1
|
Manjunatha BS, Handge KT, Shah VS, Al-Thobaiti YE, Pateel DGS. Immunohistochemical expression of matrix metalloproteinase-9 and 13 in oral squamous cell carcinoma and their role in predicting lymph node metastasis. World J Methodol 2025; 15:94514. [DOI: 10.5662/wjm.v15.i2.94514] [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: 03/19/2024] [Revised: 09/23/2024] [Accepted: 10/20/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND One of the main characteristics of oral squamous cell carcinoma (OSCC) is that it metastasizes to cervical lymph nodes frequently with a high degree of local invasiveness. A primary feature of malignant tumors is their penetration of neighboring tissues, such as lymphatic and blood arteries, due to the tumor cells' capacity to break down the extracellular matrix (ECM). Matrix metalloproteinases (MMPs) constitute a family of proteolytic enzymes that facilitate tissue remodeling and the degradation of the ECM. MMP-9 and MMP-13 belong to the group of extracellular matrix degrading enzymes and their expression has been studied in OSCC because of their specific functions. MMP-13, a collagenase family member, is thought to play an essential role in the MMP activation cascade by breaking down the fibrillar collagens, whereas MMP-9 is thought to accelerate the growth of tumors. Elevated MMP-13 expression has been associated with tumor behavior and patient prognosis in a number of malignant cases.
AIM To assess the immunohistochemical expression of MMP-9 and MMP-13 in OSCC.
METHODS A total of 40 cases with histologically confirmed OSCC by incisional biopsy were included in this cross-sectional retrospective study. The protocols for both MMP-9 and MMP-13 immunohistochemical staining were performed according to the manufacturer’s recommendations along with the normal gingival epithelium as a positive control. All the observations were recorded and Pearson’s χ² test with Fisher exact test was used for statistical analysis.
RESULTS Our study showed no significant correlation between MMP-9 and MMP-13 staining intensity and tumor size. The majority of the patients were in advanced TNM stages (III and IV), and showed intense expression of MMP-9 and MMP-13.
CONCLUSION The present study suggests that both MMP-9 and MMP-13 play an important and independent role in OSCC progression and invasiveness. Intense expression of MMP-9 and MMP-13, irrespective of histological grade of OSCC, correlates well with TNM stage. Consequently, it is evident that MMP-9 and MMP-13 are important for the invasiveness and progression of tumors. The findings may facilitate the development of new approaches for evaluating lymph node metastases and interventional therapy techniques, hence enhancing the prognosis of patients diagnosed with OSCC.
Collapse
Affiliation(s)
- Bhari Sharanesha Manjunatha
- Department of Basic Oral Medicine and Allied Dental Sciences, Taif University, At`Taif 26571, Makkah, Saudi Arabia
| | - Keshav T Handge
- Department of Oral and Maxillofacial Pathology, Dr. Vasantrao Pawar Medical College, Hospital and Research Centre, Nashik 423101, Maharashtra, India
| | - Vandana Sandeep Shah
- Department of Oral Pathology and Microbiology, KM Shah Dental College and Hospital, Sumandeep Vidyapeeth, Vadodara 391760, Gujarat, India
| | - Yasser Eid Al-Thobaiti
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, Faculty of Dentistry, Taif University, Al-Haweiah 26571 Makkah, Saudi Arabia
| | | |
Collapse
|
2
|
Tan Z, Shek HT, Li Z, Xia L, He Y, Chen P, Wong JSH, Gao B, Chan D, To MKT. An inducible mouse model of osteogenesis imperfecta type V reveals aberrant osteogenesis caused by Ifitm5 c.-14C>T mutation. J Bone Miner Res 2025; 40:577-590. [PMID: 39908237 PMCID: PMC12103725 DOI: 10.1093/jbmr/zjaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025]
Abstract
Osteogenesis imperfecta (OI) type V is typically characterized by radial head dislocation, calcification of interosseous membrane, and hyperplastic callus. It is caused by the c.-14C>T mutation in the 5' UTR of IFITM5 gene, adding 5 amino acids (MALEP) to the N-terminal of IFITM5 protein. Previous studies have suggested a neomorphic function of the MALEP-IFITM5 protein. However, the underlying mechanisms remain unclear due to embryonic lethality in previous mouse models. Therefore, we developed an inducible mouse model (Ifitm5flox c.-14C>T) that could be induced by Cre expressed at different developmental stages to explore the pathogenic effects of the neomorphic MALEP-IFITM5. The mutant Ifitm5 allele could be regulated by the endogenous regulatory elements after Cre recombination, maintaining its spatiotemporal expression pattern and physiological level. Specifically, Prx1-Cre; Ifitm5flox c.-14C>T mutant mice were born with fractures in all limbs, showing impaired ossification and enhanced chondrogenesis associated with increased SOX9 abundance. Analyses of single-cell RNA sequencing data revealed arrested osteogenesis in Prx1-Cre; Ifitm5flox c.-14C>T mouse. A major population of cells expressing both osteogenic and chondrogenic signature genes was identified in the mutant mouse. Reduced expression of SP7 and SOST in the cortical regions of mutant mice confirmed delayed osteocyte maturation and compromised osteogenesis. Elevated bone marrow adipocytes were found in the adult mutant mice. Ectopic chondrogenesis and SOX9 expression were also observed in the perichondrium regions of Col1a1-Cre; Ifitm5flox c.-14C>T and Ocn-Cre; Ifitm5flox c.-14C>T mutant mice. The inducible Ifitm5flox c.-14C>T mouse model and integrated single-cell transcriptomic analyses elucidated that ectopic expression of SOX9 and disrupted homeostatic balance among osteogenesis, chondrogenesis, and adipogenesis may contribute to the pathogenesis caused by MALEP-IFITM5, helping to gain deeper insights into the molecular mechanisms of type V OI.
Collapse
Affiliation(s)
- Zhijia Tan
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hiu Tung Shek
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zeluan Li
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Linjian Xia
- Clinical, Translational and Basic Research Laboratory, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Yanni He
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Peikai Chen
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Janus Siu Him Wong
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bo Gao
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Danny Chan
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Michael Kai Tsun To
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
3
|
Raulinaite K, Zelvyte R, Skemiene K, Monkeviciene I. Treatment tactic of canine cranial cruciate ligament rupture management: A 28-day comparative analysis of ACP and NSAID induced effects on the serum MMP-3 levels and clinical outcomes. VET MED-CZECH 2025; 70:124-133. [PMID: 40406605 PMCID: PMC12093463 DOI: 10.17221/39/2024-vetmed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/23/2025] [Indexed: 05/26/2025] Open
Abstract
Cranial cruciate ligament rupture (CrCLR) is a common stifle joint pathology among dogs, leading to osteoarthritis and painfulness. Non-surgical treatment options often represent the usage of non-steroidal anti-inflammatory drugs for 14 days (NSAIDs), but autologous conditioned plasma (ACP) shows promising results in managing various orthopaedic conditions, decreasing inflammation, and improving the clinical outcome in dogs. This study aimed to determine the differences in MMP-3 serum levels and the clinical outcomes between differently treated cranial cruciate rupture cases. For this purpose, we used two different treatment methods for managing canine cranial cruciate ligament rupture (minimally invasive ACP injection or oral NSAIDs), and evaluated the clinical outcomes, indicating the quality of life, and the MMP-3 serum levels over a period of 28 days. The findings of this investigation indicate that ACP has better efficacy than two weeks of NSAIDs in inflammation reduction, clinical outcome improvement, and the allowance of a longer duration of activity after 28 days.
Collapse
Affiliation(s)
- Kristina Raulinaite
- Department of Anatomy and Physiology, Faculty of Veterinary, Veterinary Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rasa Zelvyte
- Department of Anatomy and Physiology, Faculty of Veterinary, Veterinary Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Kristina Skemiene
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ingrida Monkeviciene
- Department of Anatomy and Physiology, Faculty of Veterinary, Veterinary Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
4
|
Nieuwstraten J, Riester R, Hofmann UK, Guilak F, Danalache M. Matrix metalloproteinases accelerate pericellular matrix breakdown and disrupt mechanotransduction in osteoarthritis. Acta Biomater 2025; 195:73-82. [PMID: 39956307 DOI: 10.1016/j.actbio.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/25/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
The pericellular matrix (PCM) is a specialized, narrow matrix surrounding each chondrocyte in articular cartilage, together constituting the chondron - the fundamental metabolic and functional unit of cartilage. The PCM plays a vital role in mediating biomechanical and biochemical signals essential for chondrocyte function. In osteoarthritis (OA), a chronic joint disorder characterized by progressive cartilage degradation, the PCM is one of the earliest sites of catabolic degradation, primarily driven by matrix metalloproteinases (MMPs). This study aims to investigate the functional relationship between PCM degradation and chondrocyte mechanosignaling, with an emphasis on MMP-driven changes in mechanotransduction in osteoarthritic cartilage. Human chondrons (N = 64) were incubated with MMP-2, MMP-3, and MMP-7, and structural changes were assessed histologically by evaluating perlecan and collagen type VI. Cellular elasticity was measured using atomic force microscopy (AFM), and mechanically evoked intracellular Ca2+ transients were assessed via AFM single-cell indentations (500 nN). All three MMPs induced pronounced catabolic effects on the PCM structure, showing distinct impacts on collagen type VI and perlecan, as well as on the biomechanical properties (p < 0.001). MMP-driven alterations in PCM integrity significantly reduced the Ca2+ transients of chondrons in response to mechanical stimuli (p < 0.001). While TRPV4 activation was elevated in intact chondrons, PIEZO channels were involved in mechanotransduction in both healthy and MMP-treated chondrons. In osteoarthritic stages, the mechanotransduction dynamics shifted significantly towards PIEZO channels. This study elucidates the interplay between MMP-mediated PCM degradation, structural-functional dynamics, and chondrocyte mechanotransduction, underscoring the critical role of the PCM in maintaining normal chondrocyte functionality and mechanosensing. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) is a prevalent degenerative joint disease affecting millions worldwide. Central to its pathology is the degradation of the pericellular matrix (PCM) by matrix metalloproteinases (MMPs), which disrupts chondrocyte mechanotransduction, altering cellular responses to mechanical stimuli. This study explores the impact of MMP-2, MMP-3, and MMP-7 on PCM structure and chondrocyte mechanosensing. Our results reveal that MMP-induced degradation significantly compromises PCM structural integrity, leading to altered mechanotransduction dynamics in chondrocytes. Degradation specifically redirects the primary function of ion channels from TRPV4 to PIEZO channels in cells impacted by MMPs. This highlights the interplay between MMP-mediated PCM degradation, chondrocyte mechanotransduction and as thus structural-functional dynamics, underscoring the critical role of the PCM in maintaining normal chondrocyte functionality and mechanosensing.
Collapse
Affiliation(s)
- Jule Nieuwstraten
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-7207 Tübingen, Germany
| | - Rosa Riester
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-7207 Tübingen, Germany
| | - Ulf Krister Hofmann
- Department of Orthopedic, Trauma, and Reconstructive Surgery, Division of Arthroplasty, RWTH Aachen University Hospital, Pauwelsstraße 30, D-52074 Aachen, Germany
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-7207 Tübingen, Germany.
| |
Collapse
|
5
|
Jiang N, Yang S, Sun Y, Zhang C, Liu K, Huang Y, Li F. The effect of exosomes from canine bone mesenchymal stem cells on IL-1β-mediated inflammatory responses in chondrocytes. Cytotechnology 2025; 77:27. [PMID: 39736844 PMCID: PMC11682030 DOI: 10.1007/s10616-024-00685-4] [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: 04/12/2024] [Accepted: 12/16/2024] [Indexed: 01/01/2025] Open
Abstract
Osteoarthritis is a degenerative disease of cartilage, and exosome derived from mesenchymal stem cells (MSCs) are considered promising for treating inflammatory musculoskeletal disorders, although their mechanisms are not fully understood. This study aimed to investigate the effects of exosomes derived from canine bone marrow mesenchymal stem cells (cBMSCs-Exos) on the expression of inflammatory factors and genes related cartilage matrix metabolism in IL-1β-induced canine chondrocytes. Canine BMSCs were isolated and characterized for surface markers and trilineage differentiation. Exosomes were then extracted and performed surface labeling detection. Canine chondrocytes were exposed to IL-1β to mimic osteoarthritis in vitro. Subsequently, the chondrocytes were treated with exosomes from BMSCs, and the expression levels of related genes and IL-6 protein were assessed. The mesenchymal stem cells isolated from bone marrow and cultured exhibited positive CD44 and CD90, negative expression of CD45 and HLA, and demonstrated potential to differentiate into adipocytes, osteoblasts and chondrocytes. Exosomes from BMSCs exhibited positivity expression of CD9, CD63 and CD81. Treatment with exosomes significantly reduced IL-6 and TNF-α mRNA levels induced by IL-1β, as well as IL-6 protein expression. Additionally, a significant decrease was observed in the mRNA levels catabolic marker genes MMP-13, ADAMTS-5, and COX2. Conversely, there was a significant increase in the mRNA levels of anti-inflammatory cytokines IL-4, IL-10, and anabolic marker genes, such as COL2A1, ACAN, and SOX9. cBMSCs-Exos play a vital role in cartilage protection by suppressing the expression of pro-inflammatory and anabolic genes while simultaneously enhancing the expression of genes involved in synthesis metabolism.
Collapse
Affiliation(s)
- Nan Jiang
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| | - Shuna Yang
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| | - Yunfei Sun
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| | - Chao Zhang
- Scholl of Biotechnology, Jiuquan Vocational Technical College, Jiuquan, 735000 China
| | - Kaicheng Liu
- Qingdao Kangdi’en Animal Pharmaceutical Co., Ltd, Qingdao, 266041 China
| | - Yufeng Huang
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| | - Fangzheng Li
- College of Veterinary Medicine, Qingdao Agricultural University, No. 700 Changcheng Road, Chengyang, Qingdao, 266109 China
| |
Collapse
|
6
|
Anderson-Watters M, Khan IM. BMP9 induces postnatal zonal stratification of immature articular cartilage through reconfiguration of the existing collagen framework. Front Cell Dev Biol 2025; 12:1511908. [PMID: 39935787 PMCID: PMC11810917 DOI: 10.3389/fcell.2024.1511908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 02/13/2025] Open
Abstract
Articular cartilage lines bones in synovial joints, and its main structural element, collagen, has an arcade-like arrangement formed from an initially random network in a process called postnatal maturation. This reshaping of the extracellular matrix is similar across all species and is critical for the lifelong strength and durability of cartilage. Collagen remodelling during maturation is difficult to study because it spans a period of time between birth and puberty, and in larger animals this can be months or years. In this study, we show that growth factor bone morphogenetic protein-9 (BMP9) induces collagen remodelling in intact immature articular cartilage explants within 21 days, generating the characteristic arcade-like structure and zonal anisotropic architecture of adult cartilage. In explants exposed to BMP9, collagen fibrils underwent angular displacement from 19° to 78° with respect to the surface, cell density decreased 1.77-fold, and chondrons were significantly larger. The absence of labelling with anti-COL2¾m, a marker of collagen turnover, showed that the existing fibril network was restructured. We found that stromelysin-1 (metalloproteinase-3, MMP3) gene expression was consistently upregulated, whilst other MMP transcript levels were unchanged or reduced. Remodelling was dependent on proteoglycan turnover and could be inhibited using PD166973. These data suggest a possible mechanism whereby MMP3 induces proteoglycan turnover and depolymerises collagen fibrils enabling them to undergo spatial reorganisation. This process may be driven by tissue swelling, which generates directional strain to align fibrils into an arcade-like pattern. The ability to induce tissue maturation advances the potential for engineering durable and functional cartilage for patients requiring joint repair due to diseases such as osteoarthritis.
Collapse
Affiliation(s)
| | - Ilyas M. Khan
- Faculty of Medicine, Health and Life Science, Swansea University, Swansea, United Kingdom
| |
Collapse
|
7
|
Hamada M, Eskelinen ASA, Florea C, Mikkonen S, Nieminen P, Grodzinsky AJ, Tanska P, Korhonen RK. Loss of collagen content is localized near cartilage lesions on the day of injurious loading and intensified on day 12. J Orthop Res 2025; 43:70-83. [PMID: 39312444 PMCID: PMC11615416 DOI: 10.1002/jor.25975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024]
Abstract
Joint injury can lead to articular cartilage damage, excessive inflammation, and post-traumatic osteoarthritis (PTOA). Collagen is an essential component for cartilage function, yet current literature has limited understanding of how biochemical and biomechanical factors contribute to collagen loss in injured cartilage. Our aim was to investigate spatially dependent changes in collagen content and collagen integrity of injured cartilage, with an explant model of early-stage PTOA. We subjected calf knee cartilage explants to combinations of injurious loading (INJ), interleukin-1α-challenge (IL) and physiological cyclic loading (CL). Using Fourier transform infrared microspectroscopy, collagen content (Amide I band) and collagen integrity (Amide II/1338 cm-1 ratio) were estimated on days 0 and 12 post-injury. We found that INJ led to lower collagen content near lesions compared to intact regions on day 0 (p < 0.001). On day 12, near-lesion collagen content was lower compared to day 0 (p < 0.05). Additionally, on day 12, INJ, IL, and INJ + IL groups exhibited lower collagen content along most of tissue depth compared to free-swelling control group (p < 0.05). CL groups showed higher collagen content along most of tissue depth compared to corresponding groups without CL (p < 0.05). Immunohistochemical analysis revealed higher MMP-1 and MMP-3 staining intensities localized within cell lacunae in INJ group compared to CTRL group on day 0. Our results suggest that INJ causes rapid loss of collagen content near lesions, which is intensified on day 12. Additionally, CL could mitigate the loss of collagen content at intact regions after 12 days.
Collapse
Affiliation(s)
- Moustafa Hamada
- Department of Technical PhysicsUniversity of Eastern FinlandKuopioFinland
| | | | - Cristina Florea
- Department of Technical PhysicsUniversity of Eastern FinlandKuopioFinland
| | - Santtu Mikkonen
- Department of Technical PhysicsUniversity of Eastern FinlandKuopioFinland
| | - Petteri Nieminen
- Institute of BiomedicineUniversity of Eastern FinlandKuopioFinland
| | - Alan J. Grodzinsky
- Departments of Biological EngineeringElectrical Engineering and Computer Science, and Mechanical Engineering, Massachusetts Institute of TechnologyMassachusetts AvenueCambridgeMassachusettsUSA
| | - Petri Tanska
- Department of Technical PhysicsUniversity of Eastern FinlandKuopioFinland
| | - Rami K. Korhonen
- Department of Technical PhysicsUniversity of Eastern FinlandKuopioFinland
| |
Collapse
|
8
|
Patnaik R, Varghese R, Jannati S, Naidoo N, Banerjee Y. Targeting PAR2-mediated inflammation in osteoarthritis: a comprehensive in vitro evaluation of oleocanthal's potential as a functional food intervention for chondrocyte protection and anti-inflammatory effects. BMC Musculoskelet Disord 2024; 25:769. [PMID: 39354427 PMCID: PMC11446003 DOI: 10.1186/s12891-024-07888-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/20/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by chronic inflammation and progressive cartilage degradation, ultimately leading to joint dysfunction and disability. Oleocanthal (OC), a bioactive phenolic compound derived from extra virgin olive oil, has garnered significant attention due to its potent anti-inflammatory properties, which are comparable to those of non-steroidal anti-inflammatory drugs (NSAIDs). This study pioneers the investigation into the effects of OC on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway in OA, aiming to validate its efficacy as a functional food-based therapeutic intervention. METHODS To simulate cartilage tissue in vitro, human bone marrow-derived mesenchymal stem cells (BMSCs) were differentiated into chondrocytes. An inflammatory OA-like environment was induced in these chondrocytes using lipopolysaccharide (LPS) to mimic the pathological conditions of OA. The therapeutic effects of OC were evaluated by treating these inflamed chondrocytes with various concentrations of OC. The study focused on assessing key inflammatory markers, catabolic enzymes, and mitochondrial function to elucidate the protective mechanisms of OC. Mitochondrial function, specifically mitochondrial membrane potential (ΔΨm), was assessed using Rhodamine 123 staining, a fluorescent dye that selectively accumulates in active mitochondria. The integrity of ΔΨm serves as an indicator of mitochondrial and bioenergetic function. Additionally, Western blotting was employed to analyze protein expression levels, while real-time polymerase chain reaction (RT-PCR) was used to quantify gene expression of inflammatory cytokines and catabolic enzymes. Flow cytometry was utilized to measure cell viability and apoptosis, providing a comprehensive evaluation of OC's therapeutic effects on chondrocytes. RESULTS The results demonstrated that OC significantly downregulated PAR-2 expression in a dose-dependent manner, leading to a substantial reduction in pro-inflammatory cytokines, including TNF-α, IL-1β, and MCP-1. Furthermore, OC attenuated the expression of catabolic markers such as SOX4 and ADAMTS5, which are critically involved in cartilage matrix degradation. Importantly, OC was found to preserve mitochondrial membrane potential (ΔΨm) in chondrocytes subjected to inflammatory stress, as evidenced by Rhodamine 123 staining, indicating a protective effect on cellular bioenergetics. Additionally, OC modulated the Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL)/Receptor Activator of Nuclear Factor Kappa-Β (RANK) pathway, suggesting a broader therapeutic action against the multifactorial pathogenesis of OA. CONCLUSIONS This study is the first to elucidate the modulatory effects of OC on the PAR-2 mediated inflammatory pathway in OA, revealing its potential as a multifaceted therapeutic agent that not only mitigates inflammation but also protects cartilage integrity. The preservation of mitochondrial function and modulation of the RANKL/RANK pathway further underscores OC's comprehensive therapeutic potential in counteracting the complex pathogenesis of OA. These findings position OC as a promising candidate for integration into nutritional interventions aimed at managing OA. However, further research is warranted to fully explore OC's therapeutic potential across different stages of OA and its long-term effects in musculoskeletal disorders.
Collapse
|
9
|
Lee SB, Abdal Dayem A, Kmiecik S, Lim KM, Seo DS, Kim HT, Kumar Biswas P, Do M, Kim DH, Cho SG. Efficient improvement of the proliferation, differentiation, and anti-arthritic capacity of mesenchymal stem cells by simply culturing on the immobilized FGF2 derived peptide, 44-ERGVVSIKGV-53. J Adv Res 2024; 62:119-141. [PMID: 37777063 PMCID: PMC11331723 DOI: 10.1016/j.jare.2023.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 08/23/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
INTRODUCTION The stem cell microenvironment has been evidenced to robustly affect its biological functions and clinical grade. Natural or synthetic growth factors, especially, are essential for modulating stem cell proliferation, metabolism, and differentiation via the interaction with specific extracellular receptors. Fibroblast growth factor-2 (FGF-2) possesses pleiotropic functions in various tissues and organs. It interacts with the FGF receptor (FGFR) and activates FGFR signaling pathways, which involve numerous biological functions, such as angiogenesis, wound healing, cell proliferation, and differentiation. OBJECTIVES Here, we aim to explore the molecular functions, mode of action, and therapeutic activity of yet undetermined function, FGF-2-derived peptide, FP2 (44-ERGVVSIKGV-53) in promoting the proliferation, differentiation, and therapeutic application of human Wharton's jelly mesenchymal stem cells (hWJ-MSCs) in comparison to other test peptides, canofin1 (FP1), hexafin2 (FP3), and canofin3 (FP4) with known functions. METHODS The immobilization of test peptides that are fused with mussel adhesive proteins (MAP) on the culture plate was carried out via EDC/NHS chemistry. Cell Proliferation assay, colony-forming unit, western blotting analysis, gene expression analysis, RNA-Seq. analysis, osteogenic, and chondrogenic differentiation capacity were applied to test the activity of the test peptides. We additionally utilized three-dimensional (3D) structural analysis and artificial intelligence (AI)-based AlphaFold2 and CABS-dock programs for receptor interaction prediction of the peptide receptor. We also verified the in vivo therapeutic capacity of FP2-cultured hWJ-MSCs using an osteoarthritis mice model. RESULTS Culture of hWJ-MSC onto an FP2-immobilized culture plate showed a significant increase in cell proliferation (n = 3; *p < 0.05, **p < 0.01) and the colony-forming unit (n = 3; *p < 0.05, **p < 0.01) compared with the test peptides. FP2 showed a significantly upregulated phosphorylation of FRS2α and FGFR1 and activated the AKT and ERK signaling pathways (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001). Interestingly, we detected efficient FP2 receptor binding that was predicted using AI-based tools. Treatment with an AKT inhibitor significantly abrogated the FP2-mediated enhancement of cell differentiation (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001). Intra-articular injection of FP2-cultured MSCs significantly mitigated arthritis symptoms in an osteoarthritis mouse model, as shown through the functional tests (n = 10; *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001), modulation of the expression level of the pro-inflammatory and anti-inflammatory genes, and improved osteochondral regeneration as demonstrated by tissue sections. CONCLUSION Our study identified the FGF-2-derived peptide FP2 as a promising candidate peptide to improve the therapeutic potential of hWJ-MSCs, especially in bone and cartilage regeneration.
Collapse
Affiliation(s)
- Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sebastian Kmiecik
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, 02-089 Warsaw, Poland
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dong Sik Seo
- Stem Cell Research Center of AMOLIFESCIENCE Co., Ltd, 91, Gimpo-daero 1950 Beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Hyeong-Taek Kim
- Stem Cell Research Center of AMOLIFESCIENCE Co., Ltd, 91, Gimpo-daero 1950 Beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Polash Kumar Biswas
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
10
|
Solomonov I, Locatelli I, Tortorella S, Unni M, Aharoni SL, Alchera E, Locatelli E, Maturi M, Venegoni C, Lucianò R, Salonia A, Corti A, Curnis F, Grasso V, Malamal G, Jose J, Comes Franchini M, Sagi I, Alfano M. Contrast enhanced photoacoustic detection of fibrillar collagen in the near infrared region-I. NANOSCALE ADVANCES 2024; 6:3655-3667. [PMID: 38989511 PMCID: PMC11232541 DOI: 10.1039/d4na00204k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/22/2024] [Indexed: 07/12/2024]
Abstract
Fibrillar collagen accumulation emerges as a promising biomarker in several diseases, such as desmoplastic tumors and unstable atherosclerotic plaque. Gold nanorods (GNRs) hold great potential as contrast agents in high-resolution, biomedically safe, and non-invasive photoacoustic imaging (PAI). This study presents the design and characterization of a specialized imaging tool which exploits GNR assisted targeted photoacoustic imaging that is tailored for the identification of fibrillar collagen. In addition to the photoacoustic characterization of collagen in the NIR 1 and 2 regions, we demonstrate the detailed steps of conjugating a decoy to GNRs. This study serves as a proof of concept, that demonstrates that conjugated collagenase-1 (MMP-1) generates a distinct and collagen-specific photoacoustic signal, facilitating real-time visualization in the wavelength range of 700-970 nm (NIR I). As most of the reported studies utilized the endogenous contrast of collagen in the NIR II wavelength that has major limitations to perform in vivo deep tissue imaging, the approach that we are proposing is unique and it highlights the promise of MMP-1 decoy-functionalized GNRs as novel contrast agents for photoacoustic imaging of collagen in the NIR 1 region. To our knowledge this is the first time functionalized GNRs are optimized for the detection of fibrillar collagen and utilized in the field of non-invasive photoacoustic imaging that can facilitate a better prognosis of desmoplastic tumors and broken atherosclerotic plaques.
Collapse
Affiliation(s)
- Inna Solomonov
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science Rehovot 76100 Israel
| | - Irene Locatelli
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
| | - Silvia Tortorella
- Department of Industrial Chemistry "Toso Montanari", University of Bologna Via P. Gobetti 85 40129 Bologna Italy
| | - Manu Unni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science Rehovot 76100 Israel
| | - Shay-Lee Aharoni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science Rehovot 76100 Israel
| | - Elisa Alchera
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
| | - Erica Locatelli
- Department of Industrial Chemistry "Toso Montanari", University of Bologna Via P. Gobetti 85 40129 Bologna Italy
| | - Mirko Maturi
- Department of Industrial Chemistry "Toso Montanari", University of Bologna Via P. Gobetti 85 40129 Bologna Italy
| | - Chiara Venegoni
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
| | - Roberta Lucianò
- Department of Pathology, IRCCS San Raffaele Hospital and Scientific Institute Milan Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
- Vita-Salute San Raffaele University Milan Italy
| | - Angelo Corti
- Vita-Salute San Raffaele University Milan Italy
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute Milan Italy
| | - Flavio Curnis
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute Milan Italy
| | - Valeria Grasso
- FUJIFILM Visualsonics Inc. Amsterdam the Netherlands
- Faculty of Engineering, Institute for Materials Science, Christian-Albrecht University of Kiel Kiel Germany
| | | | - Jithin Jose
- FUJIFILM Visualsonics Inc. Amsterdam the Netherlands
| | - Mauro Comes Franchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna Via P. Gobetti 85 40129 Bologna Italy
| | - Irit Sagi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science Rehovot 76100 Israel
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele Milan Italy
| |
Collapse
|
11
|
Dubuc J, Schneider MJ, Dubuc V, Richard H, Pinsard M, Bancelin S, Legare F, Girard C, Laverty S. Degradation of Proteoglycans and Collagen in Equine Meniscal Tissues. Int J Mol Sci 2024; 25:6439. [PMID: 38928148 PMCID: PMC11203490 DOI: 10.3390/ijms25126439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Investigate meniscal extracellular matrix degradation. Equine menisci (n = 34 from 17 horses) were studied. Site-matched sections were cut and scored from three regions (ROIs; n = 102) and stained for histology, proteoglycan (safranin O and fast green), aggrecan, and collagen cleavage (NITEGE, DIPEN, and C1,2C antibodies, respectively). Picrosirius red and second harmonic generation microscopy were performed to investigate collagen ultrastructure. A total of 42 ROIs met the inclusion criteria and were included in the final analysis. The median (range) ROI histological score was 3 (0-9), providing a large spectrum of pathology. The median (range) proteoglycan score was 1 (0-3), representing superficial and central meniscal loss. The median (range) of DIPEN, NITEGE, and C1,2C scores was 1 (0-3), revealing immunostaining of the femoral and tibial surfaces. The proteoglycan scores exhibited significant positive associations with both histologic evaluation (p = 0.03) and DIPEN scores (p = 0.02). Additionally, a robust positive association (p = 0.007) was observed between the two aggrecanolysis indicators, NITEGE and DIPEN scores. A negative association (p = 0.008) was identified between NITEGE and histological scores. The C1,2C scores were not associated with any other scores. Picrosirius red and second harmonic generation microscopy (SHGM) illustrated the loss of the collagen matrix and structure centrally. Proteoglycan and collagen degradation commonly occur superficially in menisci and less frequently centrally. The identification of central meniscal proteoglycan and collagen degradation provides novel insight into central meniscal degeneration. However, further research is needed to elucidate the etiology and sequence of degradative events.
Collapse
Affiliation(s)
- Julia Dubuc
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Melodie Jil Schneider
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Valerie Dubuc
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Helene Richard
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Maxime Pinsard
- Institut National de la Recherche Scientifique, Université du Quebec, 1650 Bd Lionel-Boulet, Varennes, Quebec, QC J3X1P7, Canada
| | - Stephane Bancelin
- Institut National de la Recherche Scientifique, Université du Quebec, 1650 Bd Lionel-Boulet, Varennes, Quebec, QC J3X1P7, Canada
| | - Francois Legare
- Institut National de la Recherche Scientifique, Université du Quebec, 1650 Bd Lionel-Boulet, Varennes, Quebec, QC J3X1P7, Canada
| | - Christiane Girard
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Sheila Laverty
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| |
Collapse
|
12
|
Danalache M, Umrath F, Riester R, Schwitalle M, Guilak F, Hofmann UK. Proteolysis of the pericellular matrix: Pinpointing the role and involvement of matrix metalloproteinases in early osteoarthritic remodeling. Acta Biomater 2024; 181:297-307. [PMID: 38710401 DOI: 10.1016/j.actbio.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/14/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024]
Abstract
The pericellular matrix (PCM) serves a critical role in signal transduction and mechanoprotection in chondrocytes. Osteoarthritis (OA) leads to a gradual deterioration of the cartilage, marked by a shift in the spatial arrangement of chondrocytes from initially isolated strands to large cell clusters in end-stage degeneration. These changes coincide with progressive enzymatic breakdown of the PCM. This study aims to assess the role and involvement of specific matrix metalloproteinases (MMPs) in PCM degradation during OA. We selected cartilage samples from 148 OA patients based on the predominant spatial chondrocyte patterns. The presence of various MMPs (-1,-2,-3,-7,-8,-9,-10,-12,-13) was identified by multiplexed immunoassays. For each pattern and identified MMP, the levels and activation states (pro-form vs. active form) were measured by zymograms and western blots. The localization of these MMPs was determined using immunohistochemical labeling. To verify these results, healthy cartilage was exposed to purified MMPs, and the consecutive structural integrity of the PCM was analyzed through immunolabeling and proximity ligation assay. Screening showed elevated levels of MMP-1,-2,-3,-7, and -13, with their expression profile showing a clear dependency of the degeneration stage. MMP-2 and -7 were localized in the PCM, whereas MMP-1,-7, and -13 were predominantly intracellular. We found that MMP-2 and -3 directly disrupt collagen type VI, and MMP-3 and -7 destroy perlecan. MMP-2, -3, and -7 emerge as central players in early PCM degradation in OA. With the disease's initial stages already displaying elevated peaks in MMP expression, this insight may guide early targeted therapies to halt abnormal PCM remodeling. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) causes a gradual deterioration of the articular cartilage, accompanied by a progressive breakdown of the pericellular matrix (PCM). The PCM's crucial function in protecting and transmitting signals within chondrocytes is impaired in OA. By studying 148 OA-patient cartilage samples, the involvement of matrix metalloproteinases (MMPs) in PCM breakdown was explored. Findings highlighted elevated levels of certain MMPs linked to different stages of degeneration. Notably, MMP-2, -3, and -7 were identified as potent contributors to early PCM degradation, disrupting key components like collagen type VI and perlecan. Understanding these MMPs' roles in initiating OA progression, especially in its early stages, provides insights into potential targets for interventions to preserve PCM integrity and potentially impeding OA advancement.
Collapse
Affiliation(s)
- Marina Danalache
- Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-72072 Tübingen, Germany.
| | - Felix Umrath
- Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-72072 Tübingen, Germany; Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, D-72076 Tübingen, Germany
| | - Rosa Riester
- Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-72072 Tübingen, Germany
| | - Maik Schwitalle
- Winghofer Medicum, Röntgenstraße 38, D-72108 Rottenburg am Neckar, Germany
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA
| | - Ulf Krister Hofmann
- Department of Orthopedic, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, Pauwelsstraße 30, D-52074 Aachen, Germany
| |
Collapse
|
13
|
Fu Y, Zhang C, Yang Y, Zhou B, Yang M, Zhu G, Zhu Y. Effect of umbilical cord blood-mononuclear cells on knee osteoarthritis in rabbits. J Orthop Surg Res 2024; 19:323. [PMID: 38811966 PMCID: PMC11138004 DOI: 10.1186/s13018-024-04815-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 05/27/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND To investigate the effect and underlying mechanism of umbilical cord blood-mononuclear cells (UCB-MNCs) in treating knee osteoarthritis (KOA) in rabbits. METHODS A rabbit KOA model was prepared by anterior cruciate ligament transection (ACLT). Fifty New Zealand white rabbits were randomly divided into the control group, model group, sodium hyaluronate (SH) group, platelet-rich plasma (PRP) group and UCB-MNC group. Knee injections were performed once a week for five consecutive weeks. The gross view of the knee joint, morphology of knee cartilage and structural changes in the knee joint were observed on CT scans, and graded by the Lequesne MG behavioral score and the Mankin score. TNF-α and IL-1β levels in the synovial fluid of the knee were measured by the enzyme-linked immunosorbent assay (ELISA). Expression levels of MMP-13 and COL-II in the knee cartilage were detected by Western blotting and qRT-PCR. RESULTS The Lequesne MG behavioral score and the Mankin score were significantly higher in the model group than those in the control group (P < 0.05). Rabbits in the SH, PRP and UCB-MNC groups had sequentially lower scores than those in the model group. Imaging features of KOA were more pronounced in the model group than in the remaining groups. CB-MNC significantly relieved KOA, compared to SH and PRP. Significantly higher levels of TNF-α and IL-1β in the synovial fluid of the knee, and up-regulated MMP-13 and down-regulated COL-II in the knee cartilage were detected in the model group than in the control group. These changes were significantly reversed by the treatment with SH, PRP and UCB-MNCs, especially UCB-MNCs. CONCLUSION Injections of UCB-MNCs into knees protect the articular cartilage and hinder the progression of KOA in rabbits by improving the local microenvironment at knee joints.
Collapse
Affiliation(s)
- Yuhang Fu
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, 264199, Shandong Province, China
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Chi Zhang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Yong Yang
- Yantai City Yantai Mountain Hospital, Yantai, 264008, Shandong Province, China
| | - Baisui Zhou
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Meng Yang
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, 264199, Shandong Province, China
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Guoshuai Zhu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China
| | - Yonglin Zhu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264199, Shandong Province, China.
| |
Collapse
|
14
|
Jo HG, Baek CY, Lee J, Hwang Y, Baek E, Hwang JH, Lee D. Anti-Inflammatory, Analgesic, Functional Improvement, and Chondroprotective Effects of Erigeron breviscapus (Vant.) Hand.-Mazz. Extract in Osteoarthritis: An In Vivo and In Vitro Study. Nutrients 2024; 16:1035. [PMID: 38613068 PMCID: PMC11013172 DOI: 10.3390/nu16071035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 03/31/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative bone disease characterized by inflammation as a primary pathology and currently lacks therapeutic interventions to impede its progression. Erigeron breviscapus (Vant.) Hand.-Mazz. (EB) is an east Asian herbal medicine with a long history of use and a wide range of confirmed efficacy against cardiovascular and central nervous system diseases. The purpose of this study is to evaluate whether EB is worthy of further investigation as a treatment for OA based on anti-inflammatory activity. This study aims to assess the potential of EB as a treatment for OA, focusing on its anti-inflammatory properties. Analgesic effects, functional improvements, and inhibition of cartilage destruction induced by EB were evaluated in acetic acid-induced peripheral pain mice and monosodium iodoacetate-induced OA rat models. Additionally, the anti-inflammatory effect of EB was assessed in serum and cartilage tissue in vivo, as well as in lipopolysaccharide-induced RAW 264.7 cells. EB demonstrated a significant alleviation of pain, functional impairment, and cartilage degradation in OA along with a notable inhibition of pro-inflammatory cytokines, including interleukin-1β, interleukin-6, matrix metalloproteinases 13, and nitric oxide synthase 2, both in vitro and in vivo, in a dose-dependent manner compared to the active control. Accordingly, EB merits further exploration as a potential disease-modifying drug for OA, capable of mitigating the multifaceted pathology of osteoarthritis through its anti-inflammatory properties. Nonetheless, additional validation through a broader experimental design is essential to substantiate the findings of this study.
Collapse
Affiliation(s)
- Hee-Geun Jo
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
- Naturalis Inc., 6, Daewangpangyo-ro, Bundang-gu, Seongnam-si 13549, Republic of Korea
| | - Chae Yun Baek
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
| | - JunI Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
| | - Yeseul Hwang
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
| | - Eunhye Baek
- RexSoft Inc., 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Ji Hye Hwang
- Department of Acupuncture and Moxibustion Medicine, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Donghun Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
| |
Collapse
|
15
|
Lu J, Bian J, Wang Y, Zhao Y, Zhao X, Wang G, Yang J. Oxymatrine protects articular chondrocytes from IL-1β-induced damage through autophagy activation via AKT/mTOR signaling pathway inhibition. J Orthop Surg Res 2024; 19:178. [PMID: 38468339 PMCID: PMC10926585 DOI: 10.1186/s13018-024-04667-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/06/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common degenerative joint disease characterized by persistent articular cartilage degeneration and synovitis. Oxymatrine (OMT) is a quinzolazine alkaloid extracted from the traditional Chinese medicine, matrine, and possesses anti-inflammatory properties that may help regulate the pathogenesis of OA; however, its mechanism has not been elucidated. This study aimed to investigate the effects of OMT on interleukin-1β (IL-1β)-induced damage and the potential mechanisms of action. METHODS Chondrocytes were isolated from Sprague-Dawley rats. Toluidine blue and Collagen II immunofluorescence staining were used to determine the purity of the chondrocytes. Thereafter, the chondrocytes were subjected to IL-1β stimulation, both in the presence and absence of OMT, or the autophagy inhibitor 3-methyladenine (3-MA). Cell viability was assessed using the MTT assay and SYTOX Green staining. Additionally, flow cytometry was used to determine cell apoptosis rate and reactive oxygen species (ROS) levels. The protein levels of AKT, mTOR, LC3, P62, matrix metalloproteinase-13, and collagen II were quantitatively analyzed using western blotting. Immunofluorescence was used to assess LC3 expression. RESULTS OMT alleviated IL-1β-induced damage in chondrocytes, by increasing the survival rate, reducing the apoptosis rates of chondrocytes, and preventing the degradation of the cartilage matrix. In addition, OMT decreased the ROS levels and inhibited the AKT/mTOR signaling pathway while promoting autophagy in IL-1β treated chondrocytes. However, the effectiveness of OMT in improving chondrocyte viability under IL-1β treatment was limited when autophagy was inhibited by 3-MA. CONCLUSIONS OMT decreases oxidative stress and inhibits the AKT/mTOR signaling pathway to enhance autophagy, thus inhibiting IL-1β-induced damage. Therefore, OMT may be a novel and effective therapeutic agent for the clinical treatment of OA.
Collapse
Affiliation(s)
- Jinying Lu
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Jiang Bian
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Yutong Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Yan Zhao
- Provincial Key Laboratory of Cardiovascular and Cerebrovascular Drug Basic Research, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Xinmin Zhao
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Gao Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Jing Yang
- Provincial Key Laboratory of Cardiovascular and Cerebrovascular Drug Basic Research, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
16
|
Jiang X, Wang Q, Wang C, Zhang Y, Wei Z, Wu Z, Yu S, Wu C. The association between genetic polymorphisms of matrix metalloproteinases and knee osteoarthritis: A systematic review and meta-analysis. Int J Rheum Dis 2024; 27:e15123. [PMID: 38514927 DOI: 10.1111/1756-185x.15123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 03/23/2024]
Abstract
AIM To investigate the linkage of matrix metalloproteinase (MMP) gene polymorphisms with the pathogenesis of knee osteoarthritis (OA). METHODS This meta-analysis study systematically retrieved relevant studies from PubMed, Embase, the Cochrane Central, Wanfang Data, CNKI, and SinoMed up to November 2020. Odds ratios (ORs) and 95% confidence intervals (CIs) were used to estimate the association between MMP gene polymorphisms and OA. RESULTS A total of nine case-control studies comprising 1719 knee OA patients and 1904 controls were included in this meta-analysis. The results revealed that MMP-1-1607 (rs1799750) 1G/2G polymorphism was not significantly associated with knee OA risk in four genetic models (OR (95% CI): allele model: 0.89 (0.57, 1.40), p = .615); dominant mode: 0.82 (0.47, 1.44), p = .486; recessive model: 0.88 (0.49, 1.57), p = .659; homozygote model: 0.79 (0.34, 1.82), p = .576. The association was significant for dominant model of MMP-3 C/T: 1.54 (1.10-2.15), p = .013, especially in Asian ethnicity (1.63 (1.11, 2.39), p = .013). Variants of MMP-13 C/T polymorphism were associated with increased risk of knee OA development based on dominant model: 1.56 (1.19, 2.06), p = .001 and homozygote model: 2.12 (1.44, 3.13), p < .001, and there were significant associations between MMP-13 C/T polymorphism and knee OA risk in Asian ethnicity under different genetic models (all p > .05). CONCLUSIONS Present evidence suggested that the gene polymorphisms of MMP-1-1607 1G/2G may not be associated with the risk of OA. But, the dominant model of MMP-3 and MMP-13 polymorphisms in Asian ethnicity was significantly correlated with knee OA.
Collapse
Affiliation(s)
- Xu Jiang
- Department of Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Qianqian Wang
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Chao Wang
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Yanzhuo Zhang
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Zhenjie Wei
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Zhimin Wu
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Shunan Yu
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| | - Chengai Wu
- Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, China
| |
Collapse
|
17
|
Mukherjee A, Das B. The role of inflammatory mediators and matrix metalloproteinases (MMPs) in the progression of osteoarthritis. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100090. [PMID: 38440290 PMCID: PMC10910010 DOI: 10.1016/j.bbiosy.2024.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is a chronic musculoskeletal disorder characterized by an imbalance between (synthesis) and catabolism (degradation) in altered homeostasis of articular cartilage mediated primarily by the innate immune system. OA degenerates the joints resulting in synovial hyperplasia, degradation of articular cartilage with damage of the structural and functional integrity of the cartilage extracellular matrix, subchondral sclerosis, osteophyte formation, and is characterized by chronic pain, stiffness, and loss of function. Inflammation triggered by factors like biomechanical stress is involved in the development of osteoarthritis. In OA apart from catabolic effects, anti-inflammatory anabolic processes also occur continually. There is also an underlying chronic inflammation present, not only in cartilage tissue but also within the synovium, which perpetuates tissue destruction of the OA joint. The consideration of inflammation in OA considers synovitis and/or other cellular and molecular events in the synovium during the progression of OA. In this review, we have presented the progression of joint degradation that results in OA. The critical role of inflammation in the pathogenesis of OA is discussed in detail along with the dysregulation within the cytokine networks composed of inflammatory and anti-inflammatory cytokines that drive catabolic pathways, inhibit matrix synthesis, and promote cellular apoptosis. OA pathogenesis, fluctuation of synovitis, and its clinical impact on disease progression are presented here along with the role of synovial macrophages in promoting inflammatory and destructive responses in OA. The role of interplay between different cytokines, structure, and function of their receptors in the inter-cellular signaling pathway is further explored. The effect of cytokines in the increased synthesis and release of matrix-decomposing proteolytic enzymes, such as matrix metalloproteinase (MMPs) and a disintegrin-like and metalloproteinase with thrombospondin motif (ADAMTS), is elaborated emphasizing the potential impact of MMPs on the chondrocytes, synovial cells, articular and periarticular tissues, and other immune system cells migrating to the site of inflammation. We also shed light on the pathogenesis of OA via oxidative damage particularly due to nitric oxide (NO) via its angiogenic response to inflammation. We concluded by presenting the current knowledge about the tissue inhibitors of metalloproteinases (TIMPs). Synthetic MMP inhibitors include zinc binding group (ZBG), non-ZBG, and mechanism-based inhibitors, all of which have the potential to be therapeutically beneficial in the treatment of osteoarthritis. Improving our understanding of the signaling pathways and molecular mechanisms that regulate the MMP gene expression, may open up new avenues for the creation of therapies that can stop the joint damage associated with OA.
Collapse
Affiliation(s)
- Anwesha Mukherjee
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| |
Collapse
|
18
|
Liphardt AM, Godonou ET, Dreiner M, Mündermann A, Tascilar K, Djalal N, Heer M, Schett G, Zaucke F, Niehoff A. Immobilization by 21 days of bed rest results in type II collagen degradation in healthy individuals. Osteoarthritis Cartilage 2024; 32:177-186. [PMID: 37989468 DOI: 10.1016/j.joca.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/15/2023] [Indexed: 11/23/2023]
Abstract
OBJECTIVE To investigate the effects of 21 days of bed rest immobilization (with and without exercise and nutrition interventions) on type II collagen biomarker concentrations in healthy individuals. DESIGN Twelve healthy male participants (age 34.2 ± 8.3 years; body mass index 22.4 ± 1.7 kg/m²) were exposed to 6 days ambulatory baseline data collection (BDC), 21 days head-down-tilt bed rest (HDT, CON) + interventions (HDT + resistive vibration exercise (2 times/week, 25 minutes): RVE; HDT + RVE + whey protein (0.6 g/kg body weight/day) and bicarbonate supplementation (90 mmol KHCO3/day: NeX), and 6 days of re-ambulation (R) in a cross-over designed study. The starting HDT condition was randomized (CON-RVE-NEX, RVE-NEX-CON, NEX-CON-RVE). Blood and urine samples were collected before, during, and after HDT. Serum concentrations (s) of CPII, C2C, C1,2C, and urinary concentrations (u) of CTX-II and Coll2-1NO2 were measured. RESULTS Twenty-one days of HDT resulted in increased sCPII (p < 0.001), sC2C (p < 0.001), and sC1,2C (p = 0.001) (highest increases: sCPII (+24.2% - HDT5), sC2C (+24.4% - HDT7), sC1,2C (+13.5% - HDT2). sC2C remained elevated at R+1 (p = 0.002) and R+6 (p < 0.001) compared to baseline. NeX led to lower sCPII (p < 0.001) and sC1,2C (p = 0.003) compared to CON. uCTX-II (second void and 24-hour urine) increased during HDT (p < 0.001, highest increase on HDT21: second void +82.8% (p < 0.001); 24-hour urine + 77.8% (p < 0.001). NeX resulted in lower uCTX-II concentrations in 24-hour urine (p = 0.012) compared to CON. CONCLUSIONS Twenty-one days of bed rest immobilization results in type II collagen degradation that does not recover within 6 days of resuming ambulation. The combination of resistive vibration exercise and protein/bicarbonate supplementation minimally counteracted this effect.
Collapse
Affiliation(s)
- Anna-Maria Liphardt
- Department of Internal Medicine 3 - Rheumatology & Immunology, Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Elie-Tino Godonou
- Department of Internal Medicine 3 - Rheumatology & Immunology, Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Maren Dreiner
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Köln, Germany.
| | - Annegret Mündermann
- Department of Orthopaedics and Traumatology, University Hospital Basel, Basel Switzerland; Department of Spine Surgery, University Hospital Basel, Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Department of Clinical Research, University of Basel, Basel, Switzerland.
| | - Koray Tascilar
- Department of Internal Medicine 3 - Rheumatology & Immunology, Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Nadja Djalal
- Department of Internal Medicine 3 - Rheumatology & Immunology, Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Martina Heer
- IU International University of Applied Sciences, Health Sciences, Erfurt, Germany; Department of Nutrition and Food Science, Nutritional Physiology, University of Bonn, Bonn, Germany.
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology & Immunology, Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen & Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopaedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt, Germany.
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Köln, Germany; Cologne Center for Musculoskeletal Biomechanics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln, Germany.
| |
Collapse
|
19
|
Sandhu A, Espin-Garcia O, Rockel JS, Lively S, Perry K, Mohamed NN, Raja Rampersaud Y, Perruccio AV, Robin Poole A, Gandhi R, Kapoor M. Association of synovial fluid and urinary C2C-HUSA levels with surgical outcomes post-total knee arthroplasty. Osteoarthritis Cartilage 2024; 32:98-107. [PMID: 37805006 DOI: 10.1016/j.joca.2023.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/14/2023] [Accepted: 09/30/2023] [Indexed: 10/09/2023]
Abstract
OBJECTIVES After total knee arthroplasty (TKA), ∼30% of knee osteoarthritis (KOA) patients show little symptomatic improvement. Earlier studies have correlated urinary (u) type 2 collagen C terminal cleavage peptide assay (C2C-HUSA), which detects a fragment of cartilage collagen breakdown, with KOA progression. This study determines whether C2C levels in urine, synovial fluid, or their ratio, are associated with post-surgical outcomes. METHODS From a large sample of 489 subjects, diagnosed with primary KOA undergoing TKA, Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain and function scores were collected at baseline (time of surgery) and one-year post-TKA. Baseline urine (u) and synovial fluid (sf) were analysed using the IBEX-C2C-HUSA assay, with higher values indicating higher amounts of cartilage degradation. For urine, results were normalised to creatinine. Furthermore, subjects' changes in WOMAC scores were categorised based on percent reduction in pain or improvement in function, compared to baseline, such that >66.7%, >33.3 to ≤66.7%, and ≤33.3% denoted "strong", "moderate" and "mild/worse" responses, respectively. Associations of individual biofluid C2C-HUSA levels, or their ratio, with change in WOMAC pain and function scores up to one-year post-TKA, or category of change, were analysed by linear, logistic, or cumulative odds models. RESULTS Higher baseline uC2C-HUSA levels or a lower ratio of baseline sfC2C-HUSA to uC2C-HUSA were associated with improvements in WOMAC pain by linear multivariable modelling [odds ratio -0.40 (95% confidence interval -0.76, -0.05) p = 0.03; 0.36 (0.01, 0.71), p = 0.04, respectively], while sfC2C-HUSA alone was not. However, lower ratios of sfC2C-HUSA to uC2C-HUSA were associated with improvements in WOMAC function [1.37 (0.18, 2.55), p = 0.02], while sfC2C-HUSA and uC2C-HUSA alone were not. Lower ratios of sfC2C-HUSA to uC2C-HUSA were also associated with an increased likelihood of a subject being categorised in a group where TKA was beneficial in both univariable [pain, 0.81 (0.68, 0.96), p = 0.02; function, 0.92 (0.85, 0.99), p = 0.035] and multivariable [pain, 0.81 (0.68, 0.97) p = 0.02; function, 0.92 (0.85, 1.00), p = 0.043] ordinal modelling, while sfC2C-HUSA and uC2C-HUSA alone were not. CONCLUSIONS Overall, ratios of baseline sfC2C-HUSA to uC2C-HUSA, and baseline uC2C-HUSA, may play an important role in studying post-TKA surgical outcomes.
Collapse
Affiliation(s)
- Amit Sandhu
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Osvaldo Espin-Garcia
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada; Dalla Lana School of Public Health and Department of Statistical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Jason S Rockel
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Starlee Lively
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kimberly Perry
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Nizar N Mohamed
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Y Raja Rampersaud
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Anthony V Perruccio
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - A Robin Poole
- Department of Surgery, Faculty of Medicine and Health Sciences, McGill University, Montréal, Quebec, Canada
| | - Rajiv Gandhi
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mohit Kapoor
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
Dong Y, Lin L, Ji Y, Cheng X, Zhang Z. Cabozantinib prevents AGEs-induced degradation of type 2 collagen and aggrecan in human chondrocytes. Aging (Albany NY) 2023; 15:13646-13654. [PMID: 38059882 PMCID: PMC10756107 DOI: 10.18632/aging.205186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/26/2023] [Indexed: 12/08/2023]
Abstract
Osteoarthritis (OA) is a joint degenerative disease commonly observed in the old population, lacks effective therapeutic methods, and markedly impacts the normal lives of patients. Degradation of extracellular matrix (ECM) is reported to participate in OA development, which is a potential target for treating OA. Cabozantinib is an inhibitor of tyrosine kinases and is recently claimed with suppressive properties against inflammation. Herein, the protective function of Cabozantinib on advanced glycation end products (AGEs)-induced damages to chondrocytes was tested. SW1353 chondrocytes were stimulated with 100 μg/ml AGEs with or without 10 and 20 μM Cabozantinib for 24 h. Signally increased reactive oxygen species (ROS) levels, declined reduced glutathione (GSH) levels, and elevated release of inflammatory cytokines were observed in AGEs-stimulated SW1353 chondrocytes, which were markedly reversed by Cabozantinib. Moreover, the notably reduced type II collagen and aggrecan levels, and increased matrix metalloproteinase-13 (MMP-13) and A Disintegrin and Metalloproteinase with Thrombospondin Motifs-5 (ADAMTS-5) levels in AGEs-stimulated SW1353 chondrocytes were largely rescued by Cabozantinib. The downregulated Sry-type high-mobility-group box 9 (SOX-9) observed in AGEs-stimulated SW1353 chondrocytes was abolished by Cabozantinib. Furthermore, the impact of Cabozantinib on type II collagen and aggrecan levels in AGEs-treated SW1353 chondrocytes was abrogated by silencing SOX-9. Collectively, Cabozantinib prevented AGEs-induced degradation of type 2 collagen and aggrecan in human chondrocytes by mediating SOX-9.
Collapse
Affiliation(s)
- Yang Dong
- Second Department of Hand Surgery, Yantaishan Hospital, Yantai 264008, Shangdong Province, China
| | - Lianfang Lin
- Second Department of Hand Surgery, Yantaishan Hospital, Yantai 264008, Shangdong Province, China
| | - Yuan Ji
- Second Department of Hand Surgery, Yantaishan Hospital, Yantai 264008, Shangdong Province, China
| | - Xu Cheng
- Second Department of Hand Surgery, Yantaishan Hospital, Yantai 264008, Shangdong Province, China
| | - Zhiwu Zhang
- Second Department of Hand Surgery, Yantaishan Hospital, Yantai 264008, Shangdong Province, China
| |
Collapse
|
21
|
Zhou C, Yang Y, Duan M, Chen C, Pi C, Zhang D, Liu X, Xie J. Biomimetic Fibers Based on Equidistant Micropillar Arrays Determines Chondrocyte Fate via Mechanoadaptability. Adv Healthc Mater 2023; 12:e2301685. [PMID: 37596884 DOI: 10.1002/adhm.202301685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/02/2023] [Indexed: 08/20/2023]
Abstract
It is recognized that the changes in the physical properties of extracellular matrix (ECM) result in fine-tuned cell responses including cell morphology, proliferation and differentiation. In this study, a novel patterned equidistant micropillar substrate based on polydimethylsiloxane (PDMS) is designed to mimic the collagen fiber-like network of the cartilage matrix. By changing the component of the curing agent to an oligomeric base, micropillar substrates with the same topology but different stiffnesses are obtained and it is found that chondrocytes seeded onto the soft micropillar substrate maintain their phenotype by gathering type II collagen and aggrecan more effectively than those seeded onto the stiff micropillar substrate. Moreover, chondrocytes sense and respond to micropillar substrates with different stiffnesses by altering the ECM-cytoskeleton-focal adhesion axis. Further, it is found that the soft substrate-preserved chondrocyte phenotype is dependent on the activation of Wnt/β-catenin signaling. Finally, it is indicated that the changes in osteoid-like region formation and cartilage phenotype loss in the stiffened sclerotic area of osteoarthritis cartilage to validate the changes triggered by micropillar substrates with different stiffnesses. This study provides the cell behavior changes that are more similar to those of real chondrocytes at tissue level during the transition from a normal state to a state of osteoarthritis.
Collapse
Affiliation(s)
- Chenchen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Yueyi Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610064, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610064, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
22
|
Lee SG, Kang H. Protective Effects of a Mixed Medicinal Herb Extract (NUC1) on Collagenase-Induced Osteoarthritis in Rabbits. J Microbiol Biotechnol 2023; 33:1484-1494. [PMID: 37482815 DOI: 10.4014/jmb.2303.03044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
NUC1 (Nutraceutical compound 1) is an ethanol extract composed of a formulation based on medicinal herbs traditionally used for the treatment of arthritis in Korea and China. This study investigated the therapeutic effects of NUC1 on osteoarthritis (OA). The protective effect of NUC1 on OA was tested in a rabbit model of collagenase-induced arthritis (CIA) for 4 weeks. Results were compared among four groups (n = 9 per group): the normal group (untreated), the CIA group (vehicle control), the NUC1 group (CIA rabbits treated with 200 mg/kg NUC1), and the JOINS group (positive control, CIA rabbits treated with 200 mg/kg JOINS tablet). NUC1 significantly inhibited NO production (p < 0.05 at 125 μg/ml, p < 0.01 at 250 μg/ml, and p < 0.001 at 500 μg/ml) and iNOS expression in macrophages, in a concentration-dependent manner. NUC1 also inhibited the release and protein expression of MMP-1, 3, and 13, in TNF-α-induced chondrosarcoma cells in a concentration-dependent manner. In vivo, the MMP-1 and MMP-3 levels in synovial fluids were significantly (p < 0.05) lower in NUC1 group (77.50 ± 20.56 and 22.50 ± 7.39 pg/ml, respectively) than in the CIA group (148.33 ± 68.58 and 77.50 ± 20.46 pg/ml, respectively). Also, in histopathological, NUC1 ameliorated articular cartilage damage in OA by increasing the abundance of chondrocytes and proteoglycan in the articular cartilage. Thus, NUC1 showed promise as a potential therapeutic agent, and it can be generalized to a broader study population in different OA animal models.
Collapse
Affiliation(s)
- Sung-Gyu Lee
- Department of Medical Laboratory Science, College of Health Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Hyun Kang
- Department of Medical Laboratory Science, College of Health Science, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
23
|
Lee T, Na CB, Kim D, Han HJ, Yun J, Park SK, Cho E. Osteoarthritis improvement effect of Chrysanthemum zawadskii var. latilobum extract in relation to genotype. INT J VITAM NUTR RES 2023; 93:410-419. [PMID: 35023382 DOI: 10.1024/0300-9831/a000745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Objectives: To determine whether SNPs of osteoarthritis (OA)-related genes predict the effect of Chrysanthemum zawadskii var. latilobum (CZ) extract in OA patients with OA. Subjects/methods: To analyze correlations between CZ extract effects in humans and their genotypes, 121 Korean patients with OA were recruited. Patients ingested 600 mg/day of the CZ extract GCWB106 (one tablet daily), including 250-mg CZ, or placebo (one tablet daily) for 12 weeks. Twenty SNPs were genotyped in 11 genes associated with OA pathogenesis, including tumor necrosis factor-alpha (TNF-α) and matrix metalloproteinases (MMPs), and 9 genes involved in OA-related dietary intervention. The Visual Analogue Scale (VAS) and Korean Western Ontario and McMaster Universities (K-WOMAC) were measured as indicators of GCWB106 effect. Statistical comparisons were performed using Kruskal-Wallis tests to identify associations between these scales and genotyped loci in patients with OA. Results: Three SNPs (PPARG rs3856806, MMP13 rs2252070, and ZIP2 rs2234632) were significantly associated with the degree of change in VAS pain score. Homozygous CC genotype carriers of rs3856806, G allele carriers (GA or GG) of rs2252070, and T allele carriers (GT or TT) of rs2234632 showed lower VAS score (i.e., less severe symptoms) in the GCWB106 group (n=53) than the placebo group (n=57) (p=0.026, p=0.009, and p=0.025, respectively). Gene-gene interaction effects on GCWB106-mediated pain relief were then examined, and it was found that the addition of each genotype resulted in a greater decrease in VAS pain score in the GCWB106 group (p=0.0024) but not the placebo group (p=0.7734). Conclusions: These novel predictive markers for the pain-relieving effects of GCWB106 may be used in the personalized treatment of patients with OA.
Collapse
Affiliation(s)
- Taeheon Lee
- Precision Medicine R&BD Center, GC Genome, Yongin, Korea
| | - Chae-Bin Na
- Genome Research Center, GC Genome, Yongin, Korea
| | - Dasom Kim
- Genome Research Center, GC Genome, Yongin, Korea
| | - Hae Jung Han
- Development Division, Green Cross WellBeing, Seoul, Korea
| | - Jongbok Yun
- Development Division, Green Cross WellBeing, Seoul, Korea
| | - Sun Kyu Park
- Development Division, Green Cross WellBeing, Seoul, Korea
| | - Eunhae Cho
- Genome Research Center, GC Genome, Yongin, Korea
| |
Collapse
|
24
|
Zhao T, Li X, Li H, Deng H, Li J, Yang Z, He S, Jiang S, Sui X, Guo Q, Liu S. Advancing drug delivery to articular cartilage: From single to multiple strategies. Acta Pharm Sin B 2023; 13:4127-4148. [PMID: 37799383 PMCID: PMC10547919 DOI: 10.1016/j.apsb.2022.11.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022] Open
Abstract
Articular cartilage (AC) injuries often lead to cartilage degeneration and may ultimately result in osteoarthritis (OA) due to the limited self-repair ability. To date, numerous intra-articular delivery systems carrying various therapeutic agents have been developed to improve therapeutic localization and retention, optimize controlled drug release profiles and target different pathological processes. Due to the complex and multifactorial characteristics of cartilage injury pathology and heterogeneity of the cartilage structure deposited within a dense matrix, delivery systems loaded with a single therapeutic agent are hindered from reaching multiple targets in a spatiotemporal matched manner and thus fail to mimic the natural processes of biosynthesis, compromising the goal of full cartilage regeneration. Emerging evidence highlights the importance of sequential delivery strategies targeting multiple pathological processes. In this review, we first summarize the current status and progress achieved in single-drug delivery strategies for the treatment of AC diseases. Subsequently, we focus mainly on advances in multiple drug delivery applications, including sequential release formulations targeting various pathological processes, synergistic targeting of the same pathological process, the spatial distribution in multiple tissues, and heterogeneous regeneration. We hope that this review will inspire the rational design of intra-articular drug delivery systems (DDSs) in the future.
Collapse
Affiliation(s)
- Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Hao Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Haoyuan Deng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jianwei Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhen Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China
| | - Songlin He
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuangpeng Jiang
- Department of Joint Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
25
|
Abstract
Collagen provides mechanical and biological support for virtually all human tissues in the extracellular matrix (ECM). Its defining molecular structure, the triple-helix, could be damaged and denatured in disease and injuries. To probe collagen damage, the concept of collagen hybridization has been proposed, revised, and validated through a series of investigations reported as early as 1973: a collagen-mimicking peptide strand may form a hybrid triple-helix with the denatured chains of natural collagen but not the intact triple-helical collagen proteins, enabling assessment of proteolytic degradation or mechanical disruption to collagen within a tissue-of-interest. Here we describe the concept and development of collagen hybridization, summarize the decades of chemical investigations on rules underlying the collagen triple-helix folding, and discuss the growing biomedical evidence on collagen denaturation as a previously overlooked ECM signature for an array of conditions involving pathological tissue remodeling and mechanical injuries. Finally, we propose a series of emerging questions regarding the chemical and biological nature of collagen denaturation and highlight the diagnostic and therapeutic opportunities from its targeting.
Collapse
Affiliation(s)
- Xiaojing Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Department of Radiology, Cardiac Surgery and Structural Heart Disease Unit of Cardiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Qi Zhang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Department of Radiology, Cardiac Surgery and Structural Heart Disease Unit of Cardiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - S. Michael Yu
- Department of Biomedical Engineering, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Yang Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Department of Radiology, Cardiac Surgery and Structural Heart Disease Unit of Cardiovascular Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| |
Collapse
|
26
|
Taninaka A, Kabata T, Hayashi K, Kajino Y, Inoue D, Ohmori T, Ueoka K, Yamamuro Y, Kataoka T, Saiki Y, Yanagi Y, Ima M, Iyobe T, Tsuchiya H. Chondroprotective Effects of Chondrogenic Differentiated Adipose-Derived Mesenchymal Stem Cells Sheet on Degenerated Articular Cartilage in an Experimental Rabbit Model. Bioengineering (Basel) 2023; 10:bioengineering10050574. [PMID: 37237645 DOI: 10.3390/bioengineering10050574] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) have been studied for many years as a therapeutic option for osteoarthritis (OA); however, their efficacy remains insufficient. Since platelet-rich plasma (PRP) induces chondrogenic differentiation in ADSCs and the formation of a sheet structure by ascorbic acid can increase the number of viable cells, we hypothesized that the injection of chondrogenic cell sheets combined with the effects of PRP and ascorbic acid may hinder the progression of OA. The effects of induction of differentiation by PRP and formation of sheet structure by ascorbic acid on changes in chondrocyte markers (collagen II, aggrecan, Sox9) in ADSCs were evaluated. Changes in mucopolysaccharide and VEGF-A secretion from cells injected intra-articularly in a rabbit OA model were also evaluated. ADSCs treated by PRP strongly chondrocyte markers, including type II collagen, Sox9, and aggrecan, and their gene expression was maintained even after sheet-like structure formation induced by ascorbic acid. In this rabbit OA model study, the inhibition of OA progression by intra-articular injection was improved by inducing chondrocyte differentiation with PRP and sheet structure formation with ascorbic acid in ADSCs.
Collapse
Affiliation(s)
- Atsushi Taninaka
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Tamon Kabata
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yoshitomo Kajino
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Daisuke Inoue
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Takaaki Ohmori
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Ken Ueoka
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yuki Yamamuro
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Tomoyuki Kataoka
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yoshitomo Saiki
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yu Yanagi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Musashi Ima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Takahiro Iyobe
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| |
Collapse
|
27
|
Grillet B, Pereira RVS, Van Damme J, Abu El-Asrar A, Proost P, Opdenakker G. Matrix metalloproteinases in arthritis: towards precision medicine. Nat Rev Rheumatol 2023; 19:363-377. [PMID: 37161083 DOI: 10.1038/s41584-023-00966-w] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2023] [Indexed: 05/11/2023]
Abstract
Proteolysis of structural molecules of the extracellular matrix (ECM) is an irreversible post-translational modification in all arthropathies. Common joint disorders, including osteoarthritis and rheumatoid arthritis, have been associated with increased levels of matrix remodelling enzymes, including matrix metalloproteinases (MMPs). MMPs, in concert with other host proteinases and glycanases, destroy proteoglycans, collagens and other ECM molecules. MMPs may also control joint remodelling indirectly by signalling through cell-surface receptors or by proteolysis of cytokines and receptor molecules. After synthesis as pro-forms, MMPs can be activated by various types of post-translational modifications, including proteolysis. Once activated, MMPs are controlled by general and specific tissue inhibitors of metalloproteinases (TIMPs). In rheumatoid arthritis, proteolysis of the ECM results in so-called remnant epitopes that enhance and perpetuate autoimmune processes in susceptible hosts. In osteoarthritis, the considerable production of MMP-13 by chondrocytes, often concurrent with mechanical overload, is a key event. Hence, information about the regulation, timing, localization and activities of MMPs in specific disease phases and arthritic entities will help to develop better diagnostics. Insights into beneficial and detrimental effects of MMPs on joint tissue inflammation are also necessary to plan and execute (pre)clinical studies for better therapy and precision medicine with MMP inhibitors. With the advances in proteomics and single-cell transcriptomics, two critical points need attention: neglected neutrophil MMP biology, and the analysis of net proteolytic activities as the result of balances between MMPs and their inhibitors.
Collapse
Affiliation(s)
- Bernard Grillet
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Rafaela Vaz Sousa Pereira
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Ahmed Abu El-Asrar
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Ophthalmology, King Saud University, Riyadh, Saudi Arabia
| | - Paul Proost
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.
- Department of Ophthalmology, King Saud University, Riyadh, Saudi Arabia.
- University Hospitals Gasthuisberg, UZ Leuven, KU Leuven, Leuven, Belgium.
| |
Collapse
|
28
|
Walsh C, Rajora MA, Ding L, Nakamura S, Endisha H, Rockel J, Chen J, Kapoor M, Zheng G. Protease-Activatable Porphyrin Molecular Beacon for Osteoarthritis Management. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:66-80. [PMID: 37122828 PMCID: PMC10131263 DOI: 10.1021/cbmi.3c00005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/06/2023] [Accepted: 02/12/2023] [Indexed: 05/02/2023]
Abstract
Despite the substantial burden posed by osteoarthritis (OA) globally, difficult challenges remain in achieving early OA diagnosis and adopting effective disease-modifying treatments. In this study, we use a biomolecular approach to address these limitations by creating an inherently theranostic molecular beacon whose imaging and therapeutic capabilities are activated by early pathological changes in OA. This platform comprised (1) a peptide linker substrate for metalloproteinase-13 (MMP-13), a pathological protease upregulated in OA, which was conjugated to (2) a porphyrin moiety with inherent multimodal imaging, photodynamic therapy, and drug delivery capabilities, and (3) a quencher that silences the porphyrin's endogenous fluorescence and photoreactivity when the beacon is intact. In diseased OA tissue with upregulated MMP-13 expression, this porphyrin molecular beacon (PPMMP13B) was expected to undergo sequence-specific cleavage, yielding porphyrin fragments with restored fluorescence and photoreactivity that could, respectively, be used as a readout of MMP-13 activity within the joint for early OA imaging and disease-targeted photodynamic therapy. This study focused on the synthesis and characterization of PPMMP13B, followed by a proof-of-concept evaluation of its OA imaging and drug delivery potential. In solution, PPMMP13B demonstrated 90% photoactivity quenching in its intact form and robust MMP-13 activation, yielding a 13-fold increase in fluorescence post-cleavage. In vitro, PPMMP13B was readily uptaken and activated in an MMP-13 cell expression-dependent manner in primary OA synoviocytes without exuding significant cytotoxicity. This translated into effective intra-articular cartilage (to a 50 μm depth) and synovial uptake and activation of PPMMP13B in a destabilization of the medial meniscus OA mouse model, yielding strong fluorescence contrast (7-fold higher signal than background) at the diseased joint site. These results provide the foundation for further exploration of porphyrin molecular beacons for image-guided OA disease stratification, effective articular delivery of disease-modify agents, and OA photodynamic therapy.
Collapse
Affiliation(s)
- Connor Walsh
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
| | - Maneesha A. Rajora
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
| | - Lili Ding
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Sayaka Nakamura
- Schroeder
Arthritis Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Krembil
Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Helal Endisha
- Schroeder
Arthritis Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Krembil
Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Jason Rockel
- Schroeder
Arthritis Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Krembil
Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Juan Chen
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Mohit Kapoor
- Schroeder
Arthritis Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Krembil
Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Gang Zheng
- Princess
Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
29
|
Zhu Y, Zhang C, Jiang B, Dong Q. MiR-760 targets HBEGF to control cartilage extracellular matrix degradation in osteoarthritis. J Orthop Surg Res 2023; 18:186. [PMID: 36894989 PMCID: PMC9999495 DOI: 10.1186/s13018-023-03664-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
The present study was developed to explore whether microRNA (miR)-760 targets heparin-binding EGF-like growth factor (HBEGF) to control cartilage extracellular matrix degradation in osteoarthritis. Both miR-760 and HBEGF expression levels were analysed in human degenerative cartilage tissues and in interleukin (IL)-1β/tumour necrosis factor (TNF)-α-treated chondrocytes in vitro. A series of knockdown and overexpression assays were then used to gauge the functional importance of miR-760 and HBEGF in OA, with qPCR and western immunoblotting analyses. Bioinformatics assays were used to identify putative miR-760 target genes, with these predictions then being validated through RNA pulldown and luciferase reporter assays. A murine anterior cruciate ligament transection model of OA was then established to prove the in vivo relevance of these findings. These experiments revealed that human degenerative cartilage tissues exhibited significant increases in miR-760 expression with a concomitant drop in HBEGF levels. IL-1β/TNF-α-treated chondrocytes also exhibited significant increases in miR-760 expression with a concomitant drop in HBEGF expression. When chondrocytes were transfected with either miR-760 inhibitor or HBEGF overexpression constructs, this was sufficient to interfere with degradation of the extracellular matrix (ECM). Moreover, miR-760 was confirmed to control chondrocyte matrix homeostasis by targeting HBEGF, and the overexpression of HBEGF partially reversed the effects of miR-760 mimic treatment on the degradation of the cartilage ECM. When OA model mice were administered an intra-articular knee injection of an adenoviral vector encoding a miR-760 mimic construct, cartilage ECM degradation was aggravated. Conversely, the overexpression of HBEGF in OA model mice partially reversed the effects of miR-760 overexpression, restoring appropriate ECM homeostasis. In summary, these data indicated that the miR-760/HBEGF axis plays a central role in orchestrating the pathogenesis of OA, making it a candidate target for therapeutic efforts in OA.
Collapse
Affiliation(s)
- Yingchun Zhu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Soochow University, Soochow, 215000, China
| | - Chi Zhang
- Department of Orthopedic Surgery, Ningbo First Hospital, No. 59, LiuTing Street, Ningbo, 315010, China
| | - Bo Jiang
- Department of Orthopedic Surgery, Ningbo First Hospital, No. 59, LiuTing Street, Ningbo, 315010, China
| | - Qirong Dong
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Soochow University, Soochow, 215000, China.
| |
Collapse
|
30
|
Fowkes MM, Troeberg L, Brennan PE, Vincent TL, Meldal M, Lim NH. Development of Selective ADAMTS-5 Peptide Substrates to Monitor Proteinase Activity. J Med Chem 2023; 66:3522-3539. [PMID: 36891740 PMCID: PMC10009750 DOI: 10.1021/acs.jmedchem.2c02090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Indexed: 02/25/2023]
Abstract
The dysregulation of proteinase activity is a hallmark of osteoarthritis (OA), a disease characterized by progressive degradation of articular cartilage by catabolic proteinases such as a disintegrin and metalloproteinase with thrombospondin type I motifs-5 (ADAMTS-5). The ability to detect such activity sensitively would aid disease diagnosis and the evaluation of targeted therapies. Förster resonance energy transfer (FRET) peptide substrates can detect and monitor disease-related proteinase activity. To date, FRET probes for detecting ADAMTS-5 activity are nonselective and relatively insensitive. We describe the development of rapidly cleaved and highly selective ADAMTS-5 FRET peptide substrates through in silico docking and combinatorial chemistry. The lead substrates 3 and 26 showed higher overall cleavage rates (∼3-4-fold) and catalytic efficiencies (∼1.5-2-fold) compared to the best current ADAMTS-5 substrate ortho-aminobenzoyl(Abz)-TESE↓SRGAIY-N-3-[2,4-dinitrophenyl]-l-2,3-diaminopropionyl(Dpa)-KK-NH2. They exhibited high selectivity for ADAMTS-5 over ADAMTS-4 (∼13-16-fold), MMP-2 (∼8-10-fold), and MMP-9 (∼548-2561-fold) and detected low nanomolar concentrations of ADAMTS-5.
Collapse
Affiliation(s)
- Milan M. Fowkes
- Centre
for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - Linda Troeberg
- Norwich
Medical School, Bob Champion Research and Education Building, Rosalind
Franklin Road, University of East Anglia, Norwich NR4 7UQ, United Kingdom
| | - Paul E. Brennan
- Alzheimer’s
Research UK Oxford Drug Discovery Institute, Centre for Medicines
Discovery, Nuffield Department of Medicine Research Building, University of Oxford, Old Road Campus, Headington, Oxford OX3 7FZ, United Kingdom
| | - Tonia L. Vincent
- Centre
for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - Morten Meldal
- Department
of Chemistry, University of Copenhagen, Universitetsparken 5, Building B304, Copenhagen DK-2100, Denmark
| | - Ngee H. Lim
- Centre
for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
31
|
Kleuskens MWA, Crispim JF, van Doeselaar M, van Donkelaar CC, Janssen RPA, Ito K. Neo-cartilage formation using human nondegenerate versus osteoarthritic chondrocyte-derived cartilage organoids in a viscoelastic hydrogel. J Orthop Res 2023. [PMID: 36866819 DOI: 10.1002/jor.25540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 03/04/2023]
Abstract
Current regenerative cartilage therapies are associated with several drawbacks such as dedifferentiation of chondrocytes during expansion and the formation of fibrocartilage. Optimized chondrocyte expansion and tissue formation could lead to better clinical results of these therapies. In this study, a novel chondrocyte suspension expansion protocol that includes the addition of porcine notochordal cell-derived matrix was used to self-assemble human chondrocytes from osteoarthritic (OA) and nondegenerate (ND) origin into cartilage organoids containing collagen type II and proteoglycans. Proliferation rate and viability were similar for OA and ND chondrocytes and organoids formed had a similar histologic appearance and gene expression profile. Organoids were then encapsulated in viscoelastic alginate hydrogels to form larger tissues. Chondrocytes on the outer bounds of the organoids produced a proteoglycan-rich matrix to bridge the space between organoids. In hydrogels containing ND organoids some collagen type I was observed between the organoids. Surrounding the bulk of organoids in the center of the gels, in both OA and ND gels a continuous tissue containing cells, proteoglycans and collagen type II had been produced. No difference was observed in sulphated glycosaminoglycan and hydroxyproline content between gels containing organoids from OA or ND origin after 28 days. It was concluded that OA chondrocytes, which can be harvested from leftover surgery tissue, perform similar to ND chondrocytes in terms of human cartilage organoid formation and matrix production in alginate gels. This opens possibilities for their potential to serve as a platform for cartilage regeneration but also as an in vitro model to study pathways, pathology, or drug development.
Collapse
Affiliation(s)
- Meike W A Kleuskens
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - João F Crispim
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Marina van Doeselaar
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Corrinus C van Donkelaar
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rob P A Janssen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Orthopaedic Surgery and Trauma, Máxima Medical Center, Eindhoven-Veldhoven, The Netherlands.,Department of Paramedical Sciences, Fontys University of Applied Sciences, Eindhoven, The Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
32
|
Rapp AE, Zaucke F. Cartilage extracellular matrix-derived matrikines in osteoarthritis. Am J Physiol Cell Physiol 2023; 324:C377-C394. [PMID: 36571440 DOI: 10.1152/ajpcell.00464.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Osteoarthritis (OA) is among the most frequent diseases of the musculoskeletal system. Degradation of cartilage extracellular matrix (ECM) is a hallmark of OA. During the degradation process, intact/full-length proteins and proteolytic fragments are released which then might induce different downstream responses via diverse receptors, therefore leading to different biological consequences. Collagen type II and the proteoglycan aggrecan are the most abundant components of the cartilage ECM. However, over the last decades, a large number of minor components have been identified and for some of those, a role in the manifold processes associated with OA has already been demonstrated. To date, there is still no therapy able to halt or cure OA. A better understanding of the matrikine landscape occurring with or even preceding obvious degenerative changes in joint tissues is needed and might help to identify molecules that could serve as biomarkers, druggable targets, or even be blueprints for disease modifying drug OA drugs. For this narrative review, we screened PubMed for relevant literature in the English language and summarized the current knowledge regarding the function of selected ECM molecules and the derived matrikines in the context of cartilage and OA.
Collapse
Affiliation(s)
- Anna E Rapp
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
33
|
Bradley PX, Thomas KN, Kratzer AL, Robinson AC, Wittstein JR, DeFrate LE, McNulty AL. The Interplay of Biomechanical and Biological Changes Following Meniscus Injury. Curr Rheumatol Rep 2023; 25:35-46. [PMID: 36479669 PMCID: PMC10267895 DOI: 10.1007/s11926-022-01093-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Meniscus injury often leads to joint degeneration and post-traumatic osteoarthritis (PTOA) development. Therefore, the purpose of this review is to outline the current understanding of biomechanical and biological repercussions following meniscus injury and how these changes impact meniscus repair and PTOA development. Moreover, we identify key gaps in knowledge that must be further investigated to improve meniscus healing and prevent PTOA. RECENT FINDINGS Following meniscus injury, both biomechanical and biological alterations frequently occur in multiple tissues in the joint. Biomechanically, meniscus tears compromise the ability of the meniscus to transfer load in the joint, making the cartilage more vulnerable to increased strain. Biologically, the post-injury environment is often characterized by an increase in pro-inflammatory cytokines, catabolic enzymes, and immune cells. These multi-faceted changes have a significant interplay and result in an environment that opposes tissue repair and contributes to PTOA development. Additionally, degenerative changes associated with OA may cause a feedback cycle, negatively impacting the healing capacity of the meniscus. Strides have been made towards understanding post-injury biological and biomechanical changes in the joint, their interplay, and how they affect healing and PTOA development. However, in order to improve clinical treatments to promote meniscus healing and prevent PTOA development, there is an urgent need to understand the physiologic changes in the joint following injury. In particular, work is needed on the in vivo characterization of the temporal biomechanical and biological changes that occur in patients following meniscus injury and how these changes contribute to PTOA development.
Collapse
Affiliation(s)
- Patrick X Bradley
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Karl N Thomas
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Avery L Kratzer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Allison C Robinson
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Jocelyn R Wittstein
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Louis E DeFrate
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Amy L McNulty
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA.
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
34
|
Gremese E, Tolusso B, Bruno D, Perniola S, Ferraccioli G, Alivernini S. The forgotten key players in rheumatoid arthritis: IL-8 and IL-17 - Unmet needs and therapeutic perspectives. Front Med (Lausanne) 2023; 10:956127. [PMID: 37035302 PMCID: PMC10073515 DOI: 10.3389/fmed.2023.956127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 02/21/2023] [Indexed: 04/11/2023] Open
Abstract
Despite the relevant advances in our understanding of the pathogenetic mechanisms regulating inflammation in rheumatoid arthritis (RA) and the development of effective therapeutics, to date, there is still a proportion of patients with RA who do not respond to treatment and end up progressing toward the development of joint damage, extra-articular complications, and disability. This is mainly due to the inter-individual heterogeneity of the molecular and cellular taxonomy of the synovial membrane, which represents the target tissue of RA inflammation. Tumor necrosis factor alpha (TNFα) and interleukin-6 (IL-6) are crucial key players in RA pathogenesis fueling the inflammatory cascade, as supported by experimental evidence derived from in vivo animal models and the effectiveness of biologic-Disease Modifying Anti-Rheumatic Drugs (b-DMARDs) in patients with RA. However, additional inflammatory soluble mediators such as IL-8 and IL-17 exert their pathogenetic actions promoting the detrimental activation of immune and stromal cells in RA synovial membrane, tendons, and extra-articular sites, as well as blood vessels and lungs, causing extra-articular complications, which might be excluded by the action of anti-TNFα and anti-IL6R targeted therapies. In this narrative review, we will discuss the role of IL-8 and IL-17 in promoting inflammation in multiple biological compartments (i.e., synovial membrane, blood vessels, and lung, respectively) in animal models of arthritis and patients with RA and how their selective targeting could improve the management of treatment resistance in patients.
Collapse
Affiliation(s)
- Elisa Gremese
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Immunology Core Facility, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
- *Correspondence: Elisa Gremese, Gianfranco Ferraccioli
| | - Barbara Tolusso
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Immunology Core Facility, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Dario Bruno
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Simone Perniola
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Gianfranco Ferraccioli
- School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
- *Correspondence: Elisa Gremese, Gianfranco Ferraccioli
| | - Stefano Alivernini
- Immunology Core Facility, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| |
Collapse
|
35
|
Baltzer AW, Casadonte R, Korff A, Baltzer LM, Kriegsmann K, Kriegsmann M, Kriegsmann J. Biological injection therapy with leukocyte-poor platelet-rich plasma induces cellular alterations, enhancement of lubricin, and inflammatory downregulation in vivo in human knees: A controlled, prospective human clinical trial based on mass spectrometry imaging analysis. Front Surg 2023; 10:1169112. [PMID: 37151865 PMCID: PMC10160617 DOI: 10.3389/fsurg.2023.1169112] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/27/2023] [Indexed: 05/09/2023] Open
Abstract
Objective To investigate the in vivo biological effects of leukocyte-poor platelet-rich plasma (LpPRP) treatment in human synovial layer to establish the cellular basis for a prolonged clinical improvement. Methods Synovial tissues (n = 367) were prospectively collected from patients undergoing arthroscopic surgery. Autologous-conditioned plasma, LpPRP, was injected into the knees of 163 patients 1-7 days before surgery to reduce operative trauma and inflammation, and to induce the onset of regeneration. A total of 204 patients did not receive any injection. All samples were analyzed by mass spectrometry imaging. Data analysis was evaluated by clustering, classification, and investigation of predictive peptides. Peptide identification was done by tandem mass spectrometry and database matching. Results Data analysis revealed two major clusters belonging to LpPRP-treated (LpPRP-1) and untreated (LpPRP-0) patients. Classification analysis showed a discrimination accuracy of 82%-90%. We identified discriminating peptides for CD45 and CD29 receptors (receptor-type tyrosine-protein phosphatase C and integrin beta 1), indicating an enhancement of musculoskeletal stem cells, as well as an enhancement of lubricin, collagen alpha-1-(I) chain, and interleukin-receptor-17-E, dampening the inflammatory reaction in the LpPRP-1 group following LpPRP injection. Conclusions We could demonstrate for the first time that injection therapy using "autologic-conditioned biologics" may lead to cellular changes in the synovial membrane that might explain the reported prolonged beneficial clinical effects. Here, we show in vivo cellular changes, possibly based on muscular skeletal stem cell alterations, in the synovial layer. The gliding capacities of joints might be improved by enhancing of lubricin, anti-inflammation by activation of interleukin-17 receptor E, and reduction of the inflammatory process by blocking interleukin-17.
Collapse
Affiliation(s)
- Axel W. Baltzer
- Center for Molecular Orthopaedics, MVZ Ortho Koenigsallee, Düsseldorf, Germany
- Correspondence: Axel W. Baltzer
| | - Rita Casadonte
- Imaging Mass Spectrometry, Proteopath GmbH, Trier, Germany
| | - Alexei Korff
- Center for Molecular Orthopaedics, MVZ Ortho Koenigsallee, Düsseldorf, Germany
| | | | - Katharina Kriegsmann
- Department for Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Germany Translational Lung Research Centre Heidelberg, Member of the German Centre for Lung Research (DZL), Heidelberg, Germany
| | - Jörg Kriegsmann
- Imaging Mass Spectrometry, Proteopath GmbH, Trier, Germany
- MVZ-Zentrum für Histologie, Zytologie und Molekulare Diagnostik, Trier, Germany
- Department of Medicine, Faculty of Medicine/Dentistry, Danube Private University, Krems, Austria
| |
Collapse
|
36
|
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: 31] [Impact Index Per Article: 10.3] [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
|
37
|
Gomathi K, Rohini M, Vairamani M, Selvamurugan N. Identification and characterization of TGF-β1-responsive Runx2 acetylation sites for matrix Metalloproteinase-13 expression in osteoblastic cells. Biochimie 2022; 201:1-6. [PMID: 35779648 DOI: 10.1016/j.biochi.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/26/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022]
Abstract
In skeletal tissues, transforming growth factor-beta 1 (TGF-β1) serves a number of activities. For example, in osteoblastic cells, TGF-β1 stimulates the expression of matrix metalloproteinase-13 (MMP-13, a bone remodeling gene), which requires the bone transcription factor Runx2. Although TGF-β1 is known to stimulate Runx2 acetylation, the sites involved in MMP-13 gene activation remain unknown. Mass spectrometry analysis revealed that Runx2 was acetylated at one site (K134) and three sites (K24, K134, and K169) following control and TGF-β1-treatment, respectively, in osteoblastic cells. In addition, we mutated the lysine residues in the Runx2 construct into arginine and transfected the construct into mouse mesenchymal stem cells (C3H10T1/2). Wild-type Runx2 expression and acetylation were significantly increased by TGF-β1-treatment, whereas this effect was decreased in the presence of the Runx2 double mutant construct (K24 + K169) in C3H10T1/2 cells. TGF-β1 enhanced MMP-13 promoter activity in cells transfected with the wild-type Runx2 construct, but this effect was considerably reduced in cells transfected with the Runx2 double mutant construct (K24 + K169), according to a luciferase reporter test. Hence, the stability of Runx2 may be mediated by TGF-β1-induced acetylation at K24 and K169 and is required for MMP-13 expression in osteoblastic cells. These findings add to our knowledge of TGF-β1, Runx2, and MMP-13's physiological roles in bone metabolism.
Collapse
Affiliation(s)
- Kanagaraj Gomathi
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Muthukumar Rohini
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Mariappan Vairamani
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India.
| |
Collapse
|
38
|
Jaabar IL, Cornette P, Miche A, Wanherdrick K, Dupres V, Ehkirch FP, Cambon Binder A, Berenbaum F, Houard X, Landoulsi J. Deciphering pathological remodelling of the human cartilage extracellular matrix in osteoarthritis at the supramolecular level. NANOSCALE 2022; 14:8691-8708. [PMID: 35673929 DOI: 10.1039/d2nr00474g] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The extracellular matrix (ECM) of articular cartilage is a three-dimensional network mainly constituted of entangled collagen fibrils and interfibrillar aggrecan aggregates. During the development of osteoarthritis (OA), the most common musculoskeletal disorder, the ECM is subjected to a combination of chemical and structural changes that play a pivotal role in the initiation and the progress of the disease. While the molecular mechanisms involved in the pathological remodelling of the ECM are considered as decisive, they remain, however, not completely elucidated. Herein, we report a relevant way for unravelling the role and nature of OA progress on human cartilage tissues, in terms of chemical composition and morphological and mechanical properties at the level of supramolecular assemblies constituting the cartilage ECM. For this purpose, we used X-ray photoelectron spectroscopy (XPS), and developed an innovative methodological approach that provides the molecular composition of the ECM. Moreover, we used atomic force microscopy (AFM) to probe the tissues at the level of individual collagen fibrils, both imaging and force spectroscopy modes being explored to this end. Taken together, these nanoscale characterization studies reveal the existence of two stages in the OA progress. At the early stage, a marked increase in the aggrecan and collagen content is observed, reflecting the homeostatic chondrocyte activity that tends to repair the cartilage ECM. At the late stage, we observe a failed attempt to stabilize and/or restore the tissue, yielding significant degradation of the supramolecular assemblies. This suggests an imbalance in the chondrocyte activity that turns in favor of catabolic events. Chemical changes are also accompanied by ECM structural changes and stiffening. Interestingly, we showed the possibility to mimic the imbalanced activities of chondrocytes by applying enzymatic digestions of healthy cartilage, through the combined action of hyaluronidase and collagenase. This yields damage strictly analogous to that observed at high OA severity. These findings bring mechanistic insights leading to a better understanding of the mechanism by which OA is initiated and progresses in the cartilage ECM. They offer guidelines for the development of curative treatments, such as targeting the homeostatic balance of chondrocyte metabolism through the control of enzymatic reactions involved in catabolic processes.
Collapse
Affiliation(s)
- Ilhem Lilia Jaabar
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Pauline Cornette
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Antoine Miche
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
| | - Kristell Wanherdrick
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Vincent Dupres
- Université Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - François-Paul Ehkirch
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Clinique Maussins-Nollet, F-75019 Paris, France
| | - Adeline Cambon Binder
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Orthopedics and Hand Surgery Department, Saint-Antoine Hospital, 184 Rue du Faubourg Saint-Antoine, Paris, 75012, France
| | - Francis Berenbaum
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Rheumatology Department, AP-HP Saint-Antoine Hospital, 184, rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Xavier Houard
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Jessem Landoulsi
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
| |
Collapse
|
39
|
Vincent TL, Alliston T, Kapoor M, Loeser RF, Troeberg L, Little CB. Osteoarthritis Pathophysiology: Therapeutic Target Discovery may Require a Multifaceted Approach. Clin Geriatr Med 2022; 38:193-219. [PMID: 35410676 PMCID: PMC9107912 DOI: 10.1016/j.cger.2021.11.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Molecular understanding of osteoarthritis (OA) has greatly increased through careful analysis of tissue samples, preclinical models, and large-scale agnostic "-omic" studies. There is broad acceptance that systemic and biomechanical signals affect multiple tissues of the joint, each of which could potentially be targeted to improve patient outcomes. In this review six experts in different aspects of OA pathogenesis provide their independent view on what they believe to be good tractable approaches to OA target discovery. We conclude that molecular discovery has been high but future transformative studies require a multidisciplinary holistic approach to develop therapeutic strategies with high clinical efficacy.
Collapse
Affiliation(s)
- Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mohit Kapoor
- Department of Surgery and Laboratory Medicine and Pathobiology, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, University of Toronto, Toronto, Canada
| | - Richard F Loeser
- Department of Medicine, Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Linda Troeberg
- University of East Anglia, Norwich Medical School, Norwich NR4 7UQ, UK
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute University of Sydney Faculty of Medicine and Health at Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| |
Collapse
|
40
|
Do Synovial Inflammation and Meniscal Degeneration Impact Clinical Outcomes of Patients Undergoing Arthroscopic Partial Meniscectomy? A Histological Study. Int J Mol Sci 2022; 23:ijms23073903. [PMID: 35409262 PMCID: PMC8999499 DOI: 10.3390/ijms23073903] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 01/14/2023] Open
Abstract
The menisci exert a prominent role in joint stabilization and in the distribution of mechanical loading. Meniscal damage is associated with increased risk of knee OA. The aim of this study was to characterize the synovial membrane and meniscal tissues in patients undergoing arthroscopic partial meniscectomy for meniscal tear and to evaluate association with clinical outcomes. A total of 109 patients were recruited. Demographic and clinical data were collected. Visual Analogic Scale (VAS) measuring pain and Knee injury and Osteoarthritis Outcome Score (KOOS) were recorded at baseline and at 2-years follow-up. Histological and immunohistochemical characterizations were performed on synovial membranes and meniscal tissues. More than half of the patients demonstrated synovial mononuclear cell infiltration and hyperplasia. Synovial fibrosis was present in most of the patients; marked vascularity and CD68 positivity were observed. Inflammation had an impact on both pain and knee symptoms. Patients with synovial inflammation had higher values of pre-operative VAS and inflammation. Higher pre-operative pain was observed in patients with meniscal MMP-13 production. In conclusion, multivariate analysis showed that synovial inflammation was associated with pre-operative total KOOS scores, knee symptoms, and pain. Moreover, meniscal MMP-13 expression was found to be associated with pre-operative pain in multivariate analysis. Thus, targeting inflammation of the synovial membrane and meniscus might reduce clinical symptoms and dysfunction at the time of surgery.
Collapse
|
41
|
Zhang W, Zheng X, Gong Y, Jiang T, Qiu J, Wu X, Lu F, Wang Z, Hong Z. VX-11e protects articular cartilage and subchondral bone in osteoarthritis by inhibiting the RIP1/RIP3/MLKL and MAPK signaling pathways. Bioorg Chem 2022; 120:105632. [DOI: 10.1016/j.bioorg.2022.105632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/16/2022] [Indexed: 12/18/2022]
|
42
|
Gomathi K, Rohini M, Partridge NC, Selvamurugan N. Regulation of transforming growth factor-β1-stimulation of Runx2 acetylation for matrix metalloproteinase 13 expression in osteoblastic cells. Biol Chem 2022; 403:305-315. [PMID: 34643076 DOI: 10.1515/hsz-2021-0292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/30/2021] [Indexed: 01/12/2023]
Abstract
Transforming growth factor beta 1 (TGF-β1) functions as a coupling factor between bone development and resorption. Matrix metalloproteinase 13 (MMP13) is important in bone remodeling, and skeletal dysplasia is caused by a deficiency in MMP13 expre-ssion. Runx2, a transcription factor is essential for bone development, and MMP13 is one of its target genes. TGF-β1 promoted Runx2 phosphorylation, which was necessary for MMP13 production in osteoblastic cells, as we previously shown. Since the phosphorylation of some proteins causes them to be degraded by the ubiquitin/proteasome pathway, we hypothesized that TGF-β1 might stabilize the phosphorylated Runx2 protein for its activity by other post-translational modification (PTM). This study demonstrated that TGF-β1-stimulated Runx2 acetylation in rat osteoblastic cells. p300, a histone acetyltransferase interacted with Runx2, and it promoted Runx2 acetylation upon TGF-β1-treatment in these cells. Knockdown of p300 decreased the TGF-β1-stimulated Runx2 acetylation and MMP13 expression in rat osteoblastic cells. TGF-β1-treatment stimulated the acetylated Runx2 bound at the MMP13 promoter, and knockdown of p300 reduced this effect in these cells. Overall, our studies identified the transcriptional regulation of MMP13 by TGF-β1 via Runx2 acetylation in rat osteoblastic cells, and these findings contribute to the knowledge of events presiding bone metabolism.
Collapse
Affiliation(s)
- Kanagaraj Gomathi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Muthukumar Rohini
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Nicola C Partridge
- Department of Molecular Pathobiology, New York University College Dentistry, New York, NY, USA
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| |
Collapse
|
43
|
Mixon A, Bahar-Moni AS, Faisal TR. Mechanical characterization of articular cartilage degraded combinedly with MMP-1 and MMP-9. J Mech Behav Biomed Mater 2022; 129:105131. [DOI: 10.1016/j.jmbbm.2022.105131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/24/2021] [Accepted: 02/11/2022] [Indexed: 11/29/2022]
|
44
|
Chen H, Cui Y, Zhang D, Xie J, Zhou X. The role of fibroblast growth factor 8 in cartilage development and disease. J Cell Mol Med 2022; 26:990-999. [PMID: 35001536 PMCID: PMC8831980 DOI: 10.1111/jcmm.17174] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factor 8 (FGF‐8), also known as androgen‐induced growth factor (AIGF), is presumed to be a potent mitogenic cytokine that plays important roles in early embryonic development, brain formation and limb development. In the bone environment, FGF‐8 produced or received by chondrocyte precursor cells binds to fibroblast growth factor receptor (FGFR), causing different levels of activation of downstream signalling pathways, such as phospholipase C gamma (PLCγ)/Ca2+, RAS/mitogen‐activated protein kinase‐extracellular regulated protein kinases (RAS/MAPK‐MEK‐ERK), and Wnt‐β‐catenin‐Axin2 signalling, and ultimately controlling chondrocyte proliferation, differentiation, cell survival and migration. However, the molecular mechanism of FGF‐8 in normal or pathological cartilage remains unclear, and thus, FGF‐8 represents a novel exploratory target for studies of chondrocyte development and cartilage disease progression. In this review, studies assessing the relationship between FGF‐8 and chondrocytes that have been published in the past 5 years are systematically summarized to determine the probable mechanism and physiological effect of FGF‐8 on chondrocytes. Based on the existing research results, a therapeutic regimen targeting FGF‐8 is proposed to explore the possibility of treating chondrocyte‐related diseases.
Collapse
Affiliation(s)
- Haoran Chen
- State Key Laboratory of Oral Diseases, 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
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, 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
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Bay-Jensen AC, Mobasheri A, Thudium CS, Kraus VB, Karsdal MA. Blood and urine biomarkers in osteoarthritis - an update on cartilage associated type II collagen and aggrecan markers. Curr Opin Rheumatol 2022; 34:54-60. [PMID: 34652292 PMCID: PMC8635261 DOI: 10.1097/bor.0000000000000845] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a painful disease for which drug development has proven difficult. One major reason for this is the heterogeneity of the disease and the current lack of operationalized means to distinguish various disease endotypes (molecular subtypes). Biomarkers measured in blood or urine, reflecting joint tissue turnover, have been developed and tested during the last decades. In this narrative review, we provide highlights on biomarkers derived from the two most studied and abundant cartilage proteins - type II collagen and aggrecan. RECENT FINDINGS Multiple biomarkers assessing type II collagen degradation and formation, and aggrecan turnover have been developed. Several markers, such as uCTX-II, have been validated for their association with disease severity and prognosis, as well as pharmacodynamically used to describe the mode of action and efficacy of drugs in development. There is a great need for biomarkers for subdividing patients (i.e., endotyping) and recent scientific advances have not yet come closer to achieving this goal. SUMMARY There is strong support for using biomarkers for understanding OA, reflecting degradation and formation of the joint tissues, focused on type II collagen and aggrecan. There is still a lack of in vitro diagnostics, in all contexts of use.
Collapse
Affiliation(s)
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- University Medical Center Utrecht, Department of Orthopedics, Rheumatology and Clinical Immunology, Utrecht, The Netherlands
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, University of Liege, Liege, Belgium
| | | | - Virginia B. Kraus
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | |
Collapse
|
46
|
Lu R, He Z, Zhang W, Wang Y, Cheng P, Lv Z, Yuan X, Guo F, You H, Chen AM, Hu W. Oroxin B alleviates osteoarthritis through anti-inflammation and inhibition of PI3K/AKT/mTOR signaling pathway and enhancement of autophagy. Front Endocrinol (Lausanne) 2022; 13:1060721. [PMID: 36531454 PMCID: PMC9751055 DOI: 10.3389/fendo.2022.1060721] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common aging-related degenerative joint disease with chronic inflammation as its possible pathogenesis. Oroxin B (OB), a flavonoid isolated from traditional Chinese herbal medicine, possesses anti-inflammation properties which may be involved in regulating the pathogenesis of OA, but its mechanism has not been elucidated. Our study was the first to explore the potential chondroprotective effect and elucidate the underlying mechanism of OB in OA. METHODS In vitro, primary mice chondrocytes were stimulated with IL-1β along with or without the administration of OB or autophagy inhibitor 3-methyladenine (3-MA). Cell viability assay was measured with a cell counting kit-8 (CCK-8). The phenotypes of anabolic-related (Aggrecan and Collagen II), catabolic-related (MMP3, MMP13, and ADAMTS5), inflammation-related (iNOS, COX-2, TNF-α, IL-6, and IL-1β), and markers of related signaling pathways in chondrocytes with different treatment were detected through western blot, RT-qPCR, and immunofluorescent staining. In vivo, the destabilized medial meniscus (DMM) operation was performed to establish the OA mice model. After knee intra-articular injection with OB for 8 weeks, the mice's knee joints were obtained for subsequent histological staining and analysis. RESULTS OB reversed the expression level of anabolic-related proteins (Aggrecan and Collagen II) and catabolic-related (MMP3, MMP13, and ADAMTS5) in IL-1β-induced chondrocytes. Mechanistically, OB suppressed the inflammatory response stimulated by IL-1β, as the inflammation-related (iNOS, COX-2, TNF-α, IL-6, and IL-1β) markers were downregulated after the administration of OB in IL-1β-induced chondrocytes. Besides, the activation of PI3K/AKT/mTOR signaling pathway induced by IL-1β could be inhibited by OB. Additionally, the autophagy process impaired by IL-1β could be rescued by OB. What's more, the introduction of 3-MA to specifically inhibit the autophagic process impairs the protective effect of OB on cartilage. In vivo, histological staining revealed that intra-articular injection of OB attenuated the cartilage degradation, as well as reversed the expression level of anabolic and catabolic-related proteins such as Aggrecan, Collagen II, and MMP13 induced in DMM-induced OA models. CONCLUSIONS The study verified that OB exhibited the chondroprotective effect by anti-inflammatory, inhibiting the PI3K/AKT/mTOR signaling pathway, and enhancing the autophagy process, indicating that OB might be a promising agent for the treatment of OA.
Collapse
Affiliation(s)
- Rui Lu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyi He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weikai Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingguang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengtao Lv
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuefeng Yuan
- Department of Traumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - An-min Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weihua Hu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Weihua Hu,
| |
Collapse
|
47
|
Groen SS, Sinkeviciute D, Bay-Jensen AC, Thudium CS, Karsdal MA, Thomsen SF, Lindemann S, Werkmann D, Blair J, Staunstrup LM, Önnerfjord P, Arendt-Nielsen L, Nielsen SH. A serological type II collagen neoepitope biomarker reflects cartilage breakdown in patients with osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100207. [PMID: 36474766 PMCID: PMC9718155 DOI: 10.1016/j.ocarto.2021.100207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/11/2021] [Indexed: 01/09/2023] Open
Abstract
Objectives There is an unmet medical need for biomarkers in OA which can be applied in clinical drug development trials. The present study describes the development of a specific and robust assay measuring type II collagen degradation (T2CM) and discusses its potential as a noninvasive translational biomarker. Methods A type II collagen specific neoepitope (T2CM) was identified by mass spectrometry and monoclonal antibodies were raised towards the epitope, employed in a chemiluminescence immunoassay. T2CM was assessed in bovine cartilage explants with or without MMP-13 inhibitor, and explant supernatants were analyzed by Western blot. T2CM was measured in plasma samples from one study (n = 48 patients) where OA patients were referred to total knee replacement (TKR). Additionally, T2CM was quantified in serum from OA patients receiving salmon calcitonin treatment (sCT) (n = 50) compared to placebo (n = 57). Results The T2CM assay was technically robust (13/4 % inter/intra-variation) and specific for the type II collagen fragment cleaved by MMP-1 and -13. The MMP-13 inhibitor reduced the T2CM release from bovine cartilage explants receiving catabolic treatment. These results were confirmed by Western blot. In human end-stage OA patients (scheduled for TKR), the T2CM levels were elevated compared to moderate OA (p<0.004). The OA patients receiving sCT had lower levels of T2CM compared to placebo group after 1, 6, and 24 months of treatment (p = 0.0285, p = 0.0484, p = 0.0035). Conclusions To our knowledge, T2CM is the first technically robust serological biomarker assay which has shown biological relevance in ex vivo models and OA cohorts. This suggests that T2CM may have potential as a translational biomarker for cartilage degradation.
Collapse
Affiliation(s)
- Solveig Skovlund Groen
- Immunoscience, Nordic Bioscience, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Dovile Sinkeviciute
- Immunoscience, Nordic Bioscience, Herlev, Denmark
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | | | | | | | | | | | - Joseph Blair
- Immunoscience, Nordic Bioscience, Herlev, Denmark
| | | | | | - Lars Arendt-Nielsen
- Center for Neuroplasticity and Pain (CNAP), SMI®, Department of Health Science and Technology, School of Medicine, Aalborg University, Aalborg, Denmark
| | - Signe Holm Nielsen
- Immunoscience, Nordic Bioscience, Herlev, Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
48
|
Yue D, Du L, Zhang B, Wu H, Yang Q, Wang M, Pan J. Time-dependently Appeared Microenvironmental Changes and Mechanism after Cartilage or Joint Damage and the Influences on Cartilage Regeneration. Organogenesis 2021; 17:85-99. [PMID: 34806543 DOI: 10.1080/15476278.2021.1991199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cartilage and joint damage easily degenerates cartilage and turns into osteoarthritis (OA), which seriously affects human life and work, and has no cure currently. The temporal and spatial changes of multiple microenvironments upon the damage of cartilage and joint are noticed, including the emergences of inflammation, bone remodeling, blood vessels, and nerves, as well as alterations of extracellular and pericellular matrix, oxygen tension, biomechanics, underneath articular cartilage tissues, and pH value. This review summarizes the existing literatures on microenvironmental changes, mechanisms, and their negative effects on cartilage regeneration following cartilage and joint damage. We conclude that time-dependently rebuilding the multiple normal microenvironments of damaged cartilage is the key for cartilage regeneration after systematic studies for the timing and correlations of various microenvironment changes.
Collapse
Affiliation(s)
- Danyang Yue
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Lin Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Bingbing Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Huan Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Qiong Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Min Wang
- Orthopedic Department, Xinqiao Hospital, Army Medical University, Chongqing, PR China
| | - Jun Pan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| |
Collapse
|
49
|
Yamamoto K, Wilkinson D, Bou-Gharios G. Targeting Dysregulation of Metalloproteinase Activity in Osteoarthritis. Calcif Tissue Int 2021; 109:277-290. [PMID: 32772139 PMCID: PMC8403128 DOI: 10.1007/s00223-020-00739-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 07/24/2020] [Indexed: 02/06/2023]
Abstract
Metalloproteinases were first identified as collagen cleaving enzymes and are now appreciated to play important roles in a wide variety of biological processes. The aberrant activity and dysregulation of the metalloproteinase family are linked to numerous diseases including cardiovascular and pulmonary diseases, chronic wounds, cancer, fibrosis and arthritis. Osteoarthritis (OA) is the most prevalent age-related joint disorder that causes pain and disability, but there are no disease-modifying drugs available. The hallmark of OA is loss of articular cartilage and elevated activities of matrix-degrading metalloproteinases are responsible. These enzymes do not exist in isolation and their activity is tightly regulated by a number of processes, such as transcription, proteolytic activation, interaction with their inhibitors, cell surface and extracellular matrix molecules, and endocytic clearance from the extracellular milieu. Here, we describe the functions and roles of metalloproteinase family in OA pathogenesis. We highlight recent studies that have illustrated novel mechanisms regulating their extracellular activity and impairment of such regulations that lead to the development of OA. We also discuss how to stop or slow down the degenerative processes by targeting aberrant metalloproteinase activity, which may in future become therapeutic interventions for the disease.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| | - David Wilkinson
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| |
Collapse
|
50
|
Mixon A, Savage A, Bahar-Moni AS, Adouni M, Faisal T. An in vitro investigation to understand the synergistic role of MMPs-1 and 9 on articular cartilage biomechanical properties. Sci Rep 2021; 11:14409. [PMID: 34257325 PMCID: PMC8277889 DOI: 10.1038/s41598-021-93744-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/30/2021] [Indexed: 11/09/2022] Open
Abstract
Matrix metalloproteinases (MMPs) play a crucial role in enzymatically digesting cartilage extracellular matrix (ECM) components, resulting in degraded cartilage with altered mechanical loading capacity. Overexpression of MMPs is often caused by trauma, physiologic conditions and by disease. To understand the synergistic impact MMPs have on cartilage biomechanical properties, MMPs from two subfamilies: collagenase (MMP-1) and gelatinase (MMP-9) were investigated in this study. Three different ratios of MMP-1 (c) and MMP-9 (g), c1:g1, c3:g1 and c1:g3 were considered to develop a degradation model. Thirty samples, harvested from bovine femoral condyles, were treated in groups of 10 with one concentration of enzyme mixture. Each sample was tested in a healthy state prior to introducing degradative enzymes to establish a baseline. Samples were subjected to indentation loading up to 20% bulk strain. Both control and treated samples were mechanically and histologically assessed to determine the impact of degradation. Young's modulus and peak load of the tissue under indentation were compared between the control and degraded cartilage explants. Cartilage degraded with the c3:g1 enzyme concentration resulted in maximum 33% reduction in stiffness and peak load compared to the other two concentrations. The abundance of collagenase is more responsible for cartilage degradation and reduced mechanical integrity.
Collapse
Affiliation(s)
- Allison Mixon
- Department of Mechanical Engineering, University of Louisiana at Lafayette, Lafayette, LA, 70503, USA
| | - Andrew Savage
- Department of Biology, University of Louisiana at Lafayette, Lafayette, LA, 70503, USA
| | - Ahmed Suparno Bahar-Moni
- Department of Orthopaedics, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Malek Adouni
- Department of Mechanical Engineering, Australian College of Kuwait, P.O. Box 1411, East Meshrif, Kuwait
| | - Tanvir Faisal
- Department of Mechanical Engineering, University of Louisiana at Lafayette, Lafayette, LA, 70503, USA.
| |
Collapse
|