1
|
Li Y, Li T, Zhu C, Li H, Fang R, Li R, Jin Y, Zhu Z, Xia L, Fang B. Stable-Dynamic Hydrogels Mimicking the Pericellular Matrix for Articular Cartilage Repair. Adv Healthc Mater 2025:e2405081. [PMID: 40159810 DOI: 10.1002/adhm.202405081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/13/2025] [Indexed: 04/02/2025]
Abstract
Cartilage regeneration requires a specialized biomechanical environment. Macroscopically, cartilage repair requires a protracted, stable mechanical environment, whereas microscopically, it involves dynamic interactions between cells and the extracellular matrix. Therefore, this study aims to design a hydrogel that meets the complex biomechanical requirements for cartilage repair. Dynamic hybrid hydrogels with temporal stability at the macroscale and dynamic properties at the microscale are successfully synthesized. The dynamic hybrid hydrogel simulates the stress relaxation and viscoelasticity of the pericellular matrix, facilitating effective interactions between the extracellular matrix and cells. The in vitro and in vivo experiments demonstrated that the hybrid hydrogel significantly promoted cartilage repair. The dynamic hybrid hydrogel alleviates abnormal actin polymerization, reduces intracellular stress, and increases the volume of individual cells. By modulating the cytoskeleton, the hybrid hydrogel inhibits Notch signal transduction in both the receptor and ligand cells, resulting in an improved cartilage phenotype. This study introduces an effective hybrid hydrogel scaffold that modulates the chondrocyte cytoskeleton and Notch signaling pathways by establishing an appropriate biomechanical environment, thus offering a promising material for cartilage repair.
Collapse
Affiliation(s)
- Yixin Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Tiancheng Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Cheng Zhu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Hairui Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Runzhe Fang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Ruomei Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Yu Jin
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Zhiyu Zhu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Lunguo Xia
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| | - Bing Fang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, 500 Quxi Road, Shanghai, 200011, China
| |
Collapse
|
2
|
Pueyo Moliner A, Ito K, Zaucke F, Kelly DJ, de Ruijter M, Malda J. Restoring articular cartilage: insights from structure, composition and development. Nat Rev Rheumatol 2025:10.1038/s41584-025-01236-7. [PMID: 40155694 DOI: 10.1038/s41584-025-01236-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/01/2025]
Abstract
Articular cartilage can withstand substantial compressive and shear forces within the joint and also reduces friction during motion. The exceptional mechanical properties of articular cartilage stem from its highly organized extracellular matrix (ECM). The ECM is composed mainly of collagen type II and is pivotal in conferring mechanical durability to the tissue within its proteoglycan-rich matrix. Articular cartilage is prone to injury and degeneration, and current treatments often fail to restore the mechanical function of this tissue. A key challenge is replicating the intricate collagen-proteoglycan network, which is essential for the long-lasting restoration and mechanical durability of the tissue. Understanding articular cartilage development, which arises between late embryonic and early juvenile development, is vital for the creation of durable therapeutic strategies. The development of the articular ECM involves the biosynthesis, fibrillogenesis and self-assembly of the collagen type II network, which, along with proteoglycans and minor ECM components, shapes the architecture of adult articular cartilage. A deeper understanding of these processes could inform biomaterial-based therapies aimed at improving therapeutic outcomes. Emerging biofabrication technologies offer new opportunities to integrate developmental principles into the creation of durable articular cartilage implants. Bridging fundamental biology with innovative engineering offers novel approaches to generating more-durable 3D implants for articular cartilage restoration.
Collapse
Affiliation(s)
- Alba Pueyo Moliner
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Keita Ito
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Frank Zaucke
- Department of Trauma Surgery and Orthopedics, Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mylène de Ruijter
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, Utrecht, the Netherlands.
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands.
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Janczi T, Böhm B, Fehrl Y, Hartl N, Behrens F, Kinne RW, Burkhardt H, Meier F. Mechanical forces trigger invasive behavior in synovial fibroblasts through N-cadherin/ADAM15 -dependent modulation of LncRNA H19. Sci Rep 2025; 15:9814. [PMID: 40118917 PMCID: PMC11928650 DOI: 10.1038/s41598-025-94012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
Joint damage caused by immune-mediated inflammation in rheumatoid arthritis (RA) preferentially affects site-specific mechano-sensitive areas. The perception of physical forces in the synovial tissue by the residing fibroblasts initiates signalling responses with impact on cellular functions. Here, we describe a mechanotransduction pathway in rheumatoid arthritis synovial fibroblasts (RASF), which is critically dependent on the disintegrin metalloproteinase ADAM15 and N-cadherin (NCAD). Both molecules co-localize in NCAD-based adherens junctions and trigger mechanosignaling events involving the activation of p21-activated kinase 2 (PAK2). The mechanically induced phosphorylation of PAK2 subsequently leads to its co-recruitment together with the adaptor molecule Nck to the NCAD/ADAM15 complex at the cell membrane. These signal transduction events initiate strain-induced downregulation of lncRNA H19 and miR-130a-3p. They finally result in an upregulation of cadherin-11 (CDH11), thereby enhancing cell invasive properties - a feature characteristic of aggressive RASFs. Accordingly, we propose a new mechano-induced pathway that causes an altered composition of cadherin expression in the adherens junctions of synovial fibroblasts and likely contributes to the site-specific variability of the aggressive RASF-phenotype in RA-pathogenesis.
Collapse
Affiliation(s)
- Tomasz Janczi
- Division of Rheumatology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| | - Beate Böhm
- Division of Rheumatology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany.
| | - Yuliya Fehrl
- Division of Rheumatology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| | - Nikolas Hartl
- Division of Rheumatology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| | - Frank Behrens
- Division of Rheumatology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60590, Frankfurt am Main, Germany
| | - Raimund W Kinne
- Experimental Rheumatology Unit, Department of Orthopedics, Jena University Hospital, Waldkliniken Eisenberg GmbH, 07607, Eisenberg, Germany
| | - Harald Burkhardt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60590, Frankfurt am Main, Germany
| | - Florian Meier
- Division of Rheumatology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| |
Collapse
|
4
|
Li M, Wei CB, Li HF, He K, Bai RJ, Zhang FJ. Osteopontin inhibits autophagy via CD44 and avβ3 integrin and promotes cell proliferation in osteoarthritic fibroblast-like synoviocytes. BMC Musculoskelet Disord 2025; 26:274. [PMID: 40102843 PMCID: PMC11916941 DOI: 10.1186/s12891-025-08509-y] [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: 08/09/2023] [Accepted: 03/07/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is closely related to aging, and autophagy is implicated in the retardation of aging. Activated synoviocytes play important roles in OA; the synoviocytes could produce osteopontin (OPN) and its main receptors CD44 and integrin, which are all involved in OA. The purpose of this study is to investigate whether OPN has an effect on autophagy in osteoarthritic synoviocytes. METHODS We cultured human OA fibroblast-like synoviocytes (FLS) and treated them with rhOPN and antibodies against CD44 and CD51/61 (αvβ3 integrin) or isotype IgG to block the interaction between receptors and ligands. Infection with lentivirus mRFP-GFP-LC3, laser confocal imaging and Western blotting were used to determine changes in the expression of autophagy markers, and cell proliferation of FLS was assessed with a CCK-8 assay. RESULTS Our results showed the expression level of autophagy marker protein LC3 II and the mRFP-GFP-LC3 puncta were significantly decreased after treatment with rhOPN when compared with the control group, when the FLS were incubated with antibodies against CD44 or CD51/61 (αvβ3 integrin) or with control isotype IgG for 1 h, followed by rhOPN treatment for 48 h, rhOPN could suppress the relative expression of LC3 II and Beclin1 via integrin and CD44 in the FLS, CCK-8 assay also showed that rhOPN significantly increased the cell proliferation and viability of FLS. CONCLUSIONS OPN could inhibit autophagy via CD44 and αvβ3 integrin and promote the proliferation of FLS, playing an important role in OA synovitis.
Collapse
Affiliation(s)
- Min Li
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Chang-Bao Wei
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Hai-Feng Li
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Rui-Jun Bai
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China.
| | - Fang-Jie Zhang
- Department of Emergency Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), No.87 Xiangya Road, Changsha, Hunan, 410008, China.
| |
Collapse
|
5
|
Zhu Y, Zhu J, Wang X, Wang P, Liu R. Molecular roles in membrane receptor signaling pathways and cascade reactions in chondrocytes: a review. J Mol Histol 2025; 56:94. [PMID: 39988650 DOI: 10.1007/s10735-025-10368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/03/2025] [Indexed: 02/25/2025]
Abstract
Articular cartilage (AC) is a specialized connective tissue with unique biological and mechanical properties, which depends on the biological effects of each resident chondrocyte and its surrounding extracellular matrix (ECM) to form a unit that operates in a constant and balanced feedback loop. The surface membrane receptors of chondrocytes play a crucial role in the feedback balance of this biological unit. Various biological signals outside chondrocytes, such as water-soluble chemical signal molecules and mechanical signals, are unable to directly enter the cell and must first bind to the plasma membrane receptors to induce changes in the level and activity of intracellular signal transduction molecules. These changes then transmit through signaling cascade pathways into the nucleus, changing the cell phenotype, and producing physiological or pathological changes. Specific chemical and mechanical signals break the feedback balance of cartilage tissue units through membrane receptors. In the ECM environment, the molecular actions of chondrocyte membrane receptors in response to these specific signals, along with associated ion channel receptors, collectively regulate the biological effects of chondrocytes. This leads to decreased chondrocyte survival and an imbalance in ECM regulation, ultimately disrupting the tissue's molecular framework and physiological feedback mechanisms, and resulting in pathological changes in cartilage tissue. To provide insights into addressing the complexities associated with cartilage tissue injury and repair engineering, this review provides a comprehensive overview of the molecular mechanisms and biological implications of chondrocyte membrane receptor-mediated signal transduction, including G protein-coupled receptors (GPCRs), enzyme-linked receptors (tyrosine kinase receptors (TKRs)), and integrin receptors.
Collapse
Affiliation(s)
- Yingkang Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jingjing Zhu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xu Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Pengbo Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ruiyu Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
6
|
Kramer NE, Byun S, Coryell P, D'Costa S, Thulson E, Kim H, Parkus SM, Bond ML, Klein ER, Shine J, Chubinskaya S, Love MI, Mohlke KL, Diekman BO, Loeser RF, Phanstiel DH. Response eQTLs, chromatin accessibility, and 3D chromatin structure in chondrocytes provide mechanistic insight into osteoarthritis risk. CELL GENOMICS 2025; 5:100738. [PMID: 39788104 PMCID: PMC11770232 DOI: 10.1016/j.xgen.2024.100738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/29/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025]
Abstract
Osteoarthritis (OA) poses a significant healthcare burden with limited treatment options. While genome-wide association studies (GWASs) have identified over 100 OA-associated loci, translating these findings into therapeutic targets remains challenging. To address this gap, we mapped gene expression, chromatin accessibility, and 3D chromatin structure in primary human articular chondrocytes in both resting and OA-mimicking conditions. We identified thousands of differentially expressed genes, including those associated with differences in sex and age. RNA sequencing in chondrocytes from 101 donors across two conditions uncovered 3,782 unique eGenes, including 420 that exhibited strong and significant condition-specific effects. Colocalization with OA GWAS signals revealed 13 putative OA risk genes, 6 of which have not been previously identified. Chromatin accessibility and 3D chromatin structure provided insights into the mechanisms and conditional specificity of these variants. Our findings shed light on OA pathogenesis and highlight potential targets for therapeutic development.
Collapse
Affiliation(s)
- Nicole E Kramer
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA; Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Seyoun Byun
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Philip Coryell
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan D'Costa
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eliza Thulson
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - HyunAh Kim
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sylvie M Parkus
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Marielle L Bond
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Emma R Klein
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susanna Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michael I Love
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA; Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27695, USA.
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA; Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Douglas H Phanstiel
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA; Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
7
|
Tao H, Feng M, Feng H, Ren H. Research advance of 3D printing for articular cartilage regeneration. Regen Med 2025; 20:45-55. [PMID: 39957623 PMCID: PMC11881833 DOI: 10.1080/17460751.2025.2466346] [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: 12/08/2024] [Accepted: 02/10/2025] [Indexed: 02/18/2025] Open
Abstract
Articular cartilage lesion frequently leads to dysfunction and the development of degenerative diseases, posing a significant public health challenge due to the limited self-healing capacity of cartilage tissue. Current surgical treatments, including marrow stimulation techniques and osteochondral autografts/allografts, have limited efficacy or have significant drawbacks, highlighting the urgent need for alternative strategies. Advances in 3D printing for cartilage regeneration have shown promising potential in creating cartilage-mimicking constructs, thereby opening new possibilities for cartilage repair. In this review, we summarize current surgical treatment methods and their limitations for addressing articular cartilage lesion, various 3D printing strategies and their features in cartilage tissue engineering, seed cells from different sources, and different types of biomaterials. We also explore the benefits, current challenges, and future research directions for 3D printing in the treatment of articular cartilage lesion within the field of cartilage tissue engineering.
Collapse
Affiliation(s)
- Haicheng Tao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Mingli Feng
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hui Feng
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hongchen Ren
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Li L, Al‐Jallad H, Sun A, Georgiopoulos M, Bokhari R, Ouellet J, Jarzem P, Cherif H, Haglund L. The proteomic landscape of extracellular vesicles derived from human intervertebral disc cells. JOR Spine 2024; 7:e70007. [PMID: 39507593 PMCID: PMC11538033 DOI: 10.1002/jsp2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024] Open
Abstract
Background Extracellular vesicles (EVs) function as biomarkers and are crucial in cell communication and regulation, with therapeutic potential for intervertebral disc (IVD)-related low back pain (LBP). EV cargo is often affected by tissue health, which may affect the therapeutic potential. There is currently limited knowledge of how the cargo of IVD cell-derived EVs varies with tissue health and how differences in proteomic profile affect the predicted biological functions. Methods Our study purified EVs from human IVD cell conditioned media by size-exclusion chromatography. Nanoparticle tracking analysis was conducted to measure EV size and concentration. Transmission electron microscopy and Western blot were performed to examine EV structure and markers. Tandem mass tag-mass spectrometry was conducted to determine protein cargo. Results Most EVs were exosomes and intermediate microvesicles with an increasing amount linked to disease progression. Of the proteins detected, 88.6% were shared across the non-degenerate, mildly-degenerate, and degenerate samples. GO and KEGG analyses revealed that cargo from the mildly-degenerate samples was the most distinct, with the proteins in high abundance strongly associated with extracellular matrix (ECM) organization and structure. Shared proteins, highly expressed in the non-degenerate and degenerate samples, showed strong associations with cell adhesion, ECM-receptor interaction, and vesicle-mediated transport, respectively. Conclusions Our findings indicate that EVs from IVD cells from tissue with different degrees of degeneration share a majority of the cargo proteins. However, the level of expression differs with degeneration grade. Cargo from the mildly-degenerate samples exhibits the most differences. A better understanding of changes in EV cargo in the degenerative process may provide novel information related to molecular mechanisms underlying IVD degeneration and suggest new potential treatment modalities for IVD-related LBP.
Collapse
Affiliation(s)
- Li Li
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
- The Orthopaedic Research Laboratory, Research Institute of the McGill University Health CentreMontrealQuebecCanada
| | | | - Aiwei Sun
- Department of Anatomy and Cell BiologyMcGill UniversityMontrealQuebecCanada
| | - Miltiadis Georgiopoulos
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
| | - Rakan Bokhari
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- Department of Surgery, Division of NeurosurgeryFaculty of Medicine, King Abdulaziz UniversityJeddahSaudi Arabia
| | - Jean Ouellet
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
- Shriners Hospital for ChildrenMontrealQuebecCanada
| | - Peter Jarzem
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
| | - Hosni Cherif
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
- The Orthopaedic Research Laboratory, Research Institute of the McGill University Health CentreMontrealQuebecCanada
| | - Lisbet Haglund
- Department of Surgery, Division of OrthopaedicsMcGill UniversityMontrealQuebecCanada
- The McGill Scoliosis and Spine Group, McGill University Health CentreMontrealQuebecCanada
- The Orthopaedic Research Laboratory, Research Institute of the McGill University Health CentreMontrealQuebecCanada
- Shriners Hospital for ChildrenMontrealQuebecCanada
| |
Collapse
|
9
|
Du C, Liu J, Liu S, Xiao P, Chen Z, Chen H, Huang W, Lei Y. Bone and Joint-on-Chip Platforms: Construction Strategies and Applications. SMALL METHODS 2024; 8:e2400436. [PMID: 38763918 DOI: 10.1002/smtd.202400436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/28/2024] [Indexed: 05/21/2024]
Abstract
Organ-on-a-chip, also known as "tissue chip," is an advanced platform based on microfluidic systems for constructing miniature organ models in vitro. They can replicate the complex physiological and pathological responses of human organs. In recent years, the development of bone and joint-on-chip platforms aims to simulate the complex physiological and pathological processes occurring in human bones and joints, including cell-cell interactions, the interplay of various biochemical factors, the effects of mechanical stimuli, and the intricate connections between multiple organs. In the future, bone and joint-on-chip platforms will integrate the advantages of multiple disciplines, bringing more possibilities for exploring disease mechanisms, drug screening, and personalized medicine. This review explores the construction and application of Organ-on-a-chip technology in bone and joint disease research, proposes a modular construction concept, and discusses the new opportunities and future challenges in the construction and application of bone and joint-on-chip platforms.
Collapse
Affiliation(s)
- Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Xiao
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
10
|
Shih SY, Grant MP, Epure LM, Alad M, Lerouge S, Huk OL, Bergeron SG, Zukor DJ, Merle G, Im HJ, Antoniou J, Mwale F. Advances in the Regulation of Periostin for Osteoarthritic Cartilage Repair Applications. Biomolecules 2024; 14:1469. [PMID: 39595645 PMCID: PMC11592007 DOI: 10.3390/biom14111469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging evidence indicates periostin (POSTN) is upregulated in patients with OA, and studies have shown that it can induce the activation of inflammatory cytokines and catabolic enzymes, making it a potential therapeutic target. Link N (LN) is a peptide fragment derived from the link protein and has been demonstrated as an anabolic-like factor and anti-catabolic and anti-inflammatory factors both in vitro and in vivo. This study aims to determine if LN can regulate POSTN expression and function in OA cartilage. Articular cartilage was recovered from donors undergoing total knee replacements to isolate chondrocytes and prepare osteochondral explants. Cells and explants were treated with POSTN and LN (1 and 100 μg) and measured for changes in POSTN expression and various matrix proteins, catabolic and proinflammatory factors, and signaling. To determine the effects of POSTN expression in vivo, a rabbit OA model was used. Immunoprecipitation and in silico modeling were used to determine peptide/POSTN interactions. Western blotting, PCR, and immunohistochemistry demonstrated that LN decreased POSTN expression both in vitro and in vivo. LN was also able to directly inhibit POSTN signaling in OA chondrocytes. In silico docking suggested the direct interaction of LN with POSTN at residues responsible for its oligomerization. Immunoprecipitation experiments confirmed the direct interaction of LN with POSTN and the destabilization of its oligomerization. This study demonstrates the ability of a peptide, LN, to suppress the overexpression and function of POSTN in OA cartilage.
Collapse
Affiliation(s)
- Sunny Y. Shih
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Michael P. Grant
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Laura M. Epure
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Department of Orthopaedics, SMBD-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Muskan Alad
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Sophie Lerouge
- Department of Mechanical Engineering, École de Technologie Supérieure (ETS), Montreal, QC H3C 1K3, Canada
- Laboratory of Endovascular Biomaterials (LBeV), Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Olga L. Huk
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Department of Orthopaedics, SMBD-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Stephane G. Bergeron
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Department of Orthopaedics, SMBD-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - David J. Zukor
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Department of Orthopaedics, SMBD-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Géraldine Merle
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Chemical Engineering Department, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada
| | - Hee-Jeong Im
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL 60612, USA
| | - John Antoniou
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Fackson Mwale
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
11
|
Byun S, Coryell P, Kramer N, D’Costa S, Thulson E, Shine J, Parkus S, Chubinskaya S, Loeser RF, Diekman BO, Phanstiel DH. Response splicing QTLs in primary human chondrocytes identifies putative osteoarthritis risk genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.622754. [PMID: 39605710 PMCID: PMC11601258 DOI: 10.1101/2024.11.11.622754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Osteoarthritis affects millions worldwide, yet effective treatments remain elusive due to poorly understood molecular mechanisms. While genome-wide association studies (GWAS) have identified over 100 OA-associated loci, identifying the genes impacted at each locus remains challenging. Several studies have mapped expression quantitative trait loci (eQTL) in chondrocytes and colocalized them with OA GWAS variants to identify putative OA risk genes; however, the degree to which genetic variants influence OA risk via alternative splicing has not been explored. We investigated the role of alternative splicing in OA pathogenesis using RNA-seq data from 101 human chondrocyte samples treated with PBS (control) or fibronectin fragment (FN-f), an OA trigger. We identified 590 differentially spliced genes between conditions, with FN-f inducing splicing events similar to those in primary OA tissue. We used CRISPR/Cas9 to mimic an SNRNP70 splicing event observed in OA and FN-f-treated chondrocytes and found that it induced an OA-like expression pattern. Integration with genotyping data revealed 7,188 splicing quantitative trait loci (sQTL) affecting 3,056 genes. While many sQTLs were shared, we identified 738 and 343 condition-specific sQTLs for control and FN-f, respectively. We identified 15 RNA binding proteins whose binding sites were enriched at sQTL splice junctions and found that expression of those RNA binding proteins correlated with exon inclusion. Colocalization with OA GWAS identified 6 putative risk genes, including a novel candidate, PBRM1. Our study highlights the significant impact of alternative splicing in OA and provides potential therapeutic targets for future research.
Collapse
Affiliation(s)
- Seyoun Byun
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Philip Coryell
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nicole Kramer
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan D’Costa
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eliza Thulson
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sylvie Parkus
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan Chubinskaya
- Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Dvision of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27695, USA
| | - Douglas H Phanstiel
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Kramer NE, Byun S, Coryell P, D’Costa S, Thulson E, Kim H, Parkus SM, Bond ML, Klein ER, Shine J, Chubinskaya S, Love MI, Mohlke KL, Diekman BO, Loeser RF, Phanstiel DH. Response eQTLs, chromatin accessibility, and 3D chromatin structure in chondrocytes provide mechanistic insight into osteoarthritis risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592567. [PMID: 38952796 PMCID: PMC11216363 DOI: 10.1101/2024.05.05.592567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Osteoarthritis (OA) poses a significant healthcare burden with limited treatment options. While genome-wide association studies (GWAS) have identified over 100 OA-associated loci, translating these findings into therapeutic targets remains challenging. Integrating expression quantitative trait loci (eQTL), 3D chromatin structure, and other genomic approaches with OA GWAS data offers a promising approach to elucidate disease mechanisms; however, comprehensive eQTL maps in OA-relevant tissues and conditions remain scarce. We mapped gene expression, chromatin accessibility, and 3D chromatin structure in primary human articular chondrocytes in both resting and OA-mimicking conditions. We identified thousands of differentially expressed genes, including those associated with differences in sex and age. RNA-seq in chondrocytes from 101 donors across two conditions uncovered 3782 unique eGenes, including 420 that exhibited strong and significant condition-specific effects. Colocalization with OA GWAS signals revealed 13 putative OA risk genes, 10 of which have not been previously identified. Chromatin accessibility and 3D chromatin structure provided insights into the mechanisms and conditional specificity of these variants. Our findings shed light on OA pathogenesis and highlight potential targets for therapeutic development.
Collapse
Affiliation(s)
- Nicole E Kramer
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Seyoun Byun
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Philip Coryell
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan D’Costa
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eliza Thulson
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - HyunAh Kim
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sylvie M Parkus
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Marielle L Bond
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Emma R Klein
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susanna Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michael I Love
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27695, USA
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Douglas H Phanstiel
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
13
|
Bian X, Liu X, Zhou M, Tang H, Wang R, Ma L, He G, Xu S, Wang Y, Tan J, Tang K, Guo L. Mechanical stimulation promotes fibrochondrocyte proliferation by activating the TRPV4 signaling pathway during tendon-bone insertion healing: CCN2 plays an important regulatory role. BURNS & TRAUMA 2024; 12:tkae028. [PMID: 39429645 PMCID: PMC11491146 DOI: 10.1093/burnst/tkae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 10/22/2024]
Abstract
Background We previously confirmed that mechanical stimulation is an important factor in the repair of tendon-bone insertion (TBI) injuries and that mechanoreceptors such as transient receptor potential ion-channel subfamily V member 4 (TRPV4; also known as transient receptor potential vanilloid 4) are key to transforming mechanical stimulation into intracellular biochemical signals. This study aims to elucidate the mechanism of mechanical stimulation regulating TRPV4. Methods Immunohistochemical staining and western blotting were used to evaluate cartilage repair at the TBI after injury. The RNA expression and protein expression of mechanoreceptors and key pathway molecules regulating cartilage proliferation were analyzed. TBI samples were collected for transcriptome sequencing to detect gene expression. Calcium-ion imaging and flow cytometry were used to evaluate the function of TPRV4 and cellular communication network factor 2 (CCN2) after the administration of siRNA, recombinant adenovirus and agonists. Results We found that treadmill training improved the quality of TBI healing and enhanced fibrochondrocyte proliferation. The transcriptome sequencing results suggested that the elevated expression of the mechanistically stimulated regulator CCN2 and the exogenous administration of recombinant human CCN2 significantly promoted TRPV4 protein expression and fibrochondrocyte proliferation. In vitro, under mechanical stimulation conditions, small interfering RNA (siRNA)-CCN2 not only inhibited the proliferation of primary fibrochondrocytes but also suppressed TRPV4 protein expression and activity. Subsequently, primary fibrochondrocytes were treated with the TRPV4 agonist GSK1016790A and the recombinant adenovirus TRPV4 (Ad-TRPV4), and GSK1016790A partially reversed the inhibitory effect of siRNA-CCN2. The phosphoinositide 3-kinase/ protein kinase B (PI3K/AKT) signaling pathway participated in the above process. Conclusions Mechanical stimulation promoted fibrochondrocyte proliferation and TBI healing by activating TRPV4 channels and the PI3K/AKT signaling pathway, and CCN2 may be a key regulatory protein in maintaining TRPV4 activation.
Collapse
Affiliation(s)
- Xuting Bian
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiao Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Mei Zhou
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Hong Tang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Rui Wang
- Chongqing Institute of Bio-Intelligent Manufacturing, No. 60, Xingguang Avenue, Yubei District, Chongqing, 400000, China
| | - Lin Ma
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Gang He
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Shibo Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yunjiao Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jindong Tan
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Kanglai Tang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Lin Guo
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
14
|
Patel K, Barter M, Soul J, Clark P, Proctor C, Clark I, Young D, Shanley DP. Systems analysis of miR-199a/b-5p and multiple miR-199a/b-5p targets during chondrogenesis. eLife 2024; 12:RP89701. [PMID: 39401064 PMCID: PMC11473111 DOI: 10.7554/elife.89701] [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: 10/15/2024] Open
Abstract
Changes in chondrocyte gene expression can contribute to the development of osteoarthritis (OA), and so recognition of the regulative processes during chondrogenesis can lead to a better understanding of OA. microRNAs (miRNAs) are key regulators of gene expression in chondrocytes/OA, and we have used a combined experimental, bioinformatic, and systems biology approach to explore the multiple miRNA-mRNA interactions that regulate chondrogenesis. A longitudinal chondrogenesis bioinformatic analysis identified paralogues miR-199a-5p and miR-199b-5p as pro-chondrogenic regulators. Experimental work in human cells demonstrated alteration of miR-199a-5p or miR-199b-5p expression led to significant inverse modulation of key chondrogenic genes and extracellular matrix production. miR-199a/b-5p targets FZD6, ITGA3 and CAV1 were identified by inhibition experiments and verified as direct targets by luciferase assay. The experimental work was used to generate and parameterise a multi-miRNA 14-day chondrogenesis kinetic model to be used as a repository for the experimental work and as a resource for further investigation of this system. This is the first multi-miRNA model of a chondrogenesis-based system, and highlights the complex relationships between regulatory miRNAs, and their target mRNAs.
Collapse
Affiliation(s)
- Krutik Patel
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle UniversityNewcastle-upon-TyneUnited Kingdom
| | - Matt Barter
- Regenerative Medicine, Stem Cells, Transplantation, Biosciences Institute, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Jamie Soul
- Regenerative Medicine, Stem Cells, Transplantation, Biosciences Institute, Newcastle UniversityNewcastle upon TyneUnited Kingdom
- Computational Biology Facility, Faculty of Health and Life Sciences, University of LiverpoolLiverpoolUnited Kingdom
| | - Peter Clark
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle UniversityNewcastle-upon-TyneUnited Kingdom
| | - Carole Proctor
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle UniversityNewcastle-upon-TyneUnited Kingdom
| | - Ian Clark
- School of Biological Sciences, University of East AngliaNorwichUnited Kingdom
| | - David Young
- Regenerative Medicine, Stem Cells, Transplantation, Biosciences Institute, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Daryl P Shanley
- Campus for Ageing and Vitality, Biosciences Institute, Newcastle UniversityNewcastle-upon-TyneUnited Kingdom
| |
Collapse
|
15
|
Mariné-Casadó R, Domenech-Coca C, Fernández S, Costa A, Segarra S, López-Andreo MJ, Puiggròs F, Cerón JJ, Martínez-Puig D, Soler C, Sifre V, Serra CI, Caimari A. Effects of the oral administration of glycosaminoglycans with or without native type II collagen on the articular cartilage transcriptome in an osteoarthritic-induced rabbit model. GENES & NUTRITION 2024; 19:19. [PMID: 39232650 PMCID: PMC11375882 DOI: 10.1186/s12263-024-00749-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND In a previous study, the 84-day administration of glycosaminoglycans (GAGs), with or without native collagen type II (NC), in an osteoarthritis (OA)-induced rabbit model slowed down OA progression, improved several micro- and macroscopic parameters and magnetic resonance imaging (MRI) biomarkers in cartilage, and increased hyaluronic acid levels in synovial fluid. To elucidate the potential underlying mechanisms, a transcriptomics approach was conducted using medial femoral condyle and trochlea samples. RESULTS The administration of chondroitin sulfate (CS), glucosamine hydrochloride (GlHCl), and hyaluronic acid (HA), with (CGH-NC) or without (CGH) NC, strongly modulated several genes involved in chondrocyte extracellular matrix (ECM) remodeling and homeostasis when compared to non-treated rabbits (CTR group). Notably, both treatments shared the main mechanism of action, which was related to ECM modulation through the down-regulation of genes encoding proteolytic enzymes, such as ADAM metallopeptidase with thrombospondin type 1 motif, 9 (Adamts9), and the overexpression of genes with a relevant role in the synthesis of ECM components, such as aggrecan (Acan) in both CGH-NC and CGH groups, and fibronectin 1 (Fn1) and collagen type II, alpha 1 (Col2A1) in the CGH group. Furthermore, there was a significant modulation at the gene expression level of the mTOR signaling pathway, which is associated with the regulation of the synthesis of ECM proteolytic enzymes, only in CGH-NC-supplemented rabbits. This modulation could account for the better outcomes concerning the microscopic and macroscopic evaluations reported in these animals. CONCLUSIONS In conclusion, the expression of key genes involved in chondrocyte ECM remodeling and homeostasis was significantly modulated in rabbits in response to both CGH and CGH-NC treatments, which would partly explain the mechanisms by which these therapies exert beneficial effects against OA.
Collapse
Affiliation(s)
- Roger Mariné-Casadó
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, Reus, 43204, Spain
| | - Cristina Domenech-Coca
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, Reus, 43204, Spain
| | - Salvador Fernández
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit, Universitat Rovira i Virgili-EURECAT, Reus, 43204, Spain
| | - Andrea Costa
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, Reus, 43204, Spain
| | - Sergi Segarra
- R&D Bioiberica S.A.U., Esplugues de Llobregat, 08950, Spain
| | - Maria José López-Andreo
- Servicio de Investigación Biosanitaria, Área Científica y Técnica de Investigación (ACTI), Universidad de Murcia, Murcia, 30100, Spain
| | - Francesc Puiggròs
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, Reus, 43204, Spain
| | - José Joaquín Cerón
- Interlab-UMU, Campus de Excelencia "Mare Nostrum", University of Murcia, Campus Espinardo, Murcia, 30071, Spain
| | | | - Carme Soler
- Hospital Veterinario UCV, Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, Valencia, 46002, Spain
| | - Vicente Sifre
- Hospital Veterinario UCV, Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, Valencia, 46002, Spain
| | - Claudio Iván Serra
- Hospital Veterinario UCV, Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, Valencia, 46002, Spain
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, Reus, 43204, Spain.
| |
Collapse
|
16
|
Lan M, Liu Y, Liu J, Zhang J, Haider MA, Zhang Y, Zhang Q. Matrix Viscoelasticity Tunes the Mechanobiological Behavior of Chondrocytes. Cell Biochem Funct 2024; 42:e4126. [PMID: 39324844 DOI: 10.1002/cbf.4126] [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: 04/18/2024] [Revised: 08/25/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
In articular cartilage, the pericellular matrix acting as a specialized mechanical microenvironment modulates environmental signals to chondrocytes through mechanotransduction. Matrix viscoelastic alterations during cartilage development and osteoarthritis (OA) degeneration play an important role in regulating chondrocyte fate and cartilage matrix homeostasis. In recent years, scientists are gradually realizing the importance of matrix viscoelasticity in regulating chondrocyte function and phenotype. Notably, this is an emerging field, and this review summarizes the existing literatures to the best of our knowledge. This review provides an overview of the viscoelastic properties of hydrogels and the role of matrix viscoelasticity in directing chondrocyte behavior. In this review, we elaborated the mechanotransuction mechanisms by which cells sense and respond to the viscoelastic environment and also discussed the underlying signaling pathways. Moreover, emerging insights into the role of matrix viscoelasticity in regulating chondrocyte function and cartilage formation shed light into designing cell-instructive biomaterial. We also describe the potential use of viscoelastic biomaterials in cartilage tissue engineering and regenerative medicine. Future perspectives on mechanobiological comprehension of the viscoelastic behaviors involved in tissue homeostasis, cellular responses, and biomaterial design are highlighted. Finally, this review also highlights recent strategies utilizing viscoelastic hydrogels for designing cartilage-on-a-chip.
Collapse
Affiliation(s)
- Minhua Lan
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Yanli Liu
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Junjiang Liu
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Jing Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Muhammad Adnan Haider
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Yanjun Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Quanyou Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
17
|
Zhu C, Zhang L, Ding X, Wu W, Zou J. Non-coding RNAs as regulators of autophagy in chondrocytes: Mechanisms and implications for osteoarthritis. Ageing Res Rev 2024; 99:102404. [PMID: 38971322 DOI: 10.1016/j.arr.2024.102404] [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: 02/23/2024] [Revised: 06/22/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease with multiple causative factors such as aging, mechanical injury, and obesity. Autophagy is a complex dynamic process that is involved in the degradation and modification of intracellular proteins and organelles under different pathophysiological conditions. Autophagy, as a cell survival mechanism under various stress conditions, plays a key role in regulating chondrocyte life cycle metabolism and cellular homeostasis. Non-coding RNAs (ncRNAs) are heterogeneous transcripts that do not possess protein-coding functions, but they can act as effective post-transcriptional and epigenetic regulators of gene and protein expression, thus participating in numerous fundamental biological processes. Increasing evidence suggests that ncRNAs, autophagy, and their crosstalk play crucial roles in OA pathogenesis. Therefore, we summarized the complex role of autophagy in OA chondrocytes and focused on the regulatory role of ncRNAs in OA-associated autophagy to elucidate the complex pathological mechanisms of the ncRNA-autophagy network in the development of OA, thus providing new research targets for the clinical diagnosis and treatment of OA.
Collapse
Affiliation(s)
- Chenyu Zhu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Lingli Zhang
- School of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaoqing Ding
- School of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China.
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
18
|
Li CJ, Park JH, Jin GS, Mandakhbayar N, Yeo D, Lee JH, Lee JH, Kim HS, Kim HW. Strontium/Silicon/Calcium-Releasing Hierarchically Structured 3D-Printed Scaffolds Accelerate Osteochondral Defect Repair. Adv Healthc Mater 2024; 13:e2400154. [PMID: 38647029 DOI: 10.1002/adhm.202400154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/24/2024] [Indexed: 04/25/2024]
Abstract
Articular cartilage defects are a global challenge, causing substantial disability. Repairing large defects is problematic, often exceeding cartilage's self-healing capacity and damaging bone structures. To tackle this problem, a scaffold-mediated therapeutic ion delivery system is developed. These scaffolds are constructed from poly(ε-caprolactone) and strontium (Sr)-doped bioactive nanoglasses (SrBGn), creating a unique hierarchical structure featuring macropores from 3D printing, micropores, and nanotopologies due to SrBGn integration. The SrBGn-embedded scaffolds (SrBGn-µCh) release Sr, silicon (Si), and calcium (Ca) ions, which improve chondrocyte activation, adhesion, proliferation, and maturation-related gene expression. This multiple ion delivery significantly affects metabolic activity and maturation of chondrocytes. Importantly, Sr ions may play a role in chondrocyte regulation through the Notch signaling pathway. Notably, the scaffold's structure and topological cues expedite the recruitment, adhesion, spreading, and proliferation of chondrocytes and bone marrow-derived mesenchymal stem cells. Si and Ca ions accelerate osteogenic differentiation and blood vessel formation, while Sr ions enhance the polarization of M2 macrophages. The findings show that SrBGn-µCh scaffolds accelerate osteochondral defect repair by delivering multiple ions and providing structural/topological cues, ultimately supporting host cell functions and defect healing. This scaffold holds great promise for osteochondral repair applications.
Collapse
Affiliation(s)
- Cheng Ji Li
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeong-Hui Park
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Gang Shi Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Donghyeon Yeo
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Dankook Physician Scientist Research Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Dankook Physician Scientist Research Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Dankook Physician Scientist Research Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Dankook Physician Scientist Research Center, Dankook University Hospital, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell and Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
19
|
Miao MZ, Lee JS, Yamada KM, Loeser RF. Integrin signalling in joint development, homeostasis and osteoarthritis. Nat Rev Rheumatol 2024; 20:492-509. [PMID: 39014254 PMCID: PMC11886400 DOI: 10.1038/s41584-024-01130-8] [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] [Accepted: 05/24/2024] [Indexed: 07/18/2024]
Abstract
Integrins are key regulators of cell-matrix interactions during joint development and joint tissue homeostasis, as well as in the development of osteoarthritis (OA). The signalling cascades initiated by the interactions of integrins with a complex network of extracellular matrix (ECM) components and intracellular adaptor proteins orchestrate cellular responses necessary for maintaining joint tissue integrity. Dysregulated integrin signalling, triggered by matrix degradation products such as matrikines, disrupts this delicate balance, tipping the scales towards an environment conducive to OA pathogenesis. The interplay between integrin signalling and growth factor pathways further underscores the multifaceted nature of OA. Moreover, emerging insights into the role of endocytic trafficking in regulating integrin signalling add a new layer of complexity to the understanding of OA development. To harness the therapeutic potential of targeting integrins for mitigation of OA, comprehensive understanding of their molecular mechanisms across joint tissues is imperative. Ultimately, deciphering the complexities of integrin signalling will advance the ability to treat OA and alleviate its global burden.
Collapse
Affiliation(s)
- Michael Z Miao
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janice S Lee
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Office of the Clinical Director, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
20
|
Gheorghe SR, Crăciun AM, Ilyés T, Tisa IB, Sur L, Lupan I, Samasca G, Silaghi CN. Converging Mechanisms of Vascular and Cartilaginous Calcification. BIOLOGY 2024; 13:565. [PMID: 39194503 DOI: 10.3390/biology13080565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Physiological calcification occurs in bones and epiphyseal cartilage as they grow, whereas ectopic calcification occurs in blood vessels, cartilage, and soft tissues. Although it was formerly thought to be a passive and degenerative process associated with aging, ectopic calcification has been identified as an active cell-mediated process resembling osteogenesis, and an increasing number of studies have provided evidence for this paradigm shift. A significant association between vascular calcification and cardiovascular risk has been demonstrated by various studies, which have shown that arterial calcification has predictive value for future coronary events. With respect to cartilaginous calcification, calcium phosphate or hydroxyapatite crystals can form asymptomatic deposits in joints or periarticular tissues, contributing to the pathophysiology of osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, tendinitis, and bursitis. The risk factors and sequence of events that initiate ectopic calcification, as well as the mechanisms that prevent the development of this pathology, are still topics of debate. Consequently, in this review, we focus on the nexus of the mechanisms underlying vascular and cartilaginous calcifications, trying to circumscribe the similarities and disparities between them to provide more clarity in this regard.
Collapse
Affiliation(s)
- Simona R Gheorghe
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandra M Crăciun
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Tamás Ilyés
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Badiu Tisa
- Department of Pediatrics III, Iuliu Hatieganu University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania
| | - Lucia Sur
- Department of Pediatrics I, Iuliu Hatieganu University of Medicine and Pharmacy, 400370 Cluj-Napoca, Romania
| | - Iulia Lupan
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Ciprian N Silaghi
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
21
|
Liu Y, Lin R, Fang H, Li L, Zhang M, Lu L, Gao X, Song J, Wei J, Xiao Q, Zhang F, Wu K, Cui L. Sargassum polysaccharide attenuates osteoarthritis in rats and is associated with the up-regulation of the ITGβ1-PI3K-AKT signaling pathway. J Orthop Translat 2024; 47:176-190. [PMID: 39040490 PMCID: PMC11260896 DOI: 10.1016/j.jot.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Background Osteoarthritis (OA) presents a formidable challenge, characterized by as-yet-unclear mechanical intricacies within cartilage and the dysregulation of bone homeostasis. Our preliminary data revealed the encouraging potential of a Sargassum polysaccharide (SP), in promoting chondrogenesis. The aim of our study is to comprehensively assess the therapeutic effects of SP on OA models and further elucidate its potential mechanism. Methods The protective effects of SP were initially evaluated in an inflammation-induced human chondrocyte (C28) cell model. CCK-8 assays, Alcian blue staining, RT-qPCR and Western blotting were used to verify the chondrogenesis of SP in vitro. To assess the efficacy of SP in vivo, surgically induced medial meniscus destabilization (DMM) OA rats underwent an 8-week SP treatment. The therapeutic effects of SP in OA rats were comprehensively evaluated using X-ray imaging, micro-computed tomography (μ-CT), histopathological analysis, as well as immunohistochemical and immunofluorescent staining. Following these assessments, we delved into the potential signaling pathways of SP in inflammatory chondrocytes utilizing RNA-seq analysis. Validation of these findings was conducted through RT-qPCR and western blotting techniques. Results SP significantly enhance the viability of C28 chondrocytes, and increased the secretion of acidic glycoproteins. Moreover, SP stimulated the expression of chondrogenic genes (Aggrecan, Sox9, Col2a1) and facilitated the synthesis of Collagen II protein in C28 inflammatory chondrocytes. In vivo experiments revealed that SP markedly ameliorated knee joint stenosis, alleviated bone and cartilage injuries, and reduced the histopathological scores in the OA rats. μ-CT analysis confirmed that SP lessened bone impairments in the medial femoral condyle and the subchondral bone of the tibial plateau, significantly improving the microarchitectural parameters of the subchondral bone. Histopathological analyses indicated that SP notably enhanced cartilage quality on the surface of the tibial plateau, leading to increased cartilage thickness and area. Immunohistochemistry staining and immunofluorescence staining corroborated these findings by showing a significant promotion of Collagen II expression in OA joints treated with SP. RNA-seq analysis suggest that SP's effects were mediated through the regulation of the ITGβ1-PI3K-AKT signaling axis, thereby stimulating chondrogenesis. Verification through RT-qPCR and Western blot analyses confirmed that SP significantly upregulated the expression of ITGβ1, p110δ, AKT1, ACAN, and Col2a1. Notably, knock-down of ITGβ1 using siRNA in C28 chondrocytes inhibited the expression of ITGβ1, p110δ, AKT1, and ACAN. However, these inhibitory effects were not completely reversed by supplemental SP intervention. Conclusions In summary, our findings reveal that SP significantly enhances chondrogenesis both in vitro and in vivo, alleviating OA progression both in bone and cartilage. The observed beneficial effects are intricately linked to the activation of the ITGβ1-PI3K-AKT signaling axis. The translational potential of this article Our research marks the first instance unveiling the advantageous effects and underlying mechanisms of SP in OA treatment. With its clinical prospects, SP presents compelling new evidence for the advancement of a next-generation polysaccharide drug for OA therapy.
Collapse
Affiliation(s)
- Yanzhi Liu
- Corresponding author. Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524045, China.
| | | | | | - Lixian Li
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Min Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Lujiao Lu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiang Gao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Jintong Song
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Jinsong Wei
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Qixian Xiao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Fucheng Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Kefeng Wu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
22
|
Hay AM, Rhoades MJ, Bangerter S, Ferguson SA, Lee H, T. Gill M, Page GL, Pope A, Measom GJ, Hager RL, Seeley MK. Serum Cartilage Oligomeric Matrix Protein Concentration Increases More After Running Than Swimming for Older People. Sports Health 2024; 16:534-541. [PMID: 37697665 PMCID: PMC11195858 DOI: 10.1177/19417381231195309] [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: 09/13/2023] Open
Abstract
BACKGROUND Knee osteoarthritis is common in older people. Serum cartilage oligomeric matrix protein (sCOMP) is a biomarker of knee articular cartilage metabolism. The purpose of this study was 2-fold: to (1) determine acute effects of running and swimming on sCOMP concentration in older people; and (2) investigate relationships between sCOMP concentration change due to running and swimming and measures of knee health in older people. HYPOTHESES Running would result in greater increase in sCOMP concentration than swimming, and increase in sCOMP concentration due to running and swimming would associate positively with measures of poor knee health. STUDY DESIGN Cross-sectional. LEVEL OF EVIDENCE Level 3. METHODS A total of 20 participants ran 5 km and 19 participants swam 1500 m. sCOMP concentration was measured immediately before, immediately after, and 15, 30, and 60 minutes after running or swimming. sCOMP concentration change due to running and swimming was compared. Correlations between sCOMP concentration change due to running and swimming, and other measures of knee health were evaluated, including the Tegner Activity Scale and Knee injury and Osteoarthritis Outcome Score. RESULTS sCOMP concentration increased 29% immediately after running, relative to baseline, but only 6% immediately after swimming (P < 0.01). No significant relationship was observed between acute sCOMP change due to running and swimming, and observed measures of knee health (P > 0.05). Participants with clinically relevant knee symptoms exhibited greater sCOMP concentration before and after running and swimming (P = 0.03) and had greater body mass (P = 0.04). CONCLUSION Running results in greater acute articular cartilage metabolism than swimming; however, the chronic effects of this are unclear. Older people with clinically relevant knee symptoms possess greater sCOMP concentration and are heavier, independent of exercise mode and physical activity level. CLINICAL RELEVANCE These results describe the effects of exercise (running and swimming) for older physically active persons, with and without knee pain.
Collapse
Affiliation(s)
- Alexandra M. Hay
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | | | | | - Seth A. Ferguson
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Hyunwook Lee
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Martha T. Gill
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Garritt L. Page
- Department of Statistics, Brigham Young University, Provo, Utah
| | - Andrew Pope
- Department of Statistics, Brigham Young University, Provo, Utah
| | - Gary J. Measom
- Department of Nursing, Utah Valley University, Orem, Utah
| | - Ronald L. Hager
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| | - Matthew K. Seeley
- Department of Exercise Sciences, Brigham Young University, Provo, Utah
| |
Collapse
|
23
|
Oyama S, Kanamoto T, Ebina K, Etani Y, Hirao M, Goshima A, Otani S, Hikida M, Yamakawa S, Ito S, Okada S, Nakata K. Cyclic compressive loading induces a mature meniscal cell phenotype in mesenchymal stem cells with an atelocollagen-based scaffold. Front Bioeng Biotechnol 2024; 12:1394093. [PMID: 38832131 PMCID: PMC11145507 DOI: 10.3389/fbioe.2024.1394093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/24/2024] [Indexed: 06/05/2024] Open
Abstract
Introduction: Biomechanical stimulation is reportedly pivotal in meniscal regeneration, although its effect on mesenchymal stem cell (MSC) meniscal differentiation remains elusive. In this study, we investigated how cyclic compressive loading (CCL) could impact MSCs using three-dimensional cultures in atelocollagen-based meniscal substitute (ACMS). Methods: We extracted MSCs from the meniscus, synovium, and articular cartilage, cultured them in three-dimensional cultures, and exposed them to CCL for 7 days. We then compared the transcriptomes of MSCs treated with and without CCL. Results: Our RNA-seq analysis revealed that CCL induced significant transcriptome changes, significantly affecting chondrocyte-related genes, including SOX9, TGFB1, and PRG4 upregulation. CCL induced transcriptional differentiation of meniscus progenitors toward mature meniscal cells. Conclusion: This study unveils the potential of mechanical stress in promoting MSC meniscal differentiation within ACMS. Our investigations provide new insights for mechanisms underlying meniscal regeneration with ACMS.
Collapse
Affiliation(s)
- Shohei Oyama
- Department of Musculoskeletal Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Takashi Kanamoto
- Department of Medicine for Sports and Performing Arts, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kosuke Ebina
- Department of Musculoskeletal Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuki Etani
- Department of Medicine for Sports and Performing Arts, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makoto Hirao
- Department of Orthopaedic Surgery, National Hospital Organization, Osaka Minami Medical Center, Osaka, Japan
| | - Atsushi Goshima
- Department of Orthopaedic Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Shunya Otani
- Department of Medicine for Sports and Performing Arts, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Minami Hikida
- Department of Medicine for Sports and Performing Arts, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Yamakawa
- Department of Sports Medical Biomechanics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shohei Ito
- Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ken Nakata
- Department of Medicine for Sports and Performing Arts, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
24
|
Bloks NGC, Dicks A, Harissa Z, Nelissen RGHH, Hajmousa G, Ramos YFM, de Almeida RC, Guilak F, Meulenbelt I. Hyper-physiologic mechanical cues, as an osteoarthritis disease-relevant environmental perturbation, cause a critical shift in set points of methylation at transcriptionally active CpG sites in neo-cartilage organoids. Clin Epigenetics 2024; 16:64. [PMID: 38730337 PMCID: PMC11087253 DOI: 10.1186/s13148-024-01676-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a complex, age-related multifactorial degenerative disease of diarthrodial joints marked by impaired mobility, joint stiffness, pain, and a significant decrease in quality of life. Among other risk factors, such as genetics and age, hyper-physiological mechanical cues are known to play a critical role in the onset and progression of the disease (Guilak in Best Pract Res Clin Rheumatol 25:815-823, 2011). It has been shown that post-mitotic cells, such as articular chondrocytes, heavily rely on methylation at CpG sites to adapt to environmental cues and maintain phenotypic plasticity. However, these long-lasting adaptations may eventually have a negative impact on cellular performance. We hypothesize that hyper-physiologic mechanical loading leads to the accumulation of altered epigenetic markers in articular chondrocytes, resulting in a loss of the tightly regulated balance of gene expression that leads to a dysregulated state characteristic of the OA disease state. RESULTS We showed that hyper-physiological loading evokes consistent changes in CpGs associated with expression changes (ML-tCpGs) in ITGA5, CAV1, and CD44, among other genes, which together act in pathways such as anatomical structure morphogenesis (GO:0009653) and response to wound healing (GO:0042060). Moreover, by comparing the ML-tCpGs and their associated pathways to tCpGs in OA pathophysiology (OA-tCpGs), we observed a modest but particular interconnected overlap with notable genes such as CD44 and ITGA5. These genes could indeed represent lasting detrimental changes to the phenotypic state of chondrocytes due to mechanical perturbations that occurred earlier in life. The latter is further suggested by the association between methylation levels of ML-tCpGs mapped to CD44 and OA severity. CONCLUSION Our findings confirm that hyper-physiological mechanical cues evoke changes to the methylome-wide landscape of chondrocytes, concomitant with detrimental changes in positional gene expression levels (ML-tCpGs). Since CAV1, ITGA5, and CD44 are subject to such changes and are central and overlapping with OA-tCpGs of primary chondrocytes, we propose that accumulation of hyper-physiological mechanical cues can evoke long-lasting, detrimental changes in set points of gene expression that influence the phenotypic healthy state of chondrocytes. Future studies are necessary to confirm this hypothesis.
Collapse
Affiliation(s)
- Niek G C Bloks
- Dept. Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Amanda Dicks
- Washington University, Saint Louis, MO, USA
- Shriners Hospitals for Children, Saint Louis, MO, USA
| | - Zainab Harissa
- Washington University, Saint Louis, MO, USA
- Shriners Hospitals for Children, Saint Louis, MO, USA
| | - Rob G H H Nelissen
- Department Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ghazaleh Hajmousa
- Dept. Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Yolande F M Ramos
- Dept. Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Rodrigo Coutinho de Almeida
- Dept. Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Farshid Guilak
- Washington University, Saint Louis, MO, USA
- Shriners Hospitals for Children, Saint Louis, MO, USA
| | - Ingrid Meulenbelt
- Dept. Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands.
| |
Collapse
|
25
|
Jia S, Liang R, Chen J, Liao S, Lin J, Li W. Emerging technology has a brilliant future: the CRISPR-Cas system for senescence, inflammation, and cartilage repair in osteoarthritis. Cell Mol Biol Lett 2024; 29:64. [PMID: 38698311 PMCID: PMC11067114 DOI: 10.1186/s11658-024-00581-x] [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: 12/29/2023] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
Osteoarthritis (OA), known as one of the most common types of aseptic inflammation of the musculoskeletal system, is characterized by chronic pain and whole-joint lesions. With cellular and molecular changes including senescence, inflammatory alterations, and subsequent cartilage defects, OA eventually leads to a series of adverse outcomes such as pain and disability. CRISPR-Cas-related technology has been proposed and explored as a gene therapy, offering potential gene-editing tools that are in the spotlight. Considering the genetic and multigene regulatory mechanisms of OA, we systematically review current studies on CRISPR-Cas technology for improving OA in terms of senescence, inflammation, and cartilage damage and summarize various strategies for delivering CRISPR products, hoping to provide a new perspective for the treatment of OA by taking advantage of CRISPR technology.
Collapse
Affiliation(s)
- Shicheng Jia
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Rongji Liang
- Shantou University Medical College, Shantou, 515041, China
| | - Jiayou Chen
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Shuai Liao
- Department of Bone and Joint, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Wei Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
26
|
Zhao X, Hu L, Liu G, Yin X, Gong Q, Li Y, Li Q, Zhou Y, Sun Y, Guo C, Du Z. Fibronectin binds integrin α5β1 to regulate macular neovascularization through the Wnt/β-catenin signaling pathway. Exp Eye Res 2024; 242:109880. [PMID: 38552713 DOI: 10.1016/j.exer.2024.109880] [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: 11/22/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
Age-related macular degeneration (AMD) is a progressive, degenerative disease of the macula. The formation of macular neovascularization (MNV) and subretinal fibrosis of AMD is the most classic cause of the loss of vision in older adults worldwide. While the underlying causes of MNV and subretinal fibrosis remain elusive, the common feature of many common retinal diseases is changes the proportions of protein deposition in extracellular matrix (ECM) when compared to normal tissue. In ECM, fibronectin (FN) is a crucial component and plays a pivotal part not only in fibrotic diseases but also in the process of angiogenesis. The study aims to understand the role of ligand FN and its common integrin receptor α5β1 on MNV, and to understand the molecular mechanism involved. To study this, the laser-induced MNV mouse model and the rhesus macaque choroid-retinal endothelial cell line (RF/6A) chemical hypoxia mode were established, and the FN-α5β1 expression levels were detected by immunohistochemistry (IHC) and quantitative real-time PCR analysis (qRT-PCR). Fibronectin expression was silenced using small interfering RNA (siRNA) targeting FN. The tube formation and vitro scratch assays were used to assess the ability to form blood vessels and cell migration. To measure the formation of MNV, immunofluorescence, and Western blot assays were used. These results revealed that the expressions of FN and integrin α5β1 were distinctly increased in the laser-induced MNV mouse model and in the RF/6A cytochemically induced hypoxia model, and the expression tendency was identical. After the use of FN siRNA, the tube formation and migration abilities of the RF/6A cells were lower, the ability of endothelial cells to proliferate was confined and the scope of damage caused by the laser in animal models was significantly cut down. In addition, FN gene knockdown dramatically inhibited the expression of Wnt/β-catenin signal. The interaction of FN with the integrin receptor α5β1 in the constructed model, which may act through the Wnt/β-catenin signaling pathway, was confirmed in this study. In conclusion, FN may be a potential new molecular target for the prevention and treatment of subretinal fibrosis and MNV.
Collapse
Affiliation(s)
- Xiaoran Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Liting Hu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Guibo Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Xiaoni Yin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Qingyun Gong
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Ying Li
- Department of Ophthalmology, Linyi People's Hospital, Jiefang Road Number 27, Lanshan District, Linyi, 276003, Shandong, China
| | - Qinghua Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Yuzheng Zhou
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Yibin Sun
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Chunyan Guo
- People's Hospital of Dingxi, Dingxi, 743000, Gansu Province, China.
| | - Zhaodong Du
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
| |
Collapse
|
27
|
Hussain MT, Austin-Williams S, Wright TD, Dhawan UK, Pinto AL, Cooper D, Norling LV. β1-Integrin-Mediated Uptake of Chondrocyte Extracellular Vesicles Regulates Chondrocyte Homeostasis. Int J Mol Sci 2024; 25:4756. [PMID: 38731975 PMCID: PMC11083596 DOI: 10.3390/ijms25094756] [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: 02/29/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent age-related degenerative disorder, which severely reduces the quality of life of those affected. Whilst management strategies exist, no cures are currently available. Virtually all joint resident cells generate extracellular vesicles (EVs), and alterations in chondrocyte EVs during OA have previously been reported. Herein, we investigated factors influencing chondrocyte EV release and the functional role that these EVs exhibit. Both 2D and 3D models of culturing C28I/2 chondrocytes were used for generating chondrocyte EVs. We assessed the effect of these EVs on chondrogenic gene expression as well as their uptake by chondrocytes. Collectively, the data demonstrated that chondrocyte EVs are sequestered within the cartilage ECM and that a bi-directional relationship exists between chondrocyte EV release and changes in chondrogenic differentiation. Finally, we demonstrated that the uptake of chondrocyte EVs is at least partially dependent on β1-integrin. These results indicate that chondrocyte EVs have an autocrine homeostatic role that maintains chondrocyte phenotype. How this role is perturbed under OA conditions remains the subject of future work.
Collapse
Affiliation(s)
- Mohammed Tayab Hussain
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Shani Austin-Williams
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Thomas Dudley Wright
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Umesh Kumar Dhawan
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Andreia L. Pinto
- Royal Brompton & Harefield NHS Foundation Trust, London SW3 6PY, UK;
| | - Dianne Cooper
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| | - Lucy V. Norling
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
28
|
Huang Y, Sun M, Lu Z, Zhong Q, Tan M, Wei Q, Zheng L. Role of integrin β1 and tenascin C mediate TGF-SMAD2/3 signaling in chondrogenic differentiation of BMSCs induced by type I collagen hydrogel. Regen Biomater 2024; 11:rbae017. [PMID: 38525326 PMCID: PMC10960929 DOI: 10.1093/rb/rbae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/24/2024] [Accepted: 02/09/2024] [Indexed: 03/26/2024] Open
Abstract
Cartilage defects may lead to severe degenerative joint diseases. Tissue engineering based on type I collagen hydrogel that has chondrogenic potential is ideal for cartilage repair. However, the underlying mechanisms of chondrogenic differentiation driven by type I collagen hydrogel have not been fully clarified. Herein, we explored potential collagen receptors and chondrogenic signaling pathways through bioinformatical analysis to investigate the mechanism of collagen-induced chondrogenesis. Results showed that the super enhancer-related genes induced by collagen hydrogel were significantly enriched in the TGF-β signaling pathway, and integrin-β1 (ITGB1), a receptor of collagen, was highly expressed in bone marrow mesenchymal stem cells (BMSCs). Further analysis showed genes such as COL2A1 and Tenascin C (TNC) that interacted with ITGB1 were significantly enriched in extracellular matrix (ECM) structural constituents in the chondrogenic induction group. Knockdown of ITGB1 led to the downregulation of cartilage-specific genes (SOX9, ACAN, COL2A1), SMAD2 and TNC, as well as the downregulation of phosphorylation of SMAD2/3. Knockdown of TNC also resulted in the decrease of cartilage markers, ITGB1 and the SMAD2/3 phosphorylation but overexpression of TNC showed the opposite trend. Finally, in vitro and in vivo experiments confirmed the involvement of ITGB1 and TNC in collagen-mediated chondrogenic differentiation and cartilage regeneration. In summary, we demonstrated that ITGB1 was a crucial receptor for chondrogenic differentiation of BMSCs induced by collagen hydrogel. It can activate TGF-SMAD2/3 signaling, followed by impacting TNC expression, which in turn promotes the interaction of ITGB1 and TGF-SMAD2/3 signaling to enhance chondrogenesis. These may provide concernful support for cartilage tissue engineering and biomaterials development.
Collapse
Affiliation(s)
- Yuanjun Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Miao Sun
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
| | - Zhenhui Lu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| | - Qiuling Zhong
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
| | - Manli Tan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| | - Qingjun Wei
- Department of Trauma Orthopedic and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, China
- Life Science Institute, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
29
|
Deng R, Zhao R, Zhang Z, Chen Y, Yang M, Lin Y, Ye J, Li N, Qin H, Yan X, Shi J, Yuan F, Song S, Xu Z, Song Y, Fu J, Xu B, Nie G, Yu JK. Chondrocyte membrane-coated nanoparticles promote drug retention and halt cartilage damage in rat and canine osteoarthritis. Sci Transl Med 2024; 16:eadh9751. [PMID: 38381849 DOI: 10.1126/scitranslmed.adh9751] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by progressive degeneration of articular cartilage. A challenge in the development of disease-modifying drugs is effective delivery to chondrocytes. The unique structure of the joint promotes rapid clearance of drugs through synovial fluid, and the dense and avascular cartilage extracellular matrix (ECM) limits drug penetration. Here, we show that poly(lactide-co-glycolic acid) nanoparticles coated in chondrocyte membranes (CM-NPs) were preferentially taken up by rat chondrocytes ex vivo compared with uncoated nanoparticles. Internalization of the CM-NPs was mediated primarily by E-cadherin, clathrin-mediated endocytosis, and micropinocytosis. These CM-NPs adhered to the cartilage ECM in rat knee joints in vivo and penetrated deeply into the cartilage matrix with a residence time of more than 34 days. Simulated synovial fluid clearance studies showed that CM-NPs loaded with a Wnt pathway inhibitor, adavivint (CM-NPs-Ada), delayed the catabolic metabolism of rat and human chondrocytes and cartilage explants under inflammatory conditions. In a surgical model of rat OA, drug-loaded CM-NPs effectively restored gait, attenuated periarticular bone remodeling, and provided chondroprotection against cartilage degeneration. OA progression was also mitigated by CM-NPs-Ada in a canine model of anterior cruciate ligament transection. These results demonstrate the feasibility of using chondrocyte membrane-coated nanoparticles to improve the pharmacokinetics and efficacy of anti-OA drugs.
Collapse
Affiliation(s)
- Ronghui Deng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zining Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Meng Yang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jing Ye
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xin Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Fuzhen Yuan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Shitang Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Zijie Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yifan Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jiangnan Fu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Bingbing Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jia-Kuo Yu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
- Orthopedic Sports Medicine Center, Beijing Tsinghua Changgung Hospital, Affiliated Hospital of Tsinghua University, Beijing 102218, P. R. China
| |
Collapse
|
30
|
Guo H, Lan M, Zhang Q, Liu Y, Zhang Y, Zhang Q, Chen W. [Piezo1 Mediates the Regulation of Substrate Stiffness on Primary Cilia in Chondrocytes]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:67-73. [PMID: 38322536 PMCID: PMC10839480 DOI: 10.12182/20240160502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Indexed: 02/08/2024]
Abstract
Objective To investigate how substrate stiffness regulates the morphology of primary cilia in chondrocytes and to illustrate how Piezo1 mediates the morphology regulation of primary cilia by substrate stiffness. Methods Polydimethylsiloxane (PDMS) curing agent and the main agent (Dow Corning, Beijing, China) were mixed at the ratio of 1∶10 (stiff), 1∶50 (medium stiffness), and 1∶70 (soft), respectively, to prepare substrate films with the thickness of 1 mm at different levels of stiffness, including stiff substrate of (2.21±0.12) MPa, medium-stiffness substrate of (54.47±6.06) kPa, and soft substrate of (2.13±0.10) kPa. Chondrocytes were cultured with the substrates of three different levels of stiffness. Then, the cells were treated with Tubastatin A (Tub A) to inhibit histone deacetylase 6 (HDAC6), Piezo1 activator Yoda1, and inhibitor GsMTx4, respectively. The effects of HDAC6, Yoda1, and GsMTx4 on chondrocyte morphology and the length of primary cilia were analyzed through immunofluorescence staining. Results The stiff substrate increased the spread area of the chondrocytes. Immunofluorescence assays showed that the cytoskeleton and the nuclear area of the cells on the stiff substrate were significantly increased (P<0.05) and the primary cilia were significantly extended (P<0.05) compared with those on the medium-stiffness and soft substrates. However, the presence rate of primary cilia was not affected. The HDAC6 activity of chondrocytes increased with the decrease in substrate stiffness. When the activity of HDAC6 was inhibited, the cytoskeletal area, the nuclei area, and the primary cilium length were increased more significantly on the stiff substrate (P<0.05). Further testing showed that Piezo1 activator and inhibitor could regulate the activity of HDAC6 in chondrocytes, and that the length of primary cilia was significantly increased after treatment with the activator Yoda1 (P<0.05). On the other hand, the length of primary cilia was significantly shortened on the stiff substrate after treatment with the inhibitor GsMTx4 (P<0.05). Conclusion Both substrate stiffness and Piezo1 may affect the morphology of chondrocyte primary cilia by regulating HDAC6 activity.
Collapse
Affiliation(s)
- Huaqing Guo
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Minhua Lan
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Qiang Zhang
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yanli Liu
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yanjun Zhang
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- ( 030009) Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030009, China
| | - Quanyou Zhang
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- ( 030009) Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030009, China
| | - Weiyi Chen
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| |
Collapse
|
31
|
Kuchynsky K, Stevens P, Hite A, Xie W, Diop K, Tang S, Pietrzak M, Khan S, Walter B, Purmessur D. Transcriptional profiling of human cartilage endplate cells identifies novel genes and cell clusters underlying degenerated and non-degenerated phenotypes. Arthritis Res Ther 2024; 26:12. [PMID: 38173036 PMCID: PMC10763221 DOI: 10.1186/s13075-023-03220-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Low back pain is a leading cause of disability worldwide and is frequently attributed to intervertebral disc (IVD) degeneration. Though the contributions of the adjacent cartilage endplates (CEP) to IVD degeneration are well documented, the phenotype and functions of the resident CEP cells are critically understudied. To better characterize CEP cell phenotype and possible mechanisms of CEP degeneration, bulk and single-cell RNA sequencing of non-degenerated and degenerated CEP cells were performed. METHODS Human lumbar CEP cells from degenerated (Thompson grade ≥ 4) and non-degenerated (Thompson grade ≤ 2) discs were expanded for bulk (N=4 non-degenerated, N=4 degenerated) and single-cell (N=1 non-degenerated, N=1 degenerated) RNA sequencing. Genes identified from bulk RNA sequencing were categorized by function and their expression in non-degenerated and degenerated CEP cells were compared. A PubMed literature review was also performed to determine which genes were previously identified and studied in the CEP, IVD, and other cartilaginous tissues. For single-cell RNA sequencing, different cell clusters were resolved using unsupervised clustering and functional annotation. Differential gene expression analysis and Gene Ontology, respectively, were used to compare gene expression and functional enrichment between cell clusters, as well as between non-degenerated and degenerated CEP samples. RESULTS Bulk RNA sequencing revealed 38 genes were significantly upregulated and 15 genes were significantly downregulated in degenerated CEP cells relative to non-degenerated cells (|fold change| ≥ 1.5). Of these, only 2 genes were previously studied in CEP cells, and 31 were previously studied in the IVD and other cartilaginous tissues. Single-cell RNA sequencing revealed 11 unique cell clusters, including multiple chondrocyte and progenitor subpopulations with distinct gene expression and functional profiles. Analysis of genes in the bulk RNA sequencing dataset showed that progenitor cell clusters from both samples were enriched in "non-degenerated" genes but not "degenerated" genes. For both bulk- and single-cell analyses, gene expression and pathway enrichment analyses highlighted several pathways that may regulate CEP degeneration, including transcriptional regulation, translational regulation, intracellular transport, and mitochondrial dysfunction. CONCLUSIONS This thorough analysis using RNA sequencing methods highlighted numerous differences between non-degenerated and degenerated CEP cells, the phenotypic heterogeneity of CEP cells, and several pathways of interest that may be relevant in CEP degeneration.
Collapse
Affiliation(s)
- Kyle Kuchynsky
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Patrick Stevens
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Amy Hite
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - William Xie
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Khady Diop
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Shirley Tang
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Safdar Khan
- Department of Orthopaedics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Benjamin Walter
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Devina Purmessur
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA.
- Department of Orthopaedics, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
32
|
Hung SY, Chen JL, Tu YK, Tsai HY, Lu PH, Jou IM, Mbuyisa L, Lin MW. Isoliquiritigenin inhibits apoptosis and ameliorates oxidative stress in rheumatoid arthritis chondrocytes through the Nrf2/HO-1-mediated pathway. Biomed Pharmacother 2024; 170:116006. [PMID: 38091640 DOI: 10.1016/j.biopha.2023.116006] [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: 09/14/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory condition known for its irreversible destructive impact on the joints. Chondrocytes play a pivotal role in the production and maintenance of the cartilage matrix. However, the presence of inflammatory cytokines can hinder chondrocyte proliferation and promote apoptosis. Isoliquiritigenin (ISL), a flavonoid, potentially exerts protective effects against various inflammatory diseases. However, its specific role in regulating the nuclear factor E2-associated factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway in chondrocytes in RA remains unclear. To investigate this, this study used human chondrocytes and Sprague-Dawley rats to construct in vitro and in vivo RA models, respectively. The study findings reveal that cytokines markedly induced oxidative stress, the activation of matrix metalloproteinases, and apoptosis both in vitro and in vivo. Notably, ISL treatment significantly mitigated these effects. Moreover, Nrf2 or HO-1 inhibitors reversed the protective effects of ISL, attenuated the expression of Nrf2/HO-1 and peroxisome proliferator-activated receptor gamma-coactivator-1α, and promoted chondrocyte apoptosis. This finding indicates that ISL primarily targets the Nrf2/HO-1 pathway in RA chondrocytes. Moreover, ISL treatment led to improved behavior scores, reduced paw thickness, and mitigated joint damage as well as ameliorated oxidative stress in skeletal muscles in an RA rat model. In conclusion, this study highlights the pivotal role of the Nrf2/HO-1 pathway in the protective effects of ISL and demonstrates the potential of ISL as a treatment option for RA.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan; Division of Surgery, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Jen-Lung Chen
- Department of Surgery, E-Da Hospital, Kaohsiung 82445, Taiwan
| | - Yuan-Kun Tu
- Department of Orthopaedic Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hsin-Yi Tsai
- Department of Medical Research, E-Da Hospital/ E-Da Cancer Hospital, Kaohsiung 82445, Taiwan
| | - Pin-Hsuan Lu
- Department of Medical Research, E-Da Hospital/ E-Da Cancer Hospital, Kaohsiung 82445, Taiwan
| | - I-Ming Jou
- Department of Orthopaedic Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Lulekiwe Mbuyisa
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Ming-Wei Lin
- Department of Medical Research, E-Da Hospital/ E-Da Cancer Hospital, Kaohsiung 82445, Taiwan; Department of Nursing, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
33
|
Colazo JM, Hoogenboezem EN, Keech MC, Francini N, Shah V, Yu F, Lo JH, Sorets AG, McCune JT, Cho H, DeJulius CR, Michell DL, Maerz T, Vickers KC, Gibson-Corley KN, Hasty KA, Crofford L, Cook RS, Duvall CL. Albumin-binding RNAi Conjugate for Carrier Free Treatment of Arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.542971. [PMID: 37333210 PMCID: PMC10274717 DOI: 10.1101/2023.05.31.542971] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are joint diseases that are associated with pain and lost quality of life. No disease modifying OA drugs are currently available. RA treatments are better established but are not always effective and can cause immune suppression. Here, an MMP13-selective siRNA conjugate was developed that, when delivered intravenously, docks onto endogenous albumin and promotes preferential accumulation in articular cartilage and synovia of OA and RA joints. MMP13 expression was diminished upon intravenous delivery of MMP13 siRNA conjugates, consequently decreasing multiple histological and molecular markers of disease severity, while also reducing clinical manifestations such as swelling (RA) and joint pressure sensitivity (RA and OA). Importantly, MMP13 silencing provided more comprehensive OA treatment efficacy than standard of care (steroids) or experimental MMP inhibitors. These data demonstrate the utility of albumin 'hitchhiking' for drug delivery to arthritic joints, and establish the therapeutic utility of systemically delivered anti-MMP13 siRNA conjugates in OA and RA. Editorial summary Lipophilic siRNA conjugates optimized for albumin binding and "hitchhiking" can be leveraged to achieve preferential delivery to and gene silencing activity within arthritic joints. Chemical stabilization of the lipophilic siRNA enables intravenous siRNA delivery without lipid or polymer encapsulation. Using siRNA sequences targeting MMP13, a key driver of arthritis-related inflammation, albumin hitchhiking siRNA diminished MMP13, inflammation, and manifestations of osteoarthritis and rheumatoid arthritis at molecular, histological, and clinical levels, consistently outperforming clinical standards of care and small molecule MMP antagonists.
Collapse
|
34
|
Bačenková D, Trebuňová M, Demeterová J, Živčák J. Human Chondrocytes, Metabolism of Articular Cartilage, and Strategies for Application to Tissue Engineering. Int J Mol Sci 2023; 24:17096. [PMID: 38069417 PMCID: PMC10707713 DOI: 10.3390/ijms242317096] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Hyaline cartilage, which is characterized by the absence of vascularization and innervation, has minimal self-repair potential in case of damage and defect formation in the chondral layer. Chondrocytes are specialized cells that ensure the synthesis of extracellular matrix components, namely type II collagen and aggregen. On their surface, they express integrins CD44, α1β1, α3β1, α5β1, α10β1, αVβ1, αVβ3, and αVβ5, which are also collagen-binding components of the extracellular matrix. This article aims to contribute to solving the problem of the possible repair of chondral defects through unique methods of tissue engineering, as well as the process of pathological events in articular cartilage. In vitro cell culture models used for hyaline cartilage repair could bring about advanced possibilities. Currently, there are several variants of the combination of natural and synthetic polymers and chondrocytes. In a three-dimensional environment, chondrocytes retain their production capacity. In the case of mesenchymal stromal cells, their favorable ability is to differentiate into a chondrogenic lineage in a three-dimensional culture.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, Letná 9, 042 00 Košice, Slovakia; (M.T.); (J.D.); (J.Ž.)
| | | | | | | |
Collapse
|
35
|
St Amant J, Michaud J, Hinds D, Coyle M, Pozzi A, Clark AL. Depleting transforming growth factor beta receptor 2 signalling in the cartilage of itga1-null mice attenuates spontaneous knee osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100399. [PMID: 37649532 PMCID: PMC10462827 DOI: 10.1016/j.ocarto.2023.100399] [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: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Objectives Integrin α1β1 protects against osteoarthritis (OA) when it is upregulated in the superficial zone of cartilage in the early stages of disease. However, the mechanism behind this protection is unknown. Integrin α1β1 moderates transforming growth factor β receptor II (TGFBR2) signalling, a critical regulator of chondrocyte anabolic activity. To this end, mice lacking integrin α1β1 have increased baseline activation of TGFBR2 signalling and overall fibrosis. The purpose of this study was to evaluate the interplay between integrin α1β1 and TGFBR2 in the development of spontaneous OA. We hypothesized that dampening TGFBR2 signalling in the cartilage of itga1-null mice would attenuate OA. Methods Behavioural and histological manifestations of spontaneous knee OA were measured at 4, 8, 12 and 16 months in mice with and without a ubiquitous itga1 deletion and with and without a tamoxifen-induced cartilage specific TGFBR2 depletion. Results Knee cartilage degeneration, collateral ligament ossification and pain responses increased with age. Itga1-null mice with intact TGFBR2 signalling developed earlier and more severe OA compared to controls. In agreement with our hypothesis, depleting TGFBR2 signalling in the cartilage of itga1-null mice attenuated OA progression. Conclusion Intact TGFBR2 signalling drives early and worse knee OA in itga1-null mice. This result supports the hypothesis that the increased expression of integrin α1β1 by superficial zone chondrocytes early in OA development dampens TGFBR2 signalling and thus protects against degeneration.
Collapse
Affiliation(s)
- Jennifer St Amant
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Jana Michaud
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Daniel Hinds
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Madison Coyle
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Andrea L. Clark
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
36
|
Bloks NG, Dicks A, Harissa Z, Nelissen RG, Hajmousa G, Ramos YF, Almeida RC, Guilak F, Meulenbelt I. Hyper-physiologic mechanical cues, as an osteoarthritis disease relevant environmental perturbation, cause a critical shift in set-points of methylation at transcriptionally active CpG sites in neo-cartilage organoids. RESEARCH SQUARE 2023:rs.3.rs-3568544. [PMID: 38014245 PMCID: PMC10680909 DOI: 10.21203/rs.3.rs-3568544/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Background Osteoarthritis (OA) is a complex, age-related multifactorial degenerative disease of diarthrodial joints marked by impaired mobility, joint stiffness, pain, and a significant decrease in quality of life. Among other risk factors, such as genetics and age, hyper-physiological mechanical cues are known to play a critical role in the onset and progression of the disease (1). It has been shown that post-mitotic cells, such as articular chondrocytes, heavily rely on methylation at CpG sites to adapt to environmental cues and maintain phenotypic plasticity. However, these long-lasting adaptations may eventually have a negative impact on cellular performance. We hypothesize that hyper-physiologic mechanical loading leads to the accumulation of altered epigenetic markers in articular chondrocytes, resulting in a loss of the tightly regulated balance of gene expression that leads to a dysregulated state characteristic of the OA disease state. Results We showed that hyper-physiological loading evokes consistent changes in ML-tCpGs associated with expression changes in ITGA5, CAV1, and CD44, among other genes, which together act in pathways such as anatomical structure morphogenesis (GO:0009653) and response to wound healing (GO:0042060). Moreover, by comparing the ML-tCpGs and their associated pathways to tCpGs in OA pathophysiology, we observed a modest but particular interconnected overlap with notable genes such as CD44 and ITGA5. These genes could indeed represent lasting detrimental changes to the phenotypic state of chondrocytes due to mechanical perturbations that occurred earlier in life. The latter is further suggested by the association between methylation levels of ML-tCpGs mapped to CD44 and OA severity. Conclusion Our findings confirm that hyper-physiological mechanical cues evoke changes to the methylome-wide landscape of chondrocytes, concomitant with detrimental changes in positional gene expression levels (ML-tCpGs). Since CAV1, ITGA5, and CD44 are subject to such changes and are central and overlapping with OA-tCPGs of primary chondrocytes, we propose that accumulation of hyper-physiological mechanical cues can evoke long-lasting, detrimental changes in set points of gene expression that influence the phenotypic healthy state of chondrocytes. Future studies are necessary to confirm this hypothesis.
Collapse
|
37
|
Ruan Q, Wang C, Zhang Y, Sun J. Brevilin A attenuates cartilage destruction in osteoarthritis mouse model by inhibiting inflammation and ferroptosis via SIRT1/Nrf2/GPX4 signaling pathway. Int Immunopharmacol 2023; 124:110924. [PMID: 37717314 DOI: 10.1016/j.intimp.2023.110924] [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: 08/18/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
Osteoarthritis (OA) is a serious orthopedic disease that affects people's quality of life. Although there are many treatment methods, the treatment effect is still not good. Brevilin A is a bioactive compound isolated from the medicinal herbCentipeda minima. The potential efficacy of brevilin A on OA was explored in this study. Mouse chondrocytes were isolated and stimulated by IL-1β and mouse OA model was induced by destabilization of the medial meniscus (DMM). The results demonstrated that brevilin A markedly inhibited IL-1β-induced MMP1 and MMP3 production. IL-1β-induced PGE2, NO, MDA, and iron production were alleviated by brevilin A. The production of GSH and the expression of SIRT1, Nrf2, HO-1, GPX4, and Ferritin were increased by brevilin A. Furthermore, the inhibition of brevilin A on IL-1β-induced inflammation and ferroptosis were prevented by SIRT1 inhibitor. In vivo, the results showed brevilin A markedly attenuated OA progression in DMM-induced mouse OA model. Also, brevilin A could alleviate MMP1, MMP3, iNOS, and COX2 expression in OA mice. In conclusion, brevilin A protected mice against OA via suppressing inflammatory response and ferroptosis by regulating SIRT1/Nrf2/GPX4 signaling.
Collapse
Affiliation(s)
- Qing Ruan
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Cuijie Wang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province 130033, China
| | - Yunfeng Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Jiayang Sun
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China.
| |
Collapse
|
38
|
Rogers N, Meng QJ. Tick tock, the cartilage clock. Osteoarthritis Cartilage 2023; 31:1425-1436. [PMID: 37230460 DOI: 10.1016/j.joca.2023.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/11/2023] [Accepted: 05/20/2023] [Indexed: 05/27/2023]
Abstract
Osteoarthritis (OA) is the most common age-related joint disease, affecting articular cartilage and other joint structures, causing severe pain and disability. Due to a limited understanding of the underlying disease pathogenesis, there are currently no disease-modifying drugs for OA. Circadian rhythms are generated by cell-intrinsic timekeeping mechanisms which are known to dampen during ageing, increasing disease risks. In this review, we focus on one emerging area of chondrocyte biology, the circadian clocks. We first provide a historical perspective of circadian clock discoveries and the molecular underpinnings. We will then focus on the expression and functions of circadian clocks in articular cartilage, including their rhythmic target genes and pathways, links to ageing, tissue degeneration, and OA, as well as tissue niche-specific entrainment pathways. Further research into cartilage clocks and ageing may have broader implications in the understanding of OA pathogenesis, the standardization of biomarker detection, and the development of novel therapeutic routes for the prevention and management of OA and other musculoskeletal diseases.
Collapse
Affiliation(s)
- Natalie Rogers
- Wellcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK; Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK
| | - Qing-Jun Meng
- Wellcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK; Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK.
| |
Collapse
|
39
|
Goldring MB. Integrin-dependent recruitment of Src to ROS-producing endosomes in osteoarthritic cartilage. Sci Signal 2023; 16:eadj9760. [PMID: 37906627 DOI: 10.1126/scisignal.adj9760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Fibronectin (FN) fragments stimulate catabolic signaling, and, by binding to integrins, they induce chondrocytes to increase the production of matrix metalloproteinases, including MMP-13. In this issue of Science Signaling, Miao et al. reveal that internalization of a FN fragment, but not intact FN, by α5β1 integrin results in the formation of ROS-producing endosomes (redoxosomes) through which chondrocytes detect and respond to damaged matrix by producing more MMP-13.
Collapse
Affiliation(s)
- Mary B Goldring
- Hospital for Special Surgery, New York, NY 10021, USA
- Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
40
|
Riegger J, Brenner RE. Increase of cell surface vimentin is associated with vimentin network disruption and subsequent stress-induced premature senescence in human chondrocytes. eLife 2023; 12:e91453. [PMID: 37855367 PMCID: PMC10622146 DOI: 10.7554/elife.91453] [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: 07/30/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023] Open
Abstract
Accumulation of dysfunctional chondrocytes has detrimental consequences on the cartilagehomeostasis and is thus thought to play a crucial role during the pathogenesis of osteoarthritis(OA). However, the underlying mechanisms of phenotypical alteration in chondrocytes areincompletely understood. Here, we provide evidence that disruption of the intracellularvimentin network and consequent phenotypical alteration in human chondrocytes results in anexternalization of the intermediate filament. The presence of the so-called cell surfacevimentin (CSV) on chondrocytes was associated with the severity of tissue degeneration inclinical OA samples and was enhanced after mechanical injury of cartilage tissue. By meansof a doxorubicine-based in vitro model of stress-induced premature senescence (SIPS), wecould confirm the connection between cellular senescence and amount of CSV. AlthoughsiRNA-mediated silencing of CDKN2A clearly reduced the senescent phenotype as well asCSV levels of human chondrocytes, cellular senescence could not be completely reversed.Interestingly, knockdown of vimentin resulted in a SIPS-like phenotype and consequentlyincreased CSV. Therefore, we concluded that the integrity of the intracellular vimentinnetwork is crucial to maintain cellular function in chondrocytes. This assumption could beconfirmed by chemically- induced collapse of the vimentin network, which resulted in cellularstress and enhanced CSV expression. Regarding its biological function, CSV was found to beassociated with enhanced chondrocyte adhesion and plasticity. While osteogenic capacitiesseemed to be enhanced in chondrocytes expressing high levels of CSV, the chondrogenicpotential was clearly compromised. Overall, our study reinforces the importance of thevimentin network in maintenance of the chondrogenic phenotype and introduces CSV as anovel membrane-bound marker of dysfunctional chondrocytes.
Collapse
Affiliation(s)
- Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of UlmUlmGermany
| | - Rolf E Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of UlmUlmGermany
| |
Collapse
|
41
|
Da Z, Guo R, Sun J, Wang A. Identification of osteoarthritis-characteristic genes and immunological micro-environment features through bioinformatics and machine learning-based approaches. BMC Med Genomics 2023; 16:236. [PMID: 37805587 PMCID: PMC10559406 DOI: 10.1186/s12920-023-01672-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/23/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a multifaceted chronic joint disease characterized by complex mechanisms. It has a detrimental impact on the quality of life for individuals in the middle-aged and elderly population while also imposing a significant socioeconomic burden. At present, there remains a lack of comprehensive understanding regarding the pathophysiology of OA. The objective of this study was to examine the genes, functional pathways, and immune infiltration characteristics associated with the development and advancement of OA. METHODS The Gene Expression Omnibus (GEO) database was utilized to acquire gene expression profiles. The R software was employed to conduct the screening of differentially expressed genes (DEGs) and perform enrichment analysis on these genes. The OA-characteristic genes were identified using the Weighted Gene Co-expression Network Analysis (WGCNA) and the Lasso algorithm. In addition, the infiltration levels of immune cells in cartilage were assessed using single-sample gene set enrichment analysis (ssGSEA). Subsequently, a correlation analysis was conducted to examine the relationship between immune cells and the OA-characteristic genes. RESULTS A total of 80 DEGs were identified. As determined by functional enrichment, these DEGs were associated with chondrocyte metabolism, apoptosis, and inflammation. Three OA-characteristic genes were identified using WGCNA and the lasso algorithm, and their expression levels were then validated using the verification set. Finally, the analysis of immune cell infiltration revealed that T cells and B cells were primarily associated with OA. In addition, Tspan2, HtrA1 demonstrated a correlation with some of the infiltrating immune cells. CONCLUSIONS The findings of an extensive bioinformatics analysis revealed that OA is correlated with a variety of distinct genes, functional pathways, and processes involving immune cell infiltration. The present study has successfully identified characteristic genes and functional pathways that hold potential as biomarkers for guiding drug treatment and facilitating molecular-level research on OA.
Collapse
Affiliation(s)
- Zheng Da
- Xingtai People's Hospital Affiliated to Hebei Medical University, Xingtai City, Hebei Province, China
| | - Rui Guo
- Xingtai People's Hospital Affiliated to Hebei Medical University, Xingtai City, Hebei Province, China.
| | - Jianjian Sun
- Ningbo Huamei Hospital, University of Chinese Academy of Sciences, Ningbo City, Zhejiang Province, China
| | - Ai Wang
- Zhongshan Hospital Affiliated to Fudan University, Shanghai City, China
| |
Collapse
|
42
|
Kwok B, Chandrasekaran P, Wang C, He L, Mauck RL, Dyment NA, Koyama E, Han L. Rapid specialization and stiffening of the primitive matrix in developing articular cartilage and meniscus. Acta Biomater 2023; 168:235-251. [PMID: 37414114 PMCID: PMC10529006 DOI: 10.1016/j.actbio.2023.06.047] [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: 02/20/2023] [Revised: 06/02/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
Understanding early patterning events in the extracellular matrix (ECM) formation can provide a blueprint for regenerative strategies to better recapitulate the function of native tissues. Currently, there is little knowledge on the initial, incipient ECM of articular cartilage and meniscus, two load-bearing counterparts of the knee joint. This study elucidated distinctive traits of their developing ECMs by studying the composition and biomechanics of these two tissues in mice from mid-gestation (embryonic day 15.5) to neo-natal (post-natal day 7) stages. We show that articular cartilage initiates with the formation of a pericellular matrix (PCM)-like primitive matrix, followed by the separation into distinct PCM and territorial/interterritorial (T/IT)-ECM domains, and then, further expansion of the T/IT-ECM through maturity. In this process, the primitive matrix undergoes a rapid, exponential stiffening, with a daily modulus increase rate of 35.7% [31.9 39.6]% (mean [95% CI]). Meanwhile, the matrix becomes more heterogeneous in the spatial distribution of properties, with concurrent exponential increases in the standard deviation of micromodulus and the slope correlating local micromodulus with the distance from cell surface. In comparison to articular cartilage, the primitive matrix of meniscus also exhibits exponential stiffening and an increase in heterogeneity, albeit with a much slower daily stiffening rate of 19.8% [14.9 24.9]% and a delayed separation of PCM and T/IT-ECM. These contrasts underscore distinct development paths of hyaline versus fibrocartilage. Collectively, these findings provide new insights into how knee joint tissues form to better guide cell- and biomaterial-based repair of articular cartilage, meniscus and potentially other load-bearing cartilaginous tissues. STATEMENT OF SIGNIFICANCE: Successful regeneration of articular cartilage and meniscus is challenged by incomplete knowledge of early events that drive the initial formation of the tissues' extracellular matrix in vivo. This study shows that articular cartilage initiates with a pericellular matrix (PCM)-like primitive matrix during embryonic development. This primitive matrix then separates into distinct PCM and territorial/interterritorial domains, undergoes an exponential daily stiffening of ≈36% and an increase in micromechanical heterogeneity. At this early stage, the meniscus primitive matrix shows differential molecular traits and exhibits a slower daily stiffening of ≈20%, underscoring distinct matrix development between these two tissues. Our findings thus establish a new blueprint to guide the design of regenerative strategies to recapitulate the key developmental steps in vivo.
Collapse
Affiliation(s)
- Bryan Kwok
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Lan He
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
43
|
Qin Y, Coleman RM. Ligand Composition and Coating Density Co-Modulate the Chondrocyte Function on Poly(glycerol-dodecanedioate). J Funct Biomater 2023; 14:468. [PMID: 37754882 PMCID: PMC10531919 DOI: 10.3390/jfb14090468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Inducing chondrocyte redifferentiation and promoting cartilaginous matrix accumulation are key challenges in the application of biomaterials in articular cartilage repair. Poly(glycerol-dodecanedioate) (PGD) is a viable candidate for scaffold design in cartilage tissue engineering (CTE). However, the surface properties of PGD are not ideal for cell attachment and growth due to its relative hydrophobicity compared with natural extracellular matrix (ECM). In this study, PGD was coated with various masses of collagen type I or hyaluronic acid, individually or in combination, to generate a cell-material interface with biological cues. The effects of ligand composition and density on the PGD surface properties and shape, metabolic activity, cell phenotype, and ECM production of human articular chondrocytes (hACs) were evaluated. Introducing ECM ligands on PGD significantly improved its hydrophilicity and promoted the chondrocyte's anabolic activity. The morphology and anabolic activity of hACs on PGD were co-modulated by ligand composition and density, suggesting a combinatorial effect of both coating parameters on chondrocyte function during monolayer culture. Hyaluronic acid and its combination with collagen maintained a round cell shape and redifferentiated phenotype. This study demonstrated the complex mechanism of ligand-guided interactions between cell and biomaterial substrate and the potential of PGD as a scaffold material in the field of CTE.
Collapse
Affiliation(s)
- Yue Qin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Rhima M. Coleman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
44
|
Peng Y, Chen X, Liu S, Wu W, Shu H, Tian S, Xiao Y, Li K, Wang B, Lin H, Qing X, Shao Z. Extracellular Vesicle-Conjugated Functional Matrix Hydrogels Prevent Senescence by Exosomal miR-3594-5p-Targeted HIPK2/p53 Pathway for Disc Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206888. [PMID: 37165721 DOI: 10.1002/smll.202206888] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/24/2023] [Indexed: 05/12/2023]
Abstract
Nucleus pulposus stem cells (NPSCs) senescence plays a critical role in the progression of intervertebral disc degeneration (IDD). Stem cell-derived extracellular vesicles (EV) alleviate cellular senescence. Whereas, the underlying mechanism remains unclear. Low stability largely limited the administration of EV in vivo. RGD, an arginine-glycine-aspartic acid tripeptide, strongly binds integrins expressed on the EV membranes, allowing RGD to anchor EV and prolong their bioavailability. An RGD-complexed nucleus pulposus matrix hydrogel (RGD-DNP) is developed to enhance the therapeutic effects of small EV (sEV). RGD-DNP prolonged sEV retention in vitro and ex vivo. sEV-RGD-DNP promoted NPSCs migration, decreased the number of SA-β-gal-positive cells, alleviated cell cycle arrest, and reduced p16, p21, and p53 activation. Small RNA-seq showed that miR-3594-5p is enriched in sEV, and targets the homeodomain-interacting protein kinase 2 (HIPK2)/p53 pathway. The HIPK2 knockdown rescues the impaired therapeutic effects of sEV with downregulated miR-3594-5p. RGD-DNP conjugate with lower amounts of sEV achieved similar disc regeneration with free sEV of higher concentrations in DNP. In conclusion, sEV-RGD-DNP increases sEV bioavailability and relieves NPSCs senescence by targeting the HIPK2/p53 pathway, thereby alleviating IDD. This work achieves better regenerative effects with fewer sEV and consolidates the theoretical basis for sEV application for IDD treatment.
Collapse
Affiliation(s)
- Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuanzuo Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Shuo Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Departments of Anesthesiology and Critical Care Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Yan Xiao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kanglu Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - BaiChuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
45
|
Cui Y, Miao MZ, Wang M, Su QP, Qiu K, Arbeeva L, Chubinskaya S, Diekman BO, Loeser RF. Yes-associated protein nuclear translocation promotes anabolic activity in human articular chondrocytes. Osteoarthritis Cartilage 2023; 31:1078-1090. [PMID: 37100374 PMCID: PMC10524185 DOI: 10.1016/j.joca.2023.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/28/2023]
Abstract
OBJECTIVE Yes-associated protein (YAP) has been widely studied as a mechanotransducer in many cell types, but its function in cartilage is controversial. The aim of this study was to identify the effect of YAP phosphorylation and nuclear translocation on the chondrocyte response to stimuli relevant to osteoarthritis (OA). DESIGN Cultured normal human articular chondrocytes from 81 donors were treated with increased osmolarity media as an in vitro model of mechanical stimulation, fibronectin fragments (FN-f) or IL-1β as catabolic stimuli, and IGF-1 as an anabolic stimulus. YAP function was assessed with gene knockdown and inhibition by verteporfin. Nuclear translocation of YAP and its transcriptional co-activator TAZ and site-specific YAP phosphorylation were determined by immunoblotting. Immunohistochemistry and immunofluorescence to detect YAP were performed on normal and OA human cartilage with different degrees of damage. RESULTS Chondrocyte YAP/TAZ nuclear translocation increased under physiological osmolarity (400 mOsm) and IGF-1 stimulation, which was associated with YAP phosphorylation at Ser128. In contrast, catabolic stimulation decreased the levels of nuclear YAP/TAZ through YAP phosphorylation at Ser127. Following YAP inhibition, anabolic gene expression and transcriptional activity decreased. Additionally, YAP knockdown reduced proteoglycan staining and levels of type II collagen. Total YAP immunostaining was greater in OA cartilage, but YAP was sequestered in the cytosol in cartilage areas with more severe damage. CONCLUSIONS YAP chondrocyte nuclear translocation is regulated by differential phosphorylation in response to anabolic and catabolic stimuli. Decreased nuclear YAP in OA chondrocytes may contribute to reduced anabolic activity and promotion of further cartilage loss.
Collapse
Affiliation(s)
- Y Cui
- Xiangya International Medical Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.
| | - M Z Miao
- Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA; Division of Oral & Craniofacial Health Sciences, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, 27599, USA.
| | - M Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, 27599, USA.
| | - Q P Su
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - K Qiu
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, 27599, USA.
| | - L Arbeeva
- Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.
| | - S Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, 60612, USA.
| | - B O Diekman
- Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27599, USA.
| | - R F Loeser
- Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
46
|
Vágó J, Takács R, Kovács P, Hajdú T, van der Veen DR, Matta C. Combining biomechanical stimulation and chronobiology: a novel approach for augmented chondrogenesis? Front Bioeng Biotechnol 2023; 11:1232465. [PMID: 37456723 PMCID: PMC10349586 DOI: 10.3389/fbioe.2023.1232465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The unique structure and composition of articular cartilage is critical for its physiological function. However, this architecture may get disrupted by degeneration or trauma. Due to the low intrinsic regeneration properties of the tissue, the healing response is generally poor. Low-grade inflammation in patients with osteoarthritis advances cartilage degradation, resulting in pain, immobility, and reduced quality of life. Generating neocartilage using advanced tissue engineering approaches may address these limitations. The biocompatible microenvironment that is suitable for cartilage regeneration may not only rely on cells and scaffolds, but also on the spatial and temporal features of biomechanics. Cell-autonomous biological clocks that generate circadian rhythms in chondrocytes are generally accepted to be indispensable for normal cartilage homeostasis. While the molecular details of the circadian clockwork are increasingly well understood at the cellular level, the mechanisms that enable clock entrainment by biomechanical signals, which are highly relevant in cartilage, are still largely unknown. This narrative review outlines the role of the biomechanical microenvironment to advance cartilage tissue engineering via entraining the molecular circadian clockwork, and highlights how application of this concept may enhance the development and successful translation of biomechanically relevant tissue engineering interventions.
Collapse
Affiliation(s)
- Judit Vágó
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Roland Takács
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Patrik Kovács
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Tibor Hajdú
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Daan R. van der Veen
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Csaba Matta
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
47
|
Lubis AMT, Aprianto P, Pawitan JA, Priosoeryanto BP, Dewi TIT, Kamal AF. Intra-articular injection of secretome, derived from umbilical cord mesenchymal stem cell, enhances the regeneration process of cartilage in early-stage osteo-arthritis: an animal study. Acta Orthop 2023; 94:300-306. [PMID: 37377012 DOI: 10.2340/17453674.2023.12359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Mesenchymal stem cells (MSCs), both endogenous and exogenous, enhance chondrocyte proliferation by stimulating collagen type II. Secretome, an MSC derivate, has shown to also provide this mechanism through a paracrine effect. We aimed to evaluate the use of secretome and MSC in the management of early osteoarthritis (OA). ANIMALS AND METHODS 19 (1 control) male sheep (Ovies aries), which were operated on with total lateral meniscectomy to induce knee OA, were divided into 3 groups: the secretome group, hyaluronic acid group, and MSC group. Each group was injected with the respective substances and was evaluated macroscopically and microscopically. The Osteoarthritis Research Society International (OARSI) score was calculated for all subjects and a descriptive and comparative statistical analysis was undertaken. RESULTS The macroscopic analysis of the treated groups revealed better OARSI score in the secretome group compared with the other 2 groups. The secretome group showed a significantly better microscopic score compared with the hyaluronic acid group (mean difference [MD] 6.0, 95% confidence interval [CI] 0.15-12), but no significant difference compared with the MSC group (MD 1.0, CI -4.8 to 6.8). CONCLUSION Intra-articular injection of secretome is effective in managing early-stage osteoarthritis in the animal model compared with hyaluronic acid and has similar efficacy to MSC injection.
Collapse
Affiliation(s)
- Andri Maruli Tua Lubis
- Department of Orthopaedics and Traumatology, Cipto Mangunkusumo General Hospital, Jakarta; Department of Orthopaedics and Traumatology, Faculty of Medicine Universitas Indonesia, Jakarta
| | - Petrus Aprianto
- Department of Orthopaedics and Traumatology, Cipto Mangunkusumo General Hospital, Jakarta; Department of Orthopaedics and Traumatology, Faculty of Medicine Universitas Indonesia, Jakarta
| | - Jeanne Adiwinata Pawitan
- Department of Histology, Cipto Mangunkusumo General Hospital - Faculty of Medicine Universitas Indonesia, Jakarta
| | | | - Tri Isyani Tungga Dewi
- Department of Veterinary Pathology, Faculty of Agriculture, IBP University, Bogor, Indonesia
| | - Achmad Fauzi Kamal
- Department of Orthopaedics and Traumatology, Cipto Mangunkusumo General Hospital, Jakarta; Department of Orthopaedics and Traumatology, Faculty of Medicine Universitas Indonesia, Jakarta.
| |
Collapse
|
48
|
Pinto-Cardoso R, Bessa-Andrês C, Correia-de-Sá P, Bernardo Noronha-Matos J. Could hypoxia rehabilitate the osteochondral diseased interface? Lessons from the interplay of hypoxia and purinergic signals elsewhere. Biochem Pharmacol 2023:115646. [PMID: 37321413 DOI: 10.1016/j.bcp.2023.115646] [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: 04/07/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
The osteochondral unit comprises the articular cartilage (90%), subchondral bone (5%) and calcified cartilage (5%). All cells present at the osteochondral unit that is ultimately responsible for matrix production and osteochondral homeostasis, such as chondrocytes, osteoblasts, osteoclasts and osteocytes, can release adenine and/or uracil nucleotides to the local microenvironment. Nucleotides are released by these cells either constitutively or upon plasma membrane damage, mechanical stress or hypoxia conditions. Once in the extracellular space, endogenously released nucleotides can activate membrane-bound purinoceptors. Activation of these receptors is fine-tuning regulated by nucleotides' breakdown by enzymes of the ecto-nucleotidase cascade. Depending on the pathophysiological conditions, both the avascular cartilage and the subchondral bone subsist to significant changes in oxygen tension, which has a tremendous impact on tissue homeostasis. Cell stress due to hypoxic conditions directly influences the expression and activity of several purinergic signalling players, namely nucleotide release channels (e.g. Cx43), NTPDase enzymes and purinoceptors. This review gathers experimental evidence concerning the interplay between hypoxia and the purinergic signalling cascade contributing to osteochondral unit homeostasis. Reporting deviations to this relationship resulting from pathological alterations of articular joints may ultimately unravel novel therapeutic targets for osteochondral rehabilitation. At this point, one can only hypothesize how hypoxia mimetic conditions can be beneficial to the ex vivo expansion and differentiation of osteo- and chondro-progenitors for auto-transplantation and tissue regenerative purposes.
Collapse
Affiliation(s)
- Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP).
| |
Collapse
|
49
|
Stam LB, Clark AL. Chondrocyte primary cilia lengthening and shortening in response to mediators of osteoarthritis; a role for integrin α1β1 and focal adhesions. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100357. [PMID: 37008821 PMCID: PMC10063384 DOI: 10.1016/j.ocarto.2023.100357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Objective Integrin α1β1 protects against osteoarthritis when it is upregulated in the early stages of disease, however, the mechanism behind this is currently unknown. Hypo-osmotic stress, interleukin-1 (IL-1) and transforming growth factor β (TGFβ) influence chondrocyte signaling and are important mediators of osteoarthritis. Evidence for primary cilia as a signaling hub for these factors and the involvement of the F-actin cytoskeleton in this response is growing. The purpose of this study was to investigate the role of integrin α1β1 in the response of primary cilia and the F-actin cytoskeleton to these osteoarthritic mediators. Design Primary cilia length and the number of F-actin peaks were measured in ex vivo wild type and itga1-null chondrocytes in response to hypo-osmotic stress, IL-1, and TGFβ alone or in combination, and with or without focal adhesion kinase inhibitor. Results We show that integrin α1β1 and focal adhesions are necessary for cilial lengthening and increases in F-actin peaks with hypo-osmotic stress and IL-1, but are not required for cilial shortening with TGFβ. Furthermore, we established that the chondrocyte primary cilium has a resting length of 2.4 μm, a minimum length of 2.1 μm corresponding to the thickness of the pericellular matrix, and a maximum length of 3.0 μm. Conclusions While integrin α1β1 is not necessary for the formation of chondrocyte primary cilia and cilial shortening in response to TGFβ, it is necessary for the mediation of cilial lengthening and the formation of F-actin peaks in response to hypo-osmotic stress and IL-1.
Collapse
|
50
|
Wan J, Zhu Z, He Z, Wu H, Chen A, Zhu W, Cheng P. Stevioside protects primary articular chondrocytes against IL-1β-induced inflammation and catabolism by targeting integrin. Int Immunopharmacol 2023; 119:110261. [PMID: 37167638 DOI: 10.1016/j.intimp.2023.110261] [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: 01/04/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Osteoarthritis (OA) is a common, progressive, and chronic disorder of the joints that is characterized by the inflammation and degradation of articular cartilage and is known to significantly impair quality of daily life. Stevioside (SVS) is a natural diterpenoid glycoside that has anti-inflammatory benefits. Hence, in the current research, it was hypothesized that SVS might exert anti-inflammatory effects on articular chondrocytes and alleviate cartilage degradation in mice with OA. The expression of inflammatory cytokines, like inducible nitric oxide synthase (iNOS), NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3), and cyclooxygenase-2 (COX-2) in chondrocytes after interleukin-1β (IL-1β) exposure, was inhibited by the pretreatment of SVS. As well, SVS inhibited the reduction of collagen II and sry-box transcription factor 9 (SOX9) in chondrocytes stimulated by IL-1β and suppressed the expression of MMP3 and MMP13. Further, after treatment with SVS, cell cytometry, autophagy flux, and related protein expression showed diminished cell apoptosis and reduced autophagy impairment. Moreover, SVS blocked the activation of phosphoinositide-3-kinase/Akt/nuclear factor-kappa beta (PI3K/Akt/NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways stimulated by IL-1β. This resulted in decreased cellular inflammation. In vivo experiments with intra-articular injections of SVS in mice with the DMM mouse model demonstrated a decrease in cartilage degradation and an improvement in subchondral bone remodeling. After the integrin αVβ3-related knockdown using siRNA, a reversed effect was observed on the anti-inflammatory, anabolic promoting, catabolic blocking, and NF-κB and MAPK signaling pathway inhibition of SVS on chondrocytes treated with IL-1β. The above findings highlighted that SVS blocked IL-1β, triggered an inflammatory response in mice chondrocytes, and prevented cartilage degradation in vivo through integrin αVβ3. This suggested that SVS might serve as a novel therapeutic option for OA.
Collapse
Affiliation(s)
- Junlai Wan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Ziqing Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhiyi He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Wentao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|