1
|
Chen Y, Li G, Ge Y, Liu S, Weng J, Lin J, Xiong A, Zeng H, Wu X, Yang J, Yu F. Repair of Cartilage Defects Using ATDC5 Cells Treated with BBR Loaded in Chitosan Hydrogel. ACS Biomater Sci Eng 2025; 11:493-505. [PMID: 39654118 DOI: 10.1021/acsbiomaterials.4c01645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
In this study, we explore the cartilage defect repair mechanism by phosphocreatine-grafted chitosan hydrogels loaded with berberine-treated ATDC5 cells (CSMP@BBR@ATDC5). Under the optimal concentrations of LPS and BBR ideal conditions, ATDC5 cell toxicity and proliferation were detected with AM/PI and EdU staining. Additionally, qPCR and Western blot were employed to detect the expression of the SIRT1/BMP4 signaling pathway and chondrogenic-related factors in ATDC5 cells. Moreover, BBR-treated ATDC5 was seeded into a phosphocreatine-grafted chitosan hydrogel system. Subsequently, the cartilage defect was established in mice. After 4, 8, and 12 weeks, knee specimens were collected to evaluate the repair of cartilage defects. According to our findings, BBR can increase ATDC5 viability by LPS treatment. Likewise, it upregulates the SIRT1/BMP4 signaling pathway expression and chondrogenic-related factors. Another, it was shown by histological observation that the cartilage defect had been repaired more effectively in the CSMP@BBR@ATDC5 group than in the other groups. Finally, the expressions of chondrogenic-related factors and SIRT1/BMP4 signaling pathway were upregulates in CSMP@BBR@ATDC5 than in other groups. In vitro, BBR protects inflammatory ATDC5 cells and maintains the expression of chondrogenic-related factors. Subsequently, we successfully use CSMP@BBR@ATDC 5 to repair knee cartilage defects in mice.
Collapse
Affiliation(s)
- Yixiao Chen
- Department of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100000, China
- National Center of Orthopaedics, Beijing 100000, China
| | - Guoqing Li
- National Center of Orthopaedics, Beijing 100000, China
- Department of Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100000, China
| | - Yufeng Ge
- Department of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100000, China
- National Center of Orthopaedics, Beijing 100000, China
| | - Su Liu
- Department of Orthopaedics Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jian Weng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Ao Xiong
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Hui Zeng
- Department of Orthopaedics, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Xinbao Wu
- Department of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100000, China
- National Center of Orthopaedics, Beijing 100000, China
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Fei Yu
- Department of Spine Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| |
Collapse
|
2
|
Dong W, Yang C, Guo D, Jia M, Wang Y, Wang J. PTX3-assembled pericellular hyaluronan matrix enhances endochondral ossification during fracture healing and heterotopic ossification. Bone 2024; 192:117385. [PMID: 39732447 DOI: 10.1016/j.bone.2024.117385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Endochondral ossification (EO) is a pivotal process during fracture healing and traumatic heterotopic ossification (HO), involving the cartilaginous matrix synthesis and mineralization. Unlike the extracellular matrix, the hyaluronan (HA)-rich pericellular matrix (PCM) directly envelops chondrocytes, serving as the frontline for extracellular signal reception and undergoing dynamic remodeling. Pentraxin 3 (PTX3), a secreted glycoprotein, facilitates HA matrix assembly and remodeling. However, it remains unclear whether PTX3 affects EO by regulating HA-rich PCM assembly of chondrocytes, thereby impacting fracture healing and traumatic HO. This study demonstrates that PTX3 deficiency impairs fracture healing and inhibits traumatic HO, but dose not affect growth plate development in mice. PTX3 expression is up-regulated during chondrocyte matrix synthesis and maturation and is localized in the HA-rich PCM. PTX3 promotes the assembly of HA-rich PCM in a serum- and TSG6-dependent manner, fostering CD44 receptor clustering, activating the FAK/AKT signaling pathway, and promoting chondrocyte matrix synthesis and maturation. Local injection of PTX3/TSG6 matrix protein mixture effectively promotes fracture healing in mice. In conclusion, PTX3-assembled HA-rich PCM promotes chondrocyte matrix synthesis and maturation via CD44/FAK/AKT signaling. This mechanism facilitates EO during fracture healing and traumatic HO in mice.
Collapse
Affiliation(s)
- Wei Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Chang Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Donghua Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Meie Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jiawei Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
3
|
Sawyer M, Semodji A, Nielson O, Rektor A, Burgoyne H, Eppel M, Eixenberger J, Montenegro-Brown R, Nelson ML, Lujan T, Estrada D. Direct Scaffold-Coupled Electrical Stimulation of Chondrogenic Progenitor Cells through Graphene Foam Bioscaffolds to Control Mechanical Properties of Graphene Foam - Cell Composites. RESEARCH SQUARE 2024:rs.3.rs-5589589. [PMID: 39764126 PMCID: PMC11703340 DOI: 10.21203/rs.3.rs-5589589/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Osteoarthritis, a major global cause of pain and disability, is driven by the irreversible degradation of hyaline cartilage in joints. Cartilage tissue engineering presents a promising therapeutic avenue, but success hinges on replicating the native physiological environment to guide cellular behavior and generate tissue constructs that mimic natural cartilage. Although electrical stimulation has been shown to enhance chondrogenesis and extracellular matrix production in 2D cultures, the mechanisms underlying these effects remain poorly understood, particularly in 3D models. Here, we report that direct scaffold-coupled electrical stimulation applied to 3D graphene foam bioscaffolds significantly enhances the mechanical properties of the resulting graphene foam - cell constructs. Using custom 3D-printed electrical stimulus chambers, we applied biphasic square impulses (20, 40, 60 mVpp at 1 kHz) for 5 minutes daily over 7 days. Stimulation at 60 mVpp increased the steady-state energy dissipation and equilibrium modulus by approximately 65% and 25%, respectively, compared to unstimulated controls, while also yielding the highest cell density among stimulated samples. In addition, our custom chambers facilitated full submersion of the hydrophobic graphene foam in media, leading to enhanced cell attachment and integration across the scaffold surface and within its hollow branches. To assess this cellular integration, we employed co-localized confocal fluorescence microscopy and X-ray microCT imaging enabled by colloidal gold nanoparticle and fluorophore staining, which allowed visualization of cell distribution within the opaque scaffold's internal structure. These findings highlight the potential of direct scaffold-coupled electrical stimulus to modulate the mechanical properties of engineered tissues and offer new insights into the emergent behavior of cells within conductive 3D bioscaffolds.
Collapse
|
4
|
Bergstrom AR, Glimm MG, Houske EA, Cooper G, Viles E, Chapman M, Bourekis K, Welhaven HD, Brahmachary PP, Hahn AK, June RK. Metabolic Profiles of Encapsulated Chondrocytes Exposed to Short-Term Simulated Microgravity. Ann Biomed Eng 2024:10.1007/s10439-024-03667-x. [PMID: 39695002 DOI: 10.1007/s10439-024-03667-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
The mechanism by which chondrocytes respond to reduced mechanical loading environments and the subsequent risk of developing osteoarthritis remains unclear. This is of particular concern for astronauts. In space the reduced joint loading forces during prolonged microgravity (10-6 g) exposure could lead to osteoarthritis (OA), compromising quality of life post-spaceflight. In this study, we encapsulated human chondrocytes in an agarose gel of similar stiffness to the pericellular matrix to mimic the cartilage microenvironment. We then exposed agarose-chondrocyte constructs to simulated microgravity (SM) for four days using a rotating wall vessel (RWV) bioreactor to better assess the cartilage health risks associated with spaceflight. Metabolites extracted from media and agarose gel constructs were analyzed on liquid chromatography-mass spectrometry. Global metabolomic profiling detected a total of 1205 metabolite features, with 497 significant metabolite features identified by ANOVA (FDR-corrected p-value < 0.05). Specific metabolic shifts detected in response to SM exposure resulted in clusters of co-regulated metabolites, with glutathione, nitrogen, histidine, vitamin B3, and aminosugars metabolism identified by variable importance in projection scores. Microgravity-induced metabolic shifts in gel constructs and media were indicative of protein synthesis, energy and nucleotide metabolism, and oxidative catabolism. Microgravity associated-metabolic shifts were consistent with our previously published early osteoarthritic metabolomic profiles in human synovial fluid, suggesting that even short-term exposure to microgravity (or other reduced mechanical loading environments) may lead to the development of OA. This work further suggests the potential to detect these metabolic perturbations in synovial fluid in vivo to ascertain osteoarthritis risk in astronauts.
Collapse
Affiliation(s)
- Annika R Bergstrom
- Department of Biological and Environmental Science, Carroll College, 1601 N Benton Ave, Helena, MT, 59625, USA
- Department of Chemical and Biological Engineering, Villanova University, Villanova, PA, 19085, USA
| | - Matthew G Glimm
- Department of Biological and Environmental Science, Carroll College, 1601 N Benton Ave, Helena, MT, 59625, USA
| | - Eden A Houske
- Department of Biological and Environmental Science, Carroll College, 1601 N Benton Ave, Helena, MT, 59625, USA
| | - Gwendolyn Cooper
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Ethan Viles
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA
- Department of Mechanical and Industrial Engineering, Montana State University, PO Box 173800, Bozeman, MT, 59717-3800, USA
| | - Marrin Chapman
- Department of Biological and Environmental Science, Carroll College, 1601 N Benton Ave, Helena, MT, 59625, USA
| | - Katherine Bourekis
- Department of Biological and Environmental Science, Carroll College, 1601 N Benton Ave, Helena, MT, 59625, USA
| | - Hope D Welhaven
- Molecular Biosciences Program, Montana State University, Bozeman, MT, 59717, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Priyanka P Brahmachary
- Department of Mechanical and Industrial Engineering, Montana State University, PO Box 173800, Bozeman, MT, 59717-3800, USA
| | - Alyssa K Hahn
- Department of Biological and Environmental Science, Carroll College, 1601 N Benton Ave, Helena, MT, 59625, USA.
| | - Ronald K June
- Department of Mechanical and Industrial Engineering, Montana State University, PO Box 173800, Bozeman, MT, 59717-3800, USA.
| |
Collapse
|
5
|
Plath AMS, de Lima PHC, Amicone A, Bissacco EG, Mosayebi M, Berton SBR, Ferguson SJ. Toward low-friction and high-adhesion solutions: Emerging strategies for nanofibrous scaffolds in articular cartilage engineering. BIOMATERIALS ADVANCES 2024; 169:214129. [PMID: 39642717 DOI: 10.1016/j.bioadv.2024.214129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
Aging, trauma, pathology, and poor natural tissue regeneration are the leading causes of osteoarthritis (OA), an articular cartilage disease. Electrospun scaffolds have gained attention as potential matrices for the treatment of OA because of their high degree of ECM mimicry, which suits chondrocyte migration, adhesion, and proliferation. However, none of the products recently introduced in the market are nanofiber-based. This study aimed to review the scope and tribology of nanofibrous articular cartilage scaffolds. Herein, we briefly discuss cartilage lubrication and strategies for promoting cell adhesion in electrospun materials. Next, we discuss the emerging need to study the biotribological properties of scaffolds. Finally, we review new perspectives on surface functionalization, surface segregation, Janus membranes, layer-by-layer fabrication, and nanofibrous composites. We conclude that cell adhesion and low-friction conciliation remain poorly explored in the recent literature. The topic intersection might create novelties in the field.
Collapse
Affiliation(s)
| | - Pedro Henrique Correia de Lima
- Department of Physics and Chemistry, São Paulo State University (UNESP), Av. Brasil, 56, 15385007 Ilha Solteira, Brazil.
| | - Alessio Amicone
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| | | | - Mahdieh Mosayebi
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| | | | - Stephen J Ferguson
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37-39, 8092 Zurich, Switzerland
| |
Collapse
|
6
|
Li T, Liu J, Guo M, Bin FC, Duan Q, Dong XZ, Jin F, Fujita K, Zheng ML. Femtosecond Laser Maskless Optical Projection Lithography of Cartilage PCM Inspired 3D Protein Matrix to Chondrocyte Phenotype. Adv Healthc Mater 2024; 13:e2400849. [PMID: 38687974 DOI: 10.1002/adhm.202400849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/28/2024] [Indexed: 05/02/2024]
Abstract
Hydrogels containing chondrocytes have exhibited excellent potential in regenerating hyaline cartilage. However, chondrocytes are vulnerable to dedifferentiation during in vitro culture, leading to fibrosis and mechanical degradation of newly formed cartilage. It is proposed to modulate cartilage formation via the developed chondrocyte pericellular matrix (PCM) -like scaffolds for the first time, in which the S, M, and L-sized scaffolds are fabricated by femtosecond laser maskless optical projection lithography (FL-MOPL) of bovine serum albumin-glyceryl methacrylate hydrogel. Chondrocytes on the M PCM-like scaffold can maintain round morphology and synthesize extracellular matrix (ECM) to induce regeneration of hyaline cartilage microtissues by geometrical restriction. A series of M PCM-like scaffolds is fabricated with different stiffness and those with a high Young's modulus are more effective in maintaining the chondrocyte phenotype. The proposed PCM-like scaffolds are effective in modulating cartilage formation influenced by pore size, depth, and stiffness, which will pave the way for a better understanding of the geometric cues of mechanotransduction interactions in regulating cell fate and open up new avenues for tissue engineering.
Collapse
Affiliation(s)
- Teng Li
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Jie Liu
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Min Guo
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Fan-Chun Bin
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Qi Duan
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Xian-Zi Dong
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Feng Jin
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Katsumasa Fujita
- Department of Applied Physics, Osaka University, 2-1Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mei-Ling Zheng
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| |
Collapse
|
7
|
Bergstrom AR, Glimm MG, Houske EA, Cooper G, Viles E, Chapman M, Bourekis K, Welhaven HD, Brahmachary PP, Hahn AK, June RK. Metabolic Profiles of Encapsulated Chondrocytes Exposed to Short-Term Simulated Microgravity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601604. [PMID: 39005264 PMCID: PMC11245029 DOI: 10.1101/2024.07.01.601604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The mechanism by which chondrocytes respond to reduced mechanical loading environments and the subsequent risk of developing osteoarthritis remains unclear. This is of particular concern for astronauts. In space the reduced joint loading forces during prolonged microgravity (10-6 g) exposure could lead to osteoarthritis (OA), compromising quality of life post-spaceflight. In this study, we encapsulated human chondrocytes in an agarose gel of similar stiffness to the pericellular matrix to mimic the cartilage microenvironment. We then exposed agarose-chondrocyte constructs to simulated microgravity (SM) using a rotating wall vessel (RWV) bioreactor to better assess the cartilage health risks associated with spaceflight. Global metabolomic profiling detected a total of 1205 metabolite features across all samples, with 497 significant metabolite features identified by ANOVA (FDR-corrected p-value < 0.05). Specific metabolic shifts detected in response to SM exposure resulted in clusters of co-regulated metabolites, as well as key metabolites identified by variable importance in projection scores. Microgravity-induced metabolic shifts in gel constructs and media were indicative of protein synthesis, energy metabolism, nucleotide metabolism, and oxidative catabolism. The microgravity associated-metabolic shifts were consistent with early osteoarthritic metabolomic profiles in human synovial fluid, which suggests that even short-term exposure to microgravity (or other reduced mechanical loading environments) may lead to the development of OA.
Collapse
Affiliation(s)
- Annika R. Bergstrom
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
- Department of Chemical & Biological Engineering, Villanova University, Villanova, PA, USA, 19085
| | - Matthew G. Glimm
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Eden A. Houske
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Gwendolyn Cooper
- Molecular Biosciences Program, Montana State University, Bozeman, MT, USA, 59717
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT, USA, 59717
| | - Ethan Viles
- Molecular Biosciences Program, Montana State University, Bozeman, MT, USA, 59717
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA, 59717
| | - Marrin Chapman
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Katherine Bourekis
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Hope D. Welhaven
- Molecular Biosciences Program, Montana State University, Bozeman, MT, USA, 59717
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT, USA, 59717
| | - Priyanka P. Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA, 59717
| | - Alyssa K. Hahn
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA, 59717
| |
Collapse
|
8
|
Mobasheri A, Matta C, Giles W, Choi H, Ivanavicius S. The interplay between inflammatory mediators and mechanotransduction is mediated by ion channels in articular chondrocytes: Functional consequences in osteoarthritis. Phys Life Rev 2024; 49:123-126. [PMID: 38692123 DOI: 10.1016/j.plrev.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Affiliation(s)
- Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, PR China; World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Wayne Giles
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Heonsik Choi
- Healthcare Research Institute, Kolon Advanced Research Cluster, Magok-dong, Gangseo-gu, Seoul, South Korea
| | | |
Collapse
|
9
|
Chen L, Yang J, Cai Z, Huang Y, Xiao P, Wang J, Wang F, Huang W, Cui W, Hu N. Electroactive Biomaterials Regulate the Electrophysiological Microenvironment to Promote Bone and Cartilage Tissue Regeneration. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202314079] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Indexed: 01/06/2025]
Abstract
AbstractThe incidence of large bone and articular cartilage defects caused by traumatic injury is increasing worldwide; the tissue regeneration process for these injuries is lengthy due to limited self‐healing ability. Endogenous bioelectrical phenomenon has been well recognized to play an important role in bone and cartilage homeostasis and regeneration. Studies have reported that electrical stimulation (ES) can effectively regulate various biological processes and holds promise as an external intervention to enhance the synthesis of the extracellular matrix, thereby accelerating the process of bone and cartilage regeneration. Hence, electroactive biomaterials have been considered a biomimetic approach to ensure functional recovery by integrating various physiological signals, including electrical, biochemical, and mechanical signals. This review will discuss the role of endogenous bioelectricity in bone and cartilage tissue, as well as the effects of ES on cellular behaviors. Then, recent advances in electroactive materials and their applications in bone and cartilage tissue regeneration are systematically overviewed, with a focus on their advantages and disadvantages as tissue repair materials and performances in the modulation of cell fate. Finally, the significance of mimicking the electrophysiological microenvironment of target tissue is emphasized and future development challenges of electroactive biomaterials for bone and cartilage repair strategies are proposed.
Collapse
Affiliation(s)
- Li Chen
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Jianye Yang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Zhengwei Cai
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Yanran Huang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Pengcheng Xiao
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Juan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Fan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Wei Huang
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| | - Wenguo Cui
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Ning Hu
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University Orthopaedic Research Laboratory, Chongqing Medical University Chongqing 400016 China
| |
Collapse
|
10
|
Fan Z, Liu Y, Lan Y, Wu Y, Li J, Xu X. CoCl 2-Induced hypoxia promotes hPDLSCs osteogenic differentiation through AKT/mTOR/4EBP-1/HIF-1α signaling and facilitates the repair of alveolar bone defects. Cell Biol Int 2024; 48:808-820. [PMID: 38433534 DOI: 10.1002/cbin.12148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/11/2024] [Accepted: 02/17/2024] [Indexed: 03/05/2024]
Abstract
Bone defects are characterized by a hypoxic environment, which affects bone tissue repair. However, the role of hypoxia in the repair of alveolar bone defects remains unclear. Human periodontal ligament stem cells (hPDLSCs) are high-quality seed cells for repairing alveolar bone defects, whose behavior changes under hypoxia. However, their mechanism of action is not known and needs to be elucidated. We hypothesized that hypoxia might be beneficial to alveolar bone defect repair and the osteogenic differentiation of hPDLSCs. To test this hypothesis, cobalt chloride (CoCl2) was used to create a hypoxic environment, both in vitro and in vivo. In vitro study, the best osteogenic effect was observed after 48 h of hypoxia in hPDLSCs, and the AKT/mammalian target of rapamycin/eukaryotic translation initiation factor 4e-binding protein 1 (AKT/mTOR/4EBP-1) signaling pathway was significantly upregulated. Inhibition of the AKT/mTOR/4EBP-1 signaling pathway decreased the osteogenic ability of hPDLSCs under hypoxia and hypoxia-inducible factor 1 alpha (HIF-1α) expression. The inhibition of HIF-1α also decreased the osteogenic capacity of hPDLSCs under hypoxia without significantly affecting the level of phosphorylation of AKT/mTOR/4EBP-1. In vitro study, Micro-CT and tissue staining results show better bone regeneration in hypoxic group than control group. These results suggested that hypoxia promoted alveolar bone defect repair and osteogenic differentiation of hPDLSCs, probably through AKT/mTOR/4EBP-1/HIF-1α signaling. These findings provided important insights into the regulatory mechanism of hypoxia in hPDLSCs and elucidated the effect of hypoxia on the healing of alveolar bone defects. This study highlighted the importance of physiological oxygen conditions for tissue engineering.
Collapse
Affiliation(s)
- Zhibo Fan
- Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Yanru Liu
- Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Yuxin Lan
- Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Yujie Wu
- Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Junyu Li
- Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Xiaomei Xu
- Department of Orthodontics, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Sulcanese L, Prencipe G, Canciello A, Cerveró-Varona A, Perugini M, Mauro A, Russo V, Barboni B. Stem-Cell-Driven Chondrogenesis: Perspectives on Amnion-Derived Cells. Cells 2024; 13:744. [PMID: 38727280 PMCID: PMC11083072 DOI: 10.3390/cells13090744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Regenerative medicine harnesses stem cells' capacity to restore damaged tissues and organs. In vitro methods employing specific bioactive molecules, such as growth factors, bio-inductive scaffolds, 3D cultures, co-cultures, and mechanical stimuli, steer stem cells toward the desired differentiation pathways, mimicking their natural development. Chondrogenesis presents a challenge for regenerative medicine. This intricate process involves precise modulation of chondro-related transcription factors and pathways, critical for generating cartilage. Cartilage damage disrupts this process, impeding proper tissue healing due to its unique mechanical and anatomical characteristics. Consequently, the resultant tissue often forms fibrocartilage, which lacks adequate mechanical properties, posing a significant hurdle for effective regeneration. This review comprehensively explores studies showcasing the potential of amniotic mesenchymal stem cells (AMSCs) and amniotic epithelial cells (AECs) in chondrogenic differentiation. These cells exhibit innate characteristics that position them as promising candidates for regenerative medicine. Their capacity to differentiate toward chondrocytes offers a pathway for developing effective regenerative protocols. Understanding and leveraging the innate properties of AMSCs and AECs hold promise in addressing the challenges associated with cartilage repair, potentially offering superior outcomes in tissue regeneration.
Collapse
Affiliation(s)
- Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Monia Perugini
- Department of Bioscience and Technology for Food, Agriculture, and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| |
Collapse
|
12
|
Agten H, Van Hoven I, Van Hoorick J, Van Vlierberghe S, Luyten FP, Bloemen V. In vitro and in vivo evaluation of periosteum-derived cells and iPSC-derived chondrocytes encapsulated in GelMA for osteochondral tissue engineering. Front Bioeng Biotechnol 2024; 12:1386692. [PMID: 38665810 PMCID: PMC11043557 DOI: 10.3389/fbioe.2024.1386692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Osteochondral defects are deep joint surface lesions that affect the articular cartilage and the underlying subchondral bone. In the current study, a tissue engineering approach encompassing individual cells encapsulated in a biocompatible hydrogel is explored in vitro and in vivo. Cell-laden hydrogels containing either human periosteum-derived progenitor cells (PDCs) or human induced pluripotent stem cell (iPSC)-derived chondrocytes encapsulated in gelatin methacryloyl (GelMA) were evaluated for their potential to regenerate the subchondral mineralized bone and the articular cartilage on the joint surface, respectively. PDCs are easily isolated and expanded progenitor cells that are capable of generating mineralized cartilage and bone tissue in vivo via endochondral ossification. iPSC-derived chondrocytes are an unlimited source of stable and highly metabolically active chondrocytes. Cell-laden hydrogel constructs were cultured for up to 28 days in a serum-free chemically defined chondrogenic medium. On day 1 and day 21 of the differentiation period, the cell-laden constructs were implanted subcutaneously in nude mice to evaluate ectopic tissue formation 4 weeks post-implantation. Taken together, the data suggest that iPSC-derived chondrocytes encapsulated in GelMA can generate hyaline cartilage-like tissue constructs with different levels of maturity, while using periosteum-derived cells in the same construct type generates mineralized tissue and cortical bone in vivo. Therefore, the aforementioned cell-laden hydrogels can be an important part of a multi-component strategy for the manufacturing of an osteochondral implant.
Collapse
Affiliation(s)
- Hannah Agten
- Department of Materials Engineering, Surface and Interface Engineered Materials (SIEM), Group T Leuven Campus, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Inge Van Hoven
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | | | - Sandra Van Vlierberghe
- BIO INX BV, Zwijnaarde, Belgium
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Frank P. Luyten
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Veerle Bloemen
- Department of Materials Engineering, Surface and Interface Engineered Materials (SIEM), Group T Leuven Campus, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Melrose J. Hippo cell signaling and HS-proteoglycans regulate tissue form and function, age-dependent maturation, extracellular matrix remodeling, and repair. Am J Physiol Cell Physiol 2024; 326:C810-C828. [PMID: 38223931 DOI: 10.1152/ajpcell.00683.2023] [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: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
This review examined how Hippo cell signaling and heparan sulfate (HS)-proteoglycans (HSPGs) regulate tissue form and function. Despite being a nonweight-bearing tissue, the brain is regulated by Hippo mechanoresponsive cell signaling pathways during embryonic development. HS-proteoglycans interact with growth factors, morphogens, and extracellular matrix components to regulate development and pathology. Pikachurin and Eyes shut (Eys) interact with dystroglycan to stabilize the photoreceptor axoneme primary cilium and ribbon synapse facilitating phototransduction and neurotransduction with bipolar retinal neuronal networks in ocular vision, the primary human sense. Another HSPG, Neurexin interacts with structural and adaptor proteins to stabilize synapses and ensure specificity of neural interactions, and aids in synaptic potentiation and plasticity in neurotransduction. HSPGs also stabilize the blood-brain barrier and motor neuron basal structures in the neuromuscular junction. Agrin and perlecan localize acetylcholinesterase and its receptors in the neuromuscular junction essential for neuromuscular control. The primary cilium is a mechanosensory hub on neurons, utilized by YES associated protein (YAP)-transcriptional coactivator with PDZ-binding motif (TAZ) Hippo, Hh, Wnt, transforming growth factor (TGF)-β/bone matrix protein (BMP) receptor tyrosine kinase cell signaling. Members of the glypican HSPG proteoglycan family interact with Smoothened and Patched G-protein coupled receptors on the cilium to regulate Hh and Wnt signaling during neuronal development. Control of glycosyl sulfotransferases and endogenous protease expression by Hippo TAZ YAP represents a mechanism whereby the fine structure of HS-proteoglycans can be potentially modulated spatiotemporally to regulate tissue morphogenesis in a similar manner to how Hippo signaling controls sialyltransferase expression and mediation of cell-cell recognition, dysfunctional sialic acid expression is a feature of many tumors.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Sydney Medical School-Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Wu M, Wang X, Shuai J, Deng L, Lu H, Zhou Y, Wu M. Identification of key miRNAs in unilateral mastication-induced disruption of cartilage homeostasis. Oral Dis 2024; 30:551-561. [PMID: 36648372 DOI: 10.1111/odi.14504] [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: 01/26/2022] [Revised: 11/12/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023]
Abstract
OBJECTIVE The present study identified potentially pivotal miRNAs contributing to chondrogenic differentiation in temporomandibular joint suffering abnormal stress. MATERIALS AND METHODS Sprague-Dawley rats were randomly divided into control and experimental unilateral mastication (EUM) group. Bone micro-structure parameters was detected by micro-CT, and FGF-1 and MMP-1 expression was examined by immunohistochemistry. Differentially expressed miRNAs of bilateral condyle cartilage were screened via miRNA microarray at 4- and 8-week EUM, then further verified using quantitative reverse-transcription PCR. Over-expression of five differentially expressed miRNAs in chondrocytes was triggered by transfecting miRNA mimics. The expression of MMP-13, Col-II, OPN, and Runx2 was verified by western blotting. RESULTS Expressions of FGF-1 and MMP-1 in right condyles gradually increased from 2 to 6 weeks after EUM. A total of 20 differentially expressed miRNAs were regulated by EUM, which related to cell proliferation, invasion, and osteoblast differentiation pathways. The over-expression of miR-148a-3p and miR-1-3p led to down-regulation of Col-II, while MMP-13 and Runx2 were up-regulated by induction of hypotrophic differentiation or IL-1β stimulation. These findings suggested that miR-148a-3p and miR-1-3p promote chondrogenic differentiation. CONCLUSIONS Several pivotal miRNAs were found to be related to chondrogenic differentiation, which provides novel insight into pathogenic mechanisms of cartilage homeostasis.
Collapse
Affiliation(s)
- Mengjie Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xuebin Wang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Jing Shuai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Liquan Deng
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haiping Lu
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiqun Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Mengrui Wu
- College of Life Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Li X, Zhao W, Zhou D, Li P, Zhao C, Zhou Q, Wang Y. Construction of Integral Decellularized Cartilage Using a Novel Hydrostatic Pressure Bioreactor. Tissue Eng Part C Methods 2024; 30:113-129. [PMID: 38183634 DOI: 10.1089/ten.tec.2023.0265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2024] Open
Abstract
The decellularized extracellular matrix (ECM) of cartilage is a widely used natural bioscaffold for constructing tissue-engineered cartilage due to its good biocompatibility and regeneration properties. However, current decellularization methods for accessing decellularized cartilaginous tissues require multiple steps and a relatively long duration to produce decellularized cartilage. In addition, most decellularization strategies lead to damage of the microstructure and loss of functional components of the cartilaginous matrix. In this study, a novel decellularization strategy based on a hydrostatic pressure (HP) bioreactor was introduced, which aimed to improve the efficiency of producing integral decellularized cartilage pieces by combining physical and chemical decellularization methods in a perfusing manner. Two types of cartilaginous tissues, auricular cartilage (AC) and nucleus pulposus (NP) fibrocartilage, were selected for comparison of the effects of ordinary, positive, and negative HP-based decellularization according to the cell clearance ratio, microstructural changes, ECM components, and mechanical properties. The results indicated that applying positive HP improved the efficiency of producing decellularized AC, but no significant differences in decellularization efficiency were found between the ordinary and negative HP-treated groups. However, compared with the ordinary HP treatment, the application of the positive or negative HP did not affect the efficiency of decellularized NP productions. Moreover, neither positive nor negative HP influenced the preservation of the microstructure and components of the AC matrix. However, applying negative HP disarranged the fibril distribution of the NP matrix and reduced glycosaminoglycans and collagen type II contents, two essential ECM components. In addition, the positive HP was beneficial for maintaining the mechanical properties of decellularized cartilage. The recellularization experiments also verified the good biocompatibility of the decellularized cartilage produced by the present bioreactor-based decellularization method under positive HP. Overall, applying positive HP-based decellularization resulted in a superior effect on the production of close-to-natural scaffolds for cartilage tissue engineering. Impact statement In this study, we successfully constructed a novel hydrostatic pressure (HP) bioreactor and used this equipment to produce decellularized cartilage by combining physical and chemical decellularization methods in a perfusing manner. We found that positive HP-based decellularization could improve the production efficiency of integral decellularized cartilage pieces and promote the maintenance of matrix components and mechanical properties. This new decellularization strategy exhibited a superior effect in the production of close-to-natural scaffolds and positively impacts cartilage tissue engineering.
Collapse
Affiliation(s)
- Xiaoxiao Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weikang Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dandan Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, Jiulongpo People's Hospital of Chongqing, Chongqing, China
| | - Pei Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chen Zhao
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang Zhou
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyang Wang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Kim B, Kelly TAN, Jung HJ, Beane OS, Bhumiratana S, Bouklas N, Cohen I, Bonassar LJ. Microscale strain concentrations in tissue-engineered osteochondral implants are dictated by local compositional thresholds and architecture. J Biomech 2024; 162:111882. [PMID: 38070296 DOI: 10.1016/j.jbiomech.2023.111882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/16/2023] [Accepted: 11/20/2023] [Indexed: 01/16/2024]
Abstract
Tissue-engineered osteochondral implants manufactured from condensed mesenchymal stem cell bodies have shown promise for treating focal cartilage defects. Notably, such manufacturing techniques have shown to successfully recapture the bulk mechanical properties of native cartilage. However, the relationships among the architectural features, local composition, and micromechanical environment within tissue-engineered cartilage from cell-based aggregates remain unclear. Understanding such relationships is crucial for identifying critical parameters that can predict in vivo performance. Therefore, this study investigated the relationship among architectural features, composition, and micromechanical behavior of tissue-engineered osteochondral implants. We utilized fast-confocal microscopy combined with a strain mapping technique to analyze the micromechanical behavior under quasi-static loading, as well as Fourier Transform Infrared Spectroscopy to analyze the local compositions. More specifically, we investigated the architectural features and compositional distributions generated from tissue maturation, along with macro- and micro-level strain distributions. Our results showed that under compression, cell-based aggregates underwent deformation followed by body movement, generating high local strain around the boundaries, where local aggrecan concentration was low and local collagen concentration was high. By analyzing the micromechanics and composition at the single aggregate length scale, we identified a strong threshold relationship between local strain and compositions. Namely at the aggrecan concentration below 0.015 arbitrary unit (A.U.) and the collagen concentration above 0.15 A.U., the constructs experienced greater than threefold increase in compressive strain. Overall, this study suggests that local compositional features are the primary driver of the local mechanical environment in tissue-engineered cartilage constructs, providing insight into potential quality control parameters for manufacturing tissue-engineered constructs.
Collapse
Affiliation(s)
- Byumsu Kim
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | | | | | | | | | - Nikolaos Bouklas
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Itai Cohen
- Department of Physics, Cornell University, Ithaca, NY, United States
| | - Lawrence J Bonassar
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
17
|
Wang XH, Liu N, Zhang H, Yin ZS, Zha ZG. From cells to organs: progress and potential in cartilaginous organoids research. J Transl Med 2023; 21:926. [PMID: 38129833 PMCID: PMC10740223 DOI: 10.1186/s12967-023-04591-9] [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: 08/10/2023] [Accepted: 10/04/2023] [Indexed: 12/23/2023] Open
Abstract
While cartilage tissue engineering has significantly improved the speed and quality of cartilage regeneration, the underlying metabolic mechanisms are complex, making research in this area lengthy and challenging. In the past decade, organoids have evolved rapidly as valuable research tools. Methods to create these advanced human cell models range from simple tissue culture techniques to complex bioengineering approaches. Cartilaginous organoids in part mimic the microphysiology of human cartilage and fill a gap in high-fidelity cartilage disease models to a certain extent. They hold great promise to elucidate the pathogenic mechanism of a diversity of cartilage diseases and prove crucial in the development of new drugs. This review will focus on the research progress of cartilaginous organoids and propose strategies for cartilaginous organoid construction, study directions, and future perspectives.
Collapse
Affiliation(s)
- Xiao-He Wang
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Ning Liu
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Hui Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Zong-Sheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Zhen-Gang Zha
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
18
|
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
|
19
|
Duan M, Xia S, Liu Y, Pu X, Chen Y, Zhou Y, Huang M, Pi C, Zhang D, Xie J. Stiffened fibre-like microenvironment based on patterned equidistant micropillars directs chondrocyte hypertrophy. Mater Today Bio 2023; 20:100682. [PMID: 37304578 PMCID: PMC10251154 DOI: 10.1016/j.mtbio.2023.100682] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/04/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Articular cartilage, composed of collagen type II as a major extracellular matrix and chondrocyte as a unique cell type, is a specialized connective tissue without blood vessels, lymphatic vessels and nerves. This distinctive characteristic of articular cartilage determines its very limited ability to repair when damaged. It is well known that physical microenvironmental signals regulate many cell behaviors such as cell morphology, adhesion, proliferation and cell communication even determine chondrocyte fate. Interestingly, with increasing age or progression of joint diseases such as osteoarthritis (OA), the major collagen fibrils in the extracellular matrix of articular cartilage become larger in diameter, leading to stiffening of articular tissue and reducing its resistance to external tension, which in turn aggravates joint damage or progression of joint diseases. Therefore, designing a physical microenvironment closer to the real tissue and thus obtaining data closer to the real cellular behaviour, and then revealing the biological mechanisms of chondrocytes in pathological states is of crucial importance for the treatment of OA disease. Here we fabricated micropillar substrates with the same topology but different stiffnesses to mimic the matrix stiffening that occurs in the transition from normal to diseased cartilage. It was first found that chondrocytes responded to stiffened micropillar substrates by showing a larger cell spreading area, a stronger enhancement of cytoskeleton rearrangement and more stability of focal adhesion plaques. The activation of Erk/MAPK signalling in chondrocytes was detected in response to the stiffened micropillar substrate. Interestingly, a larger nuclear spreading area of chondrocytes at the interface layer between the cells and top surfaces of micropillars was observed in response to the stiffened micropillar substrate. Finally, it was found that the stiffened micropillar substrate promoted chondrocyte hypertrophy. Taken together, these results revealed the cell responses of chondrocytes in terms of cell morphology, cytoskeleton, focal adhesion, nuclei and cell hypertrophy, and may be beneficial for understanding the cellular functional changes affected by the matrix stiffening that occurs during the transition from a normal state to a state of osteoarthritis.
Collapse
Affiliation(s)
- Mengmeng Duan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shuang Xia
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu, 610065, China
| | - Yang Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaohua Pu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yukun Chen
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu, 610065, China
| | - Yilin Zhou
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu, 610065, China
| | - Minglei Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
20
|
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
|
21
|
Segarra-Queralt M, Piella G, Noailly J. Network-based modelling of mechano-inflammatory chondrocyte regulation in early osteoarthritis. Front Bioeng Biotechnol 2023; 11:1006066. [PMID: 36815875 PMCID: PMC9936426 DOI: 10.3389/fbioe.2023.1006066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a debilitating joint disease characterized by articular cartilage degradation, inflammation and pain. An extensive range of in vivo and in vitro studies evidences that mechanical loads induce changes in chondrocyte gene expression, through a process known as mechanotransduction. It involves cascades of complex molecular interactions that convert physical signals into cellular response(s) that favor either chondroprotection or cartilage destruction. Systematic representations of those interactions can positively inform early strategies for OA management, and dynamic modelling allows semi-quantitative representations of the steady states of complex biological system according to imposed initial conditions. Yet, mechanotransduction is rarely integrated. Hence, a novel mechano-sensitive network-based model is proposed, in the form of a continuous dynamical system: an interactome of a set of 118 nodes, i.e., mechano-sensitive cellular receptors, second messengers, transcription factors and proteins, related among each other through a specific topology of 358 directed edges is developed. Results show that under physio-osmotic initial conditions, an anabolic state is reached, whereas initial perturbations caused by pro-inflammatory and injurious mechanical loads leads to a catabolic profile of node expression. More specifically, healthy chondrocyte markers (Sox9 and CITED2) are fully expressed under physio-osmotic conditions, and reduced under inflammation, or injurious loadings. In contrast, NF-κB and Runx2, characteristic of an osteoarthritic chondrocyte, become activated under inflammation or excessive loading regimes. A literature-based evaluation shows that the model can replicate 94% of the experiments tested. Sensitivity analysis based on a factorial design of a treatment shows that inflammation has the strongest influence on chondrocyte metabolism, along with a significant deleterious effect of static compressive loads. At the same time, anti-inflammatory therapies appear as the most promising ones, though the restoration of structural protein production seems to remain a major challenge even in beneficial mechanical environments. The newly developed mechano-sensitive network model for chondrocyte activity reveals a unique potential to reflect load-induced chondroprotection or articular cartilage degradation in different mechano-chemical-environments.
Collapse
|
22
|
Sun D, Liu X, Xu L, Meng Y, Kang H, Li Z. Advances in the Treatment of Partial-Thickness Cartilage Defect. Int J Nanomedicine 2022; 17:6275-6287. [PMID: 36536940 PMCID: PMC9758915 DOI: 10.2147/ijn.s382737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/23/2022] [Indexed: 04/17/2024] Open
Abstract
Partial-thickness cartilage defects (PTCDs) of the articular surface is the most common problem in cartilage degeneration, and also one of the main pathogenesis of osteoarthritis (OA). Due to the lack of a clear diagnosis, the symptoms are often more severe when full-thickness cartilage defect (FTCDs) is present. In contrast to FTCDs and osteochondral defects (OCDs), PTCDs does not injure the subchondral bone, there is no blood supply and bone marrow exudation, and the nearby microenvironment is unsuitable for stem cells adhesion, which completely loses the ability of self-repair. Some clinical studies have shown that partial-thickness cartilage defects is as harmful as full-thickness cartilage defects. Due to the poor effect of conservative treatment, the destructive surgical treatment is not suitable for the treatment of partial-thickness cartilage defects, and the current tissue engineering strategies are not effective, so it is urgent to develop novel strategies or treatment methods to repair PTCDs. In recent years, with the interdisciplinary development of bioscience, mechanics, material science and engineering, many discoveries have been made in the repair of PTCDs. This article reviews the current status and research progress in the treatment of PTCDs from the aspects of diagnosis and modeling of PTCDs, drug therapy, tissue transplantation repair technology and tissue engineering ("bottom-up").
Collapse
Affiliation(s)
- Daming Sun
- Wuhan Sports University, Wuhan, People’s Republic of China
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Xiangzhong Liu
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Liangliang Xu
- Wuhan Sports University, Wuhan, People’s Republic of China
| | - Yi Meng
- Wuhan Sports University, Wuhan, People’s Republic of China
| | - Haifei Kang
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, People’s Republic of China
| | - Zhanghua Li
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
23
|
Lineham B, Altaie A, Harwood P, McGonagle D, Pandit H, Jones E. A systematic review on the potential value of synovial fluid biomarkers to predict clinical outcomes in cartilage repair treatments. Osteoarthritis Cartilage 2022; 30:1035-1049. [PMID: 35618204 DOI: 10.1016/j.joca.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/03/2022] [Accepted: 05/17/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Multiple biochemical biomarkers have been previously investigated for the diagnosis, prognosis and response to treatment of articular cartilage damage, including osteoarthritis (OA). Synovial fluid (SF) biomarker measurement is a potential method to predict treatment response and effectiveness. However, the significance of different biomarkers and their correlation to clinical outcomes remains unclear. This systematic review evaluated current SF biomarkers used in investigation of cartilage degeneration or regeneration in the knee joint and correlated these biomarkers with clinical outcomes following cartilage repair or regeneration interventions. METHOD PubMed, Institute of Science Index, Scopus, Cochrane Central Register of Controlled Trials, and Embase databases were searched. Studies evaluating SF biomarkers and clinical outcomes following cartilage repair intervention were included. Two researchers independently performed data extraction and Quality Assessment of Diagnostic Accuracy Score 2 (QUADAS-2) analysis. Biomarker inclusion, change following intervention and correlation with clinical outcome was compared. RESULTS 9 studies were included. Study heterogeneity precluded meta-analysis. There was significant variation in sampling and analysis. 33 biomarkers were evaluated in addition to microRNA and catabolic/anabolic ratios. Five studies reported on correlation of biomarkers with six biomarkers significantly correlated with clinical outcomes following intervention. However, correlation was only demonstrated in isolated studies. CONCLUSION This review demonstrates significant difficulties in drawing conclusions regarding the importance of SF biomarkers based on the available literature. Improved standardisation for collection and analysis of SF samples is required. Future publications should also focus on clinical outcome scores and seek to correlate biomarkers with progression to further understand the significance of identified markers in a clinical context. REGISTRATION NUMBER PROSPERO CRD42022304298. Study protocol available on PROSPERO website.
Collapse
Affiliation(s)
- B Lineham
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK.
| | - A Altaie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - P Harwood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK; Trauma and Orthopaedics Department, Leeds Teaching Hospitals NHS Trust, UK
| | - D McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - H Pandit
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK; Trauma and Orthopaedics Department, Leeds Teaching Hospitals NHS Trust, UK
| | - E Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| |
Collapse
|
24
|
Fabrication of Tissue-Engineered Cartilage Using Decellularized Scaffolds and Chondrocytes. Polymers (Basel) 2022; 14:polym14142848. [PMID: 35890624 PMCID: PMC9316171 DOI: 10.3390/polym14142848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/25/2022] [Accepted: 07/04/2022] [Indexed: 02/07/2023] Open
Abstract
In this paper, we aim to explore the application value of tissue engineering for the construction of artificial cartilage in vitro. Chondrocytes from healthy porcine articular cartilage tissue were seeded on articular cartilage extracellular matrix (ACECM) scaffolds and cultivated. Type II collagen immunofluorescent staining was used to assess secretion from the extracellular matrix. Chondrocytes, which were mainly polygonal and cobblestone-shaped, were inoculated on ACECM-oriented scaffolding for 7 days, and the neo-tissue showed translucent shape and toughness. Using inverted and fluorescence microscopy, we found that chondrocytes on the scaffolds performed well in terms of adhesion and growth, and they secreted collagen type II. Moreover, the porcine ACECM scaffolds had good biocompatibility. The inflammatory cell detection, cellular immune response assay and humoral immune response assay showed porcine ACECM scaffolds were used for xenotransplantation without significant immune inflammatory response. All these findings reveal that ACECM-oriented scaffold is an ideal natural biomaterial for cartilage tissue engineering.
Collapse
|
25
|
Rothbauer M, Reihs EI, Fischer A, Windhager R, Jenner F, Toegel S. A Progress Report and Roadmap for Microphysiological Systems and Organ-On-A-Chip Technologies to Be More Predictive Models in Human (Knee) Osteoarthritis. Front Bioeng Biotechnol 2022; 10:886360. [PMID: 35782494 PMCID: PMC9240813 DOI: 10.3389/fbioe.2022.886360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Osteoarthritis (OA), a chronic debilitating joint disease affecting hundreds of million people globally, is associated with significant pain and socioeconomic costs. Current treatment modalities are palliative and unable to stop the progressive degeneration of articular cartilage in OA. Scientific attention has shifted from the historical view of OA as a wear-and-tear cartilage disorder to its recognition as a whole-joint disease, highlighting the contribution of other knee joint tissues in OA pathogenesis. Despite much progress in the field of microfluidic systems/organs-on-a-chip in other research fields, current in vitro models in use do not yet accurately reflect the complexity of the OA pathophenotype. In this review, we provide: 1) a detailed overview of the most significant recent developments in the field of microsystems approaches for OA modeling, and 2) an OA-pathophysiology-based bioengineering roadmap for the requirements of the next generation of more predictive and authentic microscale systems fit for the purpose of not only disease modeling but also of drug screening to potentially allow OA animal model reduction and replacement in the near future.
Collapse
Affiliation(s)
- Mario Rothbauer
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Faculty of Technical Chemistry, Vienna University of Technology, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Eva I. Reihs
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Faculty of Technical Chemistry, Vienna University of Technology, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Anita Fischer
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Reinhard Windhager
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Florien Jenner
- Veterinary Tissue Engineering and Regenerative Medicine Vienna (VETERM), Equine Surgery Unit, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Stefan Toegel
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| |
Collapse
|