1
|
Tuerlings M, Houtman E, Muusers EJH, Simon J, de Haan MW, Boone I, Ramos YFM, Mahdad R, Meulenbelt I. Exploring the therapeutic effect of human recombinant IL11 on lesioned OA human osteochondral explants. Arthritis Res Ther 2025; 27:15. [PMID: 39856704 PMCID: PMC11761764 DOI: 10.1186/s13075-025-03480-4] [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: 05/07/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE To explore IL11 co-expression profiles in our previously reported RNA-sequencing dataset of OA articular cartilage, in interaction with IL6, and to investigate the effects of hrIL11 administration as potential therapeutic strategy for OA articular cartilage using our biomimetic aged human osteochondral explant model of OA. METHODS We used RNA-sequencing datasets of macroscopically preserved and lesioned OA articular cartilage (N = 35 patients). Spearman correlations were calculated between IL11 and IL6 expression levels and genes expressed in cartilage (N = 20048 genes). Osteochondral explants were isolated from macroscopically preserved and lesioned areas of the joint and were kept in culture for two weeks, with or without exposure to 200ng/ml hrIL11. RESULTS We found no overlap in correlating genes between IL11 and IL6, indicating their distinct roles in articular cartilage. Moreover, we identified more genes being correlated to IL11 in the lesioned compared to preserved articular cartilage (N = 203 and 106 genes, respectively). Upon treatment of ex vivo OA articular cartilage with hrIL11, we overall observed unbeneficial effects on chondrocyte phenotype, as illustrated by upregulation of MMP13, EPAS1, RUNX2, and POSTN. We did not observe significant differences in Mankin scores upon addition of hrIL11. CONCLUSION The current study showed that treatment of OA articular cartilage with hrIL11 is unlikely to be beneficial despite previous indications of hrIL11 as potential druggable target. These findings underscore the importance of functionally investigating OA risk genes. Better understanding of IL11 signaling and the underlying pathways is necessary towards the development of OA treatment strategy.
Collapse
Affiliation(s)
- Margo Tuerlings
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Evelyn Houtman
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elisa J H Muusers
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Janneke Simon
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Maurice W de Haan
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Ilja Boone
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Yolande F M Ramos
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Rachid Mahdad
- Department Orthopaedics, Alrijne Hospital, Leiderdorp, Netherlands
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
2
|
Zu Y, Du J, Xu Y, Niu M, Hong C, Yang C. Change in p53 nuclear localization in response to extracellular matrix stiffness. SMART MEDICINE 2024; 3:e20240026. [PMID: 39776592 PMCID: PMC11669774 DOI: 10.1002/smmd.20240026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 01/11/2025]
Abstract
Chondrocytes are commonly applied in regenerative medicine and tissue engineering. Thus, the discovery of optimal culture conditions to obtain cells with good properties and behavior for transplantation is important. In addition to biochemical cues, physical and biomechanical changes can affect the proliferation and protein expression of chondrocytes. Here we investigated the effect of extracellular matrix stiffness on mouse articular chondrocyte phenotype, growth, and subcellular p53 localization. Chondrocytes were seeded on collagen-coated substrates varying in elasticity: 0.5 and 100 kPa. Immunocytochemical staining and immunoblotting showed that a softer substrate significantly increased p53 nuclear localization in chondrocytes. Furthermore, we identified microRNA-532 (miR-532) as a potential p53 target gene to influence cell function, indicating a new target for tissue engineering. These findings provide insight into the influence of physical cues on cell phenotype maintenance and could help improve understanding of cartilage-related pathologies such as osteoarthritis.
Collapse
Affiliation(s)
- Yan Zu
- Institute of Biomechanics and Medical EngineeringSchool of Aerospace EngineeringTsinghua UniversityBeijingChina
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| | - Jing Du
- Institute of Biomechanics and Medical EngineeringSchool of Aerospace EngineeringTsinghua UniversityBeijingChina
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationBeijing Advanced Innovation Center for Biomedical EngineeringSchool of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Yipu Xu
- Institute of Biomechanics and Medical EngineeringSchool of Aerospace EngineeringTsinghua UniversityBeijingChina
- Department of General Dentistry and Emergency Dental CareBeijing Stomatological HospitalCapital Medical UniversityBeijingChina
| | - Mengying Niu
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| | - Canlin Hong
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| | - Chun Yang
- Institute of Biomechanics and Medical EngineeringSchool of Aerospace EngineeringTsinghua UniversityBeijingChina
| |
Collapse
|
3
|
Jahn J, Ehlen QT, Kaplan L, Best TM, Meng Z, Huang CY. Interplay of Glucose Metabolism and Hippo Pathway in Chondrocytes: Pathophysiology and Therapeutic Targets. Bioengineering (Basel) 2024; 11:972. [PMID: 39451348 PMCID: PMC11505586 DOI: 10.3390/bioengineering11100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
In this review, we explore the intricate relationship between glucose metabolism and mechanotransduction pathways, with a specific focus on the role of the Hippo signaling pathway in chondrocyte pathophysiology. Glucose metabolism is a vital element in maintaining proper chondrocyte function, but it has also been implicated in the pathogenesis of osteoarthritis (OA) via the induction of pro-inflammatory signaling pathways and the establishment of an intracellular environment conducive to OA. Alternatively, mechanotransduction pathways such as the Hippo pathway possess the capacity to respond to mechanical stimuli and have an integral role in maintaining chondrocyte homeostasis. However, these mechanotransduction pathways can be dysregulated and potentially contribute to the progression of OA. We discussed how alterations in glucose levels may modulate the Hippo pathway components via a variety of mechanisms. Characterizing the interaction between glucose metabolism and the Hippo pathway highlights the necessity of balancing both metabolic and mechanical signaling to maintain chondrocyte health and optimal functionality. Furthermore, this review demonstrates the scarcity of the literature on the relationship between glucose metabolism and mechanotransduction and provides a summary of current research dedicated to this specific area of study. Ultimately, increased research into this topic may elucidate novel mechanisms and relationships integrating mechanotransduction and glucose metabolism. Through this review we hope to inspire future research into this topic to develop innovative treatments for addressing the clinical challenges of OA.
Collapse
Affiliation(s)
- Jacob Jahn
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
| | - Quinn T. Ehlen
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
| | - Lee Kaplan
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Orthopedics, University of Miami, Miami, FL 33136, USA
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
| | - Thomas M. Best
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Orthopedics, University of Miami, Miami, FL 33136, USA
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
| | - Zhipeng Meng
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chun-Yuh Huang
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA
| |
Collapse
|
4
|
Zhra M, Magableh AM, Samhan LM, Fatani LM, Qasem RJ, Aljada A. The Expression of a Subset of Aging and Antiaging Markers Following the Chondrogenic and Osteogenic Differentiation of Mesenchymal Stem Cells of Placental Origin. Cells 2024; 13:1022. [PMID: 38920652 PMCID: PMC11201886 DOI: 10.3390/cells13121022] [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: 05/01/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Mesenchymal stem cells (MSCs) of placental origin hold great promise in tissue engineering and regenerative medicine for diseases affecting cartilage and bone. However, their utility has been limited by their tendency to undergo premature senescence and phenotypic drift into adipocytes. This study aimed to explore the potential involvement of a specific subset of aging and antiaging genes by measuring their expression prior to and following in vitro-induced differentiation of placental MSCs into chondrocytes and osteoblasts as opposed to adipocytes. The targeted genes of interest included the various LMNA/C transcript variants (lamin A, lamin C, and lamin A∆10), sirtuin 7 (SIRT7), and SM22α, along with the classic aging markers plasminogen activator inhibitor 1 (PAI-1), p53, and p16INK4a. MSCs were isolated from the decidua basalis of human term placentas, expanded, and then analyzed for phenotypic properties by flow cytometry and evaluated for colony-forming efficiency. The cells were then induced to differentiate in vitro into chondrocytes, osteocytes, and adipocytes following established protocols. The mRNA expression of the targeted genes was measured by RT-qPCR in the undifferentiated cells and those fully differentiated into the three cellular lineages. Compared to undifferentiated cells, the differentiated chondrocytes demonstrated decreased expression of SIRT7, along with decreased PAI-1, lamin A, and SM22α expression, but the expression of p16INK4a and p53 increased, suggesting their tendency to undergo premature senescence. Interestingly, the cells maintained the expression of lamin C, which indicates that it is the primary lamin variant influencing the mechanoelastic properties of the differentiated cells. Notably, the expression of all targeted genes did not differ from the undifferentiated cells following osteogenic differentiation. On the other hand, the differentiation of the cells into adipocytes was associated with decreased expression of lamin A and PAI-1. The distinct patterns of expression of aging and antiaging genes following in vitro-induced differentiation of MSCs into chondrocytes, osteocytes, and adipocytes potentially reflect specific roles for these genes during and following differentiation in the fully functional cells. Understanding these roles and the network of signaling molecules involved can open opportunities to improve the handling and utility of MSCs as cellular precursors for the treatment of cartilage and bone diseases.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmad M. Magableh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Lara M. Samhan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Lein M. Fatani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Rani J. Qasem
- Department of Pharmacology and Pharmacy Practice, College of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
5
|
Kong P, Ahmad RE, Zulkifli A, Krishnan S, Nam HY, Kamarul T. The role of autophagy in mitigating osteoarthritis progression via regulation of chondrocyte apoptosis: A review. Joint Bone Spine 2024; 91:105642. [PMID: 37739213 DOI: 10.1016/j.jbspin.2023.105642] [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: 03/28/2023] [Revised: 07/22/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disease with an immense socioeconomic burden; however, no treatment has achieved complete success in effectively halting or reversing cartilage degradation, which is the central pathophysiological feature of OA. Chondrocytes loss or dysfunction is a significant contributing factor to the progressive cartilage deterioration as these sole resident cells have a crucial role to produce extracellular matrix proteins, thus maintaining cartilage structure and homeostasis. It has been previously suggested that death of chondrocytes occurring through apoptosis substantially contributes to cartilage degeneration. Although the occurrence of apoptosis in osteoarthritic cartilage and its correlation with cartilage degradation is evident, the causes of chondrocyte apoptosis leading to matrix loss are still not well-understood. Autophagy, an intracellular degradative mechanism that eliminates dysfunctional cytoplasmic components to aid cell survival in unfavourable conditions, is a potential therapeutic target to inhibit chondrocyte apoptosis and reduce OA severity. Despite accumulating evidence indicating significant cytoprotective effects of autophagy against chondrocyte apoptosis, the mechanistic link between autophagy and apoptosis in chondrocytes remains to be further explored. In this review, we summarize the relevant mechanistic events that perpetuate chondrocyte apoptosis and highlight the prominent role of autophagy in modulating these events to mitigate OA progression.
Collapse
Affiliation(s)
- Peggy Kong
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Raja Elina Ahmad
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia.
| | - Amirah Zulkifli
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Shaliny Krishnan
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Hui Yin Nam
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia; Nanotechnology and Catalysis Research Centre (NANOCAT), Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Tunku Kamarul
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia; Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas Pulau Pinang, Malaysia
| |
Collapse
|
6
|
Valerio T, Milan JL, Goislard de Monsabert B, Vigouroux L. The effect of trapeziometacarpal joint passive stiffness on mechanical loadings of cartilages. J Biomech 2024; 166:112042. [PMID: 38498967 DOI: 10.1016/j.jbiomech.2024.112042] [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: 09/05/2023] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024]
Abstract
Hypermobility of the trapeziometacarpal joint is commonly considered to be a potential risk factor for osteoarthritis. Nevertheless, the results remain controversial due to a lack of quantitative validation. The objective of this study was to evaluate the effect of joint laxity on the mechanical loadings of cartilage. A patient-specific finite element model of trapeziometacarpal joint passive stiffness was developed. The joint passive stiffness was modeled by creating linear springs all around the joint. The linear spring stiffness was determined by using an optimization process to fit force-displacement data measured during laxity tests performed on eight healthy volunteers. The estimated passive stiffness parameters were then included in a full thumb finite element simulation of a pinch grip task driven by muscle forces to evaluate the effect on trapeziometacarpal loading. The correlation between stiffness and the loading of cartilage in terms of joint contact pressure and maximum shear strain was analyzed. A significant negative correlation was found between the trapeziometacarpal joint passive stiffness and the contact pressure on trapezium cartilage during the simulated pinch grip task. These results therefore suggest that the hypermobility of the trapeziometacarpal joint could affect the contact pressure on trapezium cartilage and support the existence of an increased risk associated with hypermobility.
Collapse
Affiliation(s)
- Thomas Valerio
- Aix-Marseille University, CNRS, ISM, Marseille, France; Aix-Marseille University, APHM, CNRS, ISM, St Marguerite Hospital, Institute for Locomotion, Department of Orthopaedics and Traumatology, Marseille, France.
| | - Jean-Louis Milan
- Aix-Marseille University, CNRS, ISM, Marseille, France; Aix-Marseille University, APHM, CNRS, ISM, St Marguerite Hospital, Institute for Locomotion, Department of Orthopaedics and Traumatology, Marseille, France
| | | | | |
Collapse
|
7
|
Ma W, Tan X, Xie Z, Yu J, Li P, Lin X, Ouyang S, Liu Z, Hou Q, Xie N, Peng T, Li L, Dai Z, Chen X, Xie W. P53: A Key Target in the Development of Osteoarthritis. Mol Biotechnol 2024; 66:1-10. [PMID: 37154864 DOI: 10.1007/s12033-023-00736-9] [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: 11/29/2022] [Accepted: 03/25/2023] [Indexed: 05/10/2023]
Abstract
Osteoarthritis (OA), a chronic degenerative disease characterized mainly by damage to the articular cartilage, is increasingly relevant to the pathological processes of senescence, apoptosis, autophagy, proliferation, and differentiation of chondrocytes. Clinical strategies for osteoarthritis can only improve symptoms and even along with side effects due to age, sex, disease, and other factors. Therefore, there is an urgent need to identify new ideas and targets for current clinical treatment. The tumor suppressor gene p53, which has been identified as a potential target for tumor therapeutic intervention, is responsible for the direct induction of the pathological processes involved in OA modulation. Consequently, deciphering the characteristics of p53 in chondrocytes is essential for investigating OA pathogenesis due to p53 regulation in an array of signaling pathways. This review highlights the effects of p53 on senescence, apoptosis, and autophagy of chondrocytes and its role in the development of OA. It also elucidates the underlying mechanism of p53 regulation in OA, which may help provide a novel strategies for the clinical treatment of OA.
Collapse
Affiliation(s)
- Wentao Ma
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoqian Tan
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Zhongcheng Xie
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Jiang Yu
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Pin Li
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyan Lin
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Ouyang
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Zhiyang Liu
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Qin Hou
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Nan Xie
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhong Peng
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Liang Li
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Zhu Dai
- Department of Orthopedics, Hengyang Medical School, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China.
| | - Xi Chen
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China.
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
8
|
Chen Y, Yang H, Wang Z, Zhu R, Cheng L, Cheng Q. Low-intensity pulsed ultrasound promotes mesenchymal stem cell transplantation-based articular cartilage regeneration via inhibiting the TNF signaling pathway. Stem Cell Res Ther 2023; 14:93. [PMID: 37069673 PMCID: PMC10111837 DOI: 10.1186/s13287-023-03296-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/22/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation therapy is highly investigated for the regenerative repair of cartilage defects. Low-intensity pulsed ultrasound (LIPUS) has the potential to promote chondrogenic differentiation of MSCs. However, its underlying mechanism remains unclear. Here, we investigated the promoting effects and mechanisms underlying LIPUS stimulation on the chondrogenic differentiation of human umbilical cord mesenchymal stem cells (hUC-MSCs) and further evaluated its regenerative application value in articular cartilage defects in rats. METHODS LIPUS was applied to stimulate cultured hUC-MSCs and C28/I2 cells in vitro. Immunofluorescence staining, qPCR analysis, and transcriptome sequencing were used to detect mature cartilage-related markers of gene and protein expression for a comprehensive evaluation of differentiation. Injured articular cartilage rat models were established for further hUC-MSC transplantation and LIPUS stimulation in vivo. Histopathology and H&E staining were used to evaluate the repair effects of the injured articular cartilage with LIPUS stimulation. RESULTS The results showed that LIPUS stimulation with specific parameters effectively promoted the expression of mature cartilage-related genes and proteins, inhibited TNF-α gene expression in hUC-MSCs, and exhibited anti-inflammation in C28/I2 cells. In addition, the articular cartilage defects of rats were significantly repaired after hUC-MSC transplantation and LIPUS stimulation. CONCLUSIONS Taken together, LIPUS stimulation could realize articular cartilage regeneration based on hUC-MSC transplantation due to the inhibition of the TNF signaling pathway, which is of clinical value for the relief of osteoarthritis.
Collapse
Affiliation(s)
- Yiming Chen
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Huiyi Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
- School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
- School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Qian Cheng
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China.
- Frontiers Science Center for Intelligent Autonomous Systems, Shanghai, 201210, China.
| |
Collapse
|
9
|
Buzzatto-Leite I, Afonso J, Silva-Vignato B, Coutinho L, Alvares L. Differential gene co-expression network analyses reveal novel molecules associated with transcriptional dysregulation of key biological processes in osteoarthritis knee cartilage. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100316. [PMID: 36474801 PMCID: PMC9718204 DOI: 10.1016/j.ocarto.2022.100316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 09/09/2022] [Accepted: 10/17/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES To compare co-expression networks of normal and osteoarthritis knee cartilage to uncover molecules associated with the transcriptional misregulation compromising biological processes (BPs) critical for cartilage homeostasis. DESIGN Normal and osteoarthritis human knee cartilage RNA-seq GSE114007 dataset was obtained from the Gene Expression Omnibus database. Partial Correlation and Information Theory (PCIT) algorithm was used to build co-expression networks containing all nodes connecting to at least one differentially expressed gene (DEG) in normal and osteoarthritis networks. Hub and hub centrality genes were used to perform functional enrichment analysis. Enriched BPs known to be associated with both healthy and diseased cartilage were compared in depth. RESULTS Differential co-expression network analyses allowed the identification of DDX43 and USP42 as exclusively co-expressed with DEGs in normal and osteoarthritis networks, respectively. The top hub and hub centrality genes of these networks were HIST1H3A and SNHG12 (normal) and TAF9B and OTUD1 (osteoarthritis). Enrichment analysis revealed several shared BPs between the contrasting groups, which are well-known in osteoarthritis pathogenesis. Protein-protein interaction network analysis for these BPs showed a global down-regulation of transcription factors in osteoarthritis. Specific transcription factors were identified as pleiotropic mediators in articular cartilage maintenance since they take part in several BPs. In addition, chromatin organisation and modification proteins were found relevant for osteoarthritis development. CONCLUSION Differential gene co-expression analysis allowed the identification of novel and high priority therapeutic candidate genes that may drive modifications in the transcriptional "status" of cartilage in osteoarthritis.
Collapse
Affiliation(s)
- I. Buzzatto-Leite
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - J. Afonso
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo (ESALQ/USP), Piracicaba, Brazil
| | - B. Silva-Vignato
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo (ESALQ/USP), Piracicaba, Brazil
| | - L.L. Coutinho
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo (ESALQ/USP), Piracicaba, Brazil
| | - L.E. Alvares
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil,Corresponding author. Department of Biochemistry and Tissue Biology, University of Campinas – UNICAMP, Rua Monteiro Lobato 255, Cx. Postal 6109, CEP 13083-862, Campinas, SP, Brazil.
| |
Collapse
|
10
|
Bolcos PO, Mononen ME, Roach KE, Tanaka MS, Suomalainen JS, Mikkonen S, Nissi MJ, Töyräs J, Link TM, Souza R, Majumdar S, Ma B, Li X, Korhonen RK. Subject-specific biomechanical analysis to estimate locations susceptible to osteoarthritis-Finite element modeling and MRI follow-up of ACL reconstructed patients. J Orthop Res 2022; 40:1744-1755. [PMID: 34820897 PMCID: PMC9127000 DOI: 10.1002/jor.25218] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 09/16/2021] [Accepted: 11/09/2021] [Indexed: 02/04/2023]
Abstract
The aims of this case-control study were to: (1) Identify cartilage locations and volumes at risk of osteoarthritis (OA) using subject-specific finite element (FE) models; (2) Quantify the relationships between the simulated biomechanical parameters and T2 and T1ρ relaxation times of magnetic resonance imaging (MRI). We created subject-specific FE models for seven patients with anterior cruciate ligament (ACL) reconstruction and six controls based on a previous proof-of-concept study. We identified locations and cartilage volumes susceptible to OA, based on maximum principal stresses and absolute maximum shear strains in cartilage exceeding thresholds of 7 MPa and 32%, respectively. The locations and volumes susceptible to OA were compared qualitatively and quantitatively against 2-year longitudinal changes in T2 and T1ρ relaxation times. The degeneration volumes predicted by the FE models, based on excessive maximum principal stresses, were significantly correlated (r = 0.711, p < 0.001) with the degeneration volumes determined from T2 relaxation times. There was also a significant correlation between the predicted stress values and changes in T2 relaxation time (r = 0.649, p < 0.001). Absolute maximum shear strains and changes in T1ρ relaxation time were not significantly correlated. Five out of seven patients with ACL reconstruction showed excessive maximum principal stresses in either one or both tibial cartilage compartments, in agreement with follow-up information from MRI. Expectedly, for controls, the FE models and follow-up information showed no degenerative signs. Our results suggest that the presented modelling methodology could be applied to prospectively identify ACL reconstructed patients at risk of biomechanically driven OA, particularly by the analysis of maximum principal stresses of cartilage.
Collapse
Affiliation(s)
- Paul O. Bolcos
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland,Corresponding author: Paul Octavian Bolcos, Department of Applied Physics, University of Eastern Finland, POB 1627, FI-70211 Kuopio, Finland, Tel. +358 45 2290653,
| | - Mika E. Mononen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Koren E. Roach
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, Unites States of America
| | - Matthew S. Tanaka
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, Unites States of America
| | | | - Santtu Mikkonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Mikko J. Nissi
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland,Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, Finland
| | - Juha Töyräs
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland,School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia,Diagnostic Imaging Centre, Kuopio University Hospital, Kuopio Finland
| | - Thomas M. Link
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, Unites States of America
| | - Richard Souza
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, Unites States of America
| | - Sharmila Majumdar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, Unites States of America
| | - Benjamin Ma
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, Unites States of America
| | - Xiaojuan Li
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Unites States of America
| | - Rami K Korhonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland,Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
11
|
Trp53 controls chondrogenesis and endochondral ossification by negative regulation of TAZ activity and stability via β-TrCP-mediated ubiquitination. Cell Death Dis 2022; 8:317. [PMID: 35831272 PMCID: PMC9279315 DOI: 10.1038/s41420-022-01105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022]
Abstract
Transformation-related protein 53 (Trp53) is a critical regulator of cell fate determination by controlling cell proliferation and differentiation. Ablation of Trp53 signaling in osteoblast lineages significantly promotes osteogenesis, bone formation, and bone remodeling. However, how Trp53 regulates chondrogenesis and endochondral bone formation is undefined. In this study, we found that Trp53 expression gradually decreased in tibia growth plates during embryonic development in vivo and during chondrogenesis in vitro. By deleting Trp53 in chondrocyte lineage using Col2-Cre transgenic line, we found that loss of Trp53 in chondrocytes significantly increased growth plate growth and bone formation by increasing chondrocyte proliferation, matrix production and maturation, and bone dynamic formation rate. Mechanistically, our data revealed loss of Trp53 significantly promoted TAZ transcriptional activity through inhibition of TAZ phosphorylation and nuclear translocation, whereas its activity was pronouncedly inhibited after forced expression of Trp53. Furthermore, Co-IP data demonstrated that Trp53 associated with TAZ. Moreover, Trp53 decreased the stability of TAZ protein and promoted its degradation through β-TrCP-mediated ubiquitination. Ablation of TAZ in Col2-Cre;Trp53f/f mice rescued the phenotypes of enhanced chondrogenesis and bone formation caused by Trp53 deletion. Collectively, this study revealed that Trp53 modulates chondrogenesis and endochondral ossification through negative regulation of TAZ activity and stability, suggesting that targeting Trp53 signaling may be a potential strategy for fracture healing, heterotopic ossification, arthritis, and other bone diseases.
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Translation of genetic information encoded within mRNA molecules by ribosomes into proteins is a key part of the central dogma of molecular biology. Despite the central position of the ribosome in the translation of proteins, and considering the major proteomic changes that occur in the joint during osteoarthritis development and progression, the ribosome has received very limited attention as driver of osteoarthritis pathogenesis. RECENT FINDINGS We provide an overview of the limited literature regarding this developing topic for the osteoarthritis field. Recent key findings that connect ribosome biogenesis and activity with osteoarthritis include: ribosomal RNA transcription, processing and maturation, ribosomal protein expression, protein translation capacity and preferential translation. SUMMARY The ribosome as the central cellular protein synthesis hub is largely neglected in osteoarthritis research. Findings included in this review reveal that in osteoarthritis, ribosome aberrations have been found from early-stage ribosome biogenesis, through ribosome build-up and maturation, up to preferential translation. Classically, osteoarthritis has been explained as an imbalance between joint tissue anabolism and catabolism. We postulate that osteoarthritis can be interpreted as an acquired ribosomopathy. This hypothesis fine-tunes the dogmatic anabolism/katabolism point-of-view, and may provide novel molecular opportunities for the development of osteoarthritis disease-modifying treatments.
Collapse
Affiliation(s)
- Guus G.H. van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
| | - Marjolein M.J. Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
| | - Mandy J. Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Tim J.M. Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
13
|
Castro CM, Corciulo C, Friedman B, Li Z, Jacob S, Fenyo D, Cronstein BN. Adenosine A2A receptor null chondrocyte transcriptome resembles that of human osteoarthritic chondrocytes. Purinergic Signal 2021; 17:439-448. [PMID: 33973110 PMCID: PMC8410926 DOI: 10.1007/s11302-021-09788-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/08/2021] [Indexed: 11/25/2022] Open
Abstract
Adenosine signaling plays a critical role in the maintenance of articular cartilage and may serve as a novel therapeutic for osteoarthritis (OA), a highly prevalent and morbid disease without effective therapeutics in the current market. Mice lacking adenosine A2A receptors (A2AR) develop spontaneous OA by 16 weeks of age, a finding relevant to human OA since loss of adenosine signaling due to diminished adenosine production (NT5E deficiency) also leads to development of OA in mice and humans. To better understand the mechanism by which A2AR and adenosine generation protect from OA development, we examined differential gene expression in neonatal chondrocytes from WT and A2AR null mice. Analysis of differentially expressed genes was analyzed by KEGG pathway analysis, and oPOSSUM and the flatiron database were used to identify transcription factor binding enrichment, and tissue-specific network analyses and patterns were compared to gene expression patterns in chondrocytes from patients with OA. There was a differential expression of 2211 genes (padj<0.05). Pathway enrichment analysis revealed that pro-inflammatory changes, increased metalloprotease, reduced matrix organization, and homeostasis are upregulated in A2AR null chondrocytes. Moreover, stress responses, including autophagy and HIF-1 signaling, seem to be important drivers of OA and bear marked resemblance to the human OA transcriptome. Although A2AR null mice are born with grossly intact articular cartilage, we identify here the molecular foundations for early-onset OA in these mice, further establishing their role as models for human disease and the potential use of adenosine as a treatment for human disease.
Collapse
Affiliation(s)
- Cristina M. Castro
- Department of Medicine, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA USA
| | - Carmen Corciulo
- Division of Translational Medicine, NYUGSOM, New York, NY USA
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutritional, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Benjamin Friedman
- Department of Medicine, Division of Rheumatology, NYUGSOM, New York, NY USA
| | - Zhi Li
- Institute for Systems Genetics, NYU Langone Health, New York, NY USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY USA
| | - Samson Jacob
- Institute for Systems Genetics, NYU Langone Health, New York, NY USA
| | - David Fenyo
- Institute for Systems Genetics, NYU Langone Health, New York, NY USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY USA
| | - Bruce N. Cronstein
- Department of Medicine, Division of Rheumatology, NYUGSOM, New York, NY USA
| |
Collapse
|
14
|
Li G, Rao H, Xu W. Puerarin plays a protective role in chondrocytes by activating Beclin1-dependent autophagy. Biosci Biotechnol Biochem 2021; 85:621-625. [PMID: 33624774 DOI: 10.1093/bbb/zbaa078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/05/2020] [Indexed: 11/14/2022]
Abstract
Puerarin can protect chondrocytes, whereby ameliorating osteoarthritis. Puerarin also promotes autophagy. Autophagy maintains chondrocyte homeostasis. The role of autophagy in puerarin-protected chondrocytes is unknown. Puerarin promoted chondrocyte autophagy. Puerarin-protected chondrocytes were reversed by autophagy inhibitors and Beclin1 inhibitor. 3-MA or Beclin1 inhibitor in vivo reversed puerarin-ameliorated cartilage damage of osteoarthritis mice. Thus, puerarin can protect chondrocytes through Beclin1-dependent autophagy activation.
Collapse
Affiliation(s)
- Guishuang Li
- Department of orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Hongming Rao
- Department of orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Weihong Xu
- Department of orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
15
|
Liu Z, Mo X, Ma F, Li S, Wu G, Tang B, Lin L. Synthesis of carboxymethyl chitosan-strontium complex and its therapeutic effects on relieving osteoarthritis. Carbohydr Polym 2021; 261:117869. [PMID: 33766356 DOI: 10.1016/j.carbpol.2021.117869] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/07/2021] [Accepted: 02/22/2021] [Indexed: 10/22/2022]
Abstract
Osteoarthritis (OA) is an age-related joint disorder and one of the leading causes of physical disability. In this study, we designed and synthesized a new polysaccharide complex, carboxymethyl chitosan strontium (CMCS-Sr), which is believed to have positive effects on relieving OA. The synthesized CMCS-Sr was structurally verified by SEM, EDS, FTIR, etc. The therapeutic effects of CMCS-Sr were evaluated using various biological experiments. The cell viability and apoptosis results reveal that CMCS-Sr can significantly promote the proliferation and suppress OA chondrocytes apoptosis in vitro. The immunofluorescence staining results suggest that CMCS-Sr facilitates the promotion of the secretion of Type II collagen (Col-II). The transcriptomic results support the observed positive effects of CMCS-Sr on inhibiting chondrocytes apoptosis and alleviating inflammatory reactions. Moreover, animal study demonstrates that CMCS-Sr effectively reduced articular cartilage damage and subchondral bone degradation. Therefore, we propose the use of CMCS-Sr as a promising candidate for relieving OA.
Collapse
Affiliation(s)
- Zhengwei Liu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Cell Microenvironment, PR China
| | - Xiaoqiong Mo
- Shenzhen Key Laboratory of Cell Microenvironment, PR China; Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Fenbo Ma
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Cell Microenvironment, PR China
| | - Sijing Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Cell Microenvironment, PR China
| | - Guofeng Wu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Cell Microenvironment, PR China
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China; Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, PR China; Shenzhen Key Laboratory of Cell Microenvironment, PR China.
| | - Lijun Lin
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Shenzhen Key Laboratory of Cell Microenvironment, PR China.
| |
Collapse
|
16
|
Al-Masawa ME, Wan Kamarul Zaman WS, Chua KH. Biosafety evaluation of culture-expanded human chondrocytes with growth factor cocktail: a preclinical study. Sci Rep 2020; 10:21583. [PMID: 33299022 PMCID: PMC7725787 DOI: 10.1038/s41598-020-78395-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/27/2020] [Indexed: 01/03/2023] Open
Abstract
The scarcity of chondrocytes is a major challenge for cartilage tissue engineering. Monolayer expansion is necessary to amplify the limited number of chondrocytes needed for clinical application. Growth factors are often added to improve monolayer culture conditions, promoting proliferation, and enhancing chondrogenesis. Limited knowledge on the biosafety of the cell products manipulated with growth factors in culture has driven this study to evaluate the impact of growth factor cocktail supplements in chondrocyte culture medium on chondrocyte genetic stability and tumorigenicity. The growth factors were basic fibroblast growth factor (b-FGF), transforming growth factor β2 (TGF β2), insulin-like growth factor 1 (IGF-1), insulin-transferrin-selenium (ITS), and platelet-derived growth factor (PD-GF). Nasal septal chondrocytes cultured in growth factor cocktail exhibited a significantly high proliferative capacity. Comet assay revealed no significant DNA damage. Flow cytometry showed chondrocytes were mostly at G0-G1 phase, exhibiting normal cell cycle profile with no aneuploidy. We observed a decreased tumour suppressor genes’ expression (p53, p21, pRB) and no TP53 mutations or tumour formation after 6 months of implantation in nude mice. Our data suggest growth factor cocktail has a low risk of inducing genotoxic and tumorigenic effects on chondrocytes up to passage 6 with 16.6 population doublings. This preclinical tumorigenicity and genetic instability evaluation is crucial for further clinical works.
Collapse
Affiliation(s)
- Maimonah-Eissa Al-Masawa
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| | | | - Kien-Hui Chua
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
17
|
Identification of locations susceptible to osteoarthritis in patients with anterior cruciate ligament reconstruction: Combining knee joint computational modelling with follow-up T 1ρ and T 2 imaging. Clin Biomech (Bristol, Avon) 2020; 79:104844. [PMID: 31439361 DOI: 10.1016/j.clinbiomech.2019.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/28/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Finite element modelling can be used to evaluate altered loading conditions and failure locations in knee joint tissues. One limitation of this modelling approach has been experimental comparison. The aims of this proof-of-concept study were: 1) identify areas susceptible to osteoarthritis progression in anterior cruciate ligament reconstructed patients using finite element modelling; 2) compare the identified areas against changes in T2 and T1ρ values between 1-year and 3-year follow-up timepoints. METHODS Two patient-specific finite element models of knee joints with anterior cruciate ligament reconstruction were created. The knee geometry was based on clinical magnetic resonance imaging and joint loading was obtained via motion capture. We evaluated biomechanical parameters linked with cartilage degeneration and compared the identified risk areas against T2 and T1ρ maps. FINDINGS The risk areas identified by the finite element models matched the follow-up magnetic resonance imaging findings. For Patient 1, excessive values of maximum principal stresses and shear strains were observed in the posterior side of the lateral tibial and femoral cartilage. For Patient 2, high values of maximum principal stresses and shear strains of cartilage were observed in the posterior side of the medial joint compartment. For both patients, increased T2 and T1ρ values between the follow-up times were observed in the same areas. INTERPRETATION Finite element models with patient-specific geometries and motions and relatively simple material models of tissues were able to identify areas susceptible to post-traumatic knee osteoarthritis. We suggest that the methodology presented here may be applied in large cohort studies.
Collapse
|
18
|
Impaired chondrocyte U3 snoRNA expression in osteoarthritis impacts the chondrocyte protein translation apparatus. Sci Rep 2020; 10:13426. [PMID: 32778764 PMCID: PMC7417995 DOI: 10.1038/s41598-020-70453-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Although pathways controlling ribosome activity have been described to regulate chondrocyte homeostasis in osteoarthritis, ribosome biogenesis in osteoarthritis is unexplored. We hypothesized that U3 snoRNA, a non-coding RNA involved in ribosomal RNA maturation, is critical for chondrocyte protein translation capacity in osteoarthritis. U3 snoRNA was one of a number of snoRNAs with decreased expression in osteoarthritic cartilage and osteoarthritic chondrocytes. OA synovial fluid impacted U3 snoRNA expression by affecting U3 snoRNA gene promoter activity, while BMP7 was able to increase its expression. Altering U3 snoRNA expression resulted in changes in chondrocyte phenotype. Interference with U3 snoRNA expression led to reduction of rRNA levels and translational capacity, whilst induced expression of U3 snoRNA was accompanied by increased 18S and 28S rRNA levels and elevated protein translation. Whole proteome analysis revealed a global impact of reduced U3 snoRNA expression on protein translational processes and inflammatory pathways. For the first time we demonstrate implications of a snoRNA in osteoarthritis chondrocyte biology and investigated its role in the chondrocyte differentiation status, rRNA levels and protein translational capacity.
Collapse
|
19
|
Kang D, Shin J, Cho Y, Kim HS, Gu YR, Kim H, You KT, Chang MJ, Chang CB, Kang SB, Kim JS, Kim VN, Kim JH. Stress-activated miR-204 governs senescent phenotypes of chondrocytes to promote osteoarthritis development. Sci Transl Med 2020; 11:11/486/eaar6659. [PMID: 30944169 DOI: 10.1126/scitranslmed.aar6659] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 07/14/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022]
Abstract
A progressive loss of cartilage matrix leads to the development of osteoarthritis (OA). Matrix homeostasis is disturbed in OA cartilage as the result of reduced production of cartilage-specific matrix and increased secretion of catabolic mediators by chondrocytes. Chondrocyte senescence is a crucial cellular event contributing to such imbalance in matrix metabolism during OA development. Here, we identify miR-204 as a markedly up-regulated microRNA in OA cartilage. miR-204 is induced by transcription factors GATA4 and NF-κB in response to senescence signals. Up-regulated miR-204 simultaneously targets multiple components of the sulfated proteoglycan (PG) biosynthesis pathway, effectively shutting down PG anabolism. Ectopic expression of miR-204 in joints triggers spontaneous cartilage loss and OA development, whereas miR-204 inhibition ameliorates experimental OA, with concomitant recovery of PG synthesis and suppression of inflammatory senescence-associated secretory phenotype (SASP) factors in cartilage. Collectively, we unravel a stress-activated senescence pathway that underlies disrupted matrix homeostasis in OA cartilage.
Collapse
Affiliation(s)
- Donghyun Kang
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Jungkwon Shin
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Yongsik Cho
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Hyeon-Seop Kim
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Young-Ran Gu
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Haedong Kim
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Kwon Tae You
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Moon Jong Chang
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Boramae Hospital, 07061 Seoul, South Korea
| | - Chong Bum Chang
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Boramae Hospital, 07061 Seoul, South Korea
| | - Seung-Baik Kang
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Boramae Hospital, 07061 Seoul, South Korea
| | - Jong-Seo Kim
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - V Narry Kim
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea
| | - Jin-Hong Kim
- Center for RNA Research, Institute for Basic Science, 08826 Seoul, South Korea. .,Department of Biological Sciences, College of Natural Sciences, Seoul National University, 08826 Seoul, South Korea.,Interdisciplinary Program in Bioinformatics, Seoul National University, 08826 Seoul, South Korea
| |
Collapse
|
20
|
Xiao Z, Wang J, Chen S, Feng Y. Autophagy promotion enhances the protective effect of Morroniside on human OA chondrocyte. Biosci Biotechnol Biochem 2020; 84:989-996. [PMID: 31983285 DOI: 10.1080/09168451.2020.1717925] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Morroniside plays a therapeutic role in knee osteoarthritis (OA) by protecting chondrocytes. PI3K/AKT signaling is involved in the regulation of chondrocytes by Morroniside. PI3K/AKT suppresses autophagy through downstream signaling. However, the regulation of chondrocyte autophagy by Morroniside and the significance of the above effect on protecting chondrocytes aren't clear. The results showed that Morroniside inhibited the autophagiy of human OA chondrocytes. Besides, both PI3K inhibitors and mTOR inhibitors significantly reversed the autophagy reduced by Morroniside, but had no effect on the protective effect of Morroniside on chondrocytes. However, the enhanced autophagy caused by overexpression of autophagic genes enhanced the protective effect of Morroniside on chondrocytes. In conclusion, Morroniside represses the autophagy of human OA chondrocyte, which is related to PI3K/mTOR pathway. Moreover, the upregulation of autophagy further promoted the role of Morroniside in treating chondrocytes. Our data present a potential clue for the therapeutic strategies of Morroniside in treating OA.
Collapse
Affiliation(s)
- Zhanhao Xiao
- Department of Orthopedics, Fuzhou Second Hospital Affiliated Xiamen University Fuzhou, Fuzhou, Fujian, China
| | - Jiankun Wang
- Department of Orthopedics, Fuzhou Second Hospital Affiliated Xiamen University Fuzhou, Fuzhou, Fujian, China
| | - Sunyu Chen
- Department of Orthopedics, Fuzhou Second Hospital Affiliated Xiamen University Fuzhou, Fuzhou, Fujian, China
| | - Yang Feng
- Department of Orthopedics, Fuzhou Second Hospital Affiliated Xiamen University Fuzhou, Fuzhou, Fujian, China
| |
Collapse
|
21
|
Gao Y, Cui X, Wang M, Zhang Y, He Y, Li L, Li H, Zhang X, Cheng M. Oscillatory shear stress induces the transition of EPCs into mesenchymal cells through ROS/PKCζ/p53 pathway. Life Sci 2020; 253:117728. [PMID: 32353430 DOI: 10.1016/j.lfs.2020.117728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/14/2023]
Abstract
AIMS Studies indicate that the pattern of shear stress determines the direction of endothelial progenitor cells (EPCs) differentiation. However, the mechanism remains largely unknown. Herein, we try to identify the role of oscillatory shear stress (OSS) in the transdifferentiation of EPCs into mesenchymal cells and the mechanism involved. MATERIALS AND METHODS OSS was applied to EPCs using the flow chamber system in vitro. Matrigel, Boyden chamber, and healing assay were used to observe the changes in EPCs function. Further, 2',7'-dichlorofluorescein diacetate (DCFH-DA) probe and/or western blot were performed to detect the expression of reactive oxygen species (ROS), p53 and PKCζ in EPCs. EPCs transduced with Lentivirus carrying Tp53 were implanted into the arterial vessel in the balloon injured rat model, and neointimal thickening was verified by HE staining. KEY FINDINGS OSS enhanced the expression of mesenchymal cell markers alpha-smooth muscle actin (α-SMA) and smooth muscle 22 alpha (SM22α) on EPCs. In the meantime, OSS time-dependently decreased p53 expression in EPCs, which was partially abolished by treatment with ROS scavenger N-acetylcysteine (NAC) or protein kinase C zeta (PKCζ) inhibitor Go6983. Moreover, the p53 agonist tenovin-1 attenuated the changes of OSS-mediated the mesenchymal cell markers and EPCs function. Besides, we also found that transplanting EPCs transfected with LV-Tp53 significantly inhibited neointimal thickening and promoted reendothelialization in vivo. SIGNIFICANCE This study demonstrates OSS-induced EPC transdifferentiation into mesenchymal cells and ROS/PKCζ/p53 pathway play an essential role in it. It may serve as a promising therapeutic target for cardiovascular disease in the future.
Collapse
Affiliation(s)
- Yu Gao
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China
| | - Xiaodong Cui
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China
| | - Meiyue Wang
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China
| | - Yaowen Zhang
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China
| | - Yanting He
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China
| | - Lanlan Li
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China
| | - Hong Li
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China
| | - Xiaoyun Zhang
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China.
| | - Min Cheng
- School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, PR China.
| |
Collapse
|
22
|
Li HZ, Xu XH, Lin N, Wang DW, Lin YM, Su ZZ, Lu HD. Overexpression of miR-10a-5p facilitates the progression of osteoarthritis. Aging (Albany NY) 2020; 12:5948-5976. [PMID: 32283545 PMCID: PMC7185093 DOI: 10.18632/aging.102989] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 03/02/2020] [Indexed: 12/21/2022]
Abstract
The current study was aimed at exploring the potential roles and possible mechanisms of miR-10a-5p in osteoarthritis (OA). We performed RT-qPCR, Western blot, CCK8, EdU Assay, and flow cytometry assay to clarify the roles of miR-10a-5p in OA. Furthermore, the whole transcriptome sequencing together with integrated bioinformatics analyses were conducted to elucidate the underlying mechanisms of miR-10a-5p involving in OA. Our results demonstrated that miR-10a-5p was upregulated in OA and acted as a significant contributing factor for OA. A large number of circRNAs, lncRNAs, miRNAs, and mRNAs were identified by overexpressing miR-10a-5p. Functional enrichment analyses indicated that these differentially-expressed genes were enriched in some important terms including PPAR signaling pathway, PI3K-Akt signaling pathway, and p53 signaling pathway. A total of 42 hub genes were identified in the protein-protein interaction network including SERPINA1, TTR, APOA1, and A2M. Also, we constructed the network regulatory interactions across coding and noncoding RNAs triggered by miR-10a-5p, which revealed the powerful regulating effects of miR-10a-5p. Moreover, we found that HOXA3 acted as the targeted genes of miR-10a-5p and miR-10a-5p contributed to the progression of OA by suppressing HOXA3 expression. Our findings shed insight on regulatory mechanisms of miR-10a-5p, which might provide novel therapeutic targets for OA.
Collapse
Affiliation(s)
- Hui-Zi Li
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Department of Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Xiang-He Xu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Department of Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Nan Lin
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Department of Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Da-Wei Wang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Yi-Ming Lin
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Zhong-Zhen Su
- Department of Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Department of Medical Ultrasonics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Hua-Ding Lu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Department of Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| |
Collapse
|
23
|
Rim YA, Nam Y, Ju JH. The Role of Chondrocyte Hypertrophy and Senescence in Osteoarthritis Initiation and Progression. Int J Mol Sci 2020; 21:ijms21072358. [PMID: 32235300 PMCID: PMC7177949 DOI: 10.3390/ijms21072358] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease that causes pain and disability in the adult population. OA is primarily caused by trauma induced by an external force or by age-related cartilage damage. Chondrocyte hypertrophy or chondrocyte senescence is thought to play a role in the initiation and progression of OA. Although chondrocyte hypertrophy and cell death are both crucial steps during the natural process of endochondral bone formation, the abnormal activation of these two processes after injury or during aging seems to accelerate the progression of OA. However, the exact mechanisms of OA progression and these two processes remain poorly understood. Chondrocyte senescence and hypertrophy during OA share various markers and processes. In this study, we reviewed the changes that occur during chondrocyte hypertrophy or senescence in OA and the attempts that were made to regulate them. Regulation of hypertrophic or senescent chondrocytes might be a potential therapeutic target to slow down or stop OA progression; thus, a better understanding of the processes is required for management.
Collapse
Affiliation(s)
- Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.A.R.); (Y.N.)
| | - Yoojun Nam
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.A.R.); (Y.N.)
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (Y.A.R.); (Y.N.)
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-6895
| |
Collapse
|
24
|
Lou C, Deng A, Zheng H, Sun G, Zhao H, Li A, Liu Q, Li Y, Lv Z. Pinitol suppresses TNF-α-induced chondrocyte senescence. Cytokine 2020; 130:155047. [PMID: 32200264 DOI: 10.1016/j.cyto.2020.155047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/27/2019] [Accepted: 02/19/2020] [Indexed: 01/03/2023]
Abstract
Osteoarthritis (OA) is a highly prevalent joint disorder that is tightly correlated with age. As the body ages, cell replication and function decline until homeostasis can no longer be maintained. This process involves cellular senescence as well as replicative senescence. Telomere length, cell cycle arrest, expression of p16 and p53, and the release of senescence-associated β-galactosidase (SA-β-Gal) are all markers of cell senescence. In OA joints, chondrocytes undergo cellular senescence prematurely, thereby ceasing to synthesize and maintain cartilage tissue. Upregulation of proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α), and oxidative stress induced by overproduction of reactive oxygen species (ROS) are key events in the pathogenesis of OA. In the present study, we investigated the effects of pinitol, a naturally occurring compound, on the effects of TNF-α on chondrocyte senescence and cell cycle arrest. We found that pinitol has a favorable safety profile in terms of cell viability. Pinitol significantly inhibited cellular senescence and cell cycle arrest in the G0/G1 phase induced by TNF-α. We also found that pinitol could inhibit TNF-α-induced increased telomerase activity and expression of p16 and p53. Importantly, we found that the effects of pinitol may be mediated through rescue of Nrf2 signaling, which is recognized as a key protective factor in OA. This finding was verified through a Nrf2 silencing experiment using Nrf2 siRNA. Together, our findings reveal the potential of pinitol as a safe therapeutic option for the prevention of OA-associated chondrocyte senescence and oxidative stress.
Collapse
Affiliation(s)
- Chunbiao Lou
- Department of Orthopaedics, Heze Third People Hospital, Heze, Shandong 274031, China
| | - Aiwei Deng
- Department of Orthopaedics, Heze Third People Hospital, Heze, Shandong 274031, China
| | - Huiming Zheng
- Department of Orthopaedics, Heze Third People Hospital, Heze, Shandong 274031, China
| | - Guiying Sun
- Department of Orthopaedics, Heze Third People Hospital, Heze, Shandong 274031, China
| | - Huaqin Zhao
- Department of Orthopaedics, Heze Third People Hospital, Heze, Shandong 274031, China
| | - Aixia Li
- Department of Orthopaedics, Heze Third People Hospital, Heze, Shandong 274031, China
| | - Qian Liu
- Department of Orthopaedics, Heze Third People Hospital, Heze, Shandong 274031, China
| | - Yang Li
- Department of Orthopaedics, Heze Third People Hospital, Heze, Shandong 274031, China
| | - Zhiqiang Lv
- Department of Pharmacy, The Affiliated Hospital Of Qingdao University, Qingdao, Shandong 274031, China.
| |
Collapse
|
25
|
Zhao X, Huang P, Li G, Feng Y, Zhendong L, Zhou C, Hu G, Xu Q. Overexpression of Pitx1 attenuates the senescence of chondrocytes from osteoarthritis degeneration cartilage-A self-controlled model for studying the etiology and treatment of osteoarthritis. Bone 2020; 131:115177. [PMID: 31783149 DOI: 10.1016/j.bone.2019.115177] [Citation(s) in RCA: 10] [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: 05/25/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
To explore the role of low expression of Pitx1 in degenerative cartilage tissue. A cartilage injury model was established by using the cartilage scratch method. The newly generated tissue by BrdU labeled in injured cartilage region expressed SOX-9 and Col2A1 in 5-week-old rats. Compared with that, the number of BrdU-positive cells was lower in 4-month-old cartilage injury model rats. Compared with that in lateral cartilage, the expression of Pitx1 was lower in medial cartilage. Compared with chondrocytes derived from the lateral cartilage, chondrocytes derived from the medial cartilage exhibited significantly increased cell aging, as determined by SA-β-GAL staining; downregulated Pitx1 expression; reduced autophagy levels; and decreased Col2A1 expression in a chondrogenic differentiation assay. Inhibition of Pitx1 expression in chondrocytes from the lateral cartilage significantly increased the ratio of cell senescence. Overexpression of Pitx1 in chondrocytes derived from the medial cartilage decreased the cell senescence ratio. In a luciferase assay, Pitx1 was found to promote Sirt1 gene transcription. Decreased Pitx1 expression is an essential cause of cartilage degeneration in the medial tibial plateau. The described self-controlled model is an excellent way to study OA etiology and screen therapeutic drugs for OA.
Collapse
Affiliation(s)
- Xiang Zhao
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Huang
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gen Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yu Feng
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lv Zhendong
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chun Zhou
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guangyu Hu
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qingrong Xu
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai JiaoTong University, 160 Pujian Road, Shanghai 200127, China; Department of Orthopaedics, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2000, Jiangyue Road, Shanghai 201112, China.
| |
Collapse
|
26
|
Argote PF, Kaplan JT, Poon A, Xu X, Cai L, Emery NC, Pierce DM, Neu CP. Chondrocyte viability is lost during high-rate impact loading by transfer of amplified strain, but not stress, to pericellular and cellular regions. Osteoarthritis Cartilage 2019; 27:1822-1830. [PMID: 31526876 PMCID: PMC7028439 DOI: 10.1016/j.joca.2019.07.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Deleterious impact loading to cartilage initiates post-traumatic osteoarthritis (OA). While cytokine and enzyme levels regulate disease progression, specific mechanical cues that elucidate cellular OA origins merit further investigation. We defined the dominant pericellular and cellular strain/stress transfer mechanisms following bulk-tissue injury associated with cell death. METHOD Using an in vitro model, we investigated rate-dependent loading and spatial localization of cell viability in acute indentation and time-course studies. Atomic force microscopy (AFM) and magnetic resonance imaging (MRI) confirmed depth-wise changes in cartilage micro-/macro-mechanics and structure post-indentation. To understand the transfer of loading to cartilage domains, we computationally modeled full-field strain and stress measures in interstitial matrix, pericellular and cellular regions. RESULTS Chondrocyte viability decreased following rapid impact (80%/s) vs slow loading (0.1%/s) or unloaded controls. Viability was lost immediately during impact within regions near the indenter-tissue contact but did not change over 7 days of tissue culture. AFM studies revealed a loss of stiffness following 80%/s loading, and MRI studies confirmed an increased tensile and shear strain, but not relaxometry. Image-based patterns of chondrocyte viability closely matched computational estimates of amplified maximum principal and shear strain in interstitial matrix, pericellular and cellular regions. CONCLUSION Rapid indentation worsens chondrocyte death and degrades cartilage matrix stiffness in indentation regions. Cell death at high strain rates may be driven by elevated tensile strains, but not matrix stress. Strain amplification beyond critical thresholds in the pericellular matrix and cells may define a point of origin for early damage in post-traumatic OA.
Collapse
Affiliation(s)
- Pablo F. Argote
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jonathan T. Kaplan
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA,Biomechanics Research and Engineering, Natick Soldier RD&E Center, Natick, MA, USA
| | - Alan Poon
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Xin Xu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA,Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Luyao Cai
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Nancy C. Emery
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA
| | - David M. Pierce
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA,Department of Mechanical Engineering, University of Connecticut, Storrs, CT, USA,Corresponding Authors: Corey P. Neu, Tel: (303) 492-7330, , and David M. Pierce, Tel: (860) 486-5088,
| | - Corey P. Neu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA,Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA,Corresponding Authors: Corey P. Neu, Tel: (303) 492-7330, , and David M. Pierce, Tel: (860) 486-5088,
| |
Collapse
|
27
|
Phen HM, Schenker ML. Minimizing Posttraumatic Osteoarthritis After High-Energy Intra-Articular Fracture. Orthop Clin North Am 2019; 50:433-443. [PMID: 31466660 DOI: 10.1016/j.ocl.2019.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This article serves to provide an overview of molecular and surgical interventions to minimize the progression of posttraumatic arthritis following high-energy intra-articular fractures. The roles of cartilage and the microcellular environment are discussed, as well as the response of the joint and cartilage to injury. Molecular therapies, such as glucocorticoids, mesenchymal stem cells, and bisphosphonates, are presented as potential treatments to prevent progression to posttraumatic arthritis. High-energy intra-articular fractures of the elbow, hip, knee, and ankle are discussed, with emphasis on restoring anatomic alignment, articular reduction, and stability of the joint.
Collapse
Affiliation(s)
- Huai Ming Phen
- Emory Orthopaedic Trauma & Fracture, 49 Jesse Hill Jr. Drive South East, 3rd Floor, Atlanta, GA 30303, USA.
| | - Mara L Schenker
- Emory Orthopaedic Trauma & Fracture, 49 Jesse Hill Jr. Drive South East, 3rd Floor, Atlanta, GA 30303, USA
| |
Collapse
|
28
|
PKR Promotes Oxidative Stress and Apoptosis of Human Articular Chondrocytes by Causing Mitochondrial Dysfunction through p38 MAPK Activation-PKR Activation Causes Apoptosis in Human Chondrocytes. ANTIOXIDANTS (BASEL, SWITZERLAND) 2019. [PMID: 31484360 DOI: 10.3390/antiox8090370.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Osteoarthritis (OA) is one of the most common types of arthritis in the elderly people. It has been known that chondrocyte apoptosis occurs in OA cartilage; however, the detailed molecular mechanism remains unclear. In the current study, we aimed to elucidate the role of double-stranded RNA-dependent protein kinase R (PKR) in the TNF-α-caused apoptosis in chondrocytes. Human articular chondrocytes were digested from cartilages of OA subjects who accepted arthroplastic knee surgery. Our results showed that phosphorylation of p38 MAPK was increased after TNF-α stimulation or PKR activation using poly (I:C), and TNF-α-induced p38 MAPK upregulation was inhibited by PKR inhibition, suggesting phosphor-p38 MAPK was regulated by PKR. Moreover, we found that PKR participated in the p53-dependent destruction of AKT following activation of p38 MAPK. The inhibition of AKT led to the reduced expression of PGC-1α, which resulted in mitochondrial dysfunction and increased oxidative stress. We showed that the reduction of oxidative stress using antioxidant Mito TEMPO lowered the TNF-α-induced caspase-3 activation and TUNEL-positive apoptotic cells. The diminished apoptotic response was also observed after repression of PKR/p38 MAPK/p53/AKT/PGC-1α signaling. Taken together, we demonstrated that the aberrant mitochondrial biogenesis and increased oxidative stress in chondrocytes after TNF-α stimulation were mediated by PKR, which may contribute to the chondrocyte apoptosis and cartilage degeneration in OA.
Collapse
|
29
|
PKR Promotes Oxidative Stress and Apoptosis of Human Articular Chondrocytes by Causing Mitochondrial Dysfunction through p38 MAPK Activation-PKR Activation Causes Apoptosis in Human Chondrocytes. Antioxidants (Basel) 2019; 8:antiox8090370. [PMID: 31484360 PMCID: PMC6769915 DOI: 10.3390/antiox8090370] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/14/2019] [Accepted: 08/28/2019] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) is one of the most common types of arthritis in the elderly people. It has been known that chondrocyte apoptosis occurs in OA cartilage; however, the detailed molecular mechanism remains unclear. In the current study, we aimed to elucidate the role of double-stranded RNA-dependent protein kinase R (PKR) in the TNF-α-caused apoptosis in chondrocytes. Human articular chondrocytes were digested from cartilages of OA subjects who accepted arthroplastic knee surgery. Our results showed that phosphorylation of p38 MAPK was increased after TNF-α stimulation or PKR activation using poly (I:C), and TNF-α-induced p38 MAPK upregulation was inhibited by PKR inhibition, suggesting phosphor-p38 MAPK was regulated by PKR. Moreover, we found that PKR participated in the p53-dependent destruction of AKT following activation of p38 MAPK. The inhibition of AKT led to the reduced expression of PGC-1α, which resulted in mitochondrial dysfunction and increased oxidative stress. We showed that the reduction of oxidative stress using antioxidant Mito TEMPO lowered the TNF-α-induced caspase-3 activation and TUNEL-positive apoptotic cells. The diminished apoptotic response was also observed after repression of PKR/p38 MAPK/p53/AKT/PGC-1α signaling. Taken together, we demonstrated that the aberrant mitochondrial biogenesis and increased oxidative stress in chondrocytes after TNF-α stimulation were mediated by PKR, which may contribute to the chondrocyte apoptosis and cartilage degeneration in OA.
Collapse
|
30
|
Maier F, Lewis CG, Pierce DM. Through-thickness patterns of shear strain evolve in early osteoarthritis. Osteoarthritis Cartilage 2019; 27:1382-1391. [PMID: 31121293 DOI: 10.1016/j.joca.2019.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 04/04/2019] [Accepted: 04/27/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Given the structural changes associated with the progression of Osteoarthritis (OA), we hypothesized that patterns of through-thickness, large-strain shear evolve with early-stage OA. We therefore aimed to determine whether and how patterns of shear strains change during early-stage OA to 1) gain insight into the progression of OA by quantifying changes in local deformations; 2) gauge the potential of patterns in shear strain to serve as image-based biomarkers of early-stage OA; and 3) provide high-resolution, through-thickness data for proposing, fitting, and validating constitutive models for cartilage. DESIGN We completed displacement-driven, large-strain shear tests (5, 10, 15%) on 44 specimens of variably advanced osteoarthritic human articular cartilage as determined by both Osteoarthritis Research Society International (OARSI) grade and PLM-CO score. We recorded the through-thickness deformations with a stereo-camera system and processed these data using digital image correlation (DIC) to determine full-thickness patterns of strains and relative zonal recruitments, i.e., the average shear strain in a through-thickness zone weighted by its relative thickness and normalized by the applied strain. RESULTS We observed three general shapes for the curves of averaged through-thickness, Green-Lagrange shear strains during progression of OA. We also observed that during the progression of OA only the deep zone is recruited differently under shear in a statistically significant way. CONCLUSIONS We propose that changes in through-thickness patterns of shear strain could provide sensitive biomarkers for early clinical detection of OA. The relative zonal recruitment of the deep zone decreases with progressing OA (OARSI grade) and microstructural remodeling (PLM-CO score), which do not consistently affect recruitment of the superficial and middle zones.
Collapse
Affiliation(s)
- F Maier
- University of Connecticut, Department of Mechanical Engineering, Storrs, CT, USA
| | - C G Lewis
- Hartford Healthcare, Bone & Joint Institute, Hartford, CT, USA
| | - D M Pierce
- University of Connecticut, Department of Mechanical Engineering, Storrs, CT, USA; University of Connecticut, Department of Biomedical Engineering, Storrs, CT, USA.
| |
Collapse
|
31
|
Park MJ, Moon SJ, Baek JA, Lee EJ, Jung KA, Kim EK, Kim DS, Lee JH, Kwok SK, Min JK, Kim SJ, Park SH, Cho ML. Metformin Augments Anti-Inflammatory and Chondroprotective Properties of Mesenchymal Stem Cells in Experimental Osteoarthritis. THE JOURNAL OF IMMUNOLOGY 2019; 203:127-136. [PMID: 31142603 DOI: 10.4049/jimmunol.1800006] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/01/2019] [Indexed: 01/15/2023]
Abstract
Mesenchymal stem cells (MSCs) can protect against cartilage breakdown in osteoarthritis (OA) via their immunomodulatory capacities. However, the optimization strategy for using MSCs remains challenging. This study's objective was to identify the in vivo effects of metformin-stimulated adipose tissue-derived human MSCs (Ad-hMSCs) in OA. An animal model of OA was established by intra-articular injection of monosodium iodoacetate into rats. OA rats were divided into a control group and two therapy groups (treated with Ad-hMSCs or metformin-stimulated Ad-hMSCs). Limb nociception was assessed by measuring the paw withdrawal latency and threshold. Our data show that metformin increased IL-10 and IDO expression in Ad-hMSCs and decreased high-mobility group box 1 protein, IL-1β, and IL-6 expression. Metformin increased the migration capacity of Ad-hMSCs with upregulation of chemokine expression. In cocultures, metformin-stimulated Ad-hMSCs inhibited the mRNA expression of RUNX2, COL X, VEGF, MMP1, MMP3, and MMP13 in IL-1β-stimulated OA chondrocytes and increased the expression of TIMP1 and TIMP3. The antinociceptive activity and chondroprotective effects were greater in OA rats treated with metformin-stimulated Ad-hMSCs than in those treated with unstimulated Ad-hMSCs. TGF-β expression in subchondral bone of OA joints was attenuated more in OA rats treated with metformin-stimulated Ad-hMSCs. Our findings suggest that metformin offers a promising option for the clinical application of Ad-hMSCs as a cell therapy for OA.
Collapse
Affiliation(s)
- Min-Jung Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Jin-Ah Baek
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Eun-Jung Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Kyung-Ah Jung
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Eun-Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Da-Som Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Jung-Ho Lee
- Department of Plastic and Reconstructive Surgery, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Jun-Ki Min
- Division of Rheumatology, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea; and
| | - Seok Jung Kim
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea;
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea;
| |
Collapse
|
32
|
Bi J, Cai W, Ma T, Deng A, Ma P, Han Y, Lou C, Wu L. Protective effect of vildagliptin on TNF-α-induced chondrocyte senescence. IUBMB Life 2019; 71:978-985. [PMID: 31026379 DOI: 10.1002/iub.2049] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/18/2019] [Accepted: 02/25/2019] [Indexed: 12/21/2022]
Abstract
Osteoarthritis (OA) is a common age-related disorder. Chondrocytes in joint tissue play a critical role in normal articular cartilage function and tissue homeostasis. Local inflammatory cytokine-induced chondrocyte senescence contributes to the development and progression of OA. Various dipeptidyl peptidase-4 (DPP-4) inhibitors have been widely used to treat type 2 diabetes. Here, we report a novel pharmacological role of the DPP-4 inhibitor vildagliptin in chondrocyte senescence. Our data indicate that DPP-4 is an inducible factor responsive to tumor necrosis factor-α (TNF-α) treatment in chondrocytes. The inhibition of DPP-4 by vildagliptin ameliorates TNF-α-induced chondrocyte senescence as determined by cellular senescence-associated β-galactosidase (SA-β-Gal) activity. Vildagliptin displayed protective capabilities against TNF-α-induced chondrocyte cell cycle arrest in the G1 phase. Moreover, vildagliptin suppresses the three major TNF-α-induced chondrocyte senescence proteins including p53, p21, and plasminogen activator inhibitor-1 (PAI-1). Vildagliptin also suppresses TNF-α-induced p53 acetylation at K382. Consistently, our findings demonstrate the inhibitory effect of vildagliptin on p53 acetylation, which is mediated by sirtuin 1 (SIRT1) as the inhibition of SIRT1 negated the inhibitory action of vildagliptin on p53 acetylation. Furthermore, we found that the effect of vildagliptin on SIRT1 protection is adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) dependent, and the inhibition of AMPK activity negated the protection of vildagliptin against SIRT1 and chondrocytes senescence. In conclusion, our study explored the molecular mechanism and protective effect of the antidiabetic drug vildagliptin against chondrocyte senescence, and our findings imply that vildagliptin has a therapeutic potential in OA. © 2019 IUBMB Life, 1-2, 2019.
Collapse
Affiliation(s)
- Jianping Bi
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong, China
| | - Wusheng Cai
- Department of Orthopedics, Heze Third People Hospital, Heze, Shandong, China
| | - Teng Ma
- Department of Orthopedics, Heze Third People Hospital, Heze, Shandong, China
| | - Aiwei Deng
- Department of Bone Surgery, Heze Third People Hospital, Heze, Shandong, China
| | - Ping Ma
- School of Foreign Languages, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ye Han
- Department of Emergency, Hiser Medical Center of Qingdao, Qingdao, Shandong, China
| | - Chunbiao Lou
- Department of Bone Surgery, Heze Third People Hospital, Heze, Shandong, China
| | - Leilei Wu
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
33
|
Kornicka K, Al Naem M, Röcken M, Zmiertka M, Marycz K. Osteochondritis Dissecans (OCD)-Derived Chondrocytes Display Increased Senescence, Oxidative Stress, Chaperone-Mediated Autophagy and, in Co-Culture with Adipose-Derived Stem Cells (ASCs), Enhanced Expression of MMP-13. J Clin Med 2019; 8:jcm8030328. [PMID: 30857162 PMCID: PMC6462951 DOI: 10.3390/jcm8030328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
Osteochondritis dissecans (OCD) in equids, especially in sport horses, has become a growing issue as it contributes to the occurrence of lameness. Thus the aim of this study was to investigate the cytophysiological properties of OCD chondrocytes including expression of chondrogenic genes, apoptosis, mitochondria dynamics and autophagy. Horse chondrocytes were isolated from healthy (HE) and OCD cartilages. Properties of cells were evaluated using multiple assays e.g., polymerase chain reaction (PCR), immunofluorescence, Western blot. OCD chondrocytes were characterized by increased apoptosis and senescence. Expression of chondrogenic genes (vimentin, aggrecan) was decreased while mRNA levels of matrix metalloproteinase 13 significantly upregulated in comparison to HE cells. Moreover, OCD cells displayed increased mitochondrial fusion while fission events were diminished. Interestingly, chaperone mediated autophagy was triggered in those cells and it predominated over macroautophagy. Furthermore, co-culture of LPS-treated chondrocytes with adipose-derived stem cells (ASC) decreased p62/sequestosome 1 (SQSTM) and increases MMP-13 expression in OCD cells. Our results suggest that OCD affected horse chondrocytes are characterized by senescent phenotype due to endoplasmic reticulum stress and mitochondria dynamics deterioration. Expression of chondrogenic markers is decreased in those cells while expression of chaperone mediated autophagy (CMA)-related genes increased. Increased malfunctioning of cells leads to loss of their functionality and capacity to maintain tissue homeostasis.
Collapse
Affiliation(s)
- Katarzyna Kornicka
- International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114 Wisznia Mała, Poland.
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392 Gießen, Germany.
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392 Gießen, Germany.
| | - Marta Zmiertka
- International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114 Wisznia Mała, Poland.
| | - Krzysztof Marycz
- International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114 Wisznia Mała, Poland.
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392 Gießen, Germany.
| |
Collapse
|
34
|
Song W, Zhang Y, Wang J, Ma T, Hao L, Wang K. Antagonism of cysteinyl leukotriene receptor 1 (cysLTR1) by montelukast suppresses cell senescence of chondrocytes. Cytokine 2018; 103:83-89. [PMID: 29331588 DOI: 10.1016/j.cyto.2017.12.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/13/2017] [Accepted: 12/20/2017] [Indexed: 12/22/2022]
Abstract
Aging is closely associated with osteoarthritis (OA). Although its underlying mechanisms remain unknown, cellular senescence in chondrocytes has become an important therapeutic target for the treatment of OA. Cysteinyl leukotriene receptors (cysLTRs) mediate the pathobiological function of cysteinyl leukotrienes (cysLTs). However, the roles of cysLTRs in the pathogenesis of OA have not been reported before. In the current study, we found that cysLTR1 but not cysLTR2 is expressed in human primary chondrocytes. In addition, stimulation with tumor necrosis factor α (TNF-α) resulted in a significant increase in the expression of cysLTR1. Interestingly, montelukast, a specific cysLTR1 antagonist, attenuated TNF-α-induced up-regulation of the activity of senescence-associated β-galactosidase (SA-β-Gal). In addition, TNF-α led to cell cycle arrest at the G0/G1 phase, which was prevented by treatment with montelukast. Notably, montelukast reduced expression of the senescence markers p53, p21 and PAI-1. In addition, montelukast ameliorated TNF-α-induced K382 acetylation of p53 by promoting the expression of SIRT1. Silencing of SIRT1 using SIRT1 siRNA broke the inhibitory effects of montelukast on K382 acetylation of p53. Importantly, silencing of cysLTR1 reversed the reduction of SIRT1 expression as well as the K382 acetylation of p53. Our findings strongly implicate that cysLTR1 has the capacity to regulate cellular senescence in chondrocytes. It is suggested that montelukast may be a potential therapeutic agent for chondro-protective therapy.
Collapse
Affiliation(s)
- Wei Song
- Medical School, Xi'an Jiaotong University, Xi'an 710054, China
| | - Yumin Zhang
- Department of Joint Surgery, Xi'an Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Jun Wang
- Department of Joint Surgery, Xi'an Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Tao Ma
- Department of Joint Surgery, Xi'an Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Linjie Hao
- Department of Joint Surgery, Xi'an Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Kunzheng Wang
- First Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, China.
| |
Collapse
|
35
|
Abstract
Osteoarthritis is characterized by a chronic, progressive and irreversible degradation of the articular cartilage associated with joint inflammation and a reparative bone response. More than 100 million people are affected by this condition worldwide with significant health and welfare costs. Our available treatment options in osteoarthritis are extremely limited. Chondral or osteochondral grafts have shown some promising results but joint replacement surgery is by far the most common therapeutic approach. The difficulty lies on the limited regeneration capacity of the articular cartilage, poor blood supply and the paucity of resident progenitor stem cells. In addition, our poor understanding of the molecular signalling pathways involved in the senescence and apoptosis of chondrocytes is a major factor restricting further progress in the area. This review focuses on molecules and approaches that can be implemented to delay or even rescue chondrocyte apoptosis. Ways of modulating the physiologic response to trauma preventing chondrocyte death are proposed. The use of several cytokines, growth factors and advances made in altering several of the degenerative genetic pathways involved in chondrocyte apoptosis and degradation are also presented. The suggested approaches can help clinicians to improve cartilage tissue regeneration.
Collapse
Affiliation(s)
- Ippokratis Pountos
- Academic Department of Trauma & Orthopaedics, School of Medicine, University of Leeds, UK.
| | - Peter V Giannoudis
- Academic Department of Trauma & Orthopaedics, School of Medicine, University of Leeds, UK; NIHR Leeds Biomedical Research Center, Chapel Allerton Hospital, Leeds, UK.
| |
Collapse
|
36
|
Neidlin M, Korcari A, Macheras G, Alexopoulos LG. Cue-Signal-Response Analysis in 3D Chondrocyte Scaffolds with Anabolic Stimuli. Ann Biomed Eng 2017; 46:345-353. [PMID: 29147820 DOI: 10.1007/s10439-017-1964-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/14/2017] [Indexed: 11/25/2022]
Abstract
Articular cartilage is an avascular connective tissue responsible for bearing loads. Cell signaling plays a central role in cartilage homeostasis and tissue engineering by directing chondrocytes to synthesize/degrade the extracellular matrix or promote inflammatory responses. The aim of this paper was to investigate anabolic, catabolic and inflammatory pathways of well-known and underreported anabolic stimuli in 3D chondrocyte cultures and connect them to diverse cartilage responses including matrix regeneration and cell communication. A cue-signal-response experiment was performed in chondrocytes embedded in alginate scaffolds subjected to a 9-day treatment with 7 anabolic cues. At the signaling level diverse pathways were measured whereas at the response level glycosaminoglycan (GAG) synthesis and cytokine releases were monitored. A significant increase of GAG was observed for each stimulus and well known anabolic phosphoproteins were activated. In addition, WNK1, an underreported protein of chondrocyte signaling, was uncovered. At the extracellular level, inflammatory and regulating cytokines were measured and DEFB1 and CXCL10 were identified as novel contributors to chondrocyte responses, both closely linked to TLR signaling and inflammation. Finally, two new pro-growth factors with an inflammatory potential, Cadherin-11 and MGP were observed. Interestingly, well-known anabolic stimuli yielded inflammatory responses which pinpoints to the pleiotropic roles of individual stimuli.
Collapse
Affiliation(s)
- Michael Neidlin
- Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece
| | - Antonion Korcari
- Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece
| | | | - Leonidas G Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece.
| |
Collapse
|
37
|
Lin M, Lin Y, Li X, Liang W, Wang S, Liu J, Liu X, Chen L, Qin Y. Warm sparse-dense wave inhibits cartilage degradation in papain-induced osteoarthritis through the mitogen-activated protein kinase signaling pathway. Exp Ther Med 2017; 14:3674-3680. [PMID: 29042963 PMCID: PMC5639397 DOI: 10.3892/etm.2017.4984] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 06/02/2017] [Indexed: 11/05/2022] Open
Abstract
Cartilage degradation is an important in the pathogenesis of osteoarthritis (OA). Abnormal activation of the mitogen-activated protein kinase (MAPK) signaling pathway in chondrocytes promotes an inflammatory response, resulting in the release of chondral matrix-degrading enzymes that accelerate the degradation of cartilage. As a non-pharmaceutical and non-invasive physical therapy regimen, warm sparse-dense wave (WSDW) has been successfully used for the treatment of OA. However, it remains unclear whether WSDW inhibits cartilage degradation in OA through the MAPK signaling pathway. The present study investigated the effects of WSDW on papain-induced OA in rat knee joints. Papain-induced OA was established in rats, which were subsequently divided into a model group and three experimental groups that received a WSDW with the following ratios: WSDW=1:1, WSDW=1:2 and WSDW=2:1. After 12 weeks of treatment, cartilage degradation was evaluated by Mankin scoring of paraffin-embedded sections stained with hematoxylin and eosin. The changes in cartilage structure were observed by transmission electron microscopy, and the expressions of RAS, extracellular signal-regulated kinase (ERK), p38 and p53 were measured by reverse transcription-quantitative polymerase chain reaction and western blot analysis. WSDW was demonstrated to improve the arrangement of collagen fibers, inhibit the tidemark replication and delay cartilage degradation in papain-induced OA. The expressions of RAS, ERK, p38 and p53 in the WSDW (1:2) and (2:1) groups were significantly decreased when compared with the model group (P<0.01). Furthermore, amongst the WSDW groups, the inhibitory effects of the WSDW (1:2) group were typically greater than those of the WSDW (1:1) and (2:1) groups. The results indicate that WSDW may inhibit cartilage degradation in papain-induced OA in rat knee joints by regulating the MAPK signaling pathway.
Collapse
Affiliation(s)
- Munan Lin
- Department of Traditional Chinese Medicine, Fuzhou General Hospital of Nanjing Military Command, Fuzhou, Fujian 350025, P.R. China
| | - Yanhong Lin
- Department of Internal Medicine, Shanghang Hospital, Shanghang, Fujian 364200, P.R. China
| | - Xihai Li
- Institute of Bone Diseases, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Wenna Liang
- Research Base of Traditional Chinese Medicine Syndrome, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Shuiliang Wang
- Laboratory Department, Fuzhou General Hospital of Nanjing Military Command, Fuzhou, Fujian 350025, P.R. China
| | - Jiansheng Liu
- Institute of Bone Diseases, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xianxiang Liu
- Institute of Bone Diseases, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Lidian Chen
- Institute of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yin Qin
- Department of Traditional Chinese Medicine, Fuzhou General Hospital of Nanjing Military Command, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
38
|
Jia PT, Zhang XL, Zuo HN, Lu X, Li L. Articular cartilage degradation is prevented by tanshinone IIA through inhibiting apoptosis and the expression of inflammatory cytokines. Mol Med Rep 2017; 16:6285-6289. [PMID: 28849083 DOI: 10.3892/mmr.2017.7340] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 06/14/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effect of tanshinone IIA on the degradation of articular cartilage in a rat model of osteoarthritis (OA). The OA rat model was established by anterior cruciate ligament transection (ACLT) and medial meniscus resection (MMx). The animals were treated for 28 days with 0.25‑0.5 mg/kg doses of tanshinone IIA following ACLT + MMx. The knee joints of the rats in the ACLT + MMx group exhibited marked alterations in articular cartilage histopathology and higher Mankin scores, compared with those in the normal group. Tanshinone IIA treatment at a dose of 0.5 mg/kg significantly inhibited cartilage degradation and improved Mankin scores in the OA rat model (P<0.002). Tanshinone IIA treatment completely inhibited the ACLT + MMx‑induced accumulation of inflammatory cells and disintegration of synovial lining in the rats. An increase in the dose of tanshinone IIA between 0.25 and 0.5 mg/kg reduced the proportion of apoptotic chrondrocytes from 41 to 2% on day 29. Treatment of the rats in the ACLT + MMx group with 0.5 mg/kg doses of tanshinone IIA markedly inhibited the expression level of matrix metalloproteinase and increased the expression of tissue inhibitor of metalloproteinase in the rat articular cartilage tissues. Tanshinone IIA treatment significantly reduced the levels of inflammatory cytokines, including interleukin‑1β, tumor necrosis factor‑α and nitric oxide in rat serum samples. The protein expression levels of bone morphogenetic protein and transforming growth factor‑β were significantly increased by tanshinone IIA in the ACLT + MMx rats. Therefore, tanshinone IIA inhibited articular cartilage degradation through inhibition of apoptosis and expression levels of inflammatory cytokines, offering potential for use in the treatment of OA.
Collapse
Affiliation(s)
- Pei-Tong Jia
- Department of Orthopedics, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Xing-Lin Zhang
- Department of Orthopedics, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Hai-Ning Zuo
- Department of Orthopedics, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Xing Lu
- Department of Orthopedics, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Lin Li
- Department of Orthopedics, Shandong Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
39
|
Lawrence KM, Jones RC, Jackson TR, Baylie RL, Abbott B, Bruhn-Olszewska B, Board TN, Locke IC, Richardson SM, Townsend PA. Chondroprotection by urocortin involves blockade of the mechanosensitive ion channel Piezo1. Sci Rep 2017; 7:5147. [PMID: 28698554 PMCID: PMC5505992 DOI: 10.1038/s41598-017-04367-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/11/2017] [Indexed: 12/26/2022] Open
Abstract
Osteoarthritis (OA) is characterised by progressive destruction of articular cartilage and chondrocyte cell death. Here, we show the expression of the endogenous peptide urocortin1 (Ucn1) and two receptor subtypes, CRF-R1 and CRF-R2, in primary human articular chondrocytes (AC) and demonstrate its role as an autocrine/paracrine pro-survival factor. This effect could only be removed using the CRF-R1 selective antagonist CP-154526, suggesting Ucn1 acts through CRF-R1 when promoting chondrocyte survival. This cell death was characterised by an increase in p53 expression, and cleavage of caspase 9 and 3. Antagonism of CRF-R1 with CP-154526 caused an accumulation of intracellular calcium (Ca2+) over time and cell death. These effects could be prevented with the non-selective cation channel blocker Gadolinium (Gd3+). Therefore, opening of a non-selective cation channel causes cell death and Ucn1 maintains this channel in a closed conformation. This channel was identified to be the mechanosensitive channel Piezo1. We go on to determine that this channel inhibition by Ucn1 is mediated initially by an increase in cyclic adenosine monophosphate (cAMP) and a subsequent inactivation of phospholipase A2 (PLA2), whose metabolites are known to modulate ion channels. Knowledge of these novel pathways may present opportunities for interventions that could abrogate the progression of OA.
Collapse
Affiliation(s)
- K M Lawrence
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK.
| | - R C Jones
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - T R Jackson
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - R L Baylie
- Division of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, M13 9NT, Manchester, UK
| | - B Abbott
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - B Bruhn-Olszewska
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - T N Board
- The Center for Hip Surgery, Wrightington Hospital, Wigan, WN6 9EP, UK
| | - I C Locke
- Department of Biomedical Sciences, University of Westminster, London, W1W 6UW, UK
| | - S M Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, Centre for Tissue Injury and Repair, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, M13 9PT, UK
| | - P A Townsend
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| |
Collapse
|
40
|
Diaz-Romero J, Nesic D. S100A1 and S100B: Calcium Sensors at the Cross-Roads of Multiple Chondrogenic Pathways. J Cell Physiol 2017; 232:1979-1987. [DOI: 10.1002/jcp.25720] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 11/30/2016] [Indexed: 01/13/2023]
Affiliation(s)
- José Diaz-Romero
- Osteoarticular Research Group; Department of Clinical Research; University of Bern; Bern Switzerland
| | - Dobrila Nesic
- Osteoarticular Research Group; Department of Clinical Research; University of Bern; Bern Switzerland
| |
Collapse
|
41
|
Zhang P. Ginsenoside‑Rg5 treatment inhibits apoptosis of chondrocytes and degradation of cartilage matrix in a rat model of osteoarthritis. Oncol Rep 2017; 37:1497-1502. [PMID: 28112382 DOI: 10.3892/or.2017.5392] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 11/07/2016] [Indexed: 11/05/2022] Open
Abstract
This study investigated the effect of ginsenoside‑Rg5 on the degradation of articular cartilage in osteoarthritis rat model and on induction of chondrocyte apoptosis. Osteoarthritis rat model was prepared by ligament transection and medial meniscus resection. The rats were then treated with different doses (1, 2, 5, 10 and 15 µM) of ginsenoside‑Rg5 for 48 h. The results from histopathological analysis revealed a significant (P=0.005) prevention of cartilage degradation in OA rat model by ginsenoside‑Rg5 treatment at 15 µM. Ginsenoside‑Rg5 treatment prevented the disintegration of synovial membrane to a significant (P=0.005) extent. The proportion of apoptotic cells in the knee joints was reduced to 7% by ginsenoside‑Rg5 treatment after one month compared to the control. Treatment of the rats with ginsenoside‑Rg5 caused increase in the levels of proteoglycan, collagen and type II collagen by 5-, 3- and 4-fold compared to the control group. Immunohistochemistry revealed that the level of MMP-13 was reduced to 45% and that of TIMP‑1 was increased by 67% on treatment with ginsenoside‑Rg5. The levels of interleukin-1β, tumor necrosis factor-α, nitric oxide and inducible nitric oxide synthetase were reduced by 67, 54, 32 ad 49%, respectively after one month of treatment with 15 mg/kg dose of ginsenoside‑Rg5. The expression was increased to 67 and 52% for BMP-2 and TGF-β1, respectively on treatment with ginsenoside‑Rg5. Thus ginsenoside‑Rg5 prevents cartilage degradation in the OA rats and inhibits cartilage apoptosis, therefore it can be used for osteoarthritis treatment.
Collapse
Affiliation(s)
- Ping Zhang
- The Disease Prevention Center of Anyang Hospital of Traditional Chinese Medicine of Henan Province, Nanyang, Henan 455000, P.R. China
| |
Collapse
|
42
|
Henak CR, Ross KA, Bonnevie ED, Fortier LA, Cohen I, Kennedy JG, Bonassar LJ. Human talar and femoral cartilage have distinct mechanical properties near the articular surface. J Biomech 2016; 49:3320-3327. [PMID: 27589932 DOI: 10.1016/j.jbiomech.2016.08.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/05/2016] [Accepted: 08/16/2016] [Indexed: 12/16/2022]
Abstract
Talar osteochondral lesions (OCL) frequently occur following injury. Surgical interventions such as femoral condyle allogeneic or autogenic osteochondral transplant (AOT) are often used to treat large talar OCL. Although AOT aims to achieve OCL repair by replacing damaged cartilage with mechanically matched cartilage, the spatially inhomogeneous material behavior of the talar dome and femoral donor sites have not been evaluated or compared. The objective of this study was to characterize the depth-dependent shear properties and friction behavior of human talar and donor-site femoral cartilage. To achieve this objective, depth-dependent shear modulus, depth-dependent energy dissipation and coefficient of friction were measured on osteochondral cores from the femur and talus. Differences between anatomical regions were pronounced near the articular surface, where the femur was softer, dissipated more energy and had a lower coefficient of friction than the talus. Conversely, shear modulus near the osteochondral interface was nearly indistinguishable between anatomical regions. Differences in energy dissipation, shear moduli and friction coefficients have implications for graft survival and host cartilage wear. When the biomechanical variation is combined with known biological variation, these data suggest the use of caution in transplanting cartilage from the femur to the talus. Where alternatives exist in the form of talar allograft, donor-recipient mechanical mismatch can be greatly reduced.
Collapse
Affiliation(s)
- Corinne R Henak
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Keir A Ross
- Hospital for Special Surgery, New York, NY, United States
| | - Edward D Bonnevie
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Lisa A Fortier
- Department of Clinical Sciences, Cornell University, Ithaca, NY, United States
| | - Itai Cohen
- Department of Physics, Cornell University, Ithaca, NY, United States
| | - John G Kennedy
- Hospital for Special Surgery, New York, NY, United States
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States; Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
43
|
Maier F, Drissi H, Pierce DM. Shear deformations of human articular cartilage: Certain mechanical anisotropies apparent at large but not small shear strains. J Mech Behav Biomed Mater 2016; 65:53-65. [PMID: 27552599 DOI: 10.1016/j.jmbbm.2016.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/18/2016] [Accepted: 08/03/2016] [Indexed: 01/12/2023]
Abstract
Articular cartilage has pronounced through-the-thickness heterogeneity in both ultrastructure and mechanical function. The tissue undergoes a combination of large deformations in vivo, where shear is critical in both failure and chondrocyte death. Yet the microstructure mechanical response of cartilage to multi-axial large shear deformations is unknown. We harvested a total of 42 cartilage specimens from seven matched locations across the lateral femoral condyles and patellofemoral grooves of six human male donors (30.2±8.8yrs old, M±SD). With each specimen we applied a range of quasi-static, multi-axial large (simple) shear displacements both parallel and perpendicular to the local split-line direction (SLD). Shear stresses in cartilage specimens from the patellofemoral grooves were higher, and more energy was dissipated, at all applied strains under loading parallel to the local SLD versus perpendicular, while specimens from the lateral condyles were mechanically anisotropic only under larger strains of 20% and 25%. Cartilage also showed significant intra-donor variability at larger shear strains but no significant inter-donor variability. Overall, shear strain-energy dissipation was almost constant at 5% applied shear strain and increased nonlinearly with increasing shear magnitude. Our results suggest that full understanding of cartilage mechanics requires large-strain analyses to account for nonlinear, anisotropic and location-dependent effects not fully realized at small strains.
Collapse
Affiliation(s)
- Franz Maier
- University of Connecticut, Department of Mechanical Engineering, Storrs, CT, USA
| | - Hicham Drissi
- University of Connecticut Health Center, Orthopedic Surgery, Farmington, CT, USA
| | - David M Pierce
- University of Connecticut, Department of Mechanical Engineering, Storrs, CT, USA; University of Connecticut, Department of Biomedical Engineering, Storrs, CT, USA.
| |
Collapse
|
44
|
Fan R, Emery T, Zhang Y, Xia Y, Sun J, Wan J. Circulatory shear flow alters the viability and proliferation of circulating colon cancer cells. Sci Rep 2016; 6:27073. [PMID: 27255403 PMCID: PMC4891768 DOI: 10.1038/srep27073] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/15/2016] [Indexed: 01/17/2023] Open
Abstract
During cancer metastasis, circulating tumor cells constantly experience hemodynamic shear stress in the circulation. Cellular responses to shear stress including cell viability and proliferation thus play critical roles in cancer metastasis. Here, we developed a microfluidic approach to establish a circulatory microenvironment and studied circulating human colon cancer HCT116 cells in response to a variety of magnitude of shear stress and circulating time. Our results showed that cell viability decreased with the increase of circulating time, but increased with the magnitude of wall shear stress. Proliferation of cells survived from circulation could be maintained when physiologically relevant wall shear stresses were applied. High wall shear stress (60.5 dyne/cm(2)), however, led to decreased cell proliferation at long circulating time (1 h). We further showed that the expression levels of β-catenin and c-myc, proliferation regulators, were significantly enhanced by increasing wall shear stress. The presented study provides a new insight to the roles of circulatory shear stress in cellular responses of circulating tumor cells in a physiologically relevant model, and thus will be of interest for the study of cancer cell mechanosensing and cancer metastasis.
Collapse
Affiliation(s)
- Rong Fan
- Microsystems Engineering, Rochester Institute of Technology, Rochester, New York, USA
| | - Travis Emery
- Department of Mechanical Engineering, Rochester Institute of Technology, Rochester, New York, USA
| | - Yongguo Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yuxuan Xia
- Department of Applied Physics and Applied Mathematics/Materials Science and Engineering Program, Columbia University, New York, USA
| | - Jun Sun
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jiandi Wan
- Microsystems Engineering, Rochester Institute of Technology, Rochester, New York, USA
| |
Collapse
|
45
|
Karamchedu NP, Tofte JN, Waller KA, Zhang LX, Patel TK, Jay GD. Superficial zone cellularity is deficient in mice lacking lubricin: a stereoscopic analysis. Arthritis Res Ther 2016; 18:64. [PMID: 26975998 PMCID: PMC5477516 DOI: 10.1186/s13075-016-0967-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 03/04/2016] [Indexed: 01/09/2023] Open
Abstract
Background Lubricin, a mucinous glycoprotein secreted by synoviocytes and chondrocytes plays an important role in reducing the coefficient of friction in mammalian joints. Elevated cartilage surface friction is thought to cause chondrocyte loss; however, its quantification and methodological approaches have not been reported. We adapted a stereological method and incorporated vital cell staining to assess cellular loss in superficial and upper intermediate zones in lubricin deficient mouse cartilage. Methods The femoral condyle cartilage of the intact knees from lubricin wild type (Prg4+/+), heterozygote (Prg4+/-), and knockout (Prg4-/-) mice was imaged using fluorescein diacetate (FDA), propidium iodide (PI), and Hoechst staining, and confocal microscopy. Three dimensional reconstructions of confocal images to a depth of 14 μm were analyzed using Matlab to determine the volume fraction occupied by chondrocytes in cartilage of both medial and lateral femoral condyles. Living chondrocyte volume fraction was defined as FDA stained chondrocyte volume/total volume of superficial + upper intermediate zone. Living and dead (total) chondrocyte volume fraction was defined as FDA + PI stained chondrocyte volume/total volume of superficial + upper intermediate zone. MicroCT provided an orthogonal measure of cartilage thickness. Immunohistology for activated caspase-3 and TUNEL staining were performed to evaluate the presence of apoptotic chondrocytes in Prg4 mutant mice. Results Living chondrocyte volume fraction of the medial femoral condyle was significantly lower in Prg4-/- mice compared to Prg4+/+ (p = 0.002) and Prg4+/- (p = 0.002) littermates. There was no significant difference in medial condyle chondrocyte volume fraction between Prg4+/+ and Prg4+/- mice (p = 0.82). No significant differences were observed for the chondrocyte volume fraction for the lateral condyle (p > 0.26). Cartilage thickness increased in the medial condyle for Prg4-/- mice compared to Prg4+/+ (p = 0.02) and Prg4+/- (p = 0.03) littermates, and the lateral condyle for Prg4-/- mice compared to Prg4+/+ (p < 0.0001) and Prg4+/- (p < 0.0001) littermates, indicating that a multi-dimensional increase in cartilage volume did not artifactually lower the chondrocyte volume fraction in the medial condyle. Significantly higher number of caspase-3 positive cells were observed in the superficial and upper intermediate zone cartilage of the medial femoral condyle of Prg4-/- mice compared to Prg4+/+ (p = 0.01) and Prg4+/- (p = 0.04) littermates, and the lateral femoral condyle of Prg4-/- mice compared to Prg4+/+ (p = 0.02) and Prg4+/- (p = 0.02) littermates. There were no significant differences in TUNEL staining among different Prg4 genotypes in both condyles (p > 0.05 for all comparisons). Conclusions Increased Caspase-3 activation is observed in Prg4 deficient mice compared to Prg4 sufficient littermates. Absence of Prg4 induces loss of chondrocytes in the superficial and upper intermediate zone of mouse cartilage that is quantifiable by a novel image processing technique. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-0967-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Naga Padmini Karamchedu
- Department of Orthopedics, Rhode Island Hospital; Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Josef N Tofte
- Department of Orthopedics and Rehabilitation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Kimberly A Waller
- Department of Orthopedics, Rhode Island Hospital; Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ling X Zhang
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, 02903, USA
| | - Tarpit K Patel
- Department of Orthopedics, Rhode Island Hospital; Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Gregory D Jay
- Department of Emergency Medicine, Rhode Island Hospital, Providence, RI, 02903, USA. .,Department of Engineering, Brown University, Providence, RI, USA.
| |
Collapse
|
46
|
Hill A, Waller KA, Cui Y, Allen JM, Smits P, Zhang LX, Ayturk UM, Hann S, Lessard SG, Zurakowski D, Warman ML, Jay GD. Lubricin restoration in a mouse model of congenital deficiency. Arthritis Rheumatol 2016. [PMID: 26216721 DOI: 10.1002/art.39276] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Congenital deficiency of the principal boundary lubricant in cartilage (i.e., lubricin, encoded by the gene PRG4) increases joint friction and causes progressive joint failure. This study was undertaken to determine whether restoring lubricin expression in a mouse model would prevent, delay, or reverse the disease process caused by congenital deficiency. METHODS Using genetically engineered lubricin-deficient mice, we restored gene function before conception or at ages 3 weeks, 2 months, or 6 months after birth. The effect of restoring gene function (i.e., expression of lubricin) on the tibiofemoral patellar joints of mice was evaluated histologically and by ex vivo biomechanical testing. RESULTS Restoring gene function in mice prior to conception prevented joint disease. In 3-week-old mice, restoring gene function improved, but did not normalize, histologic features of the articular cartilage and whole-joint friction. In addition, cyclic loading of the joints produced fewer activated caspase 3-containing chondrocytes when lubricin expression was restored, as compared to that in littermate mice whose gene function was not restored (nonrestored controls). Restoration of lubricin expression in 2-month-old or 6-month-old mice had no beneficial effect on histopathologic cartilage damage, extent of whole-joint friction, or activation of caspase 3 when compared to nonrestored controls. CONCLUSION When boundary lubrication is congenitally deficient and cartilage becomes damaged, the window of opportunity for restoring lubrication and slowing disease progression is limited.
Collapse
Affiliation(s)
- Adele Hill
- Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kimberly A Waller
- Alpert Medical School of Brown University and Rhode Island Hospital, Providence
| | - Yajun Cui
- Boston Children's Hospital, Boston, Massachusetts
| | - Justin M Allen
- Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Ling X Zhang
- Alpert Medical School of Brown University and Rhode Island Hospital, Providence
| | - Ugur M Ayturk
- Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Steven Hann
- Boston Children's Hospital, Boston, Massachusetts
| | | | | | - Matthew L Warman
- Howard Hughes Medical Institute, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Gregory D Jay
- Rhode Island Hospital, Alpert Medical School of Brown University, and Brown University School of Engineering, Providence, Rhode Island
| |
Collapse
|
47
|
Ashraf S, Cha BH, Kim JS, Ahn J, Han I, Park H, Lee SH. Regulation of senescence associated signaling mechanisms in chondrocytes for cartilage tissue regeneration. Osteoarthritis Cartilage 2016; 24:196-205. [PMID: 26190795 DOI: 10.1016/j.joca.2015.07.008] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 06/11/2015] [Accepted: 07/09/2015] [Indexed: 02/06/2023]
Abstract
Adult articular chondrocytes undergo slow senescence and dedifferentiation during in vitro expansion, restricting successful cartilage regeneration. A complete understanding of the molecular signaling pathways involved in the senescence and dedifferentiation of chondrocytes is essential in order to better characterize chondrocytes for cartilage tissue engineering applications. During expansion, cell fate is determined by the change in expression of various genes in response to aspects of the microenvironment, including oxidative stress, mechanical stress, and unsuitable culture conditions. Rapid senescence or dedifferentiation not only results in the loss of the chondrocytic phenotype but also enhances production of inflammatory mediators and matrix-degrading enzymes. This review focuses on the two groups of genes that play direct and indirect roles in the induction of senescence and dedifferentiation. Numerous degenerative signaling pathways associated with these genes have been reported. Upregulation of the genes interleukin 1 beta (IL-1β), p53, p16, p21, and p38 mitogen-activated protein kinase (MAPK) is responsible for the direct induction of senescence, whereas downregulation of the genes transforming growth factor-beta (TGF-β), bone morphogenetic protein-2 (BMP-2), SRY (sex determining region Y)-box 9 (SOX9), and insulin-like growth factor-1 (IGF-1), indirectly induces senescence. In senescent and dedifferentiated chondrocytes, it was found that TGF-β, BMP-2, SOX9, and IGF-1 are downregulated, while the levels of IL-1β, p53, p16, p21, and p38 MAPK are upregulated followed by inhibition of the normal molecular functioning of the chondrocytes. This review helps to elucidate the underlying mechanism in degenerative cartilage disease, which may help to improve cartilage tissue regeneration techniques.
Collapse
Affiliation(s)
- S Ashraf
- School of Integrative Engineering, Chung-Ang University, Seoul, South Korea; Department of Biomedical Science, CHA University, Seoul, South Korea.
| | - B-H Cha
- Department of Biomedical Science, CHA University, Seoul, South Korea.
| | - J-S Kim
- Department of Biomedical Science, CHA University, Seoul, South Korea.
| | - J Ahn
- Department of Biomedical Science, CHA University, Seoul, South Korea.
| | - I Han
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, 59, Yatap-ro Bundang-gu, Seongnam-si, Kyeunggi-do, 463-712, South Korea.
| | - H Park
- School of Integrative Engineering, Chung-Ang University, Seoul, South Korea.
| | - S-H Lee
- Department of Biomedical Science, CHA University, Seoul, South Korea.
| |
Collapse
|
48
|
Zeng F, Yu X, Sherry JP, Dixon B, Duncker BP, Bols NC. The p53 inhibitor, pifithrin-α, disrupts microtubule organization, arrests growth, and induces polyploidy in the rainbow trout gill cell line, RTgill-W1. Comp Biochem Physiol C Toxicol Pharmacol 2016; 179:1-10. [PMID: 26291498 DOI: 10.1016/j.cbpc.2015.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/04/2015] [Accepted: 08/10/2015] [Indexed: 11/16/2022]
Abstract
Pifithrin-α (PFT-α) blocks p53-dependent transcription and is an example of the many drugs being developed to target the p53 pathway in humans that could be released into the environment with potential impacts on aquatic animals if they were to become successful pharmaceuticals. In order to understand how p53 drugs might act on fish, the effects of PFT-α on rainbow trout gill epithelial cell line, RTgill-W1, were studied. PFT-α was not cytotoxic to RTgill-W1 in cultures with or without fetal bovine serum (FBS), but at 5.25μg/ml, PFT-α completely arrested proliferation. When FBS was present, PFT-α increased the number of polyploid cells over 12days. Those results suggest that like in mammals, p53 appears to regulate ploidy in fish. However, several effects were seen that have not been observed with mammalian cells. PFT-α caused a transient rise in the mitotic index and a disruption in cytoskeletal microtubules. These results suggest that in fish cells PFT-α affects microtubules either directly through an off-target action on tubulin or indirectly through an on-target action on p53-regulated transcription.
Collapse
Affiliation(s)
- Fanxing Zeng
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Xiang Yu
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - James P Sherry
- Aquatic Contaminants Research Division, Environment Canada, Burlington, ON, Canada L7R 4A6
| | - Brian Dixon
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Bernard P Duncker
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Niels C Bols
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1.
| |
Collapse
|
49
|
Hayashi S, Fujishiro T, Hashimoto S, Kanzaki N, Chinzei N, Kihara S, Takayama K, Matsumoto T, Nishida K, Kurosaka M, Kuroda R. p21 deficiency is susceptible to osteoarthritis through STAT3 phosphorylation. Arthritis Res Ther 2015; 17:314. [PMID: 26546411 PMCID: PMC4636813 DOI: 10.1186/s13075-015-0828-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 10/20/2015] [Indexed: 12/18/2022] Open
Abstract
Introduction Osteoarthritis (OA) is a multifactorial disease, and recent studies have suggested that cell cycle–related proteins play a role in OA pathology. p21 was initially identified as a potent inhibitor of cell cycle progression. However, it has been proposed that p21 is a regulator of transcription factor activity. In this study, we evaluated the role of p21 in response to biomechanical stress. Methods Human chondrocytes were treated with p21-specific small interfering RNA (siRNA), and cyclic tensile strain was introduced in the presence or absence of a signal transducer and activator of transcription 3 (STAT3)-specific inhibitor. Further, we developed an in vivo OA model in a p21-knockout background for in vivo experiments. Results The expression of matrix metalloproteinase (MMP13) mRNA increased in response to cyclic tensile strain following transfection with p21 siRNA, whereas the expression of aggrecan was decreased. Phospho-STAT3 and MMP-13 protein levels increased following downregulation of p21, and this was reversed by treatment with a STAT3 inhibitor. p21-deficient mice were susceptible to OA, and this was associated with increased STAT3 phosphorylation, elevated MMP-13 expression, and elevation of synovial inflammation. The expression of p21 mRNA was decreased and phosphorylation of STAT3 was elevated in human OA chondrocytes. Conclusions The lack of p21 has catabolic effects by regulation of aggrecan and MMP-13 expression through STAT3 phosphorylation in the cartilage tissue. p21 may function as a regulator of transcriptional factors other than the inhibitor of cell cycle progression in the cartilage tissue. Thus, the regulation of p21 may be a therapeutic strategy for the treatment of OA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0828-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Takaaki Fujishiro
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Shingo Hashimoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Noriyuki Kanzaki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Nobuaki Chinzei
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Shinsuke Kihara
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Koji Takayama
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Kotaro Nishida
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
50
|
Depletion of SIRT6 causes cellular senescence, DNA damage, and telomere dysfunction in human chondrocytes. Osteoarthritis Cartilage 2015; 23:1412-20. [PMID: 25819580 DOI: 10.1016/j.joca.2015.03.024] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 03/15/2015] [Accepted: 03/18/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE SIRT6, a member of the sirtuin family of nicotinamide adenine dinucleotide (NAD(+))-dependent protein deacetylases, has been implicated as a key factor in aging-related diseases. However, the role of SIRT6 in chondrocytes has not been fully explored. The purpose of this study was to examine the role of SIRT6 in human chondrocytes by inhibiting SIRT6 in vitro. DESIGN First, the localization of SIRT6 and proliferation cell nuclear antigen (PCNA) in human cartilages was examined by immunohistochemistry. Next, SIRT6 was depleted by RNA interference (RNAi), and the effect of SIRT6 depletion on changes in gene expression, protein levels, proliferation, and senescence in human chondrocytes was assessed. Furthermore, to detect DNA damage and telomere dysfunction, γH2AX foci and telomere dysfunction-induced foci (TIFs) were examined using immunofluorescence microscopy. The protein levels of two mediators for DNA damage induced-senescence, p16 and p21, were examined by western blotting. RESULTS Immunohistochemical analysis showed SIRT6 was preferentially expressed in the superficial zone chondrocytes and PCNA-positive cluster-forming chondrocytes in the osteoarthritic cartilage tissue samples. Real-time PCR analysis showed that matrix metalloproteinase 1 (MMP-1) and MMP-13 mRNA were significantly increased by SIRT6 inhibition. Moreover, SIRT6 inhibition significantly reduced proliferation and increased senescence associated β-galactosidase (SA-β-Gal)-positive chondrocytes; it also led to increased p16 levels. Immunofluorescence microscopy showed that γH2AX foci and TIFs were increased by SIRT6 inhibition. CONCLUSION Depletion of SIRT6 in human chondrocytes caused increased DNA damage and telomere dysfunction, and subsequent premature senescence. These findings suggest that SIRT6 plays an important role in the regulation of senescence of human chondrocytes.
Collapse
|