1
|
Huang K, Cheng L, Jiang C, Zheng C, Cai H. Dehydroepiandrosterone inhibits ADAMTS expression via an ERK-dependent mechanism in chondrocytes. PLoS One 2024; 19:e0313560. [PMID: 39576807 PMCID: PMC11584127 DOI: 10.1371/journal.pone.0313560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024] Open
Abstract
Osteoarthritis (OA) is a joint disease in which cartilage degradation is the hallmark pathological change. In this study, we investigated the anti-osteoarthritic effects of DHEA in rabbit chondrocytes. Polymerase chain reaction was performed to evaluate the expression of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4, ADAMTS-5, aggrecan and collagen type 2. In addition, ERK1/2 signaling pathway components were analyzed by Western blotting. In IL-1β-induced chondrocytes, the phosphorylation of ERK1/2 was enhanced, and the downstream catabolic genes, including ADAMTS-4 and ADAMTS-5, were upregulated, while the anabolic genes aggrecan and collagen type 2 were downregulated. DHEA administration restored the IL-1β-induced imbalance in anabolic and catabolic gene expression. In addition, the phosphorylation of ERK1/2 was suppressed by DHEA. Then, PD98059 was used to block the ERK1/2 signaling pathway. The protective effect of DHEA was significantly increased when ERK1/2 signaling was inactivated. DHEA may exert its protective effect by suppressing ADAMTS in an ERK1/2-dependent manner in rabbit chondrocytes.
Collapse
Affiliation(s)
- Kai Huang
- Department of Orthopedic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Lin Cheng
- Department of Orthopedic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Cheng Jiang
- Department of Orthopedic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Chunwei Zheng
- Department of Orthopedic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Haili Cai
- Department of Ultrasound, The 903rd Hospital of PLA, Hangzhou, China
| |
Collapse
|
2
|
Ao R, Liang W, Wang Z, Li Q, Pan X, Zhen Y, An Y. Delivery Strategies of Growth Factors in Cartilage Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39345121 DOI: 10.1089/ten.teb.2024.0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cartilage plays an important role in supporting soft tissues, reducing joint friction, and distributing pressure. However, its self-repair capacity is limited due to the lack of blood vessels, nerves, and lymphatic systems. Tissue engineering offers a potential solution to promote cartilage regeneration by combining scaffolds, seed cells, and growth factors. Among these, growth factors play a critical role in regulating cell proliferation, differentiation, and extracellular matrix remodeling. However, their instability, susceptibility to degradation and potential side effects limit their effectiveness. This article reviews the main growth factors used in cartilage tissue engineering and their delivery strategies, including affinity-based delivery, carrier-assisted delivery, stimuli-responsive delivery, spatial structure-based delivery, and cell system-based delivery. Each method shows unique advantages in enhancing the delivery efficiency and specificity of growth factors but also faces challenges such as cost, biocompatibility, and safety. Future research needs to further optimize these strategies to achieve more efficient, safe, and economical delivery of growth factors, thereby advancing the clinical application of cartilage tissue engineering.
Collapse
Affiliation(s)
- Rigele Ao
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Wei Liang
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Zimo Wang
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Qiaoyu Li
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Xingyi Pan
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| |
Collapse
|
3
|
Fan Z, Zhao X, Ma J, Zhan H, Ma X. Suppression of YAP Ameliorates Cartilage Degeneration in Ankle Osteoarthritis via Modulation of the Wnt/β-Catenin Signaling Pathway. Calcif Tissue Int 2024; 115:283-297. [PMID: 38953964 DOI: 10.1007/s00223-024-01242-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
Ankle osteoarthritis is a relatively understudied condition and the molecular mechanisms involved in its development are not well understood. This investigation aimed to explore the role and underlying molecular mechanisms of Yes-associated protein (YAP) in rat ankle osteoarthritis. The results demonstrated that YAP expression levels were abnormally increased in the ankle osteoarthritis cartilage model. In addition, knockdown of YAP expression was shown to hinder the imbalance in ECM metabolism induced by IL-1β in chondrocytes, as demonstrated by the regulation of matrix metalloproteinase (MMP)-3, MMP-9, and MMP-13, a disintegrin, metalloprotease with thrombospondin motifs, aggrecan, and collagen II expression. Additional studies revealed that downregulation of YAP expression markedly inhibited the overexpression of β-catenin stimulated by IL-1β. Furthermore, inhibition of the Wnt/β-catenin signaling pathway reversed the ECM metabolism imbalance caused by YAP overexpression in chondrocytes. It is important to note that the YAP-specific inhibitor verteporfin (VP) significantly delayed the progression of ankle osteoarthritis. In conclusion, the findings highlighted the crucial role of YAP as a regulator in modulating the progression of ankle osteoarthritis via the Wnt/β-catenin signaling pathway. These findings suggest that pharmacological inhibition of YAP can be an effective and critical therapeutic target for alleviating ankle osteoarthritis.
Collapse
Affiliation(s)
- Zhengrui Fan
- The department of Orthopedics, Tianjin Hospital, Tianjin, 300070, People's Republic of China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, China
| | - Xingwen Zhao
- The department of Orthopedics, Tianjin Hospital, Tianjin, 300070, People's Republic of China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, China
| | - Jianxiong Ma
- The department of Orthopedics, Tianjin Hospital, Tianjin, 300070, People's Republic of China.
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, China.
| | - Hongqi Zhan
- The department of Orthopedics, Tianjin Hospital, Tianjin, 300070, People's Republic of China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, China
| | - Xinlong Ma
- The department of Orthopedics, Tianjin Hospital, Tianjin, 300070, People's Republic of China.
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin Hospital, Tianjin, 300050, China.
| |
Collapse
|
4
|
Lee J, Lim J, Park S, Kim S, Park J. Morphologic Response in Femoral Cartilage During and After 40-Minute Treadmill Running. J Athl Train 2024; 59:906-914. [PMID: 39320951 PMCID: PMC11440817 DOI: 10.4085/1062-6050-0659.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
CONTEXT It is unclear whether the response in femoral cartilage to running at different intensities is different. OBJECTIVE To examine the acute patterns of deformation and recovery in femoral cartilage thickness during and after running at different speeds. DESIGN Crossover study. SETTING Laboratory. PATIENTS OR OTHER PARTICIPANTS A total of 17 healthy men (age = 23.9 ± 2.3 years, height = 173.1 ± 5.5 cm, mass = 73.9 ± 8.0 kg). INTERVENTION(S) Participants performed a 40-minute treadmill run at speeds of 7.5 and 8.5 km/h. MAIN OUTCOME MEASURE(S) Ultrasonographic images of femoral cartilage thickness (intercondylar, lateral condyle, and medial condyle) were obtained every 5 minutes during the experiment (40 minutes of running followed by a 60-minute recovery period) at each session. Data were analyzed using analysis of variance and Bonferroni- and Dunnett-adjusted post hoc t tests. To identify patterns of cartilage response, we extracted principal components (PCs) from the cartilage-thickness data using PC analysis, and PC scores were analyzed using t tests. RESULTS Regardless of time, femoral cartilage thicknesses were greater for the 8.5-km/h run than the 7.5-km/h run (intercondylar: F1,656 = 24.73, P < .001, effect size, 0.15; lateral condyle: F1,649 = 16.60, P < .001, effect size, 0.16; medial condyle: F1,649 = 16.55, P < .001, effect size, 0.12). We observed a time effect in intercondylar thickness (F20,656 = 2.15, P = .003), but the Dunnett-adjusted post hoc t test revealed that none of the time point values differed from the baseline value (P > .38 for all comparisons). Although the PC1 and PC2 captured the magnitudes of cartilage thickness and time shift (eg, earlier versus later response), respectively, t tests showed that the PC scores were not different between 7.5 and 8.5 km/h (intercondylar: P ≥ .32; lateral condyle: P ≥ .78; medial condyle: P ≥ .16). CONCLUSIONS Although the 40-minute treadmill run with different speeds produced different levels of fatigue, morphologic differences (<3%) in the femoral cartilage at both speeds seemed to be negligible.
Collapse
Affiliation(s)
- Jinwoo Lee
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| | - Junhyeong Lim
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| | - Sanghyup Park
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| | - Sojin Kim
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| | - Jihong Park
- Athletic Training Laboratory, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
5
|
Wang D, Feng Z, Zeng J, Wang Q, Zheng Y, Liu X, Jiang H. Low-Temperature Extrusion of Waterborne Polyurethane-Polycaprolactone Composites for Multi-Material Bioprinting of Engineered Elastic Cartilage. Macromol Biosci 2024; 24:e2300557. [PMID: 38409648 DOI: 10.1002/mabi.202300557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/13/2024] [Indexed: 02/28/2024]
Abstract
3D bioprinting of elastic cartilage tissues that are mechanically and structurally comparable to their native counterparts, while exhibiting favorable cellular behavior, is an unmet challenge. A practical solution for this problem is the multi-material bioprinting of thermoplastic polymers and cell-laden hydrogels using multiple nozzles. However, the processing of thermoplastic polymers requires high temperatures, which can damage hydrogel-encapsulated cells. In this study, the authors developed waterborne polyurethane (WPU)-polycaprolactone (PCL) composites to allow multi-material co-printing with cell-laden gelatin methacryloyl (GelMA) hydrogels. These composites can be extruded at low temperatures (50-60 °C) and high speeds, thereby reducing heat/shear damage to the printed hydrogel-capsulated cells. Furthermore, their hydrophilic nature improved the cell behavior in vitro. More importantly, the bioprinted structures exhibited good stiffness and viscoelasticity compared to native elastic cartilage. In summary, this study demonstrated low-temperature multi-material bioprinting of WPU-PCL-based constructs with good mechanical properties, degradation time-frames, and cell viability, showcasing their potential in elastic cartilage bio-fabrication and regeneration to serve broad biomedical applications in the future.
Collapse
Affiliation(s)
- Di Wang
- Plastic Surgery Hospital of Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100144, P. R. China
| | - Zhaoxuan Feng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
| | - Jinshi Zeng
- Plastic Surgery Hospital of Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100144, P. R. China
| | - Qian Wang
- Plastic Surgery Hospital of Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100144, P. R. China
| | - Yudong Zheng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
| | - Xia Liu
- Plastic Surgery Hospital of Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100144, P. R. China
| | - Haiyue Jiang
- Plastic Surgery Hospital of Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100144, P. R. China
| |
Collapse
|
6
|
Juhász KZ, Hajdú T, Kovács P, Vágó J, Matta C, Takács R. Hypoxic Conditions Modulate Chondrogenesis through the Circadian Clock: The Role of Hypoxia-Inducible Factor-1α. Cells 2024; 13:512. [PMID: 38534356 PMCID: PMC10969332 DOI: 10.3390/cells13060512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a heterodimer transcription factor composed of an alpha and a beta subunit. HIF-1α is a master regulator of cellular response to hypoxia by activating the transcription of genes that facilitate metabolic adaptation to hypoxia. Since chondrocytes in mature articular cartilage reside in a hypoxic environment, HIF-1α plays an important role in chondrogenesis and in the physiological lifecycle of articular cartilage. Accumulating evidence suggests interactions between the HIF pathways and the circadian clock. The circadian clock is an emerging regulator in both developing and mature chondrocytes. However, how circadian rhythm is established during the early steps of cartilage formation and through what signaling pathways it promotes the healthy chondrocyte phenotype is still not entirely known. This narrative review aims to deliver a concise analysis of the existing understanding of the dynamic interplay between HIF-1α and the molecular clock in chondrocytes, in states of both health and disease, while also incorporating creative interpretations. We explore diverse hypotheses regarding the intricate interactions among these pathways and propose relevant therapeutic strategies for cartilage disorders such as osteoarthritis.
Collapse
|
7
|
Lu J, Bian J, Wang Y, Zhao Y, Zhao X, Wang G, Yang J. Oxymatrine protects articular chondrocytes from IL-1β-induced damage through autophagy activation via AKT/mTOR signaling pathway inhibition. J Orthop Surg Res 2024; 19:178. [PMID: 38468339 PMCID: PMC10926585 DOI: 10.1186/s13018-024-04667-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/06/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common degenerative joint disease characterized by persistent articular cartilage degeneration and synovitis. Oxymatrine (OMT) is a quinzolazine alkaloid extracted from the traditional Chinese medicine, matrine, and possesses anti-inflammatory properties that may help regulate the pathogenesis of OA; however, its mechanism has not been elucidated. This study aimed to investigate the effects of OMT on interleukin-1β (IL-1β)-induced damage and the potential mechanisms of action. METHODS Chondrocytes were isolated from Sprague-Dawley rats. Toluidine blue and Collagen II immunofluorescence staining were used to determine the purity of the chondrocytes. Thereafter, the chondrocytes were subjected to IL-1β stimulation, both in the presence and absence of OMT, or the autophagy inhibitor 3-methyladenine (3-MA). Cell viability was assessed using the MTT assay and SYTOX Green staining. Additionally, flow cytometry was used to determine cell apoptosis rate and reactive oxygen species (ROS) levels. The protein levels of AKT, mTOR, LC3, P62, matrix metalloproteinase-13, and collagen II were quantitatively analyzed using western blotting. Immunofluorescence was used to assess LC3 expression. RESULTS OMT alleviated IL-1β-induced damage in chondrocytes, by increasing the survival rate, reducing the apoptosis rates of chondrocytes, and preventing the degradation of the cartilage matrix. In addition, OMT decreased the ROS levels and inhibited the AKT/mTOR signaling pathway while promoting autophagy in IL-1β treated chondrocytes. However, the effectiveness of OMT in improving chondrocyte viability under IL-1β treatment was limited when autophagy was inhibited by 3-MA. CONCLUSIONS OMT decreases oxidative stress and inhibits the AKT/mTOR signaling pathway to enhance autophagy, thus inhibiting IL-1β-induced damage. Therefore, OMT may be a novel and effective therapeutic agent for the clinical treatment of OA.
Collapse
Affiliation(s)
- Jinying Lu
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Jiang Bian
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Yutong Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Yan Zhao
- Provincial Key Laboratory of Cardiovascular and Cerebrovascular Drug Basic Research, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Xinmin Zhao
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Gao Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Jing Yang
- Provincial Key Laboratory of Cardiovascular and Cerebrovascular Drug Basic Research, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
8
|
Paez‐Perez M, Kuimova MK. Molecular Rotors: Fluorescent Sensors for Microviscosity and Conformation of Biomolecules. Angew Chem Int Ed Engl 2024; 63:e202311233. [PMID: 37856157 PMCID: PMC10952837 DOI: 10.1002/anie.202311233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023]
Abstract
The viscosity and crowding of biological environment are considered vital for the correct cellular function, and alterations in these parameters are known to underly a number of pathologies including diabetes, malaria, cancer and neurodegenerative diseases, to name a few. Over the last decades, fluorescent molecular probes termed molecular rotors proved extremely useful for exploring viscosity, crowding, and underlying molecular interactions in biologically relevant settings. In this review, we will discuss the basic principles underpinning the functionality of these probes and will review advances in their use as sensors for lipid order, protein crowding and conformation, temperature and non-canonical nucleic acid structures in live cells and other relevant biological settings.
Collapse
Affiliation(s)
- Miguel Paez‐Perez
- Department of Chemistry, Imperial College London, MSRHImperial College LondonWood LaneLondonW12 0BZUK
| | - Marina K. Kuimova
- Department of Chemistry, Imperial College London, MSRHImperial College LondonWood LaneLondonW12 0BZUK
| |
Collapse
|
9
|
Zheng Z, Luo H, Xue Q. Association between niacin intake and knee osteoarthritis pain and function: a longitudinal cohort study. Clin Rheumatol 2024; 43:753-764. [PMID: 38180674 DOI: 10.1007/s10067-023-06860-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND This research investigates the relationship between niacin intake and knee osteoarthritis (OA) severity, focusing on pain and functional ability due to niacin's role as a NAD(P)+ precursor, promoting cellular energy, and offering anti-inflammatory, analgesic, and antioxidant effects. METHODS The population-based Osteoarthritis Initiative (OAI) cohort with radiographically confirmed knee OA was analyzed through a Food Frequency Questionnaire determining niacin intake and scores from the Knee Injury and Osteoarthritis Outcome Score (KOOS) and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), using generalized additive mixed models. RESULTS A significant correlation was pinpointed in 2375 OA patients (1001 men and 1374 women; 55.96% aged between 45 and 65 and 44.04% aged ≥65) between niacin intake and reduced knee pain and functional degrees after a 48-month follow-up, evident in improved KOOS and WOMAC scores (P < 0.05). The fully adjusted models estimated a decrease of 0.26 points for every additional 1 unit of Ln-niacin intake of daily niacin intake on the WOMAC pain subscale, 0.83 points on the WOMAC function subscale, and an increase of 1.71 and 1.58 on the KOOS pain and quality of life score. Strikingly, subgroups including middle-aged individuals, women, white race, obese individuals, and those with specific dietary habits showed a more substantial improvement with increased niacin. CONCLUSION The association between increased niacin intake and reduced pain and function scores, as well improved quality of life in knee OA patients, is significant. Certain cohorts, according to a stratified analysis, could see more considerable benefits with increased niacin consumption. HIGHLIGHTS • Increased niacin intake is linked to reduced knee pain and better function in OA patients. • Specific subgroups, such as middle-aged individuals, women, and those with certain dietary habits, benefit more from increased niacin consumption. • Niacin shows promise for enhancing the quality of life in knee OA patients by reducing pain and improving function.
Collapse
Affiliation(s)
- Zitian Zheng
- Department of Orthopedics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Da Hua Road, DongDan, Beijing, 100730, P.R. China
- Peking University Fifth School of Clinical Medicine, Beijing, P.R. China
| | - Huanhuan Luo
- Department of Nursing, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, P.R. China
- Graduate School of Peking Union Medical College, Beijing, P.R. China
| | - Qingyun Xue
- Department of Orthopedics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Da Hua Road, DongDan, Beijing, 100730, P.R. China.
- Peking University Fifth School of Clinical Medicine, Beijing, P.R. China.
- Graduate School of Peking Union Medical College, Beijing, P.R. China.
| |
Collapse
|
10
|
Lou C, Jiang H, Lin Z, Xia T, Wang W, Lin C, Zhang Z, Fu H, Iqbal S, Liu H, Lin J, Wang J, Pan X, Xue X. MiR-146b-5p enriched bioinspired exosomes derived from fucoidan-directed induction mesenchymal stem cells protect chondrocytes in osteoarthritis by targeting TRAF6. J Nanobiotechnology 2023; 21:486. [PMID: 38105181 PMCID: PMC10726686 DOI: 10.1186/s12951-023-02264-9] [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: 09/18/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease characterized by progressive cartilage degradation and inflammation. In recent years, mesenchymal stem cells (MSCs) derived exosomes (MSCs-Exo) have attracted widespread attention for their potential role in modulating OA pathology. However, the unpredictable therapeutic effects of exosomes have been a significant barrier to their extensive clinical application. In this study, we investigated whether fucoidan-pretreated MSC-derived exosomes (F-MSCs-Exo) could better protect chondrocytes in osteoarthritic joints and elucidate its underlying mechanisms. In order to evaluate the role of F-MSCs-Exo in osteoarthritis, both in vitro and in vivo studies were conducted. MiRNA sequencing was employed to analyze MSCs-Exo and F-MSCs-Exo, enabling the identification of differentially expressed genes and the exploration of the underlying mechanisms behind the protective effects of F-MSCs-Exo in osteoarthritis. Compared to MSCs-Exo, F-MSCs-Exo demonstrated superior effectiveness in inhibiting inflammatory responses and extracellular matrix degradation in rat chondrocytes. Moreover, F-MSCs-Exo exhibited enhanced activation of autophagy in chondrocytes. MiRNA sequencing of both MSCs-Exo and F-MSCs-Exo revealed that miR-146b-5p emerged as a promising candidate mediator for the chondroprotective function of F-MSCs-Exo, with TRAF6 identified as its downstream target. In conclusion, our research results demonstrate that miR-146b-5p encapsulated in F-MSCs-Exo effectively inhibits TRAF6 activation, thereby suppressing inflammatory responses and extracellular matrix degradation, while promoting chondrocyte autophagy for the protection of osteoarthritic cartilage cells. Consequently, the development of a therapeutic approach combining fucoidan with MSC-derived exosomes provides a promising strategy for the clinical treatment of osteoarthritis.
Collapse
Affiliation(s)
- Chao Lou
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Hongyi Jiang
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Zhongnan Lin
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Tian Xia
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, China
| | - Weidan Wang
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Chihao Lin
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Zhiguang Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Haonan Fu
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Shoaib Iqbal
- Feik School of Pharmacy, University of the Incarnate Word, Broadway, San Antonio, 4301, USA
| | - Haixiao Liu
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Jian Lin
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Jilong Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, China.
| | - Xiaoyun Pan
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| | - Xinghe Xue
- Department of Orthopedics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
11
|
Atwal A, Dale TP, Snow M, Forsyth NR, Davoodi P. Injectable hydrogels: An emerging therapeutic strategy for cartilage regeneration. Adv Colloid Interface Sci 2023; 321:103030. [PMID: 37907031 DOI: 10.1016/j.cis.2023.103030] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
The impairment of articular cartilage due to traumatic incidents or osteoarthritis has posed significant challenges for healthcare practitioners, researchers, and individuals suffering from these conditions. Due to the absence of an approved treatment strategy for the complete restoration of cartilage defects to their native state, the tissue condition often deteriorates over time, leading to osteoarthritic (OA). However, recent advancements in the field of regenerative medicine have unveiled promising prospects through the utilization of injectable hydrogels. This versatile class of biomaterials, characterized by their ability to emulate the characteristics of native articular cartilage, offers the distinct advantage of minimally invasive administration directly to the site of damage. These hydrogels can also serve as ideal delivery vehicles for a diverse range of bioactive agents, including growth factors, anti-inflammatory drugs, steroids, and cells. The controlled release of such biologically active molecules from hydrogel scaffolds can accelerate cartilage healing, stimulate chondrogenesis, and modulate the inflammatory microenvironment to halt osteoarthritic progression. The present review aims to describe the methods used to design injectable hydrogels, expound upon their applications as delivery vehicles of biologically active molecules, and provide an update on recent advances in leveraging these delivery systems to foster articular cartilage regeneration.
Collapse
Affiliation(s)
- Arjan Atwal
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Tina P Dale
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom
| | - Martyn Snow
- Department of Arthroscopy, Royal Orthopaedic Hospital NHS Foundation Trust, Birmingham B31 2AP, United Kingdom; The Robert Jones and Agnes Hunt Hospital, Oswestry, Shropshire SY10 7AG, United Kingdom
| | - Nicholas R Forsyth
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom; Vice Principals' Office, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Pooya Davoodi
- School of Pharmacy and Bioengineering, Hornbeam building, Keele University, Staffordshire ST5 5BG, United Kingdom; Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, United Kingdom.
| |
Collapse
|
12
|
Xiao J, Zhang P, Cai FL, Luo CG, Pu T, Pan XL, Tian M. IL-17 in osteoarthritis: A narrative review. Open Life Sci 2023; 18:20220747. [PMID: 37854319 PMCID: PMC10579884 DOI: 10.1515/biol-2022-0747] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/18/2023] [Accepted: 09/07/2023] [Indexed: 10/20/2023] Open
Abstract
Osteoarthritis (OA) is a painful joint disease that is common among the middle-aged and elderly populations, with an increasing prevalence. Therapeutic options for OA are limited, and the pathogenic mechanism of OA remains unclear. The roles of cytokines and signaling pathways in the development of OA is a current research hot spot. Interleukin (IL)-17 is a pleiotropic inflammatory cytokine produced mainly by T helper 17 cells that has established roles in host defense, tissue repair, lymphoid tissue metabolism, tumor progression, and pathological processes of immune diseases, and studies in recent years have identified an important role for IL-17 in the progression of OA. This narrative review focuses on the mechanisms by which IL-17 contributes to articular cartilage degeneration and synovial inflammation in OA and discusses how IL-17 and the IL-17 signaling pathway affect the pathological process of OA. Additionally, therapeutic targets that have been proposed in recent years based on IL-17 and its pathway in OA are summarized as well as recent advances in the study of IL-17 pathway inhibitors and the potential challenges of their use for OA treatment.
Collapse
Affiliation(s)
- Juan Xiao
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, Zunyi563000, China
| | - Ping Zhang
- The First School of Clinical Medicine, Zunyi Medical University, Zunyi563000, China
| | - Fang-Lan Cai
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi563000, China
| | - Cheng-Gen Luo
- The First School of Clinical Medicine, Zunyi Medical University, Zunyi563000, China
| | - Tao Pu
- Department of Nephrology and Rheumatology, Moutai Hospital, Renhuai 564500Guizhou, China
| | - Xiao-Li Pan
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, Zunyi563000, China
| | - Mei Tian
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, Zunyi563000, China
| |
Collapse
|
13
|
Zavala G, Viafara-García SM, Novoa J, Hidalgo C, Contardo I, Díaz-Calderón P, Alejandro González-Arriagada W, Khoury M, Acevedo JP. An advanced biphasic porous and injectable scaffold displays a fine balance between mechanical strength and remodeling capabilities essential for cartilage regeneration. Biomater Sci 2023; 11:6801-6822. [PMID: 37622217 DOI: 10.1039/d3bm00703k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
An important challenge in tissue engineering is the regeneration of functional articular cartilage (AC). In the field, biomimetic hydrogels are being extensively studied as scaffolds that recapitulate microenvironmental features or as mechanical supports for transplanted cells. New advanced hydrogel formulations based on salmon methacrylate gelatin (sGelMA), a cold-adapted biomaterial, are presented in this work. The psychrophilic nature of this biomaterial provides rheological advantages allowing the fabrication of scaffolds with high concentrations of the biopolymer and high mechanical strength, suitable for formulating injectable hydrogels with high mechanical strength for cartilage regeneration. However, highly intricate cell-laden scaffolds derived from highly concentrated sGelMA solutions could be deleterious for cells and scaffold remodeling. On this account, the current study proposes the use of sGelMA supplemented with a mesophilic sacrificial porogenic component. The cytocompatibility of different sGelMA-based formulations is tested through the encapsulation of osteoarthritic chondrocytes (OACs) and stimulated to synthesize extracellular matrix (ECM) components in vitro and in vivo. The sGelMA-derived scaffolds reach high levels of stiffness, and the inclusion of porogens impacts positively the scaffold degradability and molecular diffusion, improved fitness of OACs, increased the expression of cartilage-related genes, increased glycosaminoglycan (GAG) synthesis, and improved remodeling toward cartilage-like tissues. Altogether, these data support the use of sGelMA solutions in combination with mammalian solid gelatin beads for highly injectable formulations for cartilage regeneration, strengthening the importance of the balance between mechanical properties and remodeling capabilities.
Collapse
Affiliation(s)
- Gabriela Zavala
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Sergio M Viafara-García
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Javier Novoa
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Carmen Hidalgo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ingrid Contardo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Facultad de Medicina, Escuela de Nutrición y Dietética, Biopolymer Research & Engineering Laboratory (BiopREL), Universidad de los Andes, Chile
| | - Paulo Díaz-Calderón
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Facultad de Medicina, Escuela de Nutrición y Dietética, Biopolymer Research & Engineering Laboratory (BiopREL), Universidad de los Andes, Chile
| | | | - Maroun Khoury
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Juan Pablo Acevedo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
14
|
Sun K, Guo J, Guo Z, Hou L, Liu H, Hou Y, He J, Guo F, Ye Y. The roles of the Hippo-YAP signalling pathway in Cartilage and Osteoarthritis. Ageing Res Rev 2023; 90:102015. [PMID: 37454824 DOI: 10.1016/j.arr.2023.102015] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Osteoarthritis (OA) is an age-related disease, characterized by cartilage degeneration. The pathogenesis of OA is complicated and the current therapeutic approaches for OA are limited. Cartilage, an integral part of the skeletal system composed of chondrocytes, is essential for skeletal development, tissue patterning, and maintaining the normal activity of joints. The development, homeostasis and degeneration of cartilage are tightly associated with OA. Over the past decade, accumulating evidence indicates that Hippo/YAP is a vital biochemical signalling pathway that strictly governs tissue development and homeostasis. The joint tissues, especially for cartilage, are sensitive to changes of Hippo/YAP signalling. In this review, we summarize the role of Hippo/YAP signalling in cartilage and discuss its involvement in OA progression from points of cartilage degradation, subchondral bone remodeling, and synovial alteration. We also highlight the potential therapeutic implications of Hippo/YAP signalling and further discuss current limitations and controversy on Hippo/YAP-based application for OA treatment.
Collapse
Affiliation(s)
- Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiachao Guo
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhou Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Liangcai Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Haigang Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yanjun Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Junchen He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
15
|
Pettenuzzo S, Arduino A, Belluzzi E, Pozzuoli A, Fontanella CG, Ruggieri P, Salomoni V, Majorana C, Berardo A. Biomechanics of Chondrocytes and Chondrons in Healthy Conditions and Osteoarthritis: A Review of the Mechanical Characterisations at the Microscale. Biomedicines 2023; 11:1942. [PMID: 37509581 PMCID: PMC10377681 DOI: 10.3390/biomedicines11071942] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Biomechanical studies are expanding across a variety of fields, from biomedicine to biomedical engineering. From the molecular to the system level, mechanical stimuli are crucial regulators of the development of organs and tissues, their growth and related processes such as remodelling, regeneration or disease. When dealing with cell mechanics, various experimental techniques have been developed to analyse the passive response of cells; however, cell variability and the extraction process, complex experimental procedures and different models and assumptions may affect the resulting mechanical properties. For these purposes, this review was aimed at collecting the available literature focused on experimental chondrocyte and chondron biomechanics with direct connection to their biochemical functions and activities, in order to point out important information regarding the planning of an experimental test or a comparison with the available results. In particular, this review highlighted (i) the most common experimental techniques used, (ii) the results and models adopted by different authors, (iii) a critical perspective on features that could affect the results and finally (iv) the quantification of structural and mechanical changes due to a degenerative pathology such as osteoarthritis.
Collapse
Affiliation(s)
- Sofia Pettenuzzo
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
| | - Alessandro Arduino
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
| | - Elisa Belluzzi
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), Via Giustiniani 3, 35128 Padova, Italy
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), 35128 Padova, Italy
| | - Assunta Pozzuoli
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), Via Giustiniani 3, 35128 Padova, Italy
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), 35128 Padova, Italy
| | | | - Pietro Ruggieri
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), 35128 Padova, Italy
| | - Valentina Salomoni
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
- Department of Management and Engineering (DTG), Stradella S. Nicola 3, 36100 Vicenza, Italy
| | - Carmelo Majorana
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
| | - Alice Berardo
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
16
|
Zhong W, Pathak JL, Liang Y, Zhytnik L, Pals G, Eekhoff EMW, Bravenboer N, Micha D. The intricate mechanism of PLS3 in bone homeostasis and disease. Front Endocrinol (Lausanne) 2023; 14:1168306. [PMID: 37484945 PMCID: PMC10361617 DOI: 10.3389/fendo.2023.1168306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Since our discovery in 2013 that genetic defects in PLS3 lead to bone fragility, the mechanistic details of this process have remained obscure. It has been established that PLS3 variants cause syndromic and nonsyndromic osteoporosis as well as osteoarthritis. PLS3 codes for an actin-bundling protein with a broad pattern of expression. As such, it is puzzling how PLS3 specifically leads to bone-related disease presentation. Our review aims to summarize the current state of knowledge regarding the function of PLS3 in the predominant cell types in the bone tissue, the osteocytes, osteoblasts and osteoclasts. This is related to the role of PLS3 in regulating mechanotransduction, calcium regulation, vesicle trafficking, cell differentiation and mineralization as part of the complex bone pathology presented by PLS3 defects. Considering the consequences of PLS3 defects on multiple aspects of bone tissue metabolism, our review motivates the study of its mechanism in bone diseases which can potentially help in the design of suitable therapy.
Collapse
Affiliation(s)
- Wenchao Zhong
- Department of Human Genetics, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Clinical Chemistry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Movement Sciences, Tissue Function And Regeneration, Amsterdam, Netherlands
- Department of Temporomandibular Joint, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Janak L. Pathak
- Department of Temporomandibular Joint, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yueting Liang
- Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
- The Second Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Lidiia Zhytnik
- Department of Human Genetics, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Movement Sciences, Tissue Function And Regeneration, Amsterdam, Netherlands
- Department of Traumatology and Orthopaedics, Institute of Clinical Medicine, The University of Tartu, Tartu, Estonia
| | - Gerard Pals
- Department of Human Genetics, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Movement Sciences, Tissue Function And Regeneration, Amsterdam, Netherlands
| | - Elisabeth M. W. Eekhoff
- Department Internal Medicine Section Endocrinology and Metabolism, Amsterdam UMC Location Vrije Universiteit Amsterdam, Rare Bone Disease Center, AMS, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Movement Sciences, Tissue Function And Regeneration, Amsterdam, Netherlands
| | - Dimitra Micha
- Department of Human Genetics, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Movement Sciences, Tissue Function And Regeneration, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| |
Collapse
|
17
|
Muthu S, Korpershoek JV, Novais EJ, Tawy GF, Hollander AP, Martin I. Failure of cartilage regeneration: emerging hypotheses and related therapeutic strategies. Nat Rev Rheumatol 2023:10.1038/s41584-023-00979-5. [PMID: 37296196 DOI: 10.1038/s41584-023-00979-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 06/12/2023]
Abstract
Osteoarthritis (OA) is a disabling condition that affects billions of people worldwide and places a considerable burden on patients and on society owing to its prevalence and economic cost. As cartilage injuries are generally associated with the progressive onset of OA, robustly effective approaches for cartilage regeneration are necessary. Despite extensive research, technical development and clinical experimentation, no current surgery-based, material-based, cell-based or drug-based treatment can reliably restore the structure and function of hyaline cartilage. This paucity of effective treatment is partly caused by a lack of fundamental understanding of why articular cartilage fails to spontaneously regenerate. Thus, research studies that investigate the mechanisms behind the cartilage regeneration processes and the failure of these processes are critical to instruct decisions about patient treatment or to support the development of next-generation therapies for cartilage repair and OA prevention. This Review provides a synoptic and structured analysis of the current hypotheses about failure in cartilage regeneration, and the accompanying therapeutic strategies to overcome these hurdles, including some current or potential approaches to OA therapy.
Collapse
Affiliation(s)
- Sathish Muthu
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, New Delhi, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, India
| | - Jasmijn V Korpershoek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Emanuel J Novais
- Unidade Local de Saúde do Litoral Alentejano, Orthopedic Department, Santiago do Cacém, Portugal
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gwenllian F Tawy
- Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, UK
| | - Anthony P Hollander
- Institute of Lifecourse and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
18
|
Belluzzi E, Todros S, Pozzuoli A, Ruggieri P, Carniel EL, Berardo A. Human Cartilage Biomechanics: Experimental and Theoretical Approaches towards the Identification of Mechanical Properties in Healthy and Osteoarthritic Conditions. Processes (Basel) 2023. [DOI: 10.3390/pr11041014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Articular cartilage is a complex connective tissue with the fundamental functions of load bearing, shock absorption and lubrication in joints. However, traumatic events, aging and degenerative pathologies may affect its structural integrity and function, causing pain and long-term disability. Osteoarthritis represents a health issue, which concerns an increasing number of people worldwide. Moreover, it has been observed that this pathology also affects the mechanical behavior of the articular cartilage. To better understand this correlation, the here proposed review analyzes the physiological aspects that influence cartilage microstructure and biomechanics, with a special focus on the pathological changes caused by osteoarthritis. Particularly, the experimental data on human articular cartilage are presented with reference to different techniques adopted for mechanical testing and the related theoretical mechanical models usually applied to articular cartilage are briefly discussed.
Collapse
|
19
|
Gu Z, Wang J, Fu Y, Pan H, He H, Gan Q, Liu C. Smart Biomaterials for Articular Cartilage Repair and Regeneration. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202212561] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Indexed: 01/06/2025]
Abstract
AbstractArticular cartilage defects bring about disability and worldwide socioeconomic loss, therefore, articular cartilage repair and regeneration is recognized as a global issue. However, due to its avascular and nearly acellular characteristic, cartilage tissue regeneration ability is limited to some extent. Despite the availability of various treatment methods, including palliative drugs and surgical regenerative therapy, articular cartilage repair and regeneration still face major challenges due to the lack of appropriate methods and materials. Smart biomaterials can regulate cell behavior and provide excellent tissue repair and regeneration microenvironment, thus inducing articular cartilage repair and regeneration. This process is adjusted by controlling drug/bioactive factors release via responding to exogenous/endogenous stimuli, tailoring materials’ structure and function similar to native cartilage or providing physiochemical and physical signaling factors. Herein, smart biomaterials, recently applied in articular cartilage repair and regeneration, are elaborated from two aspects: smart drug release system and smart scaffolds. Furthermore, articular cartilage and its defects and advanced manufacturing techniques of smart biomaterials are discussed in brief. Finally, perspectives for smart biomaterials used in articular cartilage repair and regeneration are presented and the clinical translation of smart biomaterials is emphasized.
Collapse
Affiliation(s)
- Zhanghao Gu
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Jiayi Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yu Fu
- School of Aerospace Engineering and Applied Mechanics Tongji University Zhangwu Road 100 Shanghai 200092 P. R. China
| | - Hao Pan
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Hongyan He
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Qi Gan
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry East China University of Science and Technology Shanghai 200237 P. R. China
| |
Collapse
|
20
|
Dilogo IH, Lubis AMT, Perwida NG, Sani SA, Rasyidah RA, Hartanto BR. The Efficacy of Intra-articular Umbilical Cord-Mesenchymal Stem Cell Injection for Knee Osteoarthritis: a Systematic Review. CURRENT STEM CELL REPORTS 2023. [DOI: 10.1007/s40778-023-00223-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
21
|
Weizel A, Distler T, Detsch R, Boccaccini AR, Seitz H, Budday S. Time-dependent hyper-viscoelastic parameter identification of human articular cartilage and substitute materials. J Mech Behav Biomed Mater 2023; 138:105618. [PMID: 36566662 DOI: 10.1016/j.jmbbm.2022.105618] [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: 09/22/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022]
Abstract
Numerical simulations are a valuable tool to understand which processes during mechanical stimulations of hydrogels for cartilage replacement influence the behavior of chondrocytes and contribute to the success or failure of these materials as implants. Such simulations critically rely on the correct prediction of the material response through appropriate material models and corresponding parameters. In this study, we identify hyper-viscoelastic material parameters for numerical simulations in COMSOL Multiphysics® v. 5.6 for human articular cartilage and two replacement materials, the commercially available ChondroFillerliquid and oxidized alginate gelatin (ADA-GEL) hydrogels. We incorporate the realistic experimental boundary conditions into an inverse parameter identification scheme based on data from multiple loading modes simultaneously, including cyclic compression-tension and stress relaxation experiments. We provide individual parameter sets for the unconditioned and conditioned responses and discuss how viscoelastic effects are related to the materials' microstructure. ADA-GEL and ChondroFillerliquid exhibit faster stress relaxation than cartilage with lower relaxation time constants, while cartilage has the largest viscoelastic stress contribution. The elastic response predominates in ADA-GEL and ChondroFillerliquid, while the viscoelastic response predominates in cartilage. These results will help to simulate mechanical stimulations, support the development of suitable materials with distinct mechanical properties in the future and provide parameters and insight into the time-dependent material behavior of human articular cartilage.
Collapse
Affiliation(s)
- A Weizel
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany.
| | - T Distler
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - R Detsch
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - A R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - H Seitz
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany
| | - S Budday
- Institute of Applied Mechanics, Department of Mechanical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
22
|
Petitjean N, Canadas P, Royer P, Noël D, Le Floc'h S. Cartilage biomechanics: From the basic facts to the challenges of tissue engineering. J Biomed Mater Res A 2022; 111:1067-1089. [PMID: 36583681 DOI: 10.1002/jbm.a.37478] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/07/2022] [Accepted: 11/22/2022] [Indexed: 12/31/2022]
Abstract
Articular cartilage (AC) is the thin tissue that covers the long bone ends in the joints and that ensures the transmission of forces between adjacent bones while allowing nearly frictionless movements between them. AC repair is a technologic and scientific challenge that has been addressed with numerous approaches. A major deadlock is the capacity to take in account its complex mechanical properties in repair strategies. In this review, we first describe the major mechanical behaviors of AC for the non-specialists. Then, we show how researchers have progressively identified specific mechanical parameters using mathematical models. There are still gaps in our understanding of some of the observations concerning AC biomechanical properties, particularly the differences in extracellular matrix stiffness measured at the microscale and at the millimetric scale. Nevertheless, for bioengineering applications, AC repair strategies must take into account what are commonly considered the main mechanical features of cartilage: its ability to withstand high stresses through three main behaviors (elasticity, poroelasticity and swelling). Finally, we emphasize that future studies need to investigate AC mechanical properties at different scales, particularly the gradient of mechanical properties around cells and across the cartilage depth, and the differences in mechanical properties at different scales. This multi-scale approach could greatly enhance the success of AC restorative approaches.
Collapse
Affiliation(s)
| | | | - Pascale Royer
- LMGC, University of Montpellier, CNRS, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France.,Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, France
| | | |
Collapse
|
23
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
24
|
Martínez‐Moreno D, Venegas‐Bustos D, Rus G, Gálvez‐Martín P, Jiménez G, Marchal JA. Chondro-Inductive b-TPUe-Based Functionalized Scaffolds for Application in Cartilage Tissue Engineering. Adv Healthc Mater 2022; 11:e2200251. [PMID: 35857383 PMCID: PMC11468339 DOI: 10.1002/adhm.202200251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/13/2022] [Indexed: 01/27/2023]
Abstract
Osteoarthritis is a disease with a great socioeconomic impact and mainly affects articular cartilage, a tissue with reduced self-healing capacity. In this work, 3D printed 1,4 butanediol thermoplastic polyurethane (b-TPUe) scaffolds are functionalized and infrapatellar mesenchymal stem cells are used as the cellular source. Since b-TPUe is a biomaterial with mechanical properties similar to cartilage, but it does not provide the desired environment for cell adhesion, scaffolds are functionalized with two methods, one based on collagen type I and the other in 1-pyrenebutiric acid (PBA) as principal components. Alamar Blue and confocal assays display that PBA functionalized scaffolds support higher cell adhesion and proliferation for the first 21 days. However, collagen type I functionalization induces higher proliferation rates and similar cell viability than the PBA method. Further, both functionalization methods induce extracellular matrix synthesis, and the presence of chondrogenic markers (Sox9, Col2a, and Acan). Finally, SEM images probe that functionalized 3D printed scaffolds present much better cell/biomaterial interactions than controls and confirm early chondrogenesis. These results indicate that the two methods of functionalization in the highly hydrophobic b-TPUe enhance the cell-biomaterial interactions and the improvement in the chondro-inductive properties, which have great potential for application in cartilage tissue engineering.
Collapse
Affiliation(s)
- Daniel Martínez‐Moreno
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaE‐18071Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranadaE‐18100Spain
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranadaE‐18016Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaE‐18016Spain
- BioFab i3D‐ Biofabrication and 3D (bio)printing laboratoryUniversity of GranadaGranadaE‐18100Spain
| | - Desiré Venegas‐Bustos
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaE‐18071Spain
| | - Guillermo Rus
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaE‐18071Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaE‐18016Spain
- Department of Structural MechanicsUniversity of GranadaPolitécnico de FuentenuevaGranadaE‐18071Spain
| | - Patricia Gálvez‐Martín
- Department of Pharmacy and Pharmaceutical TechnologyFaculty of PharmacyUniversity of GranadaGranadaE‐18071Spain
| | - Gema Jiménez
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaE‐18071Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranadaE‐18100Spain
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranadaE‐18016Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaE‐18016Spain
- BioFab i3D‐ Biofabrication and 3D (bio)printing laboratoryUniversity of GranadaGranadaE‐18100Spain
| | - Juan Antonio Marchal
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaE‐18071Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranadaE‐18100Spain
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranadaE‐18016Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaE‐18016Spain
- BioFab i3D‐ Biofabrication and 3D (bio)printing laboratoryUniversity of GranadaGranadaE‐18100Spain
| |
Collapse
|
25
|
Immunohistochemical evaluation of autotaxin and lubricin in mild osteoarthritic rat model performing moderate physical activity. Acta Histochem 2022; 124:151936. [DOI: 10.1016/j.acthis.2022.151936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/20/2022]
|
26
|
Sun P, Xu W, Zhao X, Zhang C, Lin X, Gong M, Fu Z. Ozone induces autophagy by activating PPARγ/mTOR in rat chondrocytes treated with IL-1β. J Orthop Surg Res 2022; 17:351. [PMID: 35842709 PMCID: PMC9287877 DOI: 10.1186/s13018-022-03233-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/07/2022] [Indexed: 11/24/2022] Open
Abstract
Background Osteoarthritis (OA) is the main cause of older pain and disability. Intra-articular injections of ozone (O3) commonly have been found to have antioxidative and anti-inflammatory effects to reduce pain and improve function in knee osteoarthritis. It has been reported that reduced autophagy in chondrocytes plays an important role in the development of OA. This study aimed to probe the role of O3 on the autophagy in chondrocytes treated with IL-1β. Methods Primary chondrocytes were isolated from Wistar rats cartilage within 3 days. The OA chondrocytes model was induced via treatment with IL-1β for 24 h. Then the cells were treated with O3 and GW9662, the inhibitor of PPARγ. Cell viability was assessed by CCK-8. Further, the cells subjected to Western blot analysis, qRT-PCR and immunofluorescence assay. The numbers of autophagosomes were observed via transmission electron microscopy. Results 30 μg/ml O3 improved the viability of chondrocytes treated with IL-1β. The decreased level of autophagy proteins and the numbers of autophagosomes improved in IL-1β-treated chondrocytes with O3 via activating PPARγ/mTOR. In addition, the qRT-PCR results showed that O3 decreased the levels of IL-6, TNF-α and MMP-3, MMP-13 in chondrocytes treated with IL-1β. Conclusions 30 μg/ml O3 improved autophagy via activating PPARγ/mTOR signaling and suppressing inflammation in chondrocytes treated with IL-1β.
Collapse
Affiliation(s)
- Panpan Sun
- Department of Pain Management, The Second Hospital of Shandong University, Jinan, 250033, People's Republic of China.,Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China
| | - Weicheng Xu
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Xu Zhao
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Cong Zhang
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China
| | - Xiaowen Lin
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Moxuan Gong
- Department of Anesthesiology, The Second Hospital of Shandong University, Jinan, 250033, People's Republic of China
| | - Zhijian Fu
- Department of Pain Management, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China. .,Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China.
| |
Collapse
|
27
|
Autologous chondrocyte implantation provides good long-term clinical results in the treatment of knee osteoarthritis: a systematic review. Knee Surg Sports Traumatol Arthrosc 2022; 31:2338-2348. [PMID: 35716187 DOI: 10.1007/s00167-022-07030-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE To evaluate the mid- and long-term efficacy of autologous chondrocyte implantation (ACI) and matrix-assisted chondrocyte implantation (MACI) to treat patients with knee cartilage defects in the presence of osteoarthritis (OA). METHODS PubMed and Cochrane databases were systematically searched for studies describing the treatment of knee OA with ACI or MACI (Kellgren-Lawrence (KL) ≥ 1, minimum follow-up 36 months). Results were reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines and included Lysholm, Western Ontario McMaster University and International Knee Documentation Committee scores. RESULTS Of the 127 full-text articles assessed for eligibility, only five studies were selected based on inclusion/exclusion criteria (2 on ACI and 3 on MACI). In both groups, the defects were mainly located at femoral level, size 2.2-15.1 cm2 in the ACI and 2.0-7.6 cm2 in the MACI group. ACI was mostly used for patients affected by KL I, whereas MACI for patients with KL II-IV. The data obtained from 235 patients (161 ACI, 74 MACI) showed that ACI and MACI sustained stable clinical improvements up to 11 and 15 years, respectively, with a failure rate of about 10% up to 11 years. Scarce biological details regarding chondrocyte implantation were reported. CONCLUSIONS ACI and MACI procedures for the treatment of knee cartilage lesions associated to OA showed long-term success and allowed delaying arthroplasty. Additional trials reporting homogenous data and precise patient characterization are needed to conduct an effective literature meta-analysis and identify the clinical relevance of these procedures. LEVEL OF EVIDENCE IV.
Collapse
|
28
|
Assirelli E, Caravaggi P, Mazzotti A, Ursini F, Leardini A, Belvedere C, Neri S. Location-Dependent Human Osteoarthritis Cartilage Response to Realistic Cyclic Loading: Ex-Vivo Analysis on Different Knee Compartments. Front Bioeng Biotechnol 2022; 10:862254. [PMID: 35782520 PMCID: PMC9240619 DOI: 10.3389/fbioe.2022.862254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Osteoarthritis (OA) is a multifactorial musculoskeletal disorder affecting mostly weight-bearing joints. Chondrocyte response to load is modulated by inflammatory mediators and factors involved in extracellular cartilage matrix (ECM) maintenance, but regulatory mechanisms are not fully clarified yet. By using a recently proposed experimental model combining biomechanical data with cartilage molecular information, basally and following ex-vivo load application, we aimed at improving the understanding of human cartilage response to cyclic mechanical compressive stimuli by including cartilage original anatomical position and OA degree as independent factors. Methods: 19 mono-compartmental Knee OA patients undergoing total knee replacement were recruited. Cartilage explants from four different femoral condyles zones and with different degeneration levels were collected. The response of cartilage samples, pooled according to OA score and anatomical position was tested ex-vivo in a bioreactor. Mechanical stimulation was obtained via a 3-MPa 1-Hz sinusoidal compressive load for 45-min to replicate average knee loading during normal walking. Samples were analysed for chondrocyte gene expression and ECM factor release. Results: Non parametric univariate and multivariate (generalized linear mixed model) analysis was performed to evaluate the effect of compression and IL-1β stimulation in relationship to the anatomical position, local disease severity and clinical parameters with a level of significance set at 0.05. We observed an anti-inflammatory effect of compression inducing a significant downmodulation of IL-6 and IL-8 levels correlated to the anatomical regions, but not to OA score. Moreover, ADAMTS5, PIICP, COMP and CS were upregulated by compression, whereas COL-2CAV was downmodulated, all in relationship to the anatomical position and to the OA degree. Conclusion: While unconfined compression testing may not be fully representative of the in-vivo biomechanical situation, this study demonstrates the importance to consider the original cartilage anatomical position for a reliable biomolecular analysis of knee OA metabolism following mechanical stimulation.
Collapse
Affiliation(s)
- Elisa Assirelli
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Paolo Caravaggi
- Laboratory of Movement Analysis and Functional Evaluation of Prosthesis, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Antonio Mazzotti
- I Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesco Ursini
- Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Science, IRCCS Istituto Ortopedico Rizzoli, University of Bologna, Bologna, Italy
| | - Alberto Leardini
- Laboratory of Movement Analysis and Functional Evaluation of Prosthesis, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Claudio Belvedere
- Laboratory of Movement Analysis and Functional Evaluation of Prosthesis, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Simona Neri
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
29
|
A review of composition‐structure‐function properties and tissue engineering strategies of articular cartilage: compare condyle process and knee‐joint. ADVANCED ENGINEERING MATERIALS 2022. [DOI: 10.1002/adem.202200304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
30
|
Song Z, Li Y, Shang C, Shang G, Kou H, Li J, Chen S, Liu H. Sprifermin: Effects on Cartilage Homeostasis and Therapeutic Prospects in Cartilage-Related Diseases. Front Cell Dev Biol 2022; 9:786546. [PMID: 34970547 PMCID: PMC8712868 DOI: 10.3389/fcell.2021.786546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/23/2021] [Indexed: 11/15/2022] Open
Abstract
When suffering from osteoarthritis (OA), articular cartilage homeostasis is out of balance and the living quality declines. The treatment of knee OA has always been an unsolved problem in the world. At present, symptomatic treatment is mainly adopted for OA. Drug therapy is mainly used to relieve pain symptoms, but often accompanied with adverse reactions; surgical treatment involves the problem of poor integration between the repaired or transplanted tissues and the natural cartilage, leading to the failure of repair. Biotherapy which aims to promote cartilage in situ regeneration and to restore endochondral homeostasis is expected to be an effective method for the prevention and treatment of OA. Disease-modifying osteoarthritis drugs (DMOADs) are intended for targeted treatment of OA. The DMOADs prevent excessive destruction of articular cartilage through anti-catabolism and stimulate tissue regeneration via excitoanabolic effects. Sprifermin (recombinant human FGF18, rhFGF18) is an effective DMOAD, which can not only promote the proliferation of articular chondrocyte and the synthesis of extracellular matrix, increase the thickness of cartilage in a dose-dependent manner, but also inhibit the activity of proteolytic enzymes and remarkedly slow down the degeneration of cartilage. This paper reviews the unique advantages of Sprifermin in repairing cartilage injury and improving cartilage homeostasis, aiming to provide an important strategy for the effective prevention and treatment of cartilage injury-related diseases.
Collapse
Affiliation(s)
- Zongmian Song
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Chunfeng Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongwei Kou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinfeng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Li Y, Yuan Z, Yang H, Zhong H, Peng W, Xie R. Recent Advances in Understanding the Role of Cartilage Lubrication in Osteoarthritis. Molecules 2021; 26:6122. [PMID: 34684706 PMCID: PMC8540456 DOI: 10.3390/molecules26206122] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 01/15/2023] Open
Abstract
The remarkable lubrication properties of normal articular cartilage play an essential role in daily life, providing almost frictionless movements of joints. Alterations of cartilage surface or degradation of biomacromolecules within synovial fluid increase the wear and tear of the cartilage and hence determining the onset of the most common joint disease, osteoarthritis (OA). The irreversible and progressive degradation of articular cartilage is the hallmark of OA. Considering the absence of effective options to treat OA, the mechanosensitivity of chondrocytes has captured attention. As the only embedded cells in cartilage, the metabolism of chondrocytes is essential in maintaining homeostasis of cartilage, which triggers motivations to understand what is behind the low friction of cartilage and develop biolubrication-based strategies to postpone or even possibly heal OA. This review firstly focuses on the mechanism of cartilage lubrication, particularly on boundary lubrication. Then the mechanotransduction (especially shear stress) of chondrocytes is discussed. The following summarizes the recent development of cartilage-inspired biolubricants to highlight the correlation between cartilage lubrication and OA. One might expect that the restoration of cartilage lubrication at the early stage of OA could potentially promote the regeneration of cartilage and reverse its pathology to cure OA.
Collapse
Affiliation(s)
- Yumei Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Zhongrun Yuan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China;
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Hui Yang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Haijian Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Weijie Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Renjian Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
32
|
Dubail J, Cormier-Daire V. Chondrodysplasias With Multiple Dislocations Caused by Defects in Glycosaminoglycan Synthesis. Front Genet 2021; 12:642097. [PMID: 34220933 PMCID: PMC8242584 DOI: 10.3389/fgene.2021.642097] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Chondrodysplasias with multiple dislocations form a group of severe disorders characterized by joint laxity and multiple dislocations, severe short stature of pre- and post-natal onset, hand anomalies, and/or vertebral anomalies. The majority of chondrodysplasias with multiple dislocations have been associated with mutations in genes encoding glycosyltransferases, sulfotransferases, and transporters implicated in the synthesis or sulfation of glycosaminoglycans, long and unbranched polysaccharides composed of repeated disaccharide bond to protein core of proteoglycan. Glycosaminoglycan biosynthesis is a tightly regulated process that occurs mainly in the Golgi and that requires the coordinated action of numerous enzymes and transporters as well as an adequate Golgi environment. Any disturbances of this chain of reactions will lead to the incapacity of a cell to construct correct glycanic chains. This review focuses on genetic and glycobiological studies of chondrodysplasias with multiple dislocations associated with glycosaminoglycan biosynthesis defects and related animal models. Strong comprehension of the molecular mechanisms leading to those disorders, mostly through extensive phenotypic analyses of in vitro and/or in vivo models, is essential for the development of novel biomarkers for clinical screenings and innovative therapeutics for these diseases.
Collapse
Affiliation(s)
- Johanne Dubail
- Université de Paris, INSERM UMR 1163, Institut Imagine, Paris, France
| | - Valérie Cormier-Daire
- Université de Paris, INSERM UMR 1163, Institut Imagine, Paris, France.,Service de Génétique Clinique, Centre de Référence Pour Les Maladies Osseuses Constitutionnelles, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
33
|
Saghati S, Rahbarghazi R, Baradar Khoshfetrat A, Moharamzadeh K, Tayefi Nasrabadi H, Roshangar L. Phenolated alginate-collagen hydrogel induced chondrogenic capacity of human amniotic mesenchymal stem cells. J Biomater Appl 2021; 36:789-802. [PMID: 34074175 DOI: 10.1177/08853282211021692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Horseradish peroxidase (HRP)-catalyzed hydrogels are considered to be an important platform for tissue engineering applications. In this study, we investigated the chondrogenic capacity of phenolated (1.2%) alginate-(0.5%) collagen hydrogel on human amniotic mesenchymal stem cells after 21 days. Using NMR, FTIR analyses, and SEM imaging, we studied the phenolation and structure of alginate-collagen hydrogel. For physicochemical evaluations, gelation time, mechanical properties, swelling, and degradation rate were assessed. The survival rate was monitored using the MTT assay and DAPI staining. Western blotting was performed to measure the chondrogenic differentiation of cells. NMR showed successful phenolation of the alginate-collagen hydrogel. FTIR exhibited the interaction between the functional groups of collagen with phenolated alginate. SEM showed the existence of collagen microfibrils in the alginate-collagen hydrogel. Compared to phenolated alginate, the addition of collagen increased hydrogel elasticity by 10%. Both swelling rate and biodegradability were reduced in the presence of collagen. We noted an increased survival rate in phenolated alginate-collagen compared to the control cells (p < 0.05). Western blotting revealed the increase of chondrocyte-associated proteins such as SOX9 and COL2A1 in phenolated-alginate-collagen hydrogels after 21 days. These data showed that phenolated alginate-collagen hydrogel is an appropriate 3 D substrate to induce chondrogenic capacity of human mesenchymal stem cells.
Collapse
Affiliation(s)
- Sepideh Saghati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Baradar Khoshfetrat
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamid Tayefi Nasrabadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Plastin 3 in health and disease: a matter of balance. Cell Mol Life Sci 2021; 78:5275-5301. [PMID: 34023917 PMCID: PMC8257523 DOI: 10.1007/s00018-021-03843-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
For a long time, PLS3 (plastin 3, also known as T-plastin or fimbrin) has been considered a rather inconspicuous protein, involved in F-actin-binding and -bundling. However, in recent years, a plethora of discoveries have turned PLS3 into a highly interesting protein involved in many cellular processes, signaling pathways, and diseases. PLS3 is localized on the X-chromosome, but shows sex-specific, inter-individual and tissue-specific expression variability pointing towards skewed X-inactivation. PLS3 is expressed in all solid tissues but usually not in hematopoietic cells. When escaping X-inactivation, PLS3 triggers a plethora of different types of cancers. Elevated PLS3 levels are considered a prognostic biomarker for cancer and refractory response to therapies. When it is knocked out or mutated in humans and mice, it causes osteoporosis with bone fractures; it is the only protein involved in actin dynamics responsible for osteoporosis. Instead, when PLS3 is upregulated, it acts as a highly protective SMN-independent modifier in spinal muscular atrophy (SMA). Here, it seems to counteract reduced F-actin levels by restoring impaired endocytosis and disturbed calcium homeostasis caused by reduced SMN levels. In contrast, an upregulation of PLS3 on wild-type level might cause osteoarthritis. This emphasizes that the amount of PLS3 in our cells must be precisely balanced; both too much and too little can be detrimental. Actin-dynamics, regulated by PLS3 among others, are crucial in a lot of cellular processes including endocytosis, cell migration, axonal growth, neurotransmission, translation, and others. Also, PLS3 levels influence the infection with different bacteria, mycosis, and other pathogens.
Collapse
|
35
|
Richardson BM, Walker CJ, Maples MM, Randolph MA, Bryant SJ, Anseth KS. Mechanobiological Interactions between Dynamic Compressive Loading and Viscoelasticity on Chondrocytes in Hydrazone Covalent Adaptable Networks for Cartilage Tissue Engineering. Adv Healthc Mater 2021; 10:e2002030. [PMID: 33738966 PMCID: PMC8785214 DOI: 10.1002/adhm.202002030] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/17/2021] [Indexed: 12/17/2022]
Abstract
Mechanobiological cues influence chondrocyte biosynthesis and are often used in tissue engineering applications to improve the repair of articular cartilage in load-bearing joints. In this work, the biophysical effects of an applied dynamic compression on chondrocytes encapsulated in viscoelastic hydrazone covalent adaptable networks (CANs) is explored. Here, hydrazone CANs exhibit viscoelastic loss tangents ranging from (9.03 ± 0.01) 10-4 to (1.67 ± 0.09) 10-3 based on the molar percentages of alkyl-hydrazone and benzyl-hydrazone crosslinks. Notably, viscoelastic alkyl-hydrazone crosslinks improve articular cartilage specific gene expression showing higher SOX9 expression in free swelling hydrogels and dynamic compression reduces hypertrophic chondrocyte markers (COL10A1, MMP13) in hydrazone CANs. Interestingly, dynamic compression also improves matrix biosynthesis in elastic benzyl-hydrazone controls but reduces biosynthesis in viscoelastic alkyl-hydrazone CANs. Additionally, intermediate levels of viscoelastic adaptability demonstrate the highest levels of matrix biosynthesis in hydrazone CANs, demonstrating on average 70 ± 4 µg of sulfated glycosaminoglycans per day and 31 ± 3 µg of collagen per day over one month in dynamic compression bioreactors. Collectively, the results herein demonstrate the role of matrix adaptability and viscoelasticity on chondrocytes in hydrazone CANs during dynamic compression, which may prove useful for tissue engineering applications in load-bearing joints.
Collapse
Affiliation(s)
- Benjamin M Richardson
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
| | - Cierra J Walker
- The BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
- Materials Science and Engineering Program, University of Colorado Boulder, 4001 Discovery Drive, Boulder, CO, 80303, USA
| | - Mollie M Maples
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
| | - Mark A Randolph
- Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, WAC 435, Boston, MA, 02114, USA
- Division of Plastic Surgery, Massachusetts General Hospital, Harvard Medical School, 15 Parkman St, WACC 453, Boston, MA, 02114, USA
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
- Materials Science and Engineering Program, University of Colorado Boulder, 4001 Discovery Drive, Boulder, CO, 80303, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, Boulder, CO, 80303, USA
| |
Collapse
|
36
|
Poillot P, Le Maitre CL, Huyghe JM. The strain-generated electrical potential in cartilaginous tissues: a role for piezoelectricity. Biophys Rev 2021; 13:91-100. [PMID: 33747246 PMCID: PMC7930161 DOI: 10.1007/s12551-021-00779-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/01/2021] [Indexed: 12/26/2022] Open
Abstract
The strain-generated potential (SGP) is a well-established mechanism in cartilaginous tissues whereby mechanical forces generate electrical potentials. In articular cartilage (AC) and the intervertebral disc (IVD), studies on the SGP have focused on fluid- and ionic-driven effects, namely Donnan, diffusion and streaming potentials. However, recent evidence has indicated a direct coupling between strain and electrical potential. Piezoelectricity is one such mechanism whereby deformation of most biological structures, like collagen, can directly generate an electrical potential. In this review, the SGP in AC and the IVD will be revisited in light of piezoelectricity and mechanotransduction. While the evidence base for physiologically significant piezoelectric responses in tissue is lacking, difficulties in quantifying the physiological response and imperfect measurement techniques may have underestimated the property. Hindering our understanding of the SGP further, numerical models to-date have negated ferroelectric effects in the SGP and have utilised classic Donnan theory that, as evidence argues, may be oversimplified. Moreover, changes in the SGP with degeneration due to an altered extracellular matrix (ECM) indicate that the significance of ionic-driven mechanisms may diminish relative to the piezoelectric response. The SGP, and these mechanisms behind it, are finally discussed in relation to the cell response.
Collapse
Affiliation(s)
- Philip Poillot
- Bernal Institute, University of Limerick, Limerick, Ireland
| | | | - Jacques M. Huyghe
- Bernal Institute, University of Limerick, Limerick, Ireland
- Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
37
|
Disorganization of chondrocyte columns in the growth plate does not aggravate experimental osteoarthritis in mice. Sci Rep 2020; 10:10745. [PMID: 32612184 PMCID: PMC7329885 DOI: 10.1038/s41598-020-67518-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/09/2020] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is a multifactorial joint disease mainly affecting articular cartilage (AC) with a relevant biomechanical component. During endochondral ossification growth plate (GP) chondrocytes arrange in columns. GPs do not ossify in skeletally mature rodents. In neonatal mice, an altered joint loading induces GP chondrocyte disorganization. We aimed to study whether experimental OA involves GP disorganization in adult mice and to assess if it may have additional detrimental effects on AC damage. Knee OA was induced by destabilization of the medial meniscus (DMM) in wild-type (WT) adult mice, and in Tamoxifen-inducible Ellis-van-Creveld syndrome protein (Evc) knockouts (EvccKO), used as a model of GP disorganization due to Hedgehog signalling disruption. Chondrocyte column arrangement was assessed in the tibial GP and expressed as Column Index (CI). Both DMM-operated WT mice and non-operated-EvccKO showed a decreased CI, indicating GP chondrocyte column disarrangement, although in the latter, it was not associated to AC damage. The most severe GP chondrocyte disorganization occurred in DMM-EvccKO mice, in comparison to the other groups. However, this altered GP structure in DMM-EvccKO mice did not exacerbate AC damage. Further studies are needed to confirm the lack of interference of GP alterations on the analysis of AC employing OA mice.
Collapse
|