1
|
González-Cubero E, González-Fernández ML, Esteban-Blanco M, Pérez-Castrillo S, Pérez-Fernández E, Navasa N, Aransay AM, Anguita J, Villar-Suárez V. The Therapeutic Potential of Adipose-Derived Mesenchymal Stem Cell Secretome in Osteoarthritis: A Comprehensive Study. Int J Mol Sci 2024; 25:11287. [PMID: 39457070 PMCID: PMC11508730 DOI: 10.3390/ijms252011287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage degradation and inflammation. This study investigates the therapeutic potential of secretome derived from adipose tissue mesenchymal stem cells (ASCs) in mitigating inflammation and promoting cartilage repair in an in vitro model of OA. Our in vitro model comprised chondrocytes inflamed with TNF. To assess the therapeutic potential of secretome, inflamed chondrocytes were treated with it and concentrations of pro-inflammatory cytokines, metalloproteinases (MMPs) and extracellular matrix markers were measured. In addition, secretome-treated chondrocytes were subject to a microarray analysis to determine which genes were upregulated and which were downregulated. Treating TNF-inflamed chondrocytes with secretome in vitro inhibits the NF-κB pathway, thereby mediating anti-inflammatory and anti-catabolic effects. Additional protective effects of secretome on cartilage are revealed in the inhibition of hypertrophy markers such as RUNX2 and COL10A1, increased production of COL2A1 and ACAN and upregulation of SOX9. These findings suggest that ASC-derived secretome can effectively reduce inflammation, promote cartilage repair, and maintain chondrocyte phenotype. This study highlights the potential of ASC-derived secretome as a novel, non-cell-based therapeutic approach for OA, offering a promising alternative to current treatments by targeting inflammation and cartilage repair mechanisms.
Collapse
Affiliation(s)
- Elsa González-Cubero
- Department of Neurosurgery, Stanford School of Medicine, Stanford University, Palo Alto, CA 94304, USA;
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Maria Luisa González-Fernández
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Marta Esteban-Blanco
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Saúl Pérez-Castrillo
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Esther Pérez-Fernández
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
| | - Nicolás Navasa
- Department of Molecular Biology, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain;
- Center for Cooperative Research in Biosciences (CIC bioGUNE)-Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Building 801-A, 48160 Derio, Spain; (A.M.A.); (J.A.)
| | - Ana M. Aransay
- Center for Cooperative Research in Biosciences (CIC bioGUNE)-Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Building 801-A, 48160 Derio, Spain; (A.M.A.); (J.A.)
- CIBERehd, ISCIII, 28029 Madrid, Spain
| | - Juan Anguita
- Center for Cooperative Research in Biosciences (CIC bioGUNE)-Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Building 801-A, 48160 Derio, Spain; (A.M.A.); (J.A.)
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Vega Villar-Suárez
- Department of Anatomy, Faculty of Veterinary Sciences, Campus de Vegazana, University of Léon-Universidad de León, 24071 León, Spain; (M.L.G.-F.); (M.E.-B.); (S.P.-C.); (E.P.-F.)
- Institute of Biomedicine (IBIOMED), Faculty of Veterinary Sciences, Campus de Vegazana, University of León-Universidad de León, 24071 León, Spain
| |
Collapse
|
2
|
Wongin-Sangphet S, Chotiyarnwong P, Viravaidya-Pasuwat K. Reduced Cell Migration in Human Chondrocyte Sheets Increases Tissue Stiffness and Cartilage Protein Production. Tissue Eng Regen Med 2024; 21:1021-1036. [PMID: 39037474 PMCID: PMC11416440 DOI: 10.1007/s13770-024-00662-0] [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: 02/19/2024] [Revised: 05/23/2024] [Accepted: 07/02/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Chondrogenic differentiation medium (CDM) is usually used to maintain chondrogenic activity during chondrocyte sheet production. However, tissue qualities remain to be determined as to what factors improve cell functions. Moreover, the relationship between CDM and cell migration proteins has not been reported. METHOD In this study, the effect of CDM on the behavior of chondrocyte sheets was investigated. Structural analysis, mechanical testing and proteomics were performed to observe tissue qualities. The relationship between CDM and cell migration proteins were investigated using time-lapse observations and bioinformatic analysis. RESULTS During 48 h, CDM affected the chondrocyte behaviors by reducing cell migration. Compared to the basal medium, CDM impacted the contraction of monolayered chondrocyte sheets. At day 7, the contracted sheets increased tissue thickness and improved tissue stiffness. Cartilage specific proteins were also upregulated. Remarkedly, the chondrocyte sheets in CDM displayed downregulated proteins related to cell migration. Bioinformatic analysis revealed that TGFβ1 was shown to be associated with cartilage functions and cell migration. Pathway analysis of chondrocyte sheets in CDM also revealed the presence of a TGFβ pathway without activating actin production, which might be involved in synthesizing cartilage-specific proteins. Cell migration pathway showed MAPK signaling in both cultures of the chondrocyte sheets. CONCLUSION Reduced cell migration in the chondrocyte sheet affected the tissue quality. Using CDM, TGFβ1 might trigger cartilage protein production through the TGFβ pathway and be involved in cell migration via the MAPK signaling pathway. Understanding cell behaviors and their protein expression would be beneficial for developing high-quality tissue-engineered cartilage.
Collapse
Affiliation(s)
- Sopita Wongin-Sangphet
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand.
| | - Pojchong Chotiyarnwong
- Department of Orthopedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Kwanchanok Viravaidya-Pasuwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
- Department of Chemical Engineering and Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
| |
Collapse
|
3
|
Gao Y, Wang J, Dai W, Li S, Liu Q, Zhao X, Fu W, Xiao Y, Guo L, Fan Y, Zhang X. Collagen-based hydrogels induce hyaline cartilage regeneration by immunomodulation and homeostasis maintenance. Acta Biomater 2024; 186:108-124. [PMID: 39067644 DOI: 10.1016/j.actbio.2024.07.018] [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: 02/26/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
Type I collagen (Col I) and hyaluronic acid (HA), derived from the extracellular matrix (ECM), have found widespread application in cartilage tissue engineering. Nevertheless, the potential of cell-free collagen-based scaffolds to induce in situ hyaline cartilage regeneration and the related mechanisms remain undisclosed. Here, we chose Col I and HA to construct Col I hydrogel and Col I-HA composite hydrogel with similar mechanical properties, denoted as Col and ColHA, respectively. Their potential to induce cartilage regeneration was investigated. The results revealed that collagen-based hydrogels could regenerate hyaline cartilage without any additional cells or growth factors. Notably, ColHA hydrogel stood out in this regard. It elicited a moderate activation, recruitment, and reprogramming of macrophages, thus efficiently mitigating local inflammation. Additionally, ColHA hydrogel enhanced stem cell recruitment, facilitated their chondrogenic differentiation, and inhibited chondrocyte fibrosis, hypertrophy, and catabolism, thereby preserving cartilage homeostasis. This study augments our comprehension of cartilage tissue induction theory by enriching immune-related mechanisms, offering innovative prospects for the design of cartilage defect repair scaffolds. STATEMENT OF SIGNIFICANCE: The limited self-regeneration ability and post-injury inflammation pose significant challenges to articular cartilage repair. Type I collagen (Col I) and hyaluronic acid (HA) are extensively used in cartilage tissue engineering. However, their specific roles in cartilage regeneration remain poorly understood. This study aimed to elucidate the functions of Col I and Col I-HA composite hydrogels (ColHA) in orchestrating inflammatory responses and promoting cartilage regeneration. ColHA effectively activated and recruited macrophages, reprogramming them from an M1 to an M2 phenotype, thus alleviating local inflammation. Additionally, ColHA facilitated stem cell homing, induced chondrogenesis, and concurrently inhibited fibrosis, hypertrophy, and catabolism, collectively contributing to the maintenance of cartilage homeostasis. These findings underscore the clinical potential of ColHA for repairing cartilage defects.
Collapse
Affiliation(s)
- Yongli Gao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Jing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Wenling Dai
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Shikui Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Qingli Liu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Xingchen Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Weili Fu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610064, China
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Likun Guo
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China; School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| |
Collapse
|
4
|
Campbell K, Naire S, Kuiper JH. A mathematical model of signalling molecule-mediated processes during regeneration of osteochondral defects after chondrocyte implantation. J Theor Biol 2024; 592:111874. [PMID: 38908475 DOI: 10.1016/j.jtbi.2024.111874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/12/2024] [Accepted: 06/06/2024] [Indexed: 06/24/2024]
Abstract
Treating bone-cartilage defects is a fundamental clinical problem. The ability of damaged cartilage to self-repair is limited due to its avascularity. Left untreated, these defects can lead to osteoarthritis. Details of osteochondral defect repair are elusive, but animal models indicate healing occurs via an endochondral ossification-like process, similar to that in the growth plate. In the growth plate, the signalling molecules parathyroid hormone-related protein (PTHrP) and Indian Hedgehog (Ihh) form a feedback loop regulating chondrocyte hypertrophy, with Ihh inducing and PTHrP suppressing hypertrophy. To better understand this repair process and to explore the regulatory role of signalling molecules on the regeneration process, we formulate a reaction-diffusion mathematical model of osteochondral defect regeneration after chondrocyte implantation. The drivers of healing are assumed to be chondrocytes and osteoblasts, and their interaction via signalling molecules. We model cell proliferation, migration and chondrocyte hypertrophy, and matrix production and conversion, spatially and temporally. We further model nutrient and signalling molecule diffusion and their interaction with the cells. We consider the PTHrP-Ihh feedback loop as the backbone mechanisms but the model is flexible to incorporate extra signalling mechanisms if needed. Our mathematical model is able to represent repair of osteochondral defects, starting with cartilage formation throughout the defect. This is followed by chondrocyte hypertrophy, matrix calcification and bone formation deep inside the defect, while cartilage at the surface is maintained and eventually separated from the deeper bone by a thin layer of calcified cartilage. The complete process requires around 48 months. A key highlight of the model demonstrates that the PTHrP-Ihh loop alone is insufficient and an extra mechanism is required to initiate chondrocyte hypertrophy, represented by a critical cartilage density. A parameter sensitivity study reveals that the timing of the repair process crucially depends on parameters, such as the critical cartilage density, and those describing the actions of PTHrP to suppress hypertrophy, such as its diffusion coefficient, threshold concentration and degradation rate.
Collapse
Affiliation(s)
- Kelly Campbell
- School of Computing and Mathematics, Keele University, Keele, ST5 5BG, UK
| | - Shailesh Naire
- School of Computing and Mathematics, Keele University, Keele, ST5 5BG, UK
| | - Jan Herman Kuiper
- School of Pharmacy and Bioengineering, Keele University, Keele, ST5 5BG, UK; Robert Jones and Agnes Hunt Orthopaedic & District Hospital NHS Trust, Oswestry, SY10 7AG, UK.
| |
Collapse
|
5
|
Soni N, Niranjane P, Purohit A. Role of Growth Factors in Nasal Cartilage Development and Molding: A Comprehensive Review. Cureus 2024; 16:e67202. [PMID: 39295663 PMCID: PMC11409944 DOI: 10.7759/cureus.67202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 08/19/2024] [Indexed: 09/21/2024] Open
Abstract
This review aims to investigate the properties of growth factors concerning the morphogenesis and development of nasal cartilage, which is fundamentally important for facial form and appearance. Since cartilage lacks a blood supply, it is more difficult to regenerate, as cartilage tissue obtains sustenance by diffusion. Cytokines are signalling molecules that control chondrocyte metabolism and extracellular matrix formation, which is required for cartilage development, homeostasis, and healing. Some craniofacial illnesses alter the composition of the cartilage and the structural organization of growth factors, allowing for moulding. TGF-β (transforming growth factor-β) encourages chondrocyte differentiation, whereas IGF-1 (insulin-like growth factor-1) stimulates cartilage-forming collagen synthesis and chondrocyte multiplication. We used the scoping review approach to present current research on the role of growth factors in the creation and architecture of nasal cartilage. We generally observed this structure before conducting specific experiments to determine the impact of growth agents on the development of chondrocytes and cartilage. Prominent findings increase our understanding of how growth factors influence the extracellular matrix, cell activities and features, and cartilage growth rate; all are critical for cartilage tissue development and repair. Research into growth factors and their physiological interactions with cartilage may help improve treatment's functional and aesthetic outcomes and our understanding of the origins and consequences of nasal congenital anomalies. This study emphasizes the importance of expanding knowledge and experience, as well as the use of growth factors in clinical practice, to stimulate cartilage development.
Collapse
Affiliation(s)
- Nikita Soni
- Department of Orthodontics and Dentofacial Orthopaedics, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Priyanka Niranjane
- Department of Orthodontics and Dentofacial Orthopaedics, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Akanksha Purohit
- Neglected Tropical Diseases, Global Health Strategies, Delhi, IND
| |
Collapse
|
6
|
Morris JL, Letson HL, McEwen PC, Dobson GP. Adenosine, lidocaine, and magnesium therapy augments joint tissue healing following experimental anterior cruciate ligament rupture and reconstruction. Bone Joint Res 2024; 13:279-293. [PMID: 38843878 PMCID: PMC11156504 DOI: 10.1302/2046-3758.136.bjr-2023-0360.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/10/2024] Open
Abstract
Aims Adenosine, lidocaine, and Mg2+ (ALM) therapy exerts differential immuno-inflammatory responses in males and females early after anterior cruciate ligament (ACL) reconstruction (ACLR). Our aim was to investigate sex-specific effects of ALM therapy on joint tissue repair and recovery 28 days after surgery. Methods Male (n = 21) and female (n = 21) adult Sprague-Dawley rats were randomly divided into ALM or Saline control treatment groups. Three days after ACL rupture, animals underwent ACLR. An ALM or saline intravenous infusion was commenced prior to skin incision, and continued for one hour. An intra-articular bolus of ALM or saline was also administered prior to skin closure. Animals were monitored to 28 days, and joint function, pain, inflammatory markers, histopathology, and tissue repair markers were assessed. Results Despite comparable knee function, ALM-treated males had reduced systemic inflammation, synovial fluid angiogenic and pro-inflammatory mediators, synovitis, and fat pad fibrotic changes, compared to controls. Within the ACL graft, ALM-treated males had increased expression of tissue repair markers, decreased inflammation, increased collagen organization, and improved graft-bone healing. In contrast to males, females had no evidence of persistent systemic inflammation. Compared to controls, ALM-treated females had improved knee extension, gait biomechanics, and elevated synovial macrophage inflammatory protein-1 alpha (MIP-1α). Within the ACL graft, ALM-treated females had decreased inflammation, increased collagen organization, and improved graft-bone healing. In articular cartilage of ALM-treated animals, matrix metalloproteinase (MMP)-13 expression was blunted in males, while in females repair markers were increased. Conclusion At 28 days, ALM therapy reduces inflammation, augments tissue repair patterns, and improves joint function in a sex-specific manner. The study supports transition to human safety trials.
Collapse
Affiliation(s)
- Jodie L. Morris
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Hayley L. Letson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Peter C. McEwen
- Orthopaedic Research Institute of Queensland, Townsville, Australia
| | - Geoffrey P. Dobson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| |
Collapse
|
7
|
Damerau A, Rosenow E, Alkhoury D, Buttgereit F, Gaber T. Fibrotic pathways and fibroblast-like synoviocyte phenotypes in osteoarthritis. Front Immunol 2024; 15:1385006. [PMID: 38895122 PMCID: PMC11183113 DOI: 10.3389/fimmu.2024.1385006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis, characterized by osteophyte formation, cartilage degradation, and structural and cellular alterations of the synovial membrane. Activated fibroblast-like synoviocytes (FLS) of the synovial membrane have been identified as key drivers, secreting humoral mediators that maintain inflammatory processes, proteases that cause cartilage and bone destruction, and factors that drive fibrotic processes. In normal tissue repair, fibrotic processes are terminated after the damage has been repaired. In fibrosis, tissue remodeling and wound healing are exaggerated and prolonged. Various stressors, including aging, joint instability, and inflammation, lead to structural damage of the joint and micro lesions within the synovial tissue. One result is the reduced production of synovial fluid (lubricants), which reduces the lubricity of the cartilage areas, leading to cartilage damage. In the synovial tissue, a wound-healing cascade is initiated by activating macrophages, Th2 cells, and FLS. The latter can be divided into two major populations. The destructive thymocyte differentiation antigen (THY)1─ phenotype is restricted to the synovial lining layer. In contrast, the THY1+ phenotype of the sublining layer is classified as an invasive one with immune effector function driving synovitis. The exact mechanisms involved in the transition of fibroblasts into a myofibroblast-like phenotype that drives fibrosis remain unclear. The review provides an overview of the phenotypes and spatial distribution of FLS in the synovial membrane of OA, describes the mechanisms of fibroblast into myofibroblast activation, and the metabolic alterations of myofibroblast-like cells.
Collapse
Affiliation(s)
- Alexandra Damerau
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| | - Emely Rosenow
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Dana Alkhoury
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| |
Collapse
|
8
|
Foltz L, Avabhrath N, Lanchy JM, Levy T, Possemato A, Ariss M, Peterson B, Grimes M. Craniofacial chondrogenesis in organoids from human stem cell-derived neural crest cells. iScience 2024; 27:109585. [PMID: 38623327 PMCID: PMC11016914 DOI: 10.1016/j.isci.2024.109585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
Knowledge of cell signaling pathways that drive human neural crest differentiation into craniofacial chondrocytes is incomplete, yet essential for using stem cells to regenerate craniomaxillofacial structures. To accelerate translational progress, we developed a differentiation protocol that generated self-organizing craniofacial cartilage organoids from human embryonic stem cell-derived neural crest stem cells. Histological staining of cartilage organoids revealed tissue architecture and staining typical of elastic cartilage. Protein and post-translational modification (PTM) mass spectrometry and snRNA-seq data showed that chondrocyte organoids expressed robust levels of cartilage extracellular matrix (ECM) components: many collagens, aggrecan, perlecan, proteoglycans, and elastic fibers. We identified two populations of chondroprogenitor cells, mesenchyme cells and nascent chondrocytes, and the growth factors involved in paracrine signaling between them. We show that ECM components secreted by chondrocytes not only create a structurally resilient matrix that defines cartilage, but also play a pivotal autocrine cell signaling role in determining chondrocyte fate.
Collapse
Affiliation(s)
- Lauren Foltz
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Nagashree Avabhrath
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Jean-Marc Lanchy
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Tyler Levy
- Cell Signaling Technology, Danvers, MA 01923, USA
| | | | - Majd Ariss
- Cell Signaling Technology, Danvers, MA 01923, USA
| | | | - Mark Grimes
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
9
|
Xu T, Yu X, Xu K, Lin Y, Wang J, Pan Z, Fang J, Wang S, Zhou Z, Song H, Zhu S, Dai X. Comparison of the ability of exosomes and ectosomes derived from adipose-derived stromal cells to promote cartilage regeneration in a rat osteochondral defect model. Stem Cell Res Ther 2024; 15:18. [PMID: 38229196 PMCID: PMC10792834 DOI: 10.1186/s13287-024-03632-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/04/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) offer promising prospects for stimulating cartilage regeneration. The different formation mechanisms suggest that exosomes and ectosomes possess different biological functions. However, little attention has been paid to the differential effects of EV subsets on cartilage regeneration. METHODS Our study compared the effects of the two EVs isolated from adipose-derived MSCs (ASCs) on chondrocytes and bone marrow-derived MSCs (BMSCs) in vitro. Additionally, we loaded the two EVs into type I collagen hydrogels to optimize their application for the treatment of osteochondral defects in vivo. RESULTS In vitro experiments demonstrate that ASC-derived exosomes (ASC-Exos) significantly promoted the proliferation and migration of both cells more effectively than ASC-derived ectosomes (ASC-Ectos). Furthermore, ASC-Exos facilitated a stronger differentiation of BMSCs into chondrogenic cells than ASC-Ectos, but both inhibited chondrocyte apoptosis to a similar extent. In the osteochondral defect model of rats, ASC-Exos promoted cartilage regeneration in situ better than ASC-Ectos. At 8 weeks, the hydrogel containing exosomes group (Gel + Exo group) had higher macroscopic and histological scores, a higher value of trabecular bone volume fraction (BV/TV), a lower value of trabecular thickness (Tb.Sp), and a better remodeling of extracellular matrix than the hydrogel containing ectosomes group (Gel + Ecto group). At 4 and 8 weeks, the expression of CD206 and Arginase-1 in the Gel + Exo group was significantly higher than that in the Gel + Ecto group. CONCLUSION Our findings indicate that administering ASC-Exos may be a more effective EV strategy for cartilage regeneration than the administration of ASC-Ectos.
Collapse
Affiliation(s)
- Tengjing Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Xinning Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Kaiwang Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Yunting Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Jiajie Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Zongyou Pan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Jinghua Fang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Siheng Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Zhuxing Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Hongyun Song
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Sunan Zhu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China
| | - Xuesong Dai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, People's Republic of China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, People's Republic of China.
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, People's Republic of China.
| |
Collapse
|
10
|
Shi R, Li X, Xu X, Chen Z, Zhu Y, Wang N. Genome-wide analysis of BMP/GDF family and DAP-seq of YY1 suggest their roles in Cynoglossus semilaevis sexual size dimorphism. Int J Biol Macromol 2023; 253:127201. [PMID: 37793513 DOI: 10.1016/j.ijbiomac.2023.127201] [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: 08/02/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/06/2023]
Abstract
Sexual size dimorphism (SSD) characterized by different body size between females and males have been reported in various animals. Gonadectomy experiments have implied important regulatory roles of the gonad in SSD. Among multiple factors from the gonad, TGF-β superfamily (especially BMP/GDF family) attracted our interest due to its pleiotropy in growth and reproduction regulations. Thus, whether BMP/GDF family members serve as crucial regulators for SSD was studied in a typically female-biased SSD flatfish named Chinese tongue sole (Cynoglossus semilaevis). Firstly, a total of 26 BMP/GDF family members were identified. The PPI network analysis showed that they may interact with ACVR2a, ACVR2b, ACVR1, BMPR2, SMAD3, BMPR1a, and other proteins. Subsequently, DAP-seq was employed to reveal the binding sites for yin yang 1 (yy1), a transcription factor involved in gonad function and cell growth partly by regulating TGF-β superfamily. The results revealed that two yy1 homologues yy1a and yy1b in C. semilaevis could regulate Hippo signaling pathway, mTOR signaling pathway, and AMPK signaling pathway. Moreover, BMP/GDF family genes including bmp2, bmp4, bmp5, gdf6a, and gdf6b were important components of Hippo pathway. In future, the crosstalk among yy1a, yy1b, and TGF-β family would provide more insight into sexual size dimorphism in C. semilaevis.
Collapse
Affiliation(s)
- Rui Shi
- Function Laboratory for Marine Science and Food Production Process, Laoshan laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Xihong Li
- Function Laboratory for Marine Science and Food Production Process, Laoshan laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Xiwen Xu
- Function Laboratory for Marine Science and Food Production Process, Laoshan laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Zhangfan Chen
- Function Laboratory for Marine Science and Food Production Process, Laoshan laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Ying Zhu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, China.
| | - Na Wang
- Function Laboratory for Marine Science and Food Production Process, Laoshan laboratory, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China.
| |
Collapse
|
11
|
George T, Curley AJ, Saeed SK, Kuhns BD, Parsa A, Domb BG. Orthobiologics as an adjunct in treatment of femoroacetabular impingement syndrome: cell-based therapies facilitate improved postoperative outcomes in the setting of acetabular chondral lesions-a systematic review. Knee Surg Sports Traumatol Arthrosc 2023; 31:6020-6038. [PMID: 37906291 DOI: 10.1007/s00167-023-07624-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 10/09/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE To evaluate studies utilizing orthobiologics in the management of femoroacetabular impingement syndrome (FAIS) to (1) assess the indications for usage, and (2) analyze patient-reported outcome measures (PROM) following treatment. It was hypothesized that orthobiologics would (1) be utilized for symptomatic FAIS in the setting of labral or chondral pathology, and (2) improve PROM at most recent follow-up. METHODS The Pubmed, Ovid Medline, Cochrane, and Web of Science databases were searched for clinical studies evaluating orthobiologics [hyaluronic acid (HA), platelet-rich plasma (PRP), or cell-based therapy (CBT) for treatment of FAIS. Exclusion criteria included orthobiologics used in conjunction with cartilage transfer or scaffolding procedures and a primary indication other than FAIS. Data collection included patient demographics, indications, and baseline and most recent PROM. RESULTS Eleven studies (one level I, four level II, four level III, and two level IV evidence) met inclusion criteria, consisting of 440 patients with mean ages ranging from 32.8 to 47 years. All 11 studies demonstrated an improvement in PROM from baseline to most-recent follow-up. Four studies administered PRP either intraoperatively or the day after surgery as an adjunct to labral repair. CBT was used intraoperatively in the setting of acetabular chondral lesions (three studies) and labral repair (one study). When comparing to a control group at most recent follow-up, three PRP cohorts demonstrated similar PROM (n.s.), while one PRP group exhibited worse visual analog pain scores (2.5 vs. 3.4, p = 0.005) and modified Harris Hip Scores (mHHS) (82.6 vs. 78.7, p = 0.049). The four CBT studies reported favorable results compared to a control group, with a significantly higher mHHS at most recent follow-up or mean improvement from baseline in Hip Outcome Score-Activities of Daily Living (p < 0.05). Three studies reported on HA, which was utilized exclusively in the nonoperative setting. CONCLUSIONS Intraoperative PRP and CBT have been commonly reported in the setting of hip arthroscopy for labral repairs and acetabular chondral lesions, respectively. The CBT cohorts demonstrated more favorable PROM at most recent follow-up when compared to a control group, though these results should be interpreted with caution due to heterogeneity of orthobiologic preparations. LEVEL OF EVIDENCE IV.
Collapse
Affiliation(s)
- Tracy George
- American Hip Institute Research Foundation, Chicago, IL, 60018, USA
| | - Andrew J Curley
- American Hip Institute Research Foundation, Chicago, IL, 60018, USA
| | - Sheema K Saeed
- American Hip Institute Research Foundation, Chicago, IL, 60018, USA
| | - Benjamin D Kuhns
- American Hip Institute Research Foundation, Chicago, IL, 60018, USA
| | - Ali Parsa
- American Hip Institute Research Foundation, Chicago, IL, 60018, USA
| | - Benjamin G Domb
- American Hip Institute Research Foundation, Chicago, IL, 60018, USA.
- American Hip Institute, Chicago, IL, 60018, USA.
- , 999 E Touhy Ave, Suite 450, Des Plaines, IL, 60018, USA.
| |
Collapse
|
12
|
Pitou M, Papachristou E, Bratsios D, Kefala GM, Tsagkarakou AS, Leonidas DD, Aggeli A, Papadopoulos GE, Papi RM, Choli-Papadopoulou T. In Vitro Chondrogenesis Induction by Short Peptides of the Carboxy-Terminal Domain of Transforming Growth Factor β1. Biomedicines 2023; 11:3182. [PMID: 38137403 PMCID: PMC10740954 DOI: 10.3390/biomedicines11123182] [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: 09/17/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023] Open
Abstract
Τransforming growth factor β1 (TGF-β1) comprises a key regulator protein in many cellular processes, including in vivo chondrogenesis. The treatment of human dental pulp stem cells, separately, with Leu83-Ser112 (C-terminal domain of TGF-β1), as well as two very short peptides, namely, 90-YYVGRKPK-97 (peptide 8) and 91-YVGRKP-96 (peptide 6) remarkably enhanced the chondrogenic differentiation capacity in comparison to their full-length mature TGF-β1 counterpart either in monolayer cultures or 3D scaffolds. In 3D scaffolds, the reduction of the elastic modulus and viscous modulus verified the production of different amounts and types of ECM components. Molecular dynamics simulations suggested a mode of the peptides' binding to the receptor complex TβRII-ALK5 and provided a possible structural explanation for their role in inducing chondrogenesis, along with endogenous TGF-β1. Further experiments clearly verified the aforementioned hypothesis, indicating the signal transduction pathway and the involvement of TβRII-ALK5 receptor complex. Real-time PCR experiments and Western blot analysis showed that peptides favor the ERK1/2 and Smad2 pathways, leading to an articular, extracellular matrix formation, while TGF-β1 also favors the Smad1/5/8 pathway which leads to the expression of the metalloproteinases ADAMTS-5 and MMP13 and, therefore, to a hypertrophic chondrocyte phenotype. Taken together, the two short peptides, and, mainly, peptide 8, could be delivered with a scaffold to induce in vivo chondrogenesis in damaged articular cartilage, constituting, thus, an alternative therapeutic approach for osteoarthritis.
Collapse
Affiliation(s)
- Maria Pitou
- Laboratory of Biochemistry, School of Chemistry, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Eleni Papachristou
- Laboratory of Biochemistry, School of Chemistry, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Dimitrios Bratsios
- Laboratory of Biomedical Engineering, School of Chemical Engineering, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Georgia-Maria Kefala
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Anastasia S. Tsagkarakou
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Demetrios D. Leonidas
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Amalia Aggeli
- Laboratory of Biomedical Engineering, School of Chemical Engineering, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Georgios E. Papadopoulos
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Rigini M. Papi
- Laboratory of Biochemistry, School of Chemistry, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, School of Chemistry, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| |
Collapse
|
13
|
Sun K, Guo J, Guo Z, Hou L, Liu H, Hou Y, He J, Guo F, Ye Y. The roles of the Hippo-YAP signalling pathway in Cartilage and Osteoarthritis. Ageing Res Rev 2023; 90:102015. [PMID: 37454824 DOI: 10.1016/j.arr.2023.102015] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Osteoarthritis (OA) is an age-related disease, characterized by cartilage degeneration. The pathogenesis of OA is complicated and the current therapeutic approaches for OA are limited. Cartilage, an integral part of the skeletal system composed of chondrocytes, is essential for skeletal development, tissue patterning, and maintaining the normal activity of joints. The development, homeostasis and degeneration of cartilage are tightly associated with OA. Over the past decade, accumulating evidence indicates that Hippo/YAP is a vital biochemical signalling pathway that strictly governs tissue development and homeostasis. The joint tissues, especially for cartilage, are sensitive to changes of Hippo/YAP signalling. In this review, we summarize the role of Hippo/YAP signalling in cartilage and discuss its involvement in OA progression from points of cartilage degradation, subchondral bone remodeling, and synovial alteration. We also highlight the potential therapeutic implications of Hippo/YAP signalling and further discuss current limitations and controversy on Hippo/YAP-based application for OA treatment.
Collapse
Affiliation(s)
- Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiachao Guo
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhou Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Liangcai Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Haigang Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yanjun Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Junchen He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
14
|
Uzieliene I, Bialaglovyte P, Miksiunas R, Lebedis I, Pachaleva J, Vaiciuleviciute R, Ramanaviciene A, Kvederas G, Bernotiene E. Menstrual Blood-Derived Stem Cell Paracrine Factors Possess Stimulatory Effects on Chondrogenesis In Vitro and Diminish the Degradation of Articular Cartilage during Osteoarthritis. Bioengineering (Basel) 2023; 10:1001. [PMID: 37760103 PMCID: PMC10525204 DOI: 10.3390/bioengineering10091001] [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: 07/18/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Articular cartilage is an avascular tissue with a limited capacity for self-regeneration, leading the tissue to osteoarthritis (OA). Mesenchymal stem cells (MSCs) are promising for cartilage tissue engineering, as they are capable of differentiating into chondrocyte-like cells and secreting a number of active molecules that are important for cartilage extracellular matrix (ECM) synthesis. The aim of this study was to evaluate the potential of easily accessible menstrual blood-derived MSC (MenSC) paracrine factors in stimulating bone marrow MSC (BMMSCs) chondrogenic differentiation and to investigate their role in protecting cartilage from degradation in vitro. MenSCs and BMMSCs chondrogenic differentiation was induced using four different growth factors: TGF-β3, activin A, BMP-2, and IGF-1. The chondrogenic differentiation of BMMSCs was stimulated in co-cultures with MenSCs and cartilage explants co-cultured with MenSCs for 21 days. The chondrogenic capacity of BMMSCs was analyzed by the secretion of four growth factors and cartilage oligomeric matrix protein, as well as the release and synthesis of cartilage ECM proteins, and chondrogenic gene expression in cartilage explants. Our results suggest that MenSCs stimulate chondrogenic response in BMMSCs by secreting activin A and TGF-β3 and may have protective effects on cartilage tissue ECM by decreasing the release of GAGs, most likely through the modulation of activin A related molecular pathway. In conclusion, paracrine factors secreted by MenSCs may turn out to be a promising therapeutical approach for cartilage tissue protection and repair.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (P.B.); (R.M.); (I.L.); (J.P.); (R.V.); (E.B.)
| | - Paulina Bialaglovyte
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (P.B.); (R.M.); (I.L.); (J.P.); (R.V.); (E.B.)
| | - Rokas Miksiunas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (P.B.); (R.M.); (I.L.); (J.P.); (R.V.); (E.B.)
| | - Ignas Lebedis
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (P.B.); (R.M.); (I.L.); (J.P.); (R.V.); (E.B.)
| | - Jolita Pachaleva
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (P.B.); (R.M.); (I.L.); (J.P.); (R.V.); (E.B.)
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (P.B.); (R.M.); (I.L.); (J.P.); (R.V.); (E.B.)
| | - Almira Ramanaviciene
- Department of Immunology, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania;
- NanoTechnas—Center on Nanotechnology and Materials Sciences, Faculty of Chemistry and Geosciences, Vilnius University, LT-03225 Vilnius, Lithuania
| | - Giedrius Kvederas
- The Clinic of Rheumatology, Traumatology Orthopaedics and Reconstructive Surgery, Institute of Clinical Medicine of the Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania;
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (P.B.); (R.M.); (I.L.); (J.P.); (R.V.); (E.B.)
- Department of Chemistry and Bioengineering, Faculty of Fundamental Sciences, VilniusTech, Vilnius Gediminas Technical University, 10223 Vilnius, Lithuania
| |
Collapse
|
15
|
Zhou L, Xu J, Schwab A, Tong W, Xu J, Zheng L, Li Y, Li Z, Xu S, Chen Z, Zou L, Zhao X, van Osch GJ, Wen C, Qin L. Engineered biochemical cues of regenerative biomaterials to enhance endogenous stem/progenitor cells (ESPCs)-mediated articular cartilage repair. Bioact Mater 2023; 26:490-512. [PMID: 37304336 PMCID: PMC10248882 DOI: 10.1016/j.bioactmat.2023.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/21/2023] [Accepted: 03/13/2023] [Indexed: 06/13/2023] Open
Abstract
As a highly specialized shock-absorbing connective tissue, articular cartilage (AC) has very limited self-repair capacity after traumatic injuries, posing a heavy socioeconomic burden. Common clinical therapies for small- to medium-size focal AC defects are well-developed endogenous repair and cell-based strategies, including microfracture, mosaicplasty, autologous chondrocyte implantation (ACI), and matrix-induced ACI (MACI). However, these treatments frequently result in mechanically inferior fibrocartilage, low cost-effectiveness, donor site morbidity, and short-term durability. It prompts an urgent need for innovative approaches to pattern a pro-regenerative microenvironment and yield hyaline-like cartilage with similar biomechanical and biochemical properties as healthy native AC. Acellular regenerative biomaterials can create a favorable local environment for AC repair without causing relevant regulatory and scientific concerns from cell-based treatments. A deeper understanding of the mechanism of endogenous cartilage healing is furthering the (bio)design and application of these scaffolds. Currently, the utilization of regenerative biomaterials to magnify the repairing effect of joint-resident endogenous stem/progenitor cells (ESPCs) presents an evolving improvement for cartilage repair. This review starts by briefly summarizing the current understanding of endogenous AC repair and the vital roles of ESPCs and chemoattractants for cartilage regeneration. Then several intrinsic hurdles for regenerative biomaterials-based AC repair are discussed. The recent advances in novel (bio)design and application regarding regenerative biomaterials with favorable biochemical cues to provide an instructive extracellular microenvironment and to guide the ESPCs (e.g. adhesion, migration, proliferation, differentiation, matrix production, and remodeling) for cartilage repair are summarized. Finally, this review outlines the future directions of engineering the next-generation regenerative biomaterials toward ultimate clinical translation.
Collapse
Affiliation(s)
- Liangbin Zhou
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
| | - Jietao Xu
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
| | - Andrea Schwab
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
| | - Wenxue Tong
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences - CRMH, 999077, Hong Kong SAR, China
| | - Ye Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Zhuo Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Shunxiang Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Li Zou
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Xin Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology (TU Delft), 2600 AA, Delft, the Netherlands
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
- Centre for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, The Chinese Academy of Sciences, 518000, Shenzhen, China
| |
Collapse
|
16
|
Tran NT, Truong MD, Yun HW, Min BH. Potential of secretome of human fetal cartilage progenitor cells as disease modifying agent for osteoarthritis. Life Sci 2023; 324:121741. [PMID: 37149084 DOI: 10.1016/j.lfs.2023.121741] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Abstract
AIMS Osteoarthritis (OA) is caused by an imbalance in the synthesis and degradation of cartilage tissue by chondrocytes. Therefore, a therapeutic agent for OA patients that can positively affect both synthesis and degradation is needed. However, current nonsurgical treatments for OA can barely achieve satisfactory long-term outcomes in cartilage repair. Human fetal cartilage progenitor cells-secretome (ShFCPC) has shown potent anti-inflammatory and tissue-repair effects; however, its underlying mechanisms and effects on OA have rarely been systematically elucidated. This study aims to analyze and evaluate the potency of ShFCPC in modifying OA process. MAIN METHODS Herein, secreted proteins enriched in ShFCPC have been characterized, and their biological functions both in vitro and in vivo in an OA model are compared with those of human bone marrow-derived mesenchymal stem cells-secretome (ShBMSC) and hyaluronan (HA). KEY FINDINGS Secretome analysis has shown that ShFCPC is significantly enriched with extracellular matrix molecules involved in many effects of cellular processes required for homeostasis during OA progression. Biological validation in vitro has shown that ShFCPC protects chondrocyte apoptosis by suppressing the expression of inflammatory mediators and matrix-degrading proteases and promotes the secretion of pro-chondrogenic cytokines in lipopolysaccharide-induced coculture of human chondrocytes and SW982 synovial cells compared with ShBMSC. Moreover, in a rat OA model, ShFCPC protects articular cartilage by reducing inflammatory cell infiltration and M1/M2 macrophage ratio in the synovium, which directly contributes to an increase in immunomodulatory atmosphere and enhances cartilage repair compared to ShBMSC and HA. SIGNIFICANCE Our findings support clinical translations of ShFCPC as a novel agent for modifying OA process.
Collapse
Affiliation(s)
- Ngoc-Trinh Tran
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Minh-Dung Truong
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Hee-Woong Yun
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Byoung-Hyun Min
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Institute of Regenerative Medicine, Wake Forest University, NC, USA; Advanced Translational Engineering & Medical Science, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Patel J, Chen S, Katzmeyer T, Pei YA, Pei M. Sex-dependent variation in cartilage adaptation: from degeneration to regeneration. Biol Sex Differ 2023; 14:17. [PMID: 37024929 PMCID: PMC10077643 DOI: 10.1186/s13293-023-00500-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Despite acknowledgement in the scientific community of sex-based differences in cartilage biology, the implications for study design remain unclear, with many studies continuing to arbitrarily assign demographics. Clinically, it has been well-established that males and females differ in cartilage degeneration, and accumulating evidence points to the importance of sex differences in the field of cartilage repair. However, a comprehensive review of the mechanisms behind this trend and the influence of sex on cartilage regeneration has not yet been presented. This paper aims to summarize current findings regarding sex-dependent variation in knee anatomy, sex hormones' effect on cartilage, and cartilaginous degeneration and regeneration, with a focus on stem cell therapies. Findings suggest that the stem cells themselves, as well as their surrounding microenvironment, contribute to sex-based differences. Accordingly, this paper underscores the contribution of both stem cell donor and recipient sex to sex-related differences in treatment efficacy. Cartilage regeneration is a field that needs more research to optimize strategies for better clinical results; taking sex into account could be a big factor in developing more effective and personalized treatments. The compilation of this information emphasizes the importance of investing further research in sex differences in cartilage biology.
Collapse
Affiliation(s)
- Jhanvee Patel
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, 610083, Sichuan, China
| | - Torey Katzmeyer
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | - Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
18
|
Mahdavi-Jouibari F, Parseh B, Kazeminejad E, Khosravi A. Hopes and opportunities of stem cells from human exfoliated deciduous teeth (SHED) in cartilage tissue regeneration. Front Bioeng Biotechnol 2023; 11:1021024. [PMID: 36860887 PMCID: PMC9968979 DOI: 10.3389/fbioe.2023.1021024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Cartilage lesions are common conditions, affecting elderly and non-athletic populations. Despite recent advances, cartilage regeneration remains a major challenge today. The absence of an inflammatory response following damage and the inability of stem cells to penetrate into the healing site due to the absence of blood and lymph vessels are assumed to hinder joint repair. Stem cell-based regeneration and tissue engineering have opened new horizons for treatment. With advances in biological sciences, especially stem cell research, the function of various growth factors in the regulation of cell proliferation and differentiation has been established. Mesenchymal stem cells (MSCs) isolated from different tissues have been shown to increase into therapeutically relevant cell numbers and differentiate into mature chondrocytes. As MSCs can differentiate and become engrafted inside the host, they are considered suitable candidates for cartilage regeneration. Stem cells from human exfoliated deciduous teeth (SHED) provide a novel and non-invasive source of MSCs. Due to their simple isolation, chondrogenic differentiation potential, and minimal immunogenicity, they can be an interesting option for cartilage regeneration. Recent studies have reported that SHED-derived secretome contains biomolecules and compounds that efficiently promote regeneration in damaged tissues, including cartilage. Overall, this review highlighted the advances and challenges of cartilage regeneration using stem cell-based therapies by focusing on SHED.
Collapse
Affiliation(s)
- Forough Mahdavi-Jouibari
- Department of Medical Biotechnology, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Benyamin Parseh
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran,Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ezatolah Kazeminejad
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran,Dental Research Center, Golestan University of Medical Sciences, Gorgan, Iran,*Correspondence: Ezatolah Kazeminejad, Dr. ; Ayyoob Khosravi,
| | - Ayyoob Khosravi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran,Department of Molecular Medicine, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran,*Correspondence: Ezatolah Kazeminejad, Dr. ; Ayyoob Khosravi,
| |
Collapse
|
19
|
Li J, Wang G, Xv X, Li Z, Shen Y, Zhang C, Zhang X. Identification of immune-associated genes in diagnosing osteoarthritis with metabolic syndrome by integrated bioinformatics analysis and machine learning. Front Immunol 2023; 14:1134412. [PMID: 37138862 PMCID: PMC10150333 DOI: 10.3389/fimmu.2023.1134412] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Background In the pathogenesis of osteoarthritis (OA) and metabolic syndrome (MetS), the immune system plays a particularly important role. The purpose of this study was to find key diagnostic candidate genes in OA patients who also had metabolic syndrome. Methods We searched the Gene Expression Omnibus (GEO) database for three OA and one MetS dataset. Limma, weighted gene co-expression network analysis (WGCNA), and machine learning algorithms were used to identify and analyze the immune genes associated with OA and MetS. They were evaluated using nomograms and receiver operating characteristic (ROC) curves, and finally, immune cells dysregulated in OA were investigated using immune infiltration analysis. Results After Limma analysis, the integrated OA dataset yielded 2263 DEGs, and the MetS dataset yielded the most relevant module containing 691 genes after WGCNA, with a total of 82 intersections between the two. The immune-related genes were mostly enriched in the enrichment analysis, and the immune infiltration analysis revealed an imbalance in multiple immune cells. Further machine learning screening yielded eight core genes that were evaluated by nomogram and diagnostic value and found to have a high diagnostic value (area under the curve from 0.82 to 0.96). Conclusion Eight immune-related core genes were identified (FZD7, IRAK3, KDELR3, PHC2, RHOB, RNF170, SOX13, and ZKSCAN4), and a nomogram for the diagnosis of OA and MetS was established. This research could lead to the identification of potential peripheral blood diagnostic candidate genes for MetS patients who also suffer from OA.
Collapse
Affiliation(s)
- Junchen Li
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Genghong Wang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xilin Xv
- The Third Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Teaching and Research Section of Orthopedics and Traumatology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhigang Li
- The Second Department of Orthopedics and Traumatology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yiwei Shen
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Cheng Zhang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaofeng Zhang
- Teaching and Research Section of Orthopedics and Traumatology, Heilongjiang University of Chinese Medicine, Harbin, China
- The Bone Injury Teaching Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Xiaofeng Zhang,
| |
Collapse
|
20
|
Capuana E, Marino D, Di Gesù R, La Carrubba V, Brucato V, Tuan RS, Gottardi R. A High-Throughput Mechanical Activator for Cartilage Engineering Enables Rapid Screening of in vitro Response of Tissue Models to Physiological and Supra-Physiological Loads. Cells Tissues Organs 2023; 211:670-688. [PMID: 34261061 PMCID: PMC9843549 DOI: 10.1159/000514985] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/02/2021] [Indexed: 01/25/2023] Open
Abstract
Articular cartilage is crucially influenced by loading during development, health, and disease. However, our knowledge of the mechanical conditions that promote engineered cartilage maturation or tissue repair is still incomplete. Current in vitro models that allow precise control of the local mechanical environment have been dramatically limited by very low throughput, usually just a few specimens per experiment. To overcome this constraint, we have developed a new device for the high throughput compressive loading of tissue constructs: the High Throughput Mechanical Activator for Cartilage Engineering (HiT-MACE), which allows the mechanoactivation of 6 times more samples than current technologies. With HiT-MACE we were able to apply cyclic loads in the physiological (e.g., equivalent to walking and normal daily activity) and supra-physiological range (e.g., injurious impacts or extensive overloading) to up to 24 samples in one single run. In this report, we compared the early response of cartilage to physiological and supra-physiological mechanical loading to the response to IL-1β exposure, a common but rudimentary in vitro model of cartilage osteoarthritis. Physiological loading rapidly upregulated gene expression of anabolic markers along the TGF-β1 pathway. Notably, TGF-β1 or serum was not included in the medium. Supra-physiological loading caused a mild catabolic response while IL-1β exposure drove a rapid anabolic shift. This aligns well with recent findings suggesting that overloading is a more realistic and biomimetic model of cartilage degeneration. Taken together, these findings showed that the application of HiT-MACE allowed the use of larger number of samples to generate higher volume of data to effectively explore cartilage mechanobiology, which will enable the design of more effective repair and rehabilitation strategies for degenerative cartilage pathologies.
Collapse
Affiliation(s)
- Elisa Capuana
- Department of Engineering, University of Palermo, Palermo, Italy,Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Davide Marino
- Department of Engineering, University of Palermo, Palermo, Italy,Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Roberto Di Gesù
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Children's Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA,Fondazione Ri.MED, Palermo, Italy
| | - Vincenzo La Carrubba
- Department of Engineering, University of Palermo, Palermo, Italy,INSTM, Palermo Research Unit, Palermo, Italy
| | - Valerio Brucato
- Department of Engineering, University of Palermo, Palermo, Italy
| | - Rocky S. Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Children's Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA,Fondazione Ri.MED, Palermo, Italy,*Riccardo Gottardi,
| |
Collapse
|
21
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
22
|
Xiong Y, Mi BB, Lin Z, Hu YQ, Yu L, Zha KK, Panayi AC, Yu T, Chen L, Liu ZP, Patel A, Feng Q, Zhou SH, Liu GH. The role of the immune microenvironment in bone, cartilage, and soft tissue regeneration: from mechanism to therapeutic opportunity. Mil Med Res 2022; 9:65. [PMID: 36401295 PMCID: PMC9675067 DOI: 10.1186/s40779-022-00426-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Bone, cartilage, and soft tissue regeneration is a complex spatiotemporal process recruiting a variety of cell types, whose activity and interplay must be precisely mediated for effective healing post-injury. Although extensive strides have been made in the understanding of the immune microenvironment processes governing bone, cartilage, and soft tissue regeneration, effective clinical translation of these mechanisms remains a challenge. Regulation of the immune microenvironment is increasingly becoming a favorable target for bone, cartilage, and soft tissue regeneration; therefore, an in-depth understanding of the communication between immune cells and functional tissue cells would be valuable. Herein, we review the regulatory role of the immune microenvironment in the promotion and maintenance of stem cell states in the context of bone, cartilage, and soft tissue repair and regeneration. We discuss the roles of various immune cell subsets in bone, cartilage, and soft tissue repair and regeneration processes and introduce novel strategies, for example, biomaterial-targeting of immune cell activity, aimed at regulating healing. Understanding the mechanisms of the crosstalk between the immune microenvironment and regeneration pathways may shed light on new therapeutic opportunities for enhancing bone, cartilage, and soft tissue regeneration through regulation of the immune microenvironment.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Bo-Bin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yi-Qiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Kang-Kang Zha
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany
| | - Zhen-Ping Liu
- Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany.,Joint Laboratory of Optofluidic Technology and System,National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, China
| | - Anish Patel
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China.
| | - Shuan-Hu Zhou
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA. .,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
23
|
Shan DD, Zheng QX, Chen Z. Go-Ichi-Ni-San 2: A potential biomarker and therapeutic target in human cancers. World J Gastrointest Oncol 2022; 14:1892-1902. [PMID: 36310704 PMCID: PMC9611433 DOI: 10.4251/wjgo.v14.i10.1892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/15/2022] [Accepted: 09/06/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer incidence and mortality are increasing globally, leading to its rising status as a leading cause of death. The Go-Ichi-Ni-San (GINS) complex plays a crucial role in DNA replication and the cell cycle. The GINS complex consists of four subunits encoded by the GINS1, GINS2, GINS3, and GINS4 genes. Recent findings have shown that GINS2 expression is upregulated in many diseases, particularly tumors. For example, increased GINS2 expression has been found in cervical cancer, gastric adenocarcinoma, glioma, non-small cell lung cancer, and pancreatic cancer. It correlates with the clinicopathological characteristics of the tumors. In addition, high GINS2 expression plays a pro-carcinogenic role in tumor development by promoting tumor cell proliferation and migration, inhibiting tumor cell apoptosis, and blocking the cell cycle. This review describes the upregulation of GINS2 expression in most human tumors and the pathway of GINS2 in tumor development. GINS2 may serve as a new marker for tumor diagnosis and a new biological target for therapy.
Collapse
Affiliation(s)
- Dan-Dan Shan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Qiu-Xian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
24
|
Li L, Wang P, Jin J, Xie C, Xue B, Lai J, Zhu L, Jiang Q. The triply periodic minimal surface-based 3D printed engineering scaffold for meniscus function reconstruction. Biomater Res 2022; 26:45. [PMID: 36115984 PMCID: PMC9482755 DOI: 10.1186/s40824-022-00293-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The meniscus injury is a common disease in the area of sports medicine. The main treatment for this disease is the pain relief, rather than the meniscal function recovery. It may lead to a poor prognosis and accelerate the progression of osteoarthritis. In this study, we designed a meniscal scaffold to achieve the purposes of meniscal function recovery and cartilage protection.
Methods
The meniscal scaffold was designed using the triply periodic minimal surface (TPMS) method. The scaffold was simulated as a three-dimensional (3D) intact knee model using a finite element analysis software to obtain the results of different mechanical tests. The mechanical properties were gained through the universal machine. Finally, an in vivo model was established to evaluate the effects of the TPMS-based meniscal scaffold on the cartilage protection. The radiography and histological examinations were performed to assess the cartilage and bony structures. Different regions of the regenerated meniscus were tested using the universal machine to assess the biomechanical functions.
Results
The TPMS-based meniscal scaffold with a larger volume fraction and a longer functional periodicity demonstrated a better mechanical performance, and the load transmission and stress distribution were closer to the native biomechanical environment. The radiographic images and histological results of the TPMS group exhibited a better performance in terms of cartilage protection than the grid group. The regenerated meniscus in the TPMS group also had similar mechanical properties to the native meniscus.
Conclusion
The TPMS method can affect the mechanical properties by adjusting the volume fraction and functional periodicity. The TPMS-based meniscal scaffold showed appropriate features for meniscal regeneration and cartilage protection.
Collapse
|
25
|
Marsh S, Constantin-Teodosiu T, Chapman V, Sottile V. In vitro Exposure to Inflammatory Mediators Affects the Differentiation of Mesenchymal Progenitors. Front Bioeng Biotechnol 2022; 10:908507. [PMID: 35813997 PMCID: PMC9257013 DOI: 10.3389/fbioe.2022.908507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/23/2022] [Indexed: 11/27/2022] Open
Abstract
The increasing prevalence of joint disease, and in particular osteoarthritis (OA), calls for novel treatment strategies to prevent disease progression in addition to existing approaches focusing mainly on the relief of pain symptoms. The inherent properties of mesenchymal stem cells (MSCs) make them an attractive candidate for novel tissue repair strategies, as these progenitors have the potential to differentiate into chondrocytes needed to replace degraded cartilage and can exert a modulating effect on the inflammatory environment of the diseased joint. However, the inflammatory environment of the joint may affect the ability of these cells to functionally integrate into the host tissue and exert beneficial effects, as hinted by a lack of success seen in clinical trials. Identification of factors and cell signalling pathways that influence MSC function is therefore critical for ensuring their success in the clinic, and here the effects of inflammatory mediators on bone marrow-derived MSCs were evaluated. Human MSCs were cultured in the presence of inflammatory mediators typically associated with OA pathology (IL-1β, IL-8, IL-10). While exposure to these factors did not produce marked effects on MSC proliferation, changes were observed when the mediators were added under differentiating conditions. Results collected over 21 days showed that exposure to IL-1β significantly affected the differentiation response of these cells exposed to chondrogenic and osteogenic conditions, with gene expression analysis indicating changes in MAPK, Wnt and TLR signalling pathways, alongside an increased expression of pro-inflammatory cytokines and cartilage degrading enzymes. These results highlight the value of MSCs as a preclinical model to study OA and provide a basis to define the impact of factors driving OA pathology on the therapeutic potential of MSCs for novel OA treatments.
Collapse
Affiliation(s)
- S. Marsh
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom
| | - T. Constantin-Teodosiu
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - V. Chapman
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - V. Sottile
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: V. Sottile,
| |
Collapse
|
26
|
Housmans BAC, Neefjes M, Surtel DAM, Vitík M, Cremers A, van Rhijn LW, van der Kraan PM, van den Akker GGH, Welting TJM. Synovial fluid from end-stage osteoarthritis induces proliferation and fibrosis of articular chondrocytes via MAPK and RhoGTPase signaling. Osteoarthritis Cartilage 2022; 30:862-874. [PMID: 35176481 DOI: 10.1016/j.joca.2021.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Alterations in the composition of synovial fluid have been associated with adverse effects on cartilage integrity and function. Here, we examined the phenotypic and proliferative behavior of human articular chondrocytes when cultured in vitro for 13 days with synovial fluid derived from end-stage osteoarthritis patients. MATERIALS AND METHODS Chondrocyte proliferation and phenotypical changes induced by osteoarthritic synovial fluid were analyzed using DNA staining, RT-qPCR, immunostainings, and immunoblotting. The molecular mechanisms by which osteoarthritic synovial fluid induced fibrosis and proliferation were studied using a phospho-protein antibody array and luciferase-based transcription factor activity assays. Specific pathway inhibitors were used to probe the involvement of pathways in fibrosis and proliferation. RESULTS Prolonged stimulation with osteoarthritic synovial fluid sustained chondrocyte proliferation and induced profound phenotypic changes, favoring a fibrotic over a chondrogenic or hypertrophic phenotype. A clear loss of chondrogenic markers at both the transcriptional and protein level was observed, while expression of several fibrosis-associated markers were upregulated over time. Phospho-kinase analysis revealed activation of MAPK and RhoGTPase signaling pathways by osteoarthritic synovial fluid, which was confirmed by elevated transcriptional activity of Elk-1 and SRF. Inhibitor studies revealed that ERK played a central role in the loss of chondrocyte phenotype, while EGFR and downstream mediators p38, JNK and Rac/Cdc42 were essential for fibrosis-associated collagen expression. Finally, we identified EGF signaling as a key activator of chondrocyte proliferation. CONCLUSIONS Osteoarthritic synovial fluid promoted chondrocyte fibrosis and proliferation through EGF receptor activation and downstream MAPK and RhoGTPase signaling.
Collapse
Affiliation(s)
- B A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M Neefjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - D A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M Vitík
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - A Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - L W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - P M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - G G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - T J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands; Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
27
|
Thielen NGM, Neefjes M, Vitters EL, van Beuningen HM, Blom AB, Koenders MI, van Lent PLEM, van de Loo FAJ, Blaney Davidson EN, van Caam APM, van der Kraan PM. Identification of Transcription Factors Responsible for a Transforming Growth Factor-β-Driven Hypertrophy-like Phenotype in Human Osteoarthritic Chondrocytes. Cells 2022; 11:cells11071232. [PMID: 35406794 PMCID: PMC8998018 DOI: 10.3390/cells11071232] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 12/18/2022] Open
Abstract
During osteoarthritis (OA), hypertrophy-like chondrocytes contribute to the disease process. TGF-β's signaling pathways can contribute to a hypertrophy(-like) phenotype in chondrocytes, especially at high doses of TGF-β. In this study, we examine which transcription factors (TFs) are activated and involved in TGF-β-dependent induction of a hypertrophy-like phenotype in human OA chondrocytes. We found that TGF-β, at levels found in synovial fluid in OA patients, induces hypertrophic differentiation, as characterized by increased expression of RUNX2, COL10A1, COL1A1, VEGFA and IHH. Using luciferase-based TF activity assays, we observed that the expression of these hypertrophy genes positively correlated to SMAD3:4, STAT3 and AP1 activity. Blocking these TFs using specific inhibitors for ALK-5-induced SMAD signaling (5 µM SB-505124), JAK-STAT signaling (1 µM Tofacitinib) and JNK signaling (10 µM SP-600125) led to the striking observation that only SB-505124 repressed the expression of hypertrophy factors in TGF-β-stimulated chondrocytes. Therefore, we conclude that ALK5 kinase activity is essential for TGF-β-induced expression of crucial hypertrophy factors in chondrocytes.
Collapse
|
28
|
Modulation of miR-204 Expression during Chondrogenesis. Int J Mol Sci 2022; 23:ijms23042130. [PMID: 35216245 PMCID: PMC8874780 DOI: 10.3390/ijms23042130] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/03/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
RUNX2 and SOX9 are two pivotal transcriptional regulators of chondrogenesis. It has been demonstrated that RUNX2 and SOX9 physically interact; RUNX2 transactivation may be inhibited by SOX9. In addition, RUNX2 exerts reciprocal inhibition on SOX9 transactivity. Epigenetic control of gene expression plays a major role in the alternative differentiation fates of stem cells; in particular, it has been reported that SOX9 can promote the expression of miRNA (miR)-204. Our aim was therefore to investigate the miR-204-5p role during chondrogenesis and to identify the relationship between this miR and the transcription factors plus downstream genes involved in chondrogenic commitment and differentiation. To evaluate the role of miR-204 in chondrogenesis, we performed in vitro transfection experiments by using Mesenchymal Stem Cells (MSCs). We also evaluated miR-204-5p expression in zebrafish models (adults and larvae). By silencing miR-204 during the early differentiation phase, we observed the upregulation of SOX9 and chondrogenic related genes compared to controls. In addition, we observed the upregulation of COL1A1 (a RUNX2 downstream gene), whereas RUNX2 expression of RUNX2 was slightly affected compared to controls. However, RUNX2 protein levels increased in miR-204-silenced cells. The positive effects of miR204 silencing on osteogenic differentiation were also observed in the intermediate phase of osteogenic differentiation. On the contrary, chondrocytes’ maturation was considerably affected by miR-204 downregulation. In conclusion, our results suggest that miR-204 negatively regulates the osteochondrogenic commitment of MSCs, while it positively regulates chondrocytes’ maturation.
Collapse
|
29
|
Liu X, Xiang D, Jin W, Zhao G, Li H, Xie B, Gu X. Timosaponin B-II alleviates osteoarthritis-related inflammation and extracellular matrix degradation through inhibition of mitogen-activated protein kinases and nuclear factor-κB pathways in vitro. Bioengineered 2022; 13:3450-3461. [PMID: 35094658 PMCID: PMC8973927 DOI: 10.1080/21655979.2021.2024685] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Osteoarthritis (OA), an inflammatory response in chondrocytes, leads to extracellular matrix (ECM) degradation and cartilage destruction. Timosaponin B-II (TB-II) is the main bioactive component of Rhizoma Anemarrhenae with reported antioxidant and anti-inflammatory effects. This study investigated the anti-OA function and mechanism of TB-II on IL-1β-stimulated SW1353 cells and primary rat chondrocytes. We firstly screened the concentration of TB-II in SW1353 cells and primary rat chondrocytes using CCK-8 assay. Thereafter, SW1353 cells and chondrocytes were, respectively, pretreated with TB-II (20 and 40 μg/mL) and TB-II (10 and 30 μg/mL) for 24 h and then stimulated with interleukin 1β (IL-1β, 10 ng/mL) for another 24 hours. Results showed that TB-II suppressed the production of reactive oxygen species, the protein levels of inducible nitric oxide synthase and cyclooxygenase-2 in IL-1β-stimulated SW1353 cells and chondrocytes. IL-1β-induced high secretion levels of nitric oxide and prostaglandin 2, TNF-α, IL-6 and MCP-1 were down-regulated by TB-II treatment, indicating an anti-inflammatory effect of TB-II on OA in vitro condition. Moreover, TB-II weakened the mRNA and protein expression of (matrix metalloproteinase) MMPs including MMP-1, MMP-3, and MMP-13, indicating the protection of TB-II against ECM degradation. Mechanically, TB-II suppressed MAPKs and NF-κB pathways under IL-1β stimulation evidenced by the down-regulated protein expression of p-ERK, p-p38, p-JNK, p-p65 and the reduced translocation of p65 subunit to the nucleus. The present study demonstrated that TB-II might become a novel therapeutic agent for OA treatment through repressing IL-1β-stimulated inflammation, oxidative stress and ECM degradation via suppressing the MAPKs and NF-κB pathways.
Collapse
Affiliation(s)
- Xinwei Liu
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang People’s Republic of China
| | - Dulei Xiang
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang People’s Republic of China
- Graduate School, Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Wenming Jin
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang People’s Republic of China
- Graduate School, China Medical University, Shenyang, People’s Republic of China
| | - Gen Zhao
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang People’s Republic of China
- Graduate School, Dalian Medical University, Dalian, People’s Republic of China
| | - Han Li
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang People’s Republic of China
- Graduate School, Jinzhou Medical University, Jinzhou, People’s Republic of China
| | - Bing Xie
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang People’s Republic of China
| | - Xiaochuan Gu
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
30
|
Martin SD, Kucharik MP, Abraham PF, Nazal MR, Meek WM, Varady NH. Functional Outcomes of Arthroscopic Acetabular Labral Repair with and without Bone Marrow Aspirate Concentrate. J Bone Joint Surg Am 2022; 104:4-14. [PMID: 34648479 DOI: 10.2106/jbjs.20.01740] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Osteoarthritis (OA) of the hip is a debilitating condition associated with inferior outcomes in patients undergoing hip arthroscopy. To provide symptom relief and improve outcomes in these patients, bone marrow aspirate concentrate (BMAC) has been applied as an adjuvant therapy with the hope of halting progression of cartilage damage. The current study examined the clinical efficacy of BMAC application in patients undergoing arthroscopic acetabular labral repair by comparing patient-reported outcome measures (PROMs) between groups with and without BMAC application. METHODS Patients who received BMAC during arthroscopic acetabular labral repair from December 2016 to June 2019 were compared with a control cohort that underwent the same procedure but did not receive BMAC from November 2013 to November 2016. Patients in both cohorts were asked to prospectively complete PROMs prior to surgery and at 3, 6, 12, and 24-month follow-up intervals; those who completed the PROMs at enrollment and the 12-month follow-up were included in the study. An a priori subgroup analysis was performed among patients with moderate cartilage damage (Outerbridge grade 2 or 3). The analyses were adjusted for any differences in baseline factors between groups. RESULTS Sixty-two patients with BMAC application were compared with 62 control patients without BMAC application. When compared with the no-BMAC cohort, the BMAC cohort did not report significantly different mean International Hip Outcome Tool-33 (iHOT-33) scores at any postoperative time point. However, when patients with moderate cartilage damage were compared across groups, the BMAC cohort reported significantly greater mean (95% confidence interval) scores than the no-BMAC cohort at the 12-month (78.6 [72.4 to 84.8] versus 69.2 [63.3 to 75.2]; p = 0.035) and 24-month (82.5 [73.4 to 91.6] versus 69.5 [62.1 to 76.8]; p = 0.030) follow-up. Similarly, these patients reported greater score improvements at 12 months (37.3 [30.3 to 44.3] versus 25.4 [18.7 to 32.0]; p = 0.017) and 24 months (39.6 [30.4 to 48.7] versus 26.4 [19.1 to 33.8]; p = 0.029). CONCLUSIONS Patients with moderate cartilage injury undergoing arthroscopic acetabular labral repair with BMAC application reported significantly greater functional improvements when compared with similar patients without BMAC application. LEVEL OF EVIDENCE Therapeutic Level III. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Scott D Martin
- Sports Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Mass General Brigham Integrated Health Care System, Boston, Massachusetts
| | - Michael P Kucharik
- Sports Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Mass General Brigham Integrated Health Care System, Boston, Massachusetts
| | - Paul F Abraham
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, California
| | - Mark R Nazal
- Department of Orthopedic Surgery, University of Kentucky, Lexington, Kentucky
| | - Wendy M Meek
- Sports Medicine, Department of Orthopedic Surgery, Massachusetts General Hospital, Mass General Brigham Integrated Health Care System, Boston, Massachusetts
| | | |
Collapse
|
31
|
Chilbule SK, Rajagopal K, Walter N, Dutt V, Madhuri V. Role of WNT Agonists, BMP and VEGF Antagonists in Rescuing Osteoarthritic Knee Cartilage in a Rat Model. Indian J Orthop 2022; 56:24-33. [PMID: 35070139 PMCID: PMC8748585 DOI: 10.1007/s43465-021-00434-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/27/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The superficial zone of articular cartilage (AC) is vital for its function and biomechanics. The damaged AC gets vascularized and undergoes hypertrophy and ossification. Studies have highlighted these two as the major causative factors in osteoarthritis (OA). We aimed at preventing the OA progression in a rat knee instability model by inhibiting the vascular ingrowth and ossification using VEGF and BMP antagonist. A WNT agonist was also used to promote AC regeneration because of its protective effect on the superficial layer. METHODS Rat knee OA was created by surgical excision of the medial meniscus and medial collateral ligament. Forty rats were divided into two groups of twenty each for surgical control and tests (surgery + intra-articular injection of drugs every two weeks). Ten animals from each group were sacrificed at four and eight weeks. Histology was mainly used to evaluate the outcome. RESULTS A surgical OA model was successfully created with higher histological scores for operated knees, both in short- (P = 0.0001) and long-term (P = 0.001). Modified Mankin score was lesser in the test animals as compared to control (P = 0.17) in the short-term, but the trend was reversed in the long-term (P = 0.13). Subgroup analysis revealed that repeated injections in the anterolateral compartment contributed to higher scores in the lateral (P = 0.03) and anterior (P = 0.03) compartment of the knee in the long-term. CONCLUSION The combinatorial approach was effective in controlling the OA in short-term. Further studies are needed to test the sustained drug delivery system to improve the outcome.
Collapse
Affiliation(s)
- Sanjay K. Chilbule
- Department of Paediatric Orthopaedics, Christian Medical College, Vellore, 632004 India
| | - Karthikeyan Rajagopal
- Department of Paediatric Orthopaedics, Christian Medical College, Vellore, 632004 India
- Centre for Stem Cell Research, Christian Medical College, Vellore, 632002 India
| | - Noel Walter
- Department of Forensic Medicine, Christian Medical College, Vellore, 632004 India
| | - Vivek Dutt
- Department of Paediatric Orthopaedics, Christian Medical College, Vellore, 632004 India
| | - Vrisha Madhuri
- Department of Paediatric Orthopaedics, Christian Medical College, Vellore, 632004 India
- Centre for Stem Cell Research, Christian Medical College, Vellore, 632002 India
| |
Collapse
|
32
|
Bakhtiary N, Liu C, Ghorbani F. Bioactive Inks Development for Osteochondral Tissue Engineering: A Mini-Review. Gels 2021; 7:274. [PMID: 34940334 PMCID: PMC8700778 DOI: 10.3390/gels7040274] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 01/02/2023] Open
Abstract
Nowadays, a prevalent joint disease affecting both cartilage and subchondral bone is osteoarthritis. Osteochondral tissue, a complex tissue unit, exhibited limited self-renewal potential. Furthermore, its gradient properties, including mechanical property, bio-compositions, and cellular behaviors, present a challenge in repairing and regenerating damaged osteochondral tissues. Here, tissue engineering and translational medicine development using bioprinting technology provided a promising strategy for osteochondral tissue repair. In this regard, personalized stratified scaffolds, which play an influential role in osteochondral regeneration, can provide potential treatment options in early-stage osteoarthritis to delay or avoid the use of joint replacements. Accordingly, bioactive scaffolds with possible integration with surrounding tissue and controlling inflammatory responses have promising future tissue engineering perspectives. This minireview focuses on introducing biologically active inks for bioprinting the hierarchical scaffolds, containing growth factors and bioactive materials for 3D printing of regenerative osteochondral substitutes.
Collapse
Affiliation(s)
- Negar Bakhtiary
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran 14115-114, Iran;
| | - Chaozong Liu
- Institute of Orthopaedic & Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK;
| | - Farnaz Ghorbani
- Institute of Biomaterials, Department of Material Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany
| |
Collapse
|
33
|
Zhou C, Cui Y, Yang Y, Guo D, Zhang D, Fan Y, Li X, Zou J, Xie J. Runx1 protects against the pathological progression of osteoarthritis. Bone Res 2021; 9:50. [PMID: 34876557 PMCID: PMC8651727 DOI: 10.1038/s41413-021-00173-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/02/2021] [Accepted: 09/12/2021] [Indexed: 02/05/2023] Open
Abstract
Runt-related transcription factor-1 (Runx1) is required for chondrocyte-to-osteoblast lineage commitment by enhancing both chondrogenesis and osteogenesis during vertebrate development. However, the potential role of Runx1 in joint diseases is not well known. In the current study, we aimed to explore the role of Runx1 in osteoarthritis induced by anterior cruciate ligament transaction (ACLT) surgery. We showed that chondrocyte-specific Runx1 knockout (Runx1f/fCol2a1-Cre) aggravated cartilage destruction by accelerating the loss of proteoglycan and collagen II in early osteoarthritis. Moreover, we observed thinning and ossification of the growth plate, a decrease in chondrocyte proliferative capacity and the loss of bone matrix around the growth plate in late osteoarthritis. We overexpressed Runx1 by adeno-associated virus (AAV) in articular cartilage and identified its protective effect by slowing the destruction of osteoarthritis in cartilage in early osteoarthritis and alleviating the pathological progression of growth plate cartilage in late osteoarthritis. ChIP-seq analysis identified new targets that interacted with Runx1 in cartilage pathology, and we confirmed the direct interactions of these factors with Runx1 by ChIP-qPCR. This study helps us to understand the function of Runx1 in osteoarthritis and provides new clues for targeted osteoarthritis therapy.
Collapse
Affiliation(s)
- Chenchen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yueyi Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Daimo Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaobing Li
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Zou
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
34
|
Trompeter N, Gardinier JD, DeBarros V, Boggs M, Gangadharan V, Cain WJ, Hurd L, Duncan RL. Insulin-like growth factor-1 regulates the mechanosensitivity of chondrocytes by modulating TRPV4. Cell Calcium 2021; 99:102467. [PMID: 34530313 PMCID: PMC8541913 DOI: 10.1016/j.ceca.2021.102467] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Both mechanical and biochemical stimulation are required for maintaining the integrity of articular cartilage. However, chondrocytes respond differently to mechanical stimuli in osteoarthritic cartilage when biochemical signaling pathways, such as Insulin-like Growth Factor-1 (IGF-1), are altered. The Transient Receptor Potential Vanilloid 4 (TRPV4) channel is central to chondrocyte mechanotransduction and regulation of cartilage homeostasis. Here, we propose that changes in IGF-1 can modulate TRPV4 channel activity. We demonstrate that physiologic levels of IGF-1 suppress hypotonic-induced TRPV4 currents and intracellular calcium flux by increasing apparent cell stiffness that correlates with actin stress fiber formation. Disruption of F-actin following IGF-1 treatment results in the return of the intracellular calcium response to hypotonic swelling. Using point mutations of the TRPV4 channel at the microtubule-associated protein 7 (MAP-7) site shows that regulation of TRPV4 by actin is mediated via the interaction of actin with the MAP-7 domain of TRPV4. We further highlight that ATP release, a down-stream response to mechanical stimulation in chondrocytes, is mediated by TRPV4 during hypotonic challenge. This response is significantly abrogated with IGF-1 treatment. As chondrocyte mechanosensitivity is greatly altered during osteoarthritis progression, IGF-1 presents as a promising candidate for prevention and treatment of articular cartilage damage.
Collapse
Affiliation(s)
- Nicholas Trompeter
- Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Joseph D Gardinier
- Biomechanics and Movement Science Program, University of Delaware, Newark, DE, United States; Bone and Joint Center, Henry Ford Hospital, Detroit, MI, United States
| | - Victor DeBarros
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Mary Boggs
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Vimal Gangadharan
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - William J Cain
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Lauren Hurd
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Randall L Duncan
- Biomedical Engineering, University of Delaware, Newark, DE, United States; Biomechanics and Movement Science Program, University of Delaware, Newark, DE, United States; Department of Biological Sciences, University of Delaware, Newark, DE, United States; Department of Biology, University of Michigan-Flint, Flint, MI, United States.
| |
Collapse
|
35
|
Liu Z, Hou Y, Zhang P, Lu H, Wang W, Ma W. Changes of the condylar cartilage and subchondral bone in the temporomandibular joints of rats under unilateral mastication and expression of Insulin-like Growth Factor-1. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2021; 123:405-416. [PMID: 34601167 DOI: 10.1016/j.jormas.2021.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVES This study aimed to define changes in the rat condylar cartilage and subchondral bone using the unilateral mastication model. MATERIALS AND METHODS In this study, forty 4-week-old Wistar rats were randomly divided into experimental (n = 20) and control group (n = 20). In the experimental group, unilateral dental splints were placed on the occlusal surface of left maxillary molars. The rats were sacrificed at 1, 2, 3, and 4 weeks after placement of the splint. Micro-CT scanning and histological staining were performed to observe the changes in the mandibular condylar cartilage and subchondral bone. Levels of insulin-like growth factor-1 (IGF-1) were determined via immunohistochemistry to analyse the occurrence of osteogenic changes. RESULTS Micro-CT scanning findings demonstrated the occurrence of asymmetric growth of condyle in the experimental group. The condylar cartilage and subchondral bone exhibited degradation on the chewing side of the experimental group and showed decreased bone mineral density, thinner cartilage thickness, and increased degree of degeneration and osteoclast activity. Compared with the control group, the expression of IGF-1 was remarkably higher on the non-chewing side. CONCLUSION Long-term unilateral mastication can lead to the occurrence of degenerative changes in the condylar cartilage and subchondral bone during growth and development. IGF-1 may play a role in promoting the process of osteogenesis.
Collapse
Affiliation(s)
- Ziyang Liu
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Yali Hou
- Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Department of Oral Pathology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Pengfei Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Haiyan Lu
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Wen Wang
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Wensheng Ma
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
36
|
Park S, Bello A, Arai Y, Ahn J, Kim D, Cha KY, Baek I, Park H, Lee SH. Functional Duality of Chondrocyte Hypertrophy and Biomedical Application Trends in Osteoarthritis. Pharmaceutics 2021; 13:pharmaceutics13081139. [PMID: 34452101 PMCID: PMC8400409 DOI: 10.3390/pharmaceutics13081139] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
Chondrocyte hypertrophy is one of the key indicators in the progression of osteoarthritis (OA). However, compared with other OA indications, such as cartilage collapse, sclerosis, inflammation, and protease activation, the mechanisms by which chondrocyte hypertrophy contributes to OA remain elusive. As the pathological processes in the OA cartilage microenvironment, such as the alterations in the extracellular matrix, are initiated and dictated by the physiological state of the chondrocytes, in-depth knowledge of chondrocyte hypertrophy is necessary to enhance our understanding of the disease pathology and develop therapeutic agents. Chondrocyte hypertrophy is a factor that induces OA progression; it is also a crucial factor in the endochondral ossification. This review elaborates on this dual functionality of chondrocyte hypertrophy in OA progression and endochondral ossification through a description of the characteristics of various genes and signaling, their mechanism, and their distinguishable physiological effects. Chondrocyte hypertrophy in OA progression leads to a decrease in chondrogenic genes and destruction of cartilage tissue. However, in endochondral ossification, it represents an intermediate stage at the process of differentiation of chondrocytes into osteogenic cells. In addition, this review describes the current therapeutic strategies and their mechanisms, involving genes, proteins, cytokines, small molecules, three-dimensional environments, or exosomes, against the OA induced by chondrocyte hypertrophy. Finally, this review proposes that the contrasting roles of chondrocyte hypertrophy are essential for both OA progression and endochondral ossification, and that this cellular process may be targeted to develop OA therapeutics.
Collapse
Affiliation(s)
- Sunghyun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea
| | - Alvin Bello
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- School of Integrative Engineering, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Yoshie Arai
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Jinsung Ahn
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Dohyun Kim
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Kyung-Yup Cha
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Inho Baek
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Hansoo Park
- School of Integrative Engineering, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- Correspondence: ; Tel.: +82-31-961-5153; Fax: +82-31-961-5108
| |
Collapse
|
37
|
Baddam P, Young D, Dunsmore G, Nie C, Eaton F, Elahi S, Jovel J, Adesida AB, Dufour A, Graf D. Nasal Septum Deviation as the Consequence of BMP-Controlled Changes to Cartilage Properties. Front Cell Dev Biol 2021; 9:696545. [PMID: 34249945 PMCID: PMC8265824 DOI: 10.3389/fcell.2021.696545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/24/2021] [Indexed: 11/29/2022] Open
Abstract
The nasal septum cartilage is a specialized hyaline cartilage important for normal midfacial growth. Abnormal midfacial growth is associated with midfacial hypoplasia and nasal septum deviation (NSD). However, the underlying genetics and associated functional consequences of these two anomalies are poorly understood. We have previously shown that loss of Bone Morphogenetic Protein 7 (BMP7) from neural crest (BMP7 ncko ) leads to midfacial hypoplasia and subsequent septum deviation. In this study we elucidate the cellular and molecular abnormalities underlying NSD using comparative gene expression, quantitative proteomics, and immunofluorescence analysis. We show that reduced cartilage growth and septum deviation are associated with acquisition of elastic cartilage markers and share similarities with osteoarthritis (OA) of the knee. The genetic reduction of BMP2 in BMP7 ncko mice was sufficient to rescue NSD and suppress elastic cartilage markers. To our knowledge this investigation provides the first genetic example of an in vivo cartilage fate switch showing that this is controlled by the relative balance of BMP2 and BMP7. Cellular and molecular changes similar between NSD and knee OA suggest a related etiology underlying these cartilage abnormalities.
Collapse
Affiliation(s)
- Pranidhi Baddam
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Daniel Young
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Garett Dunsmore
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Chunpeng Nie
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Farah Eaton
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Juan Jovel
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | | | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Daniel Graf
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
38
|
Bartolotti I, Roseti L, Petretta M, Grigolo B, Desando G. A Roadmap of In Vitro Models in Osteoarthritis: A Focus on Their Biological Relevance in Regenerative Medicine. J Clin Med 2021; 10:1920. [PMID: 33925222 PMCID: PMC8124812 DOI: 10.3390/jcm10091920] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a multifaceted musculoskeletal disorder, with a high prevalence worldwide. Articular cartilage and synovial membrane are among the main biological targets in the OA microenvironment. Gaining more knowledge on the accuracy of preclinical in vitro OA models could open innovative avenues in regenerative medicine to bridge major gaps, especially in translation from animals to humans. Our methodological approach entailed searches on Scopus, the Web of Science Core Collection, and EMBASE databases to select the most relevant preclinical in vitro models for studying OA. Predicting the biological response of regenerative strategies requires developing relevant preclinical models able to mimic the OA milieu influencing tissue responses and organ complexity. In this light, standard 2D culture models lack critical properties beyond cell biology, while animal models suffer from several limitations due to species differences. In the literature, most of the in vitro models only recapitulate a tissue compartment, by providing fragmented results. Biotechnological advances may enable scientists to generate new in vitro models that combine easy manipulation and organ complexity. Here, we review the state-of-the-art of preclinical in vitro models in OA and outline how the different preclinical systems (inflammatory/biomechanical/microfluidic models) may be valid tools in regenerative medicine, describing their pros and cons. We then discuss the prospects of specific and combinatorial models to predict biological responses following regenerative approaches focusing on mesenchymal stromal cells (MSCs)-based therapies to reduce animal testing.
Collapse
Affiliation(s)
- Isabella Bartolotti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Livia Roseti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Mauro Petretta
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
- RegenHu Company, Z.I Du Vivier 22, 1690 Villaz-St-Pierre, Switzerland
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| |
Collapse
|
39
|
Sousa EBD, Moura Neto V, Aguiar DP. BMP-4, TGF-β e Smad3 como moduladores da viabilidade das células do líquido sinovial. Rev Bras Ortop 2021; 57:314-320. [PMID: 35652012 PMCID: PMC9142237 DOI: 10.1055/s-0041-1724076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/16/2020] [Indexed: 11/24/2022] Open
Abstract
Objective
Our goal was to evaluate the modulation of the synovial fluid cells (SFC) from patients with and without osteoarthritis (OA) by bone morphogenetic protein 4 (BMP-4), Smad-3 and
transforming growth factor beta
(TGF-β).
Methods
Synovial fluid was collected from patients submitted to knee arthroscopy or replacement and were centrifuged to isolate cells from the fluid. Cells were cultured for 21 days and characterized as mesenchymal stem cells (MSCs) according to the criteria of the International Society of Cell Therapy. Then, we performed an [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay (MTT) assay after exposing cells with and without OA to TGF-β, Smad3 and BMP-4 pathway inhibitors and to different concentrations of BMP4.
Results
Exposure to the TGF-β, Smad3 and BMP-4 inhibitors modifies the mitochondrial activity of the SFCs. The activity of the SFCs is modified by influences of increasing concentrations of BMP4, but there is no difference in cellular activity between patients with and without OA.
Conclusion
TGF-β, Smad3 and BMP-4 modulate the activity of SFCs from patients with and without knee OA.
Collapse
Affiliation(s)
- Eduardo Branco de Sousa
- Divisão de Ensino e Pesquisa, Instituto Nacional de Ortopedia e Traumatologia Jamil Haddad, Rio de Janeiro, RJ, Brasil
| | - Vivaldo Moura Neto
- Laboratório de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Rio de Janeiro, RJ, Brasil
| | - Diego Pinheiro Aguiar
- Laboratório de Biomodelos e Protótipos, Universidade Estadual da Zona Oeste, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
40
|
Zheng L, Zhang Z, Sheng P, Mobasheri A. The role of metabolism in chondrocyte dysfunction and the progression of osteoarthritis. Ageing Res Rev 2021; 66:101249. [PMID: 33383189 DOI: 10.1016/j.arr.2020.101249] [Citation(s) in RCA: 309] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by low-grade inflammation and high levels of clinical heterogeneity. Aberrant chondrocyte metabolism is a response to changes in the inflammatory microenvironment and may play a key role in cartilage degeneration and OA progression. Under conditions of environmental stress, chondrocytes tend to adapt their metabolism to microenvironmental changes by shifting from one metabolic pathway to another, for example from oxidative phosphorylation to glycolysis. Similar changes occur in other joint cells, including synoviocytes. Switching between these pathways is implicated in metabolic alterations that involve mitochondrial dysfunction, enhanced anaerobic glycolysis, and altered lipid and amino acid metabolism. The shift between oxidative phosphorylation and glycolysis is mainly regulated by the AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) pathways. Chondrocyte metabolic changes are likely to be a feature of different OA phenotypes. Determining the role of chondrocyte metabolism in OA has revealed key features of disease pathogenesis. Future research should place greater emphasis on immunometabolism and altered metabolic pathways as a means to understand the pathophysiology of age-related OA. This knowledge will advance the development of new drugs against therapeutic targets of metabolic significance.
Collapse
Affiliation(s)
- Linli Zheng
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080 China
| | - Ziji Zhang
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080 China
| | - Puyi Sheng
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080 China.
| | - Ali Mobasheri
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080 China; Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, PO Box 5000, FI-90014 Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania; Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508 GA, Utrecht, The Netherlands.
| |
Collapse
|
41
|
McDonough RC, Gilbert RM, Gleghorn JP, Price C. Targeted Gq-GPCR activation drives ER-dependent calcium oscillations in chondrocytes. Cell Calcium 2021; 94:102363. [PMID: 33550208 DOI: 10.1016/j.ceca.2021.102363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 11/28/2022]
Abstract
The temporal dynamics of calcium signaling are critical regulators of chondrocyte homeostasis and chondrogenesis. Calcium oscillations regulate differentiation and anabolic processes in chondrocytes and their precursors. Attempts to control chondrocyte calcium signaling have been achieved through mechanical perturbations and synthetic ion channel modulators. However, such stimuli can lack both local and global specificity and precision when evoking calcium signals. Synthetic signaling platforms can more precisely and selectively activate calcium signaling, enabling improved dissection of the roles of intracellular calcium ([Ca2+]i) in chondrocyte behavior. One such platform is hM3Dq, a chemogenetic DREADD (Designer Receptors Exclusively Activated by Designer Drugs) that activates calcium signaling via the Gαq-PLCβ-IP3-ER pathway upon administration of clozapine N-oxide (CNO). We previously described the first-use of hM3Dq to precisely mediate targeted, synthetic calcium signals in chondrocyte-like ATDC5 cells. Here, we generated stably expressing hM3Dq-ATDC5 cells to investigate the dynamics of Gαq-GPCR calcium signaling in depth. CNO drove robust calcium responses in a temperature- and concentration-dependent (1 pM-100 μM) manner and elicited elevated levels of oscillatory calcium signaling above 10 nM. hM3Dq-mediated calcium oscillations in ATDC5 cells were reliant on ER calcium stores for both initiation and sustenance, and the downregulation and recovery dynamics of hM3Dq after CNO stimulation align with traditionally reported GPCR recycling kinetics. This study successfully generated a stable hM3Dq cell line to precisely drive Gαq-GPCR-mediated and ER-dependent oscillatory calcium signaling in ATDC5 cells and established a novel tool to elucidate the role that GPCR-mediated calcium signaling plays in chondrocyte biology, cartilage pathology, and cartilage tissue engineering.
Collapse
Affiliation(s)
- Ryan C McDonough
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Rachel M Gilbert
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Christopher Price
- Department of Biomedical Engineering, University of Delaware, United States.
| |
Collapse
|
42
|
Sun NN, He DM, Yang C, Zhou Q. Posttraumatic Osteoarthritis of Temporomandibular Joint in Miniature Pigs. J HARD TISSUE BIOL 2021. [DOI: 10.2485/jhtb.30.79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Ning-Ning Sun
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases
| | - Dong-Mei He
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People’s Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine
| | - Chi Yang
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People’s Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine
| | - Qing Zhou
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases
| |
Collapse
|
43
|
Masutani T, Yamada S, Hara A, Takahashi T, Green PG, Niwa M. Exogenous Application of Proteoglycan to the Cell Surface Microenvironment Facilitates to Chondrogenic Differentiation and Maintenance. Int J Mol Sci 2020; 21:ijms21207744. [PMID: 33086766 PMCID: PMC7589071 DOI: 10.3390/ijms21207744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/10/2020] [Accepted: 10/14/2020] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA), a disease that greatly impacts quality of life, has increasing worldwide prevalence as the population ages. However, its pathogenic mechanisms have not been fully elucidated and current therapeutic treatment strategies are inadequate. In recent years, abnormal endochondral ossification in articular cartilage has received attention as a pathophysiological mechanism in OA. Cartilage is composed of abundant extracellular matrix components, which are involved in tissue maintenance and regeneration, but how these factors affect endochondral ossification is not clear. Here, we show that the application of aggrecan-type proteoglycan from salmon nasal cartilage (sPG) exhibited marked proliferative capacity through receptor tyrosine kinases in chondroprogenitor cells, and also exhibited differentiation and three-dimensional structure formation via phosphorylation of Insulin-like Growth Factor-1 Receptor and Growth Differentiation Factor 5 expression. Furthermore, sPG inhibited calcification via expression of Runx2 and Col10 (factors related to induction of calcification), while increasing Mgp, a mineralization inhibitory factor. As a result of analyzing the localization of sPG applied to the cells, it was localized on the surface of the cell membrane. In this study, we found that sPG, as a biomaterial, could regulate cell proliferation, differentiation and calcification inhibition by acting on the cell surface microenvironment. Therefore, sPG may be the foundation for a novel therapeutic approach for cartilage maintenance and for improved symptoms in OA.
Collapse
Affiliation(s)
- Teruaki Masutani
- Medical Education Development Center, Gifu University School of Medicine, 1-1 Yanagido, Gifu City 501-1194, Japan;
- Research & Development Dept., Ichimaru Pharcos Co., Ltd., 318-1 Asagi, Motosu City 501-0475, Japan;
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan;
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu City 501-1194, Japan;
| | - Tatsuji Takahashi
- Research & Development Dept., Ichimaru Pharcos Co., Ltd., 318-1 Asagi, Motosu City 501-0475, Japan;
| | - Paul G Green
- Department of Oral and Maxillofacial Surgery, UCSF, San Francisco, CA 94143, USA;
| | - Masayuki Niwa
- Medical Education Development Center, Gifu University School of Medicine, 1-1 Yanagido, Gifu City 501-1194, Japan;
- Correspondence: ; Tel.: +81-58-230-6470
| |
Collapse
|
44
|
Neidlin M, Chantzi E, Macheras G, Gustafsson MG, Alexopoulos LG. A Novel Multiplex Based Platform for Osteoarthritis Drug Candidate Evaluation. Ann Biomed Eng 2020; 48:2438-2448. [PMID: 32472364 DOI: 10.1007/s10439-020-02539-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/26/2020] [Indexed: 01/10/2023]
Abstract
Osteoarthritis (OA) is characterized by irreversible cartilage degradation with very limited therapeutic interventions. Drug candidates targeted at prototypic players had limited success until now and systems based approaches might be necessary. Consequently, drug evaluation platforms should consider the biological complexity looking beyond well-known contributors of OA. In this study an ex vivo model of cartilage degradation, combined with measuring releases of 27 proteins, was utilized to study 9 drug candidates. After an initial single drug evaluation step the 3 most promising compounds were selected and employed in an exhaustive combinatorial experiment. The resulting most and least promising treatment candidates were selected and validated in an independent study. This included estimation of mechanical properties via finite element modelling (FEM) and quantification of cartilage degradation as glycosaminoglycan (GAG) release. The most promising candidate showed increase of Young's modulus, decrease of hydraulic permeability and decrease of GAG release. The least promising candidate exhibited the opposite behaviour. The study shows the potential of a novel drug evaluation platform in identifying treatments that might reduce cartilage degradation. It also demonstrates the promise of exhaustive combination experiments and a connection between chondrocyte responses at the molecular level with changes of biomechanical properties at the tissue level.
Collapse
Affiliation(s)
- Michael Neidlin
- Department of Mechanical Engineering, National Technical University of Athens, Heroon Polytechniou 9, 15780, Zografou, Greece
| | - Efthymia Chantzi
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | - Leonidas G Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, Heroon Polytechniou 9, 15780, Zografou, Greece.
| |
Collapse
|
45
|
Qu Z, Zhang F, Chen W, Lin T, Sun Y. High-dose TGF-β1 degrades human nucleus pulposus cells via ALK1-Smad1/5/8 activation. Exp Ther Med 2020; 20:3661-3668. [PMID: 32855718 PMCID: PMC7444386 DOI: 10.3892/etm.2020.9088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/01/2020] [Indexed: 01/07/2023] Open
Abstract
Transforming growth factor β1 (TGF-β1) can promote the proliferation and differentiation of intervertebral disc cells and participates in its repair process. However, whether TGF-β1 engages in the process of disc degeneration has not yet been fully elucidated. The present study aimed to investigate the function of high-dose TGF-β1 on the metabolism of nucleus pulposus cells (NPCs). TGF-β1 levels in human degenerative intervertebral disc tissues and tumor necrosis factor (TNF)-α-induced degenerative NPCs were analyzed. Furthermore, NPCs were treated with TGF-β1 and inhibitors of TGF-β1 receptors [ALK tyrosine kinase receptor (ALK) 1 and ALK5] to determine the effect of the receptors in the mediation of NPC degeneration. The NPC state was determined by the components of secretory collagen I/II, tissue inhibitor of metalloproteinase-3 (TIMP-3) and matrix metalloproteinase (MMP)-13. The mRNA expression of Smad1/2/3/5/8, the downstream gene of TGF-β1 mediated by ALK, was also measured. Results showed that TGF-β1 and ALK1 were positively associated with the degree of degeneration of NP or NPCs in vitro, but negatively associated with ALK5. Furthermore, high-doses of TGF-β1 suppressed collagen II, but enhanced collagen I, TIMP-3, MMP-13, ALK1/5 and Smad1/2/3/5/8 expression. ALK5 inhibition induced the suppression of Smad2/3 and aggravated high-dose TGF-β1-induced NPC degeneration, as shown by the reduction in collagen II and increase in collagen I, TIMP-3 and MMP-13. By contrast, ALK1 inhibition resulted in Smad1/5/8 suppression and alleviated high-dose TGF-β1-induced NPC degeneration. Taken together, it was concluded that high-doses of TGF-β1 contributed to the degeneration of NPCs via the upregulation of ALK1 and Smad1/5/8.
Collapse
Affiliation(s)
- Zhiqiang Qu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China.,Department of Orthopedics, Tongliao City Hospital, Tongliao, Inner Mongolia 028000, P.R. China
| | - Fengxiang Zhang
- Department of General Surgery, Tongliao City Hospital, Tongliao, Inner Mongolia 028000, P.R. China
| | - Weiwei Chen
- Department of Disinfecting Supply Division, Tongliao City Hospital, Tongliao, Inner Mongolia 028000, P.R. China
| | - Tao Lin
- Department of Disinfecting Supply Division, Tongliao City Hospital, Tongliao, Inner Mongolia 028000, P.R. China
| | - Yongming Sun
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
46
|
Bone marrow concentrate injections for the treatment of osteoarthritis: evidence from preclinical findings to the clinical application. INTERNATIONAL ORTHOPAEDICS 2020; 45:525-538. [PMID: 32661635 PMCID: PMC7843474 DOI: 10.1007/s00264-020-04703-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023]
Abstract
Purpose To investigate the available literature on the use of bone marrow aspirate concentrate (BMAC) and summarize the current evidence supporting its potential for the injective treatment of joints affected by osteoarthritis (OA). Methods A systematic literature search was conducted on three electronic databases (PubMed, Embase, and Cochrane Library) in April 2020, using the following string: “((bone marrow concentrate) OR (BMC) OR (bone marrow aspirate concentrate) OR (BMAC)) AND (osteoarthritis)”, and inclusion criteria: clinical and preclinical (animal) studies of any level of evidence, written in English language, and evaluating the intra-articular or subchondral use of BMAC for the injective treatment of OA joints. Results The publication trend remarkably increased over time. A total of 22 studies were included in the qualitative data synthesis: four preclinical studies and 18 clinical studies, for a total number of 4626 patients. Safety was documented by all studies, with a low number of adverse events. An overall improvement in pain and function was documented in most of the studies, but the clinical studies present significant heterogeneity, few patients, short-term follow-up, and overall poor methodology. Conclusion There is a growing interest in the field of BMAC injections for the treatment of OA, with promising results in preclinical and clinical studies in terms of safety and effectiveness. Nevertheless, the current knowledge is still preliminary. Preclinical research is still needed to optimize BMAC use, as well as high-level large controlled trials to better understand the real potential of BMAC injections for the treatment of patients affected by OA.
Collapse
|
47
|
Anti-Inflammatory and Chondroprotective Effects of Vanillic Acid and Epimedin C in Human Osteoarthritic Chondrocytes. Biomolecules 2020; 10:biom10060932. [PMID: 32575510 PMCID: PMC7356262 DOI: 10.3390/biom10060932] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 01/15/2023] Open
Abstract
In osteoarthritis (OA), inhibition of excessively expressed pro-inflammatory cytokines in the OA joint and increasing the anabolism for cartilage regeneration are necessary. In this ex-vivo study, we used an inflammatory model of human OA chondrocytes microtissues, consisting of treatment with cytokines (interleukin 1β (IL-1β)/tumor necrosis factor α (TNF-α)) with or without supplementation of six herbal compounds with previously identified chondroprotective effect. The compounds were assessed for their capacity to modulate the key catabolic and anabolic factors using several molecular analyses. We selectively investigated the mechanism of action of the two most potent compounds Vanillic acid (VA) and Epimedin C (Epi C). After identification of the anti-inflammatory and anabolic properties of VA and Epi C, the Ingenuity Pathway Analysis showed that in both treatment groups, osteoarthritic signaling pathways were inhibited. In the treatment group with VA, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling was inhibited by attenuation of the nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (IκBα) phosphorylation. Epi C showed a significant anabolic effect by increasing the expression of collagenous and non-collagenous matrix proteins. In conclusion, VA, through inhibition of phosphorylation in NF-κB signaling pathway and Epi C, by increasing the expression of extracellular matrix components, showed significant anti-inflammatory and anabolic properties and might be potentially used in combination to treat or prevent joint OA.
Collapse
|
48
|
Zhang S, Hamid MR, Wang T, Liao J, Wen L, Zhou Y, Wei P, Zou X, Chen G, Chen J, Zhou G. RSK-3 promotes cartilage regeneration via interacting with rpS6 in cartilage stem/progenitor cells. Theranostics 2020; 10:6915-6927. [PMID: 32550912 PMCID: PMC7295041 DOI: 10.7150/thno.44875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Cartilage stem/progenitor cells (CSPC) are a promising cellular source to promote endogenous cartilage regeneration in osteoarthritis (OA). Our previous work indicates that ribosomal s6 kinase 3 (RSK-3) is a target of 4-aminobiphenyl, a chemical enhancing CSPC-mediated cartilage repair in OA. However, the primary function and mechanism of RSK-3 in CSPC-mediated cartilage pathobiology remain undefined. Methods: We systematically assessed the association of RSK-3 with OA in three mouse strains with varying susceptibility to OA (MRL/MpJ>CBA>STR/Ort), and also RSK-3-/- mice. Bioinformatic analysis was used to identify the possible mechanism of RSK-3 affecting CSPC, which was further verified in OA mice and CSPC with varying RSK-3 expression induced by chemicals or gene modification. Results: We demonstrated that the level of RSK-3 in cartilage was positively correlated with cartilage repair capacities in three mouse strains (MRL/MpJ>CBA>STR/Ort). Enhanced RSK-3 expression by 4-aminobiphenyl markedly attenuated cartilage injury in OA mice and inhibition or deficiency of RSK-3 expression, on the other hand, significantly aggravated cartilage damage. Transcriptional profiling of CSPC from mice suggested the potential role of RSK-3 in modulating cell proliferation. It was further shown that the in vivo and in vitro manipulation of the RSK-3 expression indeed affected the CSPC proliferation. Mechanistically, ribosomal protein S6 (rpS6) was activated by RSK-3 to accelerate CSPC growth. Conclusion: RSK-3 is identified as a key regulator to enhance cartilage repair, at least partly by regulating the functionality of the cartilage-resident stem/progenitor cells.
Collapse
|
49
|
Bellini M, Pest MA, Miranda-Rodrigues M, Qin L, Jeong JW, Beier F. Overexpression of MIG-6 in the cartilage induces an osteoarthritis-like phenotype in mice. Arthritis Res Ther 2020; 22:119. [PMID: 32430054 PMCID: PMC7236969 DOI: 10.1186/s13075-020-02213-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common form of arthritis and characterized by degeneration of the articular cartilage. Mitogen-inducible gene 6 (Mig-6) has been identified as a negative regulator of the epidermal growth factor receptor (EGFR). Cartilage-specific Mig-6 knockout (KO) mice display increased EGFR signaling, an anabolic buildup of the articular cartilage, and formation of chondro-osseous nodules. Since our understanding of the EGFR/Mig-6 network in the cartilage remains incomplete, we characterized mice with cartilage-specific overexpression of Mig-6 in this study. METHODS Utilizing knee joints from cartilage-specific Mig-6-overexpressing (Mig-6over/over) mice (at multiple time points), we evaluated the articular cartilage using histology, immunohistochemical staining, and semi-quantitative histopathological scoring (OARSI) at multiple ages. MicroCT analysis was employed to examine skeletal morphometry, body composition, and bone mineral density. RESULTS Our data show that cartilage-specific Mig-6 overexpression did not cause any major developmental abnormalities in the articular cartilage, although Mig-6over/over mice have slightly shorter long bones compared to the control group. Moreover, there was no significant difference in bone mineral density and body composition in any of the groups. However, our results indicate that Mig-6over/over male mice show accelerated cartilage degeneration at 12 and 18 months of age. Immunohistochemistry for SOX9 demonstrated that the number of positively stained cells in Mig-6over/over mice was decreased relative to controls. Immunostaining for MMP13 appeared increased in areas of cartilage degeneration in Mig-6over/over mice. Moreover, staining for phospho-EGFR (Tyr-1173) and lubricin (PRG4) was decreased in the articular cartilage of Mig-6over/over mice. CONCLUSION Overexpression of Mig-6 in the articular cartilage causes no major developmental phenotype; however, these mice develop earlier OA during aging. These data demonstrate that Mig-6/EGFR pathways are critical for joint homeostasis and might present a promising therapeutic target for OA.
Collapse
Affiliation(s)
- Melina Bellini
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
| | - Michael A Pest
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
| | - Manuela Miranda-Rodrigues
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Frank Beier
- Department of Physiology and Pharmacology, Western University, London, ON, Canada.
- Western University Bone and Joint Institute, London, ON, Canada.
- Children's Health Research Institute, London, ON, Canada.
| |
Collapse
|
50
|
Toosi S, Behravan J. Osteogenesis and bone remodeling: A focus on growth factors and bioactive peptides. Biofactors 2020; 46:326-340. [PMID: 31854489 DOI: 10.1002/biof.1598] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/30/2019] [Indexed: 12/14/2022]
Abstract
Bone is one of the most frequently transplanted tissues. The bone structure and its physiological function and stem cells biology were known to be closely related to each other for many years. Bone is considered a home to the well-known systems of postnatal mesenchymal stem cells (MSCs). These bone resident MSCs provide a range of growth factors (GF) and cytokines to support cell growth following injury. These GFs include a group of proteins and peptides produced by different cells which are regulators of important cell functions such as division, migration, and differentiation. GF signaling controls the formation and development of the MSCs condensation and plays a critical role in regulating osteogenesis, chondrogenesis, and bone/mineral homeostasis. Thus, a combination of both MSCs and GFs receives high expectations in regenerative medicine, particularly in bone repair applications. It is known that the delivery of exogenous GFs to the non-union bone fracture site remarkably improves healing results. Here we present updated information on bone tissue engineering with a specific focus on GF characteristics and their application in cellular functions and tissue healing. Moreover, the interrelation of GFs with the damaged bone microenvironment and their mechanistic functions are discussed.
Collapse
Affiliation(s)
- Shirin Toosi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical, Mashhad, Iran
- Food and Drug Administration, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical, Mashhad, Iran
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|