diverse skeletal disorders.
Collapse
Grants
- 2022YFA1103202 National Key Research and Development Program of China
- 82272507 National Natural Science Foundation of China
- 32270887 National Natural Science Foundation of China
- 32200654 National Natural Science Foundation of China
- CSTB2023NSCQ-ZDJO008 Natural Science Foundation of Chongqing
- BX20220397 Postdoctoral Innovative Talent Support Program
- SFLKF202201 Independent Research Project of State Key Laboratory of Trauma and Chemical Poisoning
- 2021-XZYG-B10 General Hospital of Western Theater Command Research Project
- 14113723 University Grants Committee, Research Grants Council of Hong Kong, China
- N_CUHK472/22 University Grants Committee, Research Grants Council of Hong Kong, China
- C7030-18G University Grants Committee, Research Grants Council of Hong Kong, China
- T13-402/17-N University Grants Committee, Research Grants Council of Hong Kong, China
- AoE/M-402/20 University Grants Committee, Research Grants Council of Hong Kong, China
Collapse
Affiliation(s)
- Peng Lin
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yi-Bo Gan
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jian He
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, the General Hospital of Western Theater Command, Chengdu, 610031, China
| | - Si-En Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, 999077, China
| | - Jian-Kun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, 999077, China
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, 999077, China
| | - Li-Ming Zhao
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Sacramento, CA, 94305, USA
| | - Jun Zhu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liang Zhang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Sha Huang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ou Hu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ying-Bo Wang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huai-Jian Jin
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yang-Yang Li
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Pu-Lin Yan
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma and Chemical Poisoning, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jian-Xin Jiang
- Wound Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Peng Liu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
30
|
Dai Y, Chen L, Zhang Z, Liu X. Identification and validation of immune-related genes in osteoarthritic synovial fibroblasts. Heliyon 2024; 10:e28330. [PMID: 38571590 PMCID: PMC10988018 DOI: 10.1016/j.heliyon.2024.e28330] [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: 07/10/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024] Open
Abstract
Objective OA was generally considered as a non-inflammatory disease dominated by articular cartilage degeneration. However, the role of synovitis in OA pathogenesis has received increasing attention. Recent studies support that OA patients have a pro-inflammatory/catabolic synovial environment similar to RA patients, promoting the occurrence and development of OA. Therefore, we investigated the co-immune-related genes and pathways of OA and RA to explore whether part of the pathogenesis of RA synovitis can be used to explain OA synovitis. Methods Data of GSE29746 and GSE12021 were downloaded from the Gene Expression Omnibus (GEO) database. Compared with control group, differentially expressed genes (DEGs) of OA and RA groups were screened separately by R software, Venny website was used to screen co-DEGs. Metascape was used to screen the common enriched terms and pathways between OA and RA. STRING website and Cytoscape software were used to map protein-protein interaction (PPI) networks and screen co-hub genes. GSE29746 was selected as the test dataset, and GSE12021 as the validation dataset for validate the co-hub genes. The results were validated by western blotting (WB) and real-time quantitative polymerase chain reaction (qPCR) of clinical synovial samples. Results We identified 573 OA-related DEGs, 148 RA-related DEGs, and 52 co-DEGs, revealing 14 common enriched terms, most of which were related to immune inflammation. IL7R was the only upregulated co-hub gene between OA and RA in the PPI network, consistent with the validation dataset. IL7R was highly expressed in clinical osteoarthritic synovial samples (P < 0.001). Conclusion Our findings suggested that IL7R is a critical co-DEG in OA and RA and confirmed the involvement of immune inflammation in disease pathogenesis. Furthermore, it confirms the role of IL7R in synovial inflammation in RA and OA synovitis and provides evidence for further investigation of OA immune inflammation.
Collapse
Affiliation(s)
- Yaduan Dai
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lin Chen
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhan Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xueyong Liu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Bai Z, Bartelo N, Aslam M, Murphy EA, Hale CR, Blachere NE, Parveen S, Spolaore E, DiCarlo E, Gravallese EM, Smith MH, Accelerating Medicines Partnership RA/SLE Network, Frank MO, Jiang CS, Zhang H, Pyrgaki C, Lewis MJ, Sikandar S, Pitzalis C, Lesnak JB, Mazhar K, Price TJ, Malfait AM, Miller RE, Zhang F, Goodman S, Darnell RB, Wang F, Orange DE. Synovial fibroblast gene expression is associated with sensory nerve growth and pain in rheumatoid arthritis. Sci Transl Med 2024; 16:eadk3506. [PMID: 38598614 PMCID: PMC11931728 DOI: 10.1126/scitranslmed.adk3506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 03/21/2024] [Indexed: 04/12/2024]
Abstract
It has been presumed that rheumatoid arthritis (RA) joint pain is related to inflammation in the synovium; however, recent studies reveal that pain scores in patients do not correlate with synovial inflammation. We developed a machine-learning approach (graph-based gene expression module identification or GbGMI) to identify an 815-gene expression module associated with pain in synovial biopsy samples from patients with established RA who had limited synovial inflammation at arthroplasty. We then validated this finding in an independent cohort of synovial biopsy samples from patients who had early untreated RA with little inflammation. Single-cell RNA sequencing analyses indicated that most of these 815 genes were most robustly expressed by lining layer synovial fibroblasts. Receptor-ligand interaction analysis predicted cross-talk between human lining layer fibroblasts and human dorsal root ganglion neurons expressing calcitonin gene-related peptide (CGRP+). Both RA synovial fibroblast culture supernatant and netrin-4, which is abundantly expressed by lining fibroblasts and was within the GbGMI-identified pain-associated gene module, increased the branching of pain-sensitive murine CGRP+ dorsal root ganglion neurons in vitro. Imaging of solvent-cleared synovial tissue with little inflammation from humans with RA revealed CGRP+ pain-sensing neurons encasing blood vessels growing into synovial hypertrophic papilla. Together, these findings support a model whereby synovial lining fibroblasts express genes associated with pain that enhance the growth of pain-sensing neurons into regions of synovial hypertrophy in RA.
Collapse
Affiliation(s)
- Zilong Bai
- Weill Cornell Medicine, New York, NY 10065, USA
| | | | | | | | - Caryn R. Hale
- Rockefeller University, New York, NY 10065, USA
- Memorial Sloan Kettering Cancer Center, New York, NY 10065,
USA
| | - Nathalie E. Blachere
- Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, Rockefeller University,
New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | - Myles J. Lewis
- Queen Mary University of London & NIHR BRC Barts Health
NHS Trust, London E1 4NS, UK
| | - Shafaq Sikandar
- Queen Mary University of London & NIHR BRC Barts Health
NHS Trust, London E1 4NS, UK
| | - Costantino Pitzalis
- Queen Mary University of London & NIHR BRC Barts Health
NHS Trust, London E1 4NS, UK
- Department of Biomedical Sciences, Humanitas University
& IRCC Humanitas Research Hospital, Milan 20072, Italy
| | | | | | | | | | | | - Fan Zhang
- University of Colorado School of Medicine, Aurora, CO
80045, USA
| | - Susan Goodman
- Hospital for Special Surgery, New York, NY 10021,
USA
| | - Robert B. Darnell
- Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, Rockefeller University,
New York, NY 10065, USA
| | - Fei Wang
- Weill Cornell Medicine, New York, NY 10065, USA
| | - Dana E. Orange
- Rockefeller University, New York, NY 10065, USA
- Hospital for Special Surgery, New York, NY 10021,
USA
| |
Collapse
|
32
|
Barakat A, Munro G, Heegaard AM. Finding new analgesics: Computational pharmacology faces drug discovery challenges. Biochem Pharmacol 2024; 222:116091. [PMID: 38412924 DOI: 10.1016/j.bcp.2024.116091] [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: 10/02/2023] [Revised: 01/10/2024] [Accepted: 02/23/2024] [Indexed: 02/29/2024]
Abstract
Despite the worldwide prevalence and huge burden of pain, pain is an undertreated phenomenon. Currently used analgesics have several limitations regarding their efficacy and safety. The discovery of analgesics possessing a novel mechanism of action has faced multiple challenges, including a limited understanding of biological processes underpinning pain and analgesia and poor animal-to-human translation. Computational pharmacology is currently employed to face these challenges. In this review, we discuss the theory, methods, and applications of computational pharmacology in pain research. Computational pharmacology encompasses a wide variety of theoretical concepts and practical methodological approaches, with the overall aim of gaining biological insight through data acquisition and analysis. Data are acquired from patients or animal models with pain or analgesic treatment, at different levels of biological organization (molecular, cellular, physiological, and behavioral). Distinct methodological algorithms can then be used to analyze and integrate data. This helps to facilitate the identification of biological molecules and processes associated with pain phenotype, build quantitative models of pain signaling, and extract translatable features between humans and animals. However, computational pharmacology has several limitations, and its predictions can provide false positive and negative findings. Therefore, computational predictions are required to be validated experimentally before drawing solid conclusions. In this review, we discuss several case study examples of combining and integrating computational tools with experimental pain research tools to meet drug discovery challenges.
Collapse
Affiliation(s)
- Ahmed Barakat
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, Egypt.
| | | | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Rai MF, Collins KH, Lang A, Maerz T, Geurts J, Ruiz-Romero C, June RK, Ramos Y, Rice SJ, Ali SA, Pastrello C, Jurisica I, Thomas Appleton C, Rockel JS, Kapoor M. Three decades of advancements in osteoarthritis research: insights from transcriptomic, proteomic, and metabolomic studies. Osteoarthritis Cartilage 2024; 32:385-397. [PMID: 38049029 DOI: 10.1016/j.joca.2023.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a complex disease involving contributions from both local joint tissues and systemic sources. Patient characteristics, encompassing sociodemographic and clinical variables, are intricately linked with OA rendering its understanding challenging. Technological advancements have allowed for a comprehensive analysis of transcripts, proteomes and metabolomes in OA tissues/fluids through omic analyses. The objective of this review is to highlight the advancements achieved by omic studies in enhancing our understanding of OA pathogenesis over the last three decades. DESIGN We conducted an extensive literature search focusing on transcriptomics, proteomics and metabolomics within the context of OA. Specifically, we explore how these technologies have identified individual transcripts, proteins, and metabolites, as well as distinctive endotype signatures from various body tissues or fluids of OA patients, including insights at the single-cell level, to advance our understanding of this highly complex disease. RESULTS Omic studies reveal the description of numerous individual molecules and molecular patterns within OA-associated tissues and fluids. This includes the identification of specific cell (sub)types and associated pathways that contribute to disease mechanisms. However, there remains a necessity to further advance these technologies to delineate the spatial organization of cellular subtypes and molecular patterns within OA-afflicted tissues. CONCLUSIONS Leveraging a multi-omics approach that integrates datasets from diverse molecular detection technologies, combined with patients' clinical and sociodemographic features, and molecular and regulatory networks, holds promise for identifying unique patient endophenotypes. This holistic approach can illuminate the heterogeneity among OA patients and, in turn, facilitate the development of tailored therapeutic interventions.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Annemarie Lang
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jeroen Geurts
- Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Cristina Ruiz-Romero
- Grupo de Investigación de Reumatología (GIR), Unidad de Proteómica, INIBIC -Hospital Universitario A Coruña, SERGAS, Spain
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA
| | - Yolande Ramos
- Dept. Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Sarah J Rice
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Shabana Amanda Ali
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Chiara Pastrello
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada; Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
| | - C Thomas Appleton
- Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Jason S Rockel
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada.
| |
Collapse
|
34
|
Mukherjee A, Das B. The role of inflammatory mediators and matrix metalloproteinases (MMPs) in the progression of osteoarthritis. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100090. [PMID: 38440290 PMCID: PMC10910010 DOI: 10.1016/j.bbiosy.2024.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is a chronic musculoskeletal disorder characterized by an imbalance between (synthesis) and catabolism (degradation) in altered homeostasis of articular cartilage mediated primarily by the innate immune system. OA degenerates the joints resulting in synovial hyperplasia, degradation of articular cartilage with damage of the structural and functional integrity of the cartilage extracellular matrix, subchondral sclerosis, osteophyte formation, and is characterized by chronic pain, stiffness, and loss of function. Inflammation triggered by factors like biomechanical stress is involved in the development of osteoarthritis. In OA apart from catabolic effects, anti-inflammatory anabolic processes also occur continually. There is also an underlying chronic inflammation present, not only in cartilage tissue but also within the synovium, which perpetuates tissue destruction of the OA joint. The consideration of inflammation in OA considers synovitis and/or other cellular and molecular events in the synovium during the progression of OA. In this review, we have presented the progression of joint degradation that results in OA. The critical role of inflammation in the pathogenesis of OA is discussed in detail along with the dysregulation within the cytokine networks composed of inflammatory and anti-inflammatory cytokines that drive catabolic pathways, inhibit matrix synthesis, and promote cellular apoptosis. OA pathogenesis, fluctuation of synovitis, and its clinical impact on disease progression are presented here along with the role of synovial macrophages in promoting inflammatory and destructive responses in OA. The role of interplay between different cytokines, structure, and function of their receptors in the inter-cellular signaling pathway is further explored. The effect of cytokines in the increased synthesis and release of matrix-decomposing proteolytic enzymes, such as matrix metalloproteinase (MMPs) and a disintegrin-like and metalloproteinase with thrombospondin motif (ADAMTS), is elaborated emphasizing the potential impact of MMPs on the chondrocytes, synovial cells, articular and periarticular tissues, and other immune system cells migrating to the site of inflammation. We also shed light on the pathogenesis of OA via oxidative damage particularly due to nitric oxide (NO) via its angiogenic response to inflammation. We concluded by presenting the current knowledge about the tissue inhibitors of metalloproteinases (TIMPs). Synthetic MMP inhibitors include zinc binding group (ZBG), non-ZBG, and mechanism-based inhibitors, all of which have the potential to be therapeutically beneficial in the treatment of osteoarthritis. Improving our understanding of the signaling pathways and molecular mechanisms that regulate the MMP gene expression, may open up new avenues for the creation of therapies that can stop the joint damage associated with OA.
Collapse
Affiliation(s)
- Anwesha Mukherjee
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| |
Collapse
|
35
|
Gordon C, Trainor J, Shah RJ, Studholme K, Gelman A, Doswell F, Sadar F, Giovannetti A, Gershenson J, Khan A, Nicholson J, Huang Z, Spurgat M, Tang SJ, Wang H, Ojima I, Carlson D, Komatsu DE, Kaczocha M. Fatty acid binding protein 5 inhibition attenuates pronociceptive cytokine/chemokine expression and suppresses osteoarthritis pain: A comparative human and rat study. Osteoarthritis Cartilage 2024; 32:266-280. [PMID: 38035977 PMCID: PMC11283882 DOI: 10.1016/j.joca.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 10/20/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is often accompanied by debilitating pain that is refractory to available analgesics due in part to the complexity of signaling molecules that drive OA pain and our inability to target these in parallel. Fatty acid binding protein 5 (FABP5) is a lipid chaperone that regulates inflammatory pain; however, its contribution to OA pain has not been characterized. DESIGN This combined clinical and pre-clinical study utilized synovial tissues obtained from subjects with end-stage OA and rats with monoiodoacetate-induced OA. Cytokine and chemokine release from human synovia incubated with a selective FABP5 inhibitor was profiled with cytokine arrays and ELISA. Immunohistochemical analyses were conducted for FABP5 in human and rat synovium. The efficacy of FABP5 inhibitors on pain was assessed in OA rats using incapacitance as an outcome. RNA-seq was then performed to characterize the transcriptomic landscape of synovial gene expression in OA rats treated with FABP5 inhibitor or vehicle. RESULTS FABP5 was expressed in human synovium and FABP5 inhibition reduced the secretion of pronociceptive cytokines (interleukin-6 [IL6], IL8) and chemokines (CCL2, CXCL1). In rats, FABP5 was upregulated in the OA synovium and its inhibition alleviated incapacitance. The transcriptome of the rat OA synovium exhibited >6000 differentially expressed genes, including the upregulation of numerous pronociceptive cytokines and chemokines. FABP5 inhibition blunted the upregulation of the majority of these pronociceptive mediators. CONCLUSIONS FABP5 is expressed in the OA synovium and its inhibition suppresses pronociceptive signaling and pain, indicating that FABP5 inhibitors may constitute a novel class of analgesics to treat OA.
Collapse
Affiliation(s)
- Chris Gordon
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - James Trainor
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Rohan J Shah
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Keith Studholme
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Alex Gelman
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Faniya Doswell
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Faisal Sadar
- Department of Orthopaedics and Rehabilitation, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Allessio Giovannetti
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Josh Gershenson
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Ayesha Khan
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - James Nicholson
- Department of Orthopaedics and Rehabilitation, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - ZeYu Huang
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Michael Spurgat
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Shao-Jun Tang
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA; Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Hehe Wang
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
| | - Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA; Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - David Carlson
- Genomics Core Facility and Institute for Advanced Computational Sciences, Stony Brook University, Stony Brook, NY, USA
| | - David E Komatsu
- Department of Orthopaedics and Rehabilitation, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Martin Kaczocha
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA; Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA; Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
36
|
Xu H, Wang Z, Wang Z, Zhou H, Guo J, Li W, Zhou Y. Cerebral Mechanism of Tuina on the Descending Pain Inhibitory System in Knee Osteoarthritis: Protocol for a Randomized Controlled Trial. JMIR Res Protoc 2024; 13:e52820. [PMID: 38238645 PMCID: PMC10897796 DOI: 10.2196/52820] [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: 09/16/2023] [Revised: 12/17/2023] [Accepted: 01/17/2024] [Indexed: 02/13/2024] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is reputedly the most common musculoskeletal disease of the lower limbs and the main cause of pain and disability among older individuals. Pain is the most significant and widespread symptom of KOA. The descending pain inhibitory system has a cardinal role in normal pain consciousness, and its malfunction may be one of the pathophysiological mechanisms in KOA. Crucially, the rostral ventromedial medulla (RVM) and periaqueductal gray (PAG), as important components of the descending pain inhibitory system, directly modulate the activity of the spinal neurons involved in pain transmission. Tuina, a manual therapy, is effective and safe for reducing clinical symptoms of KOA; however, the mechanism that influences pain through the descending pain inhibitory system in KOA is unclear. OBJECTIVE This study aims to investigate the modulatory implications of Tuina on the RVM and PAG, which have critical roles in the descending pain inhibitory system in patients with KOA. METHODS This randomized controlled parallel trial will be conducted at the Tuina Clinic of the Third Affiliated Hospital of Henan University of Chinese Medicine (Zhengzhou, China). Patients with KOA will be randomly assigned (1:1) to 6 weeks of health education or Tuina. All patients in both groups will accept a resting-state functional magnetic resonance scan at the beginning and end of the experiment, and the resting-state functional connectivity and the voxel-based morphometry analysis will be performed to detect the RVM and PAG function and structure changes. The clinical outcome assessments will be (1) the pressure pain thresholds, (2) the Numerical Rating Scale, (3) the Hamilton Depression Scale (HAMD), and (4) the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC). Considering that this trial is a study of resting-state functional magnetic resonance imaging technology, resting-state functional connectivity and voxel-based morphometry are the primary outcomes, and clinical outcome assessments are secondary outcomes. Adverse events will be documented and assessed throughout. All main analyses will be carried out on the basis of the intention-to-treat principle. The outcome evaluators and data statisticians will be masked to the treatment group assignment to reduce the risk of bias. RESULTS This trial was approved by the ethics committee of the Third Affiliated Hospital of Henan University of Chinese Medicine. Enrollment began in December 2023, and the results of this trial are expected to be submitted for publication in May 2025. CONCLUSIONS This trial will identify a possible relationship between function and structure changes of RVM and PAG and the improvement of clinical variables, elucidating the effect of Tuina on the descending pain inhibitory system of patients with KOA. This trial will provide much-needed knowledge for Tuina for patients with KOA. TRIAL REGISTRATION Chinese Clinical Trial Registry ChiCTR2300070289; https://www.chictr.org.cn/showproj.html?proj=182570. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/52820.
Collapse
Affiliation(s)
- Hui Xu
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zheng Wang
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhen Wang
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Hang Zhou
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Juan Guo
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Wanyu Li
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yunfeng Zhou
- School of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
- Tuina Department, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
37
|
Boutet MA, Nerviani A, Fossati-Jimack L, Hands-Greenwood R, Ahmed M, Rivellese F, Pitzalis C. Comparative analysis of late-stage rheumatoid arthritis and osteoarthritis reveals shared histopathological features. Osteoarthritis Cartilage 2024; 32:166-176. [PMID: 37984558 DOI: 10.1016/j.joca.2023.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/20/2023] [Accepted: 10/24/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVES Osteoarthritis (OA) is a debilitating and heterogeneous condition, characterized by various levels of articular cartilage degradation, osteophytes formation, and synovial inflammation. Multiple evidences suggest that synovitis may appear early in the disease development and correlates with disease severity and pain, therefore representing a relevant therapeutic target. In a typical synovitis-driven joint disease, namely rheumatoid arthritis (RA), several pathotypes have been described by our group and associated with clinical phenotypes, disease progression, and response to therapy. However, whether these pathotypes can be also observed in the OA synovium is currently unknown. METHODS Here, using histological approaches combined with semi-quantitative scoring and quantitative digital image analyses, we comparatively characterize the immune cell infiltration in a large cohort of OA and RA synovial tissue samples collected at the time of total joint replacement. RESULTS We demonstrate that OA synovium can be categorized also into three pathotypes and characterized by disease- and stage-specific features. Moreover, we revealed that pathotypes specifically reflect distinct levels of peripheral inflammation. CONCLUSIONS In this study, we provide a novel and relevant pathological classification of OA synovial inflammation. Further studies investigating synovial molecular pathology in OA may contribute to the development of disease-modifying therapies.
Collapse
Affiliation(s)
- Marie-Astrid Boutet
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom; Nantes Université, Oniris, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France.
| | - Alessandra Nerviani
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Liliane Fossati-Jimack
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rebecca Hands-Greenwood
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Manzoor Ahmed
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Felice Rivellese
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Costantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
38
|
Maatuf YH, Marco M, Unger-Gelman S, Farhat E, Zobrab A, Roy A, Kumar A, Carmon I, Reich E, Dvir-Ginzberg M. Diverse Response to Local Pharmacological Blockade of Sirt1 Cleavage in Age-Induced versus Trauma-Induced Osteoarthritis Female Mice. Biomolecules 2024; 14:81. [PMID: 38254681 PMCID: PMC10813022 DOI: 10.3390/biom14010081] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Objective: Previous studies have shown that the cleavage of Sirt1 contributes to the development of osteoarthritis (OA). In fact, OA was effectively abrogated by the intra-articular (IA) administration of two compounds, one blocking Sirt1 cleavage (CA074me) and the other activating Sirt1 (SRT1720), using a post-traumatically induced model (PTOA) in young female mice. In this study, we attempted to understand if this local treatment is effective in preventing age-associated OA (AOA) progression and symptoms. Design: A group of 17-month-old female C57BL/6J mice were IA administered with CA074me and/or SRT1720 or their combination. Joint histopathological analysis and bone histomorphometry were carried out, with an assessment of knee mechanical hyperalgesia. A serum analysis for NT/CT Sirt1 was carried out along with immunohistochemistry for articular cartilage to detect p16INK4A or γH2A.X. Similarly, meniscal cartilage was monitored for Lef1 and Col1a1 deposition. The data were compared for young female mice subjected to post-traumatic OA (PTOA). Results: Similar to PTOA, combination-treated AOA exhibited improved knee hyperalgesia, yet structural improvements were undetected, corresponding to unchanged NT/CT Sirt1 serum levels. Both AOA and PTOA exhibited unchanged staining for nuclear p16INK4A or γH2A.X and lacked a correlation with OA severity. Contrarily to PTOA, the combination treatment with AOA did not exhibit a local reduction in the Lef1 and Col1 targets. Conclusions: When targeting Sirt1 cleavage, the PTOA and AOA models exhibited a similar pain response to the combination treatment; however, they displayed diverse structural outcomes for joint-related damage, related to Lef1-dependent signaling. Interestingly, nuclear p16INK4A was unaffected in both models, regardless of the treatment's effectiveness. Finally, these findings highlight the variations in the responses between two highly researched OA preclinical models, reflecting OA pathophysiology heterogeneity and variations in gender-related drug-response mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Mona Dvir-Ginzberg
- Institute of Biomedical and Oral research, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem 9112102, Israel; (Y.H.M.); (M.M.); (S.U.-G.); (E.F.); (A.Z.); (A.R.); (A.K.); (I.C.); (E.R.)
| |
Collapse
|
39
|
Wu S, Guo W, Chen L, Lin X, Tang M, Lin C, Guo H, Zhang T, Gao Y. Downregulation of Gadd45β alleviates osteoarthritis by repressing lipopolysaccharide-induced fibroblast-like synoviocyte inflammation, proliferation and migration. Int Immunopharmacol 2024; 126:111202. [PMID: 37988908 DOI: 10.1016/j.intimp.2023.111202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023]
Abstract
OBJECTIVE Gadd45β have a regulatory role in cellular inflammation, proliferation and migration. However, the role of Gadd45β in synovial inflammation in osteoarthritis (OA) remains to be explored. This study aimed to ascertain whether Gadd45β is involved in OA synovial inflammation. METHODS The rat model was induced by sodium iodoacetate and the cellular model was constructed with lipopolysaccharide (LPS)-induced fibroblast-like synoviocytes (FLSs). siRNA was applied to interfere with the expression of intracellular Gadd45β. Real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting were used to detect the expression of Gadd45β mRNA and protein. The inflammation, proliferation, and migration of OA-FLSs were detected by enzyme-linked immunosorbent assay, cell scratch assay, 5-ethynyl-2'-deoxyuridine assay, etc. The effect of downregulation of Gadd45β on the nuclear factor-κB (NF-κB) pathway was investigated. RESULTS Expression of Gadd45β in OA rat synovial tissues and OA-FLSs was increased, and LPS treatment promoted cell proliferation and enhanced cell migration. Gadd45β interference inhibited the inflammation, proliferation and migration of cells induced by LPS. LPS promoted P65 expression in the nucleus and activated the NF-κB signaling pathway, whereas si-Gadd45β reversed this situation. CONCLUSIONS si-Gadd45β inhibited the inflammatory response, proliferation and migration of FLSs, and activation of the NF-κB signaling pathway, which could delay the progression of OA. Hence, it may become a potential therapeutic target for OA.
Collapse
Affiliation(s)
- Suyu Wu
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Wenwen Guo
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Ling Chen
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Xinxin Lin
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China; Department of Pathology, Fuzhou Second Hospital, Fuzhou 350007, Fujian, China
| | - Minjie Tang
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Cheng Lin
- The School of Health, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Hanzhi Guo
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Tianwen Zhang
- Fujian Fishery Resources Monitoring Center, Fuzhou 350003, Fujian, China
| | - Yao Gao
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350122, Fujian, China; Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fuzhou 350004, Fujian, China.
| |
Collapse
|
40
|
Gu Y, Hu Y, Zhang H, Wang S, Xu K, Su J. Single-cell RNA sequencing in osteoarthritis. Cell Prolif 2023; 56:e13517. [PMID: 37317049 PMCID: PMC10693192 DOI: 10.1111/cpr.13517] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/30/2023] [Accepted: 05/26/2023] [Indexed: 06/16/2023] Open
Abstract
Osteoarthritis is a progressive and heterogeneous joint disease with complex pathogenesis. The various phenotypes associated with each patient suggest that better subgrouping of tissues associated with genotypes in different phases of osteoarthritis may provide new insights into the onset and progression of the disease. Recently, single-cell RNA sequencing was used to describe osteoarthritis pathogenesis on a high-resolution view surpassing traditional technologies. Herein, this review summarizes the microstructural changes in articular cartilage, meniscus, synovium and subchondral bone that are mainly due to crosstalk amongst chondrocytes, osteoblasts, fibroblasts and endothelial cells during osteoarthritis progression. Next, we focus on the promising targets discovered by single-cell RNA sequencing and its potential applications in target drugs and tissue engineering. Additionally, the limited amount of research on the evaluation of bone-related biomaterials is reviewed. Based on the pre-clinical findings, we elaborate on the potential clinical values of single-cell RNA sequencing for the therapeutic strategies of osteoarthritis. Finally, a perspective on the future development of patient-centred medicine for osteoarthritis therapy combining other single-cell multi-omics technologies is discussed. This review will provide new insights into osteoarthritis pathogenesis on a cellular level and the field of applications of single-cell RNA sequencing in personalized therapeutics for osteoarthritis in the future.
Collapse
Affiliation(s)
- Yuyuan Gu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- School of MedicineShanghai UniversityShanghaiChina
| | - Yan Hu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
| | - Hao Zhang
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
| | - Sicheng Wang
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- Department of OrthopedicsShanghai Zhongye HospitalShanghaiChina
| | - Ke Xu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- Wenzhou Institute of Shanghai UniversityWenzhouChina
| | - Jiacan Su
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
| |
Collapse
|
41
|
Ma Z, Wei Y, Liao T, Jie L, Yang N, Yu L, Wang P. Activation of vascular endothelial cells by synovial fibrosis promotes Netrin-1-induced sensory nerve sprouting and exacerbates pain sensitivity. J Cell Mol Med 2023; 27:3773-3785. [PMID: 37702437 PMCID: PMC10718133 DOI: 10.1111/jcmm.17950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023] Open
Abstract
Synovial fibrosis is one of the most dominant histopathological changes in osteoarthritis of the knee (KOA), and activation of vascular endothelial cells in synovial fibrosis is both an important factor in mediating pain in KOA and a major contributor to the generation of pain signals. At the same time, angiogenesis and nerve fibres are more likely to underlie the pathology of pain induced by synovial fibrosis. In the present study, we established a co-culture model of human umbilical vein endothelial cells (HUVECs) with dorsal root ganglion (DRG) and detected tissue and cellular Netrin-1, vascular cell adhesion molecule-1 (VCAM-1), intercellular cell adhesion molecule-1 (ICAM-1), growth-associated protein-43 (GAP43), colorectal cancer deleted (DCC), uncoordinated 5 (UNC5), and the related expression of calcitonin gene-related peptide (CGRP), substance P (SP) and nerve growth factor (NGF) in supernatant by ELISA to investigate the intervention of vascular endothelial cell activation on sensory nerve sprouting exacerbating peripheral pain sensitivity and to investigate the effect of Netrin-1 from the perspective of Netrin-1 secretion to illustrate its effector mechanism.
Collapse
Affiliation(s)
- Zhenyuan Ma
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Yibao Wei
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Taiyang Liao
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Lishi Jie
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Nan Yang
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Likai Yu
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Peimin Wang
- Department of Orthopaedics and TraumatologyAffiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese MedicineNanjingChina
- Key Laboratory for Metabolic Diseases in Chinese MedicineFirst College of Clinical Medicine, Nanjing University of Chinese MedicineNanjingChina
- Jiangsu Province Hospital of Chinese MedicineNanjingChina
- Jiangsu Provincial Engineering Research Center of TCM External Medication Development and ApplicationNanjingChina
| |
Collapse
|
42
|
Li Z, Zhang S, Mao G, Xu Y, Kang Y, Zheng L, Long D, Chen W, Gu M, Zhang Z, Kang Y, Sheng P, Zhang Z. Identification of anterior cruciate ligament fibroblasts and their contribution to knee osteoarthritis progression using single-cell analyses. Int Immunopharmacol 2023; 125:111109. [PMID: 37883816 DOI: 10.1016/j.intimp.2023.111109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
The mechanical properties of the anterior cruciate ligament (ACL) in the knee have been highlighted, but its role in the regulation of the joint microenvironment remains unclear, especially in the progression of Knee Osteoarthritis (KOA). Here, single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) data were integrated to reveal the transcriptional and epigenomic landscape of ACL in normal and OA states. We identified a novel subpopulation of fibroblasts in ACL, which provides new insights into the role of the ACL in knee homeostasis and disease. Degeneration of the ACL during OA mechanically alters the knee joint homeostasis and influences the microenvironment by regulating inflammatory- and osteogenic-related factors, thereby contributing to the progression of KOA. Additionally, the specific mechanism by which these Inflammation-associated Fibroblasts (IAFs) regulate KOA progression was uncovered, providing new foundation for the development of targeted treatments for KOA.
Collapse
Affiliation(s)
- Zhiwen Li
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Shiyong Zhang
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Guping Mao
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yiyang Xu
- Department of Orthopaedics, Fujian Provincial Hospital, Shengli Clinical Medical College, Fujian Medical University, China
| | - Yunze Kang
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Linli Zheng
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Dianbo Long
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weishen Chen
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Minghui Gu
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhiqi Zhang
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Yan Kang
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Puyi Sheng
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Ziji Zhang
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
43
|
Tsuchiya M, Ohashi Y, Kodera Y, Satoh M, Matsui T, Fukushima K, Iwase D, Aikawa J, Mukai M, Inoue G, Takaso M, Uchida K. CD39+CD55- Fb Subset Exhibits Myofibroblast-Like Phenotype and Is Associated with Pain in Osteoarthritis of the Knee. Biomedicines 2023; 11:3047. [PMID: 38002046 PMCID: PMC10669511 DOI: 10.3390/biomedicines11113047] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Recent studies utilizing single-cell analysis have unveiled the presence of various fibroblast (Fb) subsets within the synovium under inflammatory conditions in osteoarthritis (OA), distinguishing them from those in rheumatoid arthritis (RA). Moreover, it has been reported that pain in knee OA patients is linked to specific fibroblast subsets. Single-cell expression profiling methods offer an incredibly detailed view of the molecular states of individual cells. However, one limitation of these methods is that they require the destruction of cells during the analysis process, rendering it impossible to directly assess cell function. In our study, we employ flow cytometric analysis, utilizing cell surface markers CD39 and CD55, in an attempt to isolate fibroblast subsets and investigate their relationship with OA pathology. Synovial tissues were obtained from 25 knee OA (KOA) patients. Of these, six samples were analyzed by RNA-seq (n = 3) and LC/MS analysis (n = 3). All 25 samples were analyzed to estimate the proportion of Fb (CD45-CD31-CD90+) subset by flow cytometry. The proportion of Fb subsets (CD39+CD55- and CD39-CD55+) and their association with osteoarthritis pathology were evaluated. CD39+CD55- Fb highly expressed myogenic markers such as CNN1, IGFBP7, MYH11, and TPM1 compared to CD39-CD55+ Fb. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of upregulated differentially expressed genes (DEGs) in CD39+CD55- Fb identified the Apelin pathway and cGMP-PKC-signaling pathway as possibly contributing to pain. LC/MS analysis indicated that proteins encoded by myogenic marker genes, including CNN1, IGFBP7, and MYH11, were also significantly higher than in CD39-CD55+ Fb. CD39-CD55+ Fb highly expressed PRG4 genes and proteins. Upregulated DEGs were enriched for pathways associated with proinflammatory states ('RA', 'TNF signaling pathway', 'IL-17 signaling pathway'). The proportion of CD39+CD55- Fb in synovium significantly correlated with both resting and active pain levels in knee OA (KOA) patients (resting pain, ρ = 0.513, p = 0.009; active pain, ρ = 0.483, p = 0.015). There was no correlation between joint space width (JSW) and the proportion of CD39+CD55- Fb. In contrast, there was no correlation between the proportion of CD39-CD55+ Fb and resting pain, active pain, or JSW. In conclusion, CD39+CD55- cells exhibit a myofibroblast phenotype, and its proportion is associated with KOA pain. Our study sheds light on the potential significance of CD39+CD55- synovial fibroblasts in osteoarthritis, their myofibroblast-like phenotype, and their association with joint pain. These findings provide a foundation for further research into the mechanisms underlying fibrosis, the impact of altered gene expression on osteoarthritic joints, and potential therapeutic strategies.
Collapse
Affiliation(s)
- Maho Tsuchiya
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Yoshihisa Ohashi
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Yoshio Kodera
- Department of Physics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Kanagawa, Japan; (Y.K.); (T.M.)
- Center for Disease Proteomics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Kanagawa, Japan
| | - Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan;
| | - Takashi Matsui
- Department of Physics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Kanagawa, Japan; (Y.K.); (T.M.)
- Center for Disease Proteomics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Kanagawa, Japan
| | - Kensuke Fukushima
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Dai Iwase
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Jun Aikawa
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Manabu Mukai
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| |
Collapse
|
44
|
Zou Z, Li H, Yu K, Ma K, Wang Q, Tang J, Liu G, Lim K, Hooper G, Woodfield T, Cui X, Zhang W, Tian K. The potential role of synovial cells in the progression and treatment of osteoarthritis. EXPLORATION (BEIJING, CHINA) 2023; 3:20220132. [PMID: 37933282 PMCID: PMC10582617 DOI: 10.1002/exp.20220132] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/15/2023] [Indexed: 11/08/2023]
Abstract
Osteoarthritis (OA), the commonest arthritis, is characterized by the progressive destruction of cartilage, leading to disability. The Current early clinical treatment strategy for OA often centers on anti-inflammatory or analgesia medication, weight loss, improved muscular function and articular cartilage repair. Although these treatments can relieve symptoms, OA tends to be progressive, and most patients require arthroplasty at the terminal stages of OA. Recent studies have shown a close correlation between joint pain, inflammation, cartilage destruction and synovial cells. Consequently, understanding the potential mechanisms associated with the action of synovial cells in OA could be beneficial for the clinical management of OA. Therefore, this review comprehensively describes the biological functions of synovial cells, the synovium, together with the pathological changes of synovial cells in OA, and the interaction between the cartilage and synovium, which is lacking in the present literature. Additionally, therapeutic approaches based on synovial cells for OA treatment are further discussed from a clinical perspective, highlighting a new direction in the treatment of OA.
Collapse
Affiliation(s)
- Zaijun Zou
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Han Li
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Kai Yu
- Department of Bone and JointCentral Hospital of Zhuang He CityDalianLiaoningChina
| | - Ke Ma
- Department of Clinical MedicineChina Medical UniversityShenyangLiaoningChina
| | - Qiguang Wang
- National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuanChina
| | - Junnan Tang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Guozhen Liu
- School of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdongChina
| | - Khoon Lim
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Gary Hooper
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Tim Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Xiaolin Cui
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdongChina
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Weiguo Zhang
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- Key Laboratory of Molecular Mechanisms for Repair and Remodeling of Orthopaedic DiseasesLiaoning ProvinceDalianLiaoningChina
| | - Kang Tian
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- Key Laboratory of Molecular Mechanisms for Repair and Remodeling of Orthopaedic DiseasesLiaoning ProvinceDalianLiaoningChina
| |
Collapse
|
45
|
Morgan M, Nazemian V, Ooi LS, Burger S, Thai J, Ivanusic J. Artemin sensitizes nociceptors that innervate the osteoarthritic joint to produce pain. Osteoarthritis Cartilage 2023; 31:1342-1352. [PMID: 37353141 DOI: 10.1016/j.joca.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/14/2023] [Accepted: 06/09/2023] [Indexed: 06/25/2023]
Abstract
OBJECTIVE There have been significant developments in understanding artemin/GFRα3 signaling in recent years, and there is now accumulating evidence that artemin has important roles to play in pain signaling, including that derived from joint and bone, and that associated with osteorthritis (OA). METHODS A total of 163 Sprague-Dawley rats were used in this study. We used an animal model of mono-iodoacetate (MIA)-induced OA, in combination with electrophysiology, behavioral testing, Western blot analysis, and retrograde tracing and immunohistochemistry, to identify roles for artemin/GFRα3 signaling in the pathogenesis of OA pain. RESULTS We have found that: 1) GFRα3 is expressed in a substantial proportion of knee joint afferent neurons; 2) exogenous artemin sensitizes knee joint afferent neurons in naïve rats; 3) artemin is expressed in articular tissues of the joint, but not surrounding bone, early in MIA-induced OA; 4) artemin expression increases in bone later in MIA-induced OA when pathology involves subchondral bone; and 5) sequestration of artemin reverses MIA-induced sensitization of both knee joint and bone afferent neurons late in disease when there is inflammation of knee joint tissues and damage to the subchondral bone. CONCLUSIONS Our findings show that artemin/GFRα3 signaling has a role to play in the pathogenesis of OA pain, through effects on both knee joint and bone afferent neurons, and suggest that targeted manipulation of artemin/GFRα3 signaling may provide therapeutic benefit for the management of OA pain. DATA AVAILABILITY Data are available on request of the corresponding author.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Vida Nazemian
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Li Sha Ooi
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Sarah Burger
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Jason Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
46
|
Smith ESJ. A new art to treating osteoarthritis pain? Osteoarthritis Cartilage 2023; 31:1275-1277. [PMID: 37460008 DOI: 10.1016/j.joca.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/10/2023] [Indexed: 07/31/2023]
Affiliation(s)
- Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| |
Collapse
|
47
|
Jin Z, Zhang H, Bai L, Yue L, Zhang W, Liang J, Chang B, Yang Y, Hu Z, Chen L, Guo C. Synovium is a sensitive tissue for mapping the negative effects of systemic iron overload in osteoarthritis: identification and validation of two potential targets. J Transl Med 2023; 21:661. [PMID: 37741987 PMCID: PMC10518090 DOI: 10.1186/s12967-023-04541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/16/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND The prevention and treatment of osteoarthritis (OA) pose a major challenge in its research. The synovium is a critical tissue in the systematic treatment of OA. The present study aimed to investigate potential target genes and their correlation with iron overload in OA patients. METHODS The internal datasets for analysis included the microarray datasets GSE46750, GSE55457, and GSE56409, while the external datasets for validation included GSE12021 and GSE55235. The GSE176308 dataset was used to generate single-cell RNA sequencing profiles. To investigate the expression of the target genes in synovial samples, quantitative reverse transcription-PCR, western blotting, and immunohistochemical assay were conducted. ELISA was used to detect the levels of ferritin and Fe2+ in both serum and synovium. RESULTS JUN and ZFP36 were screened from the differentially expressed genes, and their mRNA were significantly reduced in the OA synovium compared to that in normal synovium. Subsequently, complex and dynamically evolving cellular components were observed in the OA synovium. The mRNA level of JUN and ZFP36 differed across various cell clusters of OA synovium and correlated with immune cell infiltration. Moreover, ferritin and Fe2+ were significantly increased in the serum and synovium of OA patients. Further, we found that JUN elevated and ZFP36 decreased at protein level. CONCLUSIONS The synovium is a sensitive tissue for mapping the adverse effects of systemic iron overload in OA. JUN and ZFP36 represent potential target genes for attenuating iron overload during OA treatment. Some discrepancies between the transcription and protein levels of JUN suggest that post-transcriptional modifications may be implicated. Future studies should also focus on the roles of JUN and ZFP36 in inducing changes in cellular components in the synovium during OA pathogenesis.
Collapse
Affiliation(s)
- Zhuangzhuang Jin
- Department of Emergence Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Zhang
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lingyu Yue
- Beijing AnDing Hospital of Capital Medical University, Beijing, China
| | - Weiming Zhang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiajian Liang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bohan Chang
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Yang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhehan Hu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liang Chen
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chuanji Guo
- Hospital Administration Office, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang City, Liaoning Province, China.
| |
Collapse
|
48
|
Chen X, Gong L, Li C, Wang S, Wang Z, Chu M, Zhou Y. Single-cell and bulk tissue sequencing unravels the heterogeneity of synovial microenvironment in arthrofibrosis. iScience 2023; 26:107379. [PMID: 37705954 PMCID: PMC10495645 DOI: 10.1016/j.isci.2023.107379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/06/2023] [Accepted: 07/07/2023] [Indexed: 09/15/2023] Open
Abstract
Arthrofibrosis (AF) is a debilitating complication that occurs after trauma or surgery, leading to functional impairment and surgical failures worldwide. This study aimed to uncover the underlying mechanism of AF. A total of 141 patients were enrolled, and synovial samples were collected from both patients and animal models at different time points. Single-cell RNA-sequencing (scRNA-seq) and bulk tissue RNA sequencing (bulk-seq) were employed to profile the distinct synovial microenvironment. This study revealed changes in cell proportions during AF pathogenesis and identified Engrailed-1 (EN1) as a key transcription factor strongly associated with disease severity and clinical prognosis. Additionally, the researchers discovered a specific type of synovial fibroblast called DKK3-SLF, which played a critical role in driving AF development. These findings shed light on the composition and heterogeneity of the synovial microenvironment in AF, offering potential avenues for identifying therapeutic targets and developing clinical treatments for AF and other fibrotic diseases.
Collapse
Affiliation(s)
- Xi Chen
- Department of Adult Joint Reconstructive Surgery, Beijing Jishuitan Hospital, Capital Medical University, 31 East Xinjiekou Street, Beijing 100035, China
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Lihua Gong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Cheng Li
- Department of Adult Joint Reconstructive Surgery, Beijing Jishuitan Hospital, Capital Medical University, 31 East Xinjiekou Street, Beijing 100035, China
| | - Siyuan Wang
- Department of Adult Joint Reconstructive Surgery, Beijing Jishuitan Hospital, Capital Medical University, 31 East Xinjiekou Street, Beijing 100035, China
| | - Ziyuan Wang
- Department of Adult Joint Reconstructive Surgery, Beijing Jishuitan Hospital, Capital Medical University, 31 East Xinjiekou Street, Beijing 100035, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Yixin Zhou
- Department of Adult Joint Reconstructive Surgery, Beijing Jishuitan Hospital, Capital Medical University, 31 East Xinjiekou Street, Beijing 100035, China
| |
Collapse
|
49
|
Chen Z, Wang W, Zhang Y, Xue X, Hua Y. Identification of four-gene signature to diagnose osteoarthritis through bioinformatics and machine learning methods. Cytokine 2023; 169:156300. [PMID: 37454542 DOI: 10.1016/j.cyto.2023.156300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/02/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Although osteoarthritis (OA) is one of the most prevalent joint disorders, effective biomarkers to diagnose OA are still unavailable. This study aimed to acquire some key synovial biomarkers (hub genes) and analyze their correlation with immune infiltration in OA. METHODS Gene expression profiles and clinical characteristics of OA and healthy synovial samples were retrieved from the Gene Expression Omnibus (GEO) database. Hub genes for OA were mined based on a combination of weighted gene co-expression network analysis (WGCNA), the least absolute shrinkage and selection operator (LASSO), support vector machine recursive feature elimination (SVM-RFE), and random forest (RF) algorithms. A diagnostic nomogram model for OA prediction was developed based on the hub genes. Receiver operating characteristic curves (ROC) were performed to confirm the abnormal expression of hub genes in the experimemtal and validation datasets. qRT-PCR using patients' samples were conducted as well. In addition, the infiltration level of 28 immune cells in the expression profile and their relationship with hub genes were analyzed using single-sample GSEA (ssGSEA). RESULTS 4 hub genes (ZBTB16, TNFSF11, SCRG1 and KDELR3) were obtained by WGCNA, lasso, SVM-RFE, RF algorithms as potential biomarkers for OA. The immune infiltration analyses revealed that hub genes were most correlated with regulatory T cell and natural killer cell. CONCLUSION A machine learning model to diagnose OA based on ZBTB16, TNFSF11, SCRG1 and KDELR3 using synovial tissue was constructed, providing theoretical foundation and guideline for diagnostic and treatment targets in OA.
Collapse
Affiliation(s)
- Ziyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenjuan Wang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuwen Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Xiao'ao Xue
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yinghui Hua
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
50
|
Sugimura N, Aso K, Wada H, Izumi M, Ikeuchi M. Association Between Power Doppler Ultrasound Signals and Chronic Pain After Total Knee Arthroplasty: A Cross-Sectional Explorative Study. J Pain Res 2023; 16:2981-2992. [PMID: 37664487 PMCID: PMC10474857 DOI: 10.2147/jpr.s403641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
Purpose Some patients experience chronic postsurgical pain (CPSP) after total knee arthroplasty (TKA) despite the absence of clinical or radiographic abnormalities. Postoperative synovitis as a cause of CPSP after TKA has received limited research attention. This study aimed to investigate the relationship between synovitis after TKA and CPSP. Patients and Methods A total of 111 knees of 85 patients, with at least 1-year post-TKA follow-up, were assessed retrospectively and cross-sectionally. Power Doppler (PD) ultrasonography was used to detect the synovial hypervascularity associated with synovitis. The knee joint was divided into 15 areas, and PD signals were graded semi-quantitatively (0-3) in each area, the sum of which was defined as the total PD score. Clinical information regarding CPSP, including the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain subscales, was recorded. The relationship between pain and PD ultrasonography findings was accessed. Patients were divided into two groups (CPSP+ and CPSP- groups) based on pain severity. Clinical information, including PD ultrasonography findings and other possible causes, was compared between the groups. Results The WOMAC pain subscale was significantly correlated with the total PD score and maximum PD grade (r=0.3977, p<0.0001; r=0.2797, p=0.0029; respectively). The CPSP+ group had a significantly higher maximum PD grade and total PD score than the CPSP- group (median [interquartile range]: 2 [1, 2] vs 1 [1, 2], p=0.0001; 6 [2, 11] vs 2 [1, 4], p=0.0002; respectively). Multiple and logistic regression analyses showed that the total PD score was an independent factor for the WOMAC pain subscale (β=0.3822, 95% confidence interval [CI]=0.1460, 0.6184, p=0.00176) and CPSP (odds ratio=1.19, 95% CI=1.01, 1.41, p=0.0424). Conclusion This study indicated a possible association between the total PD score and chronic pain after TKA; however, further studies are needed to corroborate these findings.
Collapse
Affiliation(s)
- Natsuki Sugimura
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
- Department of Orthopedic Surgery, Hata Prefectural Hospital, Sukumo, Kochi, 788-0785, Japan
| | - Koji Aso
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Hiroyuki Wada
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Masashi Izumi
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Masahiko Ikeuchi
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|