1
|
van Mourik M, Abinzano F, Ito K. The Regulation of Pericellular Matrix Synthesis During Articular Cartilage Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40402857 DOI: 10.1089/ten.teb.2024.0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Articular cartilage, vital to the health and functioning of joints, remains challenging to regenerate. The pericellular matrix (PCM) is critical for transducing biophysical stimuli to the articular chondrocytes (ACs) that it envelops. Given the mechanobiological sensitivity of ACs, it is pivotal in maintaining the chondrogenic phenotype and the production of extracellular matrix (ECM) during articular cartilage tissue engineering. While the maintenance of the native PCM significantly improves the quality of neocartilage, current isolation methods are limited. A solution to this challenge is facilitating ACs to regenerate their PCM. However, the regulation of PCM synthesis remains poorly understood, hindering the development of effective tissue engineering strategies. This narrative review aims to provide a comprehensive analysis of the complex interplay between extracellular cues and intracellular pathways regulating PCM synthesis during articular cartilage tissue engineering. Our analysis reveals that mechanical cues, such as material stiffness and mechanical stimulation, are the primary regulators of PCM synthesis. Additionally, the use of scaffold-free techniques potentially affects the structuring of newly created PCM. Tuning these stimuli can significantly impact the quality of the formed PCM, ultimately influencing neocartilage quality. Furthermore, we highlight intracellular mechanisms involved in the transduction of these extracellular cues, including actin polymerization, yes-associated protein and transcriptional coactivator with PDZ-binding motif localization, and transforming growth factor beta-induced Smad signaling. Although the current literature suggests the involvement of these signaling pathways in regulating the synthesis of PCM components, we found that studies investigating these processes in ACs are lacking. Elucidating the relationships between extracellular stimuli, intracellular signaling, and the expression of PCM components could provide a framework for designing new cartilage tissue engineering approaches that facilitate proper PCM synthesis. Ultimately, this can improve ECM quality and advance articular cartilage regeneration.
Collapse
Affiliation(s)
- Marloes van Mourik
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Florencia Abinzano
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
2
|
Scholpp S, Hoffmann L, Schätzlein E, Gries T, Emonts C, Blaeser A. Interlacing biology and engineering: An introduction to textiles and their application in tissue engineering. Mater Today Bio 2025; 31:101617. [PMID: 40124339 PMCID: PMC11926717 DOI: 10.1016/j.mtbio.2025.101617] [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: 12/19/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Tissue engineering (TE) aims to provide personalized solutions for tissue loss caused by trauma, tumors, or congenital defects. While traditional methods like autologous and homologous tissue transplants face challenges such as donor shortages and risk of donor site morbidity, TE provides a viable alternative using scaffolds, cells, and biologically active molecules. Textiles represent a promising scaffold option for both in-vitro and in-situ TE applications. Textile engineering is a broad field and can be divided into fiber-based textiles and yarn-based textiles. In fiber-based textiles the textile fabric is produced in the same step as the fibers (e.g. non-wovens, electrospun mats and 3D-printed). For yarn-based textiles, yarns are produced from fibers or filaments first and then, a textile fabric is produced (e.g. woven, weft-knitted, warp-knitted and braided fabrics). The selection of textile scaffold technology depends on the target tissue, mechanical requirements, and fabrication methods, with each approach offering distinct advantages. Braided scaffolds, with their high tensile strength, are ideal for load-bearing tissues like tendons and ligaments, while their ability to form stable hollow lumens makes them suitable for vascular applications. Weaving, weft-, and warp-knitting provide tunable structural properties, with warp-knitting offering the greatest design flexibility. Spacer fabrics enable complex 3D architecture, benefiting applications such as skin grafts and multilayered tissues. Electrospinning, though highly effective in mimicking the ECM, is structurally limited. The complex interactions between materials, fiber properties, and textile technologies allows for scaffolds with a wide range of morphological and mechanical characteristics (e.g., tensile strength of woven textiles ranging from 0.64 to 180.4 N/mm2). With in-depth knowledge, textiles can be tailored to obtain specific mechanical properties as accurately as possible and aid the formation of functional tissue. However, as textile structures inherently differ from biological tissues, careful optimization is required to enhance cell behavior, mechanical performance, and clinical applicability. This review is intended for TE experts interested in using textiles as scaffolds and provides a detailed analysis of the available options, their characteristics and known applications. For this, first the major fiber formation methods are introduced, then subsequent used automated textile technologies are presented, highlighting their strengths and limitations. Finally, we analyze how these textile and fiber structures are utilized in TE, organized by the use of textiles in TE across major organ systems, including the nervous, skin, cardiovascular, respiratory, urinary, digestive, and musculoskeletal systems.
Collapse
Affiliation(s)
- S. Scholpp
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany
| | - L.A. Hoffmann
- Institut für Textiltechnik, RWTH Aachen University, Aachen, Germany
| | - E. Schätzlein
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany
| | - T. Gries
- Institut für Textiltechnik, RWTH Aachen University, Aachen, Germany
| | - C. Emonts
- Institut für Textiltechnik, RWTH Aachen University, Aachen, Germany
| | - A. Blaeser
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
3
|
Semitela A, Marques PAAP, Completo A. Strategies to engineer articular cartilage with biomimetic zonal features: a review. Biomater Sci 2024; 12:5961-6005. [PMID: 39463257 DOI: 10.1039/d4bm00579a] [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: 10/29/2024]
Abstract
Articular cartilage (AC) is a highly specialized tissue with restricted ability for self-regeneration, given its avascular and acellular nature. Although a considerable number of surgical treatments is available for the repair, reconstruction, and regeneration of AC defects, most of them do not prioritize the development of engineered cartilage with zonal stratification derived from biomimetic biochemical, biomechanical and topographic cues. In the absence of these zonal elements, engineered cartilage will exhibit increased susceptibility to failure and will neither be able to withstand the mechanical loading to which AC is subjected nor will it integrate well with the surrounding tissue. In this regard, new breakthroughs in the development of hierarchical stratified engineered cartilage are highly sought after. Initially, this review provides a comprehensive analysis of the composition and zonal organization of AC, aiming to enhance our understanding of the significance of the structure of AC for its function. Next, we direct our attention towards the existing in vitro and in vivo studies that introduce zonal elements in engineered cartilage to elicit appropriate AC regeneration by employing tissue engineering strategies. Finally, the advantages, challenges, and future perspectives of these approaches are presented.
Collapse
Affiliation(s)
- Angela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
4
|
Liu Y, Chen P, Hu B, Xiao Y, Su T, Luo X, Tu M, Cai G. Excessive mechanical loading promotes osteoarthritis development by upregulating Rcn2. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167251. [PMID: 38795835 DOI: 10.1016/j.bbadis.2024.167251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/28/2024]
Abstract
Exposure of articular cartilage to excessive mechanical loading is closely related to the pathogenesis of osteoarthritis (OA). However, the exact molecular mechanism by which excessive mechanical loading drives OA remains unclear. In vitro, primary chondrocytes were exposed to cyclic tensile strain at 0.5 Hz and 10 % elongation for 30 min to simulate excessive mechanical loading in OA. In vivo experiments involved mice undergoing anterior cruciate ligament transection (ACLT) to model OA, followed by interventions on Rcn2 expression through adeno-associated virus (AAV) injection and tamoxifen-induced gene deletion. 10 μL AAV2/5 containing AAV-Rcn2 or AAV-shRcn2 was administered to the mice by articular injection at 1 week post ACLT surgery, and Col2a1-creERT: Rcn2flox/flox mice were injected with tamoxifen intraperitoneally to obtain Rcn2-conditional knockout mice. Finally, we explored the mechanism of Rcn2 affecting OA. Here, we identified reticulocalbin-2 (Rcn2) as a mechanosensitive factor in chondrocytes, which was significantly elevated in chondrocytes under mechanical overloading. PIEZO type mechanosensitive ion channel component 1 (Piezo1) is a critical mechanosensitive ion channel, which mediates the effect of mechanical loading on chondrocytes, and we found that increased Rcn2 could be suppressed through knocking down Piezo1 under excessive mechanical loading. Furthermore, chondrocyte-specific deletion of Rcn2 in adult mice alleviated OA progression in the mice receiving the surgery of ACLT. On the contrary, articular injection of Rcn2-expressing adeno-associated virus (AAV) accelerated the progression of ACLT-induced OA in mice. Mechanistically, Rcn2 accelerated the progression of OA through promoting the phosphorylation and nuclear translocation of signal transducer and activator of transcription 3 (Stat3).
Collapse
Affiliation(s)
- Yalin Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Peng Chen
- Department of Orthopedic, Xiangya Hospital of Central South University, Changsha, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Manli Tu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, China; Jiangxi Branch of National Clinical Research Center for metabolic Disease, China.
| | - Guangping Cai
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
5
|
Bordbar S, Li Z, Lotfibakhshaiesh N, Ai J, Tavassoli A, Beheshtizadeh N, Vainieri L, Khanmohammadi M, Sayahpour FA, Baghaban Eslaminejad M, Azami M, Grad S, Alini M. Cartilage tissue engineering using decellularized biomatrix hydrogel containing TGF-β-loaded alginate microspheres in mechanically loaded bioreactor. Sci Rep 2024; 14:11991. [PMID: 38796487 PMCID: PMC11127927 DOI: 10.1038/s41598-024-62474-5] [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: 07/29/2023] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Physiochemical tissue inducers and mechanical stimulation are both efficient variables in cartilage tissue fabrication and regeneration. In the presence of biomolecules, decellularized extracellular matrix (ECM) may trigger and enhance stem cell proliferation and differentiation. Here, we investigated the controlled release of transforming growth factor beta (TGF-β1) as an active mediator of mesenchymal stromal cells (MSCs) in a biocompatible scaffold and mechanical stimulation for cartilage tissue engineering. ECM-derived hydrogel with TGF-β1-loaded alginate-based microspheres (MSs) was created to promote human MSC chondrogenic development. Ex vivo explants and a complicated multiaxial loading bioreactor replicated the physiological conditions. Hydrogels with/without MSs and TGF-β1 were highly cytocompatible. MSCs in ECM-derived hydrogel containing TGF-β1/MSs showed comparable chondrogenic gene expression levels as those hydrogels with TGF-β1 added in culture media or those without TGF-β1. However, constructs with TGF-β1 directly added within the hydrogel had inferior properties under unloaded conditions. The ECM-derived hydrogel group including TGF-β1/MSs under loading circumstances formed better cartilage matrix in an ex vivo osteochondral defect than control settings. This study demonstrates that controlled local delivery of TGF-β1 using MSs and mechanical loading is essential for neocartilage formation by MSCs and that further optimization is needed to prevent MSC differentiation towards hypertrophy.
Collapse
Affiliation(s)
- Sima Bordbar
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- AO Research Institute Davos, Davos, Switzerland
| | - Zhen Li
- AO Research Institute Davos, Davos, Switzerland
| | - Nasrin Lotfibakhshaiesh
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Jafar Ai
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Tavassoli
- Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Mehdi Khanmohammadi
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Wołoska 141, 02-507, Warsaw, Poland
| | | | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mahmoud Azami
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland.
| |
Collapse
|
6
|
Jia Y, Le H, Wang X, Zhang J, Liu Y, Ding J, Zheng C, Chang F. Double-edged role of mechanical stimuli and underlying mechanisms in cartilage tissue engineering. Front Bioeng Biotechnol 2023; 11:1271762. [PMID: 38053849 PMCID: PMC10694366 DOI: 10.3389/fbioe.2023.1271762] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/11/2023] [Indexed: 12/07/2023] Open
Abstract
Mechanical stimuli regulate the chondrogenic differentiation of mesenchymal stem cells and the homeostasis of chondrocytes, thus affecting implant success in cartilage tissue engineering. The mechanical microenvironment plays fundamental roles in the maturation and maintenance of natural articular cartilage, and the progression of osteoarthritis Hence, cartilage tissue engineering attempts to mimic this environment in vivo to obtain implants that enable a superior regeneration process. However, the specific type of mechanical loading, its optimal regime, and the underlying molecular mechanisms are still under investigation. First, this review delineates the composition and structure of articular cartilage, indicating that the morphology of chondrocytes and components of the extracellular matrix differ from each other to resist forces in three top-to-bottom overlapping zones. Moreover, results from research experiments and clinical trials focusing on the effect of compression, fluid shear stress, hydrostatic pressure, and osmotic pressure are presented and critically evaluated. As a key direction, the latest advances in mechanisms involved in the transduction of external mechanical signals into biological signals are discussed. These mechanical signals are sensed by receptors in the cell membrane, such as primary cilia, integrins, and ion channels, which next activate downstream pathways. Finally, biomaterials with various modifications to mimic the mechanical properties of natural cartilage and the self-designed bioreactors for experiment in vitro are outlined. An improved understanding of biomechanically driven cartilage tissue engineering and the underlying mechanisms is expected to lead to efficient articular cartilage repair for cartilage degeneration and disease.
Collapse
Affiliation(s)
- Yao Jia
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Hanxiang Le
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
- The Fourth Treatment Area of Trauma Hip Joint Surgery Department, Tianjin Hospital, Tianjin, China
| | - Xianggang Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Jiaxin Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Yan Liu
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Jiacheng Ding
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Changjun Zheng
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Fei Chang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| |
Collapse
|
7
|
Paul S, Schrobback K, Tran PA, Meinert C, Davern JW, Weekes A, Nedunchezhiyan U, Klein TJ. GelMA-glycol chitosan hydrogels for cartilage regeneration: The role of uniaxial mechanical stimulation in enhancing mechanical, adhesive, and biochemical properties. APL Bioeng 2023; 7:036114. [PMID: 37692373 PMCID: PMC10492648 DOI: 10.1063/5.0160472] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Untreated osteochondral defects are a leading cause of osteoarthritis, a condition that places a heavy burden on both patients and orthopedic surgeons. Although tissue engineering has shown promise for creating mechanically similar cartilage-like constructs, their integration with cartilage remains elusive. Therefore, a formulation of biodegradable, biocompatible biomaterial with sufficient mechanical and adhesive properties for cartilage repair is required. To accomplish this, we prepared biocompatible, photo-curable, mechanically robust, and highly adhesive GelMA-glycol chitosan (GelMA-GC) hydrogels. GelMA-GC hydrogels had a modulus of 283 kPa and provided a biocompatible environment (>70% viability of embedded chondrocytes) in long-term culture within a bovine cartilage ring. The adhesive strength of bovine chondrocyte-laden GelMA-GC hydrogel to bovine cartilage increased from 38 to 52 kPa over four weeks of culture. Moreover, intermittent uniaxial mechanical stimulation enhanced the adhesive strength to ∼60 kPa, indicating that the cartilage-hydrogel integration could remain secure and functional under dynamic loading conditions. Furthermore, gene expression data and immunofluorescence staining revealed the capacity of chondrocytes in GelMA-GC hydrogel to synthesize chondrogenic markers (COL2A1 and ACAN), suggesting the potential for tissue regeneration. The promising in vitro results of this work motivate further exploration of the potential of photo-curable GelMA-GC bioadhesive hydrogels for cartilage repair and regeneration.
Collapse
Affiliation(s)
| | - Karsten Schrobback
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | | | | | | | | | | | | |
Collapse
|
8
|
Alizadeh Sardroud H, Chen X, Eames BF. Reinforcement of Hydrogels with a 3D-Printed Polycaprolactone (PCL) Structure Enhances Cell Numbers and Cartilage ECM Production under Compression. J Funct Biomater 2023; 14:313. [PMID: 37367278 DOI: 10.3390/jfb14060313] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/30/2023] [Accepted: 06/04/2023] [Indexed: 06/28/2023] Open
Abstract
Hydrogels show promise in cartilage tissue engineering (CTE) by supporting chondrocytes and maintaining their phenotype and extracellular matrix (ECM) production. Under prolonged mechanical forces, however, hydrogels can be structurally unstable, leading to cell and ECM loss. Furthermore, long periods of mechanical loading might alter the production of cartilage ECM molecules, including glycosaminoglycans (GAGs) and collagen type 2 (Col2), specifically with the negative effect of stimulating fibrocartilage, typified by collagen type 1 (Col1) secretion. Reinforcing hydrogels with 3D-printed Polycaprolactone (PCL) structures offer a solution to enhance the structural integrity and mechanical response of impregnated chondrocytes. This study aimed to assess the impact of compression duration and PCL reinforcement on the performance of chondrocytes impregnated with hydrogel. Results showed that shorter loading periods did not significantly affect cell numbers and ECM production in 3D-bioprinted hydrogels, but longer periods tended to reduce cell numbers and ECM compared to unloaded conditions. PCL reinforcement enhanced cell numbers under mechanical compression compared to unreinforced hydrogels. However, the reinforced constructs seemed to produce more fibrocartilage-like, Col1-positive ECM. These findings suggest that reinforced hydrogel constructs hold potential for in vivo cartilage regeneration and defect treatment by retaining higher cell numbers and ECM content. To further enhance hyaline cartilage ECM formation, future studies should focus on adjusting the mechanical properties of reinforced constructs and exploring mechanotransduction pathways.
Collapse
Affiliation(s)
- Hamed Alizadeh Sardroud
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - B Frank Eames
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
9
|
Alizadeh Sardroud H, Chen X, Eames BF. Applied Compressive Strain Governs Hyaline-like Cartilage versus Fibrocartilage-like ECM Produced within Hydrogel Constructs. Int J Mol Sci 2023; 24:ijms24087410. [PMID: 37108575 PMCID: PMC10138702 DOI: 10.3390/ijms24087410] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
The goal of cartilage tissue engineering (CTE) is to regenerate new hyaline cartilage in joints and treat osteoarthritis (OA) using cell-impregnated hydrogel constructs. However, the production of an extracellular matrix (ECM) made of fibrocartilage is a potential outcome within hydrogel constructs when in vivo. Unfortunately, this fibrocartilage ECM has inferior biological and mechanical properties when compared to native hyaline cartilage. It was hypothesized that compressive forces stimulate fibrocartilage development by increasing production of collagen type 1 (Col1), an ECM protein found in fibrocartilage. To test the hypothesis, 3-dimensional (3D)-bioprinted hydrogel constructs were fabricated from alginate hydrogel impregnated with ATDC5 cells (a chondrogenic cell line). A bioreactor was used to simulate different in vivo joint movements by varying the magnitude of compressive strains and compare them with a control group that was not loaded. Chondrogenic differentiation of the cells in loaded and unloaded conditions was confirmed by deposition of cartilage specific molecules including glycosaminoglycans (GAGs) and collagen type 2 (Col2). By performing biochemical assays, the production of GAGs and total collagen was also confirmed, and their contents were quantitated in unloaded and loaded conditions. Furthermore, Col1 vs. Col2 depositions were assessed at different compressive strains, and hyaline-like cartilage vs. fibrocartilage-like ECM production was analyzed to investigate how applied compressive strain affects the type of cartilage formed. These assessments showed that fibrocartilage-like ECM production tended to reduce with increasing compressive strain, though its production peaked at a higher compressive strain. According to these results, the magnitude of applied compressive strain governs the production of hyaline-like cartilage vs. fibrocartilage-like ECM and a high compressive strain stimulates fibrocartilage-like ECM formation rather than hyaline cartilage, which needs to be addressed by CTE approaches.
Collapse
Affiliation(s)
- Hamed Alizadeh Sardroud
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - B Frank Eames
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
10
|
Tee CA, Han J, Hui JHP, Lee EH, Yang Z. Perspective in Achieving Stratified Articular Cartilage Repair Using Zonal Chondrocytes. TISSUE ENGINEERING. PART B, REVIEWS 2023. [PMID: 36416231 DOI: 10.1089/ten.teb.2022.0142] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Articular cartilage is composed of superficial, medial, and deep zones, which endow the tissue with biphasic mechanical properties to withstand shearing force and compressional loading. The tissue has very limited self-healing capacity once it is damaged due to its avascular nature. To prevent the early onset of osteoarthritis, surgical intervention is often needed to repair the injured cartilage. Current noncell-based and cell-based treatments focus on the regeneration of homogeneous cartilage to achieve bulk compressional properties without recapitulating the zonal matrix and mechanical properties, and often oversight in aiding cartilage integration between host and repair cartilage. It is hypothesized that achieving zonal architecture in articular cartilage tissue repair could improve the structural and mechanical integrity and thus the life span of the regenerated tissue. Engineering stratified cartilage constructs using zonal chondrocytes have been hypothesized to improve the functionality and life span of the regenerated tissues. However, stratified articular cartilage repair has yet to be realized to date due to the lack of an efficient zonal chondrocyte isolation method and an expansion platform that would allow both cell propagation and phenotype maintenance. Various attempts and challenges in achieving stratified articular cartilage repair in a clinical setting are evaluated. In this review, different perspectives on achieving stratified articular cartilage repair using zonal chondrocytes are described. The effectiveness of different zonal chondrocyte isolation and zonal chondrocyte phenotype maintenance methodologies during expansion are compared, with the focus on recent advancements in zonal chondrocyte isolation and expansion that could present a possible strategy to overcome the limitation of applying zonal chondrocytes to facilitate zonal architecture development in articular cartilage regeneration. Impact Statement The zonal properties of articular cartilage contribute to the biphasic mechanical properties of the tissues. Recapitulation of the zonal architecture in regenerated articular cartilage has been hypothesized to improve the mechanical integrity and life span of the regenerated tissue. This review provides a comprehensive discussion on the current state of research relevant to achieving stratified articular cartilage repair using zonal chondrocytes from different perspectives. This review further elaborates on a zonal chondrocyte production pipeline that can potentially overcome the current clinical challenges and future work needed to realize stratified zonal chondrocyte implantation in a clinical setting.
Collapse
Affiliation(s)
- Ching Ann Tee
- Critical Analytics for Manufacturing Personalised-Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, Singapore, Singapore.,Department of Orthopaedic Surgery, National University of Singapore, Singapore, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing Personalised-Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, Singapore, Singapore.,Department of Electrical Engineering and Computer Science, Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Eng Hin Lee
- Critical Analytics for Manufacturing Personalised-Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, Singapore, Singapore.,Department of Orthopaedic Surgery, National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Zheng Yang
- Critical Analytics for Manufacturing Personalised-Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, Singapore, Singapore.,Department of Orthopaedic Surgery, National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Pendyala M, Woods PS, Brubaker DK, Blaber EA, Schmidt TA, Chan DD. Endogenous production of hyaluronan, PRG4, and cytokines is sensitive to cyclic loading in synoviocytes. PLoS One 2022; 17:e0267921. [PMID: 36576921 PMCID: PMC9797074 DOI: 10.1371/journal.pone.0267921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 12/09/2022] [Indexed: 12/29/2022] Open
Abstract
Synovial fluid is composed of hyaluronan and proteoglycan-4 (PRG4 or lubricin), which work synergistically to maintain joint lubrication. In diseases like osteoarthritis, hyaluronan and PRG4 concentrations can be altered, resulting in lowered synovial fluid viscosity, and pro-inflammatory cytokine concentrations within the synovial fluid increase. Synovial fibroblasts within the synovium are responsible for contributing to synovial fluid and can be targeted to improve endogenous production of hyaluronan and PRG4 and to alter the cytokine profile. We cyclically loaded SW982 synoviocytes to 0%, 5%, 10%, or 20% strain for three hours at 1 Hz. To assess the impact of substrate stiffness, we compared the 0% strain group to cells grown on tissue culture plastic. We measured the expression of hyaluronan turnover genes, hyaluronan localization within the cell layer, hyaluronan concentration, PRG4 concentration, and the cytokine profile within the media. Our results show that the addition of cyclic loading increased HAS3 expression, but not in a magnitude-dependent response. Hyaluronidase expression was impacted by strain magnitude, which is exemplified by the decrease in hyaluronan concentration due to cyclic loading. We also show that PRG4 concentration is increased at 5% strain, while higher strain magnitude decreases overall PRG4 concentration. Finally, 10% and 20% strain show a distinct, more pro-inflammatory cytokine profile when compared to the unloaded group. Multivariate analysis showed distinct separation between certain strain groups in being able to predict strain group, hyaluronan concentration, and PRG4 concentration from gene expression or cytokine concentration data, highlighting the complexity of the system. Overall, this study shows that cyclic loading can be used tool to modulate the endogenous production of hyaluronan, PRG4, and cytokines from synovial fibroblasts.
Collapse
Affiliation(s)
- Meghana Pendyala
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Paige S Woods
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Douglas K Brubaker
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Elizabeth A Blaber
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Blue Marble Space Institute of Science at NASA Ames Research Center, Moffett Field, California, United States of America
| | - Tannin A Schmidt
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Deva D Chan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
12
|
Comellas E, Farkas JE, Kleinberg G, Lloyd K, Mueller T, Duerr TJ, Muñoz JJ, Monaghan JR, Shefelbine SJ. Local mechanical stimuli correlate with tissue growth in axolotl salamander joint morphogenesis. Proc Biol Sci 2022; 289:20220621. [PMID: 35582804 PMCID: PMC9114971 DOI: 10.1098/rspb.2022.0621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/22/2022] [Indexed: 01/04/2023] Open
Abstract
Movement-induced forces are critical to correct joint formation, but it is unclear how cells sense and respond to these mechanical cues. To study the role of mechanical stimuli in the shaping of the joint, we combined experiments on regenerating axolotl (Ambystoma mexicanum) forelimbs with a poroelastic model of bone rudiment growth. Animals either regrew forelimbs normally (control) or were injected with a transient receptor potential vanilloid 4 (TRPV4) agonist during joint morphogenesis. We quantified growth and shape in regrown humeri from whole-mount light sheet fluorescence images of the regenerated limbs. Results revealed significant differences in morphology and cell proliferation between groups, indicating that TRPV4 desensitization has an effect on joint shape. To link TRPV4 desensitization with impaired mechanosensitivity, we developed a finite element model of a regenerating humerus. Local tissue growth was the sum of a biological contribution proportional to chondrocyte density, which was constant, and a mechanical contribution proportional to fluid pressure. Computational predictions of growth agreed with experimental outcomes of joint shape, suggesting that interstitial pressure driven from cyclic mechanical stimuli promotes local tissue growth. Predictive computational models informed by experimental findings allow us to explore potential physical mechanisms involved in tissue growth to advance our understanding of the mechanobiology of joint morphogenesis.
Collapse
Affiliation(s)
- Ester Comellas
- Serra Húnter Fellow, Department of Physics, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA USA
| | | | - Giona Kleinberg
- Department of Bioengineering, Northeastern University, Boston, MA USA
| | - Katlyn Lloyd
- Department of Bioengineering, Northeastern University, Boston, MA USA
| | - Thomas Mueller
- Department of Bioengineering, Northeastern University, Boston, MA USA
| | | | - Jose J. Muñoz
- Department of Mathematics, Laboratori de Càlcul Numeric (LaCàN), Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
- Centre Internacional de Mètodes Numèrics en Enginyeria (CIMNE), Barcelona, Spain
- Institut de Matemàtiques de la UPC-BarcelonaTech (IMTech), Barcelona, Spain
| | - James R. Monaghan
- Department of Biology, Northeastern University, Boston, MA USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, MA USA
| | - Sandra J. Shefelbine
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA USA
- Department of Bioengineering, Northeastern University, Boston, MA USA
| |
Collapse
|
13
|
Paggi CA, Hendriks J, Karperien M, Le Gac S. Emulating the chondrocyte microenvironment using multi-directional mechanical stimulation in a cartilage-on-chip. LAB ON A CHIP 2022; 22:1815-1828. [PMID: 35352723 DOI: 10.1039/d1lc01069g] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The multi-directional mechanical stimulation experienced by articular cartilage during motion is transferred to the chondrocytes through a thin layer of pericellular matrix around each cell; chondrocytes in turn respond by releasing matrix proteins and/or matrix-degrading enzymes. In the present study we investigated how different types of mechanical stimulation can affect a chondrocyte's phenotype and extracellular matrix (ECM) production. To this end, we employed a cartilage-on-chip system which allows exerting well-defined compressive and multi-directional mechanical stimulation on a 3D chondrocyte-laden agarose hydrogel using a thin deformable membrane and three individually addressed actuation chambers. First, the 3D chondrocyte culture in agarose responded to exposure to mechanical stimulation by an initial increase in IL-6 production and little-to-no change in IL-1β and TNF-α secretion after one day of on-chip culture. Exposure to mechanical stimulation enhanced COL2A1 (hyaline cartilage marker) and decreased COL1A1 (fibrotic cartilage) expression, this being more marked for the multi-directional stimulation. Remarkably, the production of glycosaminoglycans (GAGs), one of the main components of native cartilage ECM, was significantly increased after 15 days of on-chip culture and 14 days of mechanical stimulation. Specifically, a thin pericellular matrix shell (1-5 μm) surrounding the chondrocytes as well as an interstitial matrix, both reminiscent of the in vivo situation, were deposited. Matrix deposition was highest in chips exposed to multi-directional mechanical stimulation. Finally, exposure to mechanical cues enhanced the production of essential cartilage ECM markers, such as aggrecan, collagen II and collagen VI, a marker for the pericellular matrix. Altogether our results highlight the importance of mechanical cues, and using the right type of stimulation, to emulate in vitro, the chondrocyte microenvironment.
Collapse
Affiliation(s)
- Carlo Alberto Paggi
- Department of Developmental BioEngineering, TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
| | - Jan Hendriks
- Department of Developmental BioEngineering, TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
| | - Marcel Karperien
- Department of Developmental BioEngineering, TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
14
|
Application of Alginate Hydrogels for Next-Generation Articular Cartilage Regeneration. Int J Mol Sci 2022; 23:ijms23031147. [PMID: 35163071 PMCID: PMC8835677 DOI: 10.3390/ijms23031147] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022] Open
Abstract
The articular cartilage has insufficient intrinsic healing abilities, and articular cartilage injuries often progress to osteoarthritis. Alginate-based scaffolds are attractive biomaterials for cartilage repair and regeneration, allowing for the delivery of cells and therapeutic drugs and gene sequences. In light of the heterogeneity of findings reporting the benefits of using alginate for cartilage regeneration, a better understanding of alginate-based systems is needed in order to improve the approaches aiming to enhance cartilage regeneration with this compound. This review provides an in-depth evaluation of the literature, focusing on the manipulation of alginate as a tool to support the processes involved in cartilage healing in order to demonstrate how such a material, used as a direct compound or combined with cell and gene therapy and with scaffold-guided gene transfer procedures, may assist cartilage regeneration in an optimal manner for future applications in patients.
Collapse
|
15
|
Alizadeh Sardroud H, Wanlin T, Chen X, Eames BF. Cartilage Tissue Engineering Approaches Need to Assess Fibrocartilage When Hydrogel Constructs Are Mechanically Loaded. Front Bioeng Biotechnol 2022; 9:787538. [PMID: 35096790 PMCID: PMC8790514 DOI: 10.3389/fbioe.2021.787538] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/19/2022] Open
Abstract
Chondrocytes that are impregnated within hydrogel constructs sense applied mechanical force and can respond by expressing collagens, which are deposited into the extracellular matrix (ECM). The intention of most cartilage tissue engineering is to form hyaline cartilage, but if mechanical stimulation pushes the ratio of collagen type I (Col1) to collagen type II (Col2) in the ECM too high, then fibrocartilage can form instead. With a focus on Col1 and Col2 expression, the first part of this article reviews the latest studies on hyaline cartilage regeneration within hydrogel constructs that are subjected to compression forces (one of the major types of the forces within joints) in vitro. Since the mechanical loading conditions involving compression and other forces in joints are difficult to reproduce in vitro, implantation of hydrogel constructs in vivo is also reviewed, again with a focus on Col1 and Col2 production within the newly formed cartilage. Furthermore, mechanotransduction pathways that may be related to the expression of Col1 and Col2 within chondrocytes are reviewed and examined. Also, two recently-emerged, novel approaches of load-shielding and synchrotron radiation (SR)–based imaging techniques are discussed and highlighted for future applications to the regeneration of hyaline cartilage. Going forward, all cartilage tissue engineering experiments should assess thoroughly whether fibrocartilage or hyaline cartilage is formed.
Collapse
Affiliation(s)
- Hamed Alizadeh Sardroud
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- *Correspondence: Hamed Alizadeh Sardroud,
| | - Tasker Wanlin
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - B. Frank Eames
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
16
|
Caravaggi P, Assirelli E, Ensini A, Ortolani M, Mariani E, Leardini A, Neri S, Belvedere C. Biomechanical-Based Protocol for in vitro Study of Cartilage Response to Cyclic Loading: A Proof-of-Concept in Knee Osteoarthritis. Front Bioeng Biotechnol 2021; 9:634327. [PMID: 34012954 PMCID: PMC8126668 DOI: 10.3389/fbioe.2021.634327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/08/2021] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is an evolving disease and a major cause of pain and impaired mobility. A deeper understanding of cartilage metabolism in response to loading is critical to achieve greater insight into OA mechanisms. While physiological joint loading helps maintain cartilage integrity, reduced or excessive loading have catabolic effects. The main scope of this study is to present an original methodology potentially capable to elucidate the effect of cyclic joint loading on cartilage metabolism, to identify mechanisms involved in preventing or slowing down OA progression, and to provide preliminary data on its application. In the proposed protocol, the combination of biomechanical data and medical imaging are integrated with molecular information about chondrocyte mechanotransduction and tissue homeostasis. The protocol appears to be flexible and suitable to analyze human OA knee cartilage explants, with different degrees of degeneration, undergoing ex vivo realistic cyclic joint loading estimated via gait analysis in patients simulating mild activities of daily living. The modulation of molecules involved in cartilage homeostasis, mechanotransduction, inflammation, pain and wound healing can be analyzed in chondrocytes and culture supernatants. A thorough analysis performed with the proposed methodology, combining in vivo functional biomechanical evaluations with ex vivo molecular assessments is expected to provide new insights on the beneficial effects of physiological loading and contribute to the design and optimization of non-pharmacological treatments limiting OA progression.
Collapse
Affiliation(s)
- Paolo Caravaggi
- Movement Analysis Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Assirelli
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Andrea Ensini
- I Orthopaedic and Traumatologic Clinic, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Maurizio Ortolani
- Movement Analysis Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Erminia Mariani
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy.,Department of Medical and Surgical Sciences, Alma Mater Studiorum-Università di Bologna, Bologna, Italy
| | - Alberto Leardini
- Movement Analysis Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Simona Neri
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Claudio Belvedere
- Movement Analysis Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
17
|
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: 25] [Impact Index Per Article: 6.3] [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
|
18
|
Ji X, Ito A, Nakahata A, Nishitani K, Kuroki H, Aoyama T. Effects of in vivo cyclic compressive loading on the distribution of local Col2 and superficial lubricin in rat knee cartilage. J Orthop Res 2021; 39:543-552. [PMID: 32716572 DOI: 10.1002/jor.24812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/20/2020] [Accepted: 07/09/2020] [Indexed: 02/04/2023]
Abstract
This study aimed to examine the effects of an episode of in vivo cyclic loading on rat knee articular cartilage (AC) under medium-term observation, while also investigating relevant factors associated with the progression of post-traumatic osteoarthritis (PTOA). Twelve-week-old Wistar rats underwent one episode comprising 60 cycles of 20 N or 50 N dynamic compression on the right knee joint. Spatiotemporal changes in the AC after loading were evaluated using histology and immunohistochemistry at 3 days and 1, 2, 4, and 8 weeks after loading (n = 6 for each condition). Chondrocyte vitality was assessed at 1, 3, 6, and 12 hours after loading (n = 2 for each condition). A localized AC lesion on the lateral femoral condyle was confirmed in all subjects. The surface and intermediate cartilage in the affected area degenerated after loading, but the calcified cartilage remained intact. Expression of type II collagen in the lesion cartilage was upregulated after loading, whereas the superficial lubricin layer was eroded in response to cyclic compression. However, the distribution of superficial lubricin gradually recovered to the normal level 4 weeks after loading-induced injury. We confirmed that 60 repetitions of cyclic loading exceeding 20 N could result in cartilage damage in the rat knee. Endogenous repairs in well-structured joints work well to rebuild protective layers on the lesion cartilage surface, which may be the latent factor delaying the progression of PTOA.
Collapse
Affiliation(s)
- Xiang Ji
- Department of Development and Rehabilitation of Motor Function, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiro Nakahata
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kohei Nishitani
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Kuroki
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Development and Rehabilitation of Motor Function, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
19
|
Boosting in vitro cartilage tissue engineering through the fabrication of polycaprolactone-gelatin 3D scaffolds with specific depth-dependent fiber alignments and mechanical stimulation. J Mech Behav Biomed Mater 2021; 117:104373. [PMID: 33618241 DOI: 10.1016/j.jmbbm.2021.104373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/13/2021] [Accepted: 01/28/2021] [Indexed: 11/21/2022]
Abstract
Due to the limited self-healing ability of natural cartilage, several tissue engineering strategies have been explored to develop functional replacements. Still, most of these approaches do not attempt to recreate in vitro the anisotropic organization of its extracellular matrix, which is essential for a suitable load-bearing function. In this work, different depth-dependent alignments of polycaprolactone-gelatin electrospun fibers were assembled into three-dimensional scaffold architectures to assess variations on chondrocyte response under static, unconfined compressed and perfused culture conditions. The in vitro results confirmed that not only the 3D scaffolds specific depth-dependent fiber alignments potentiated chondrocyte proliferation and migration towards the fibrous systems, but also the mechanical stimulation protocols applied were able to enhance significantly cell metabolic activity and extracellular matrix deposition, respectively.
Collapse
|
20
|
Monaco G, El Haj AJ, Alini M, Stoddart MJ. Ex Vivo Systems to Study Chondrogenic Differentiation and Cartilage Integration. J Funct Morphol Kinesiol 2021; 6:E6. [PMID: 33466400 PMCID: PMC7838775 DOI: 10.3390/jfmk6010006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022] Open
Abstract
Articular cartilage injury and repair is an issue of growing importance. Although common, defects of articular cartilage present a unique clinical challenge due to its poor self-healing capacity, which is largely due to its avascular nature. There is a critical need to better study and understand cellular healing mechanisms to achieve more effective therapies for cartilage regeneration. This article aims to describe the key features of cartilage which is being modelled using tissue engineered cartilage constructs and ex vivo systems. These models have been used to investigate chondrogenic differentiation and to study the mechanisms of cartilage integration into the surrounding tissue. The review highlights the key regeneration principles of articular cartilage repair in healthy and diseased joints. Using co-culture models and novel bioreactor designs, the basis of regeneration is aligned with recent efforts for optimal therapeutic interventions.
Collapse
Affiliation(s)
- Graziana Monaco
- AO Research Institute Davos, Clavadelerstrasse 8, CH-7270 Davos Platz, Switzerland; (G.M.); (M.A.)
- School of Pharmacy & Bioengineering Research, University of Keele, Keele ST5 5BG, UK;
| | - Alicia J. El Haj
- School of Pharmacy & Bioengineering Research, University of Keele, Keele ST5 5BG, UK;
- Healthcare Technology Institute, Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TH, UK
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, CH-7270 Davos Platz, Switzerland; (G.M.); (M.A.)
| | - Martin J. Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, CH-7270 Davos Platz, Switzerland; (G.M.); (M.A.)
- School of Pharmacy & Bioengineering Research, University of Keele, Keele ST5 5BG, UK;
| |
Collapse
|
21
|
Santos S, Richard K, Fisher MC, Dealy CN, Pierce DM. Chondrocytes respond both anabolically and catabolically to impact loading generally considered non-injurious. J Mech Behav Biomed Mater 2020; 115:104252. [PMID: 33385951 DOI: 10.1016/j.jmbbm.2020.104252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 11/25/2020] [Accepted: 12/03/2020] [Indexed: 11/24/2022]
Abstract
We aimed to determine the longitudinal effects of low-energy (generally considered non-injurious) impact loading on (1) chondrocyte proliferation, (2) chondroprogenitor cell activity, and (3) EGFR signaling. In an in vitro study, we assessed 127 full-thickness, cylindrical osteochondral plugs of bovine cartilage undergoing either single, uniaxial unconfined impact loads with energy densities in the range of 1.5-3.2mJ/mm3 or no impact (controls). We quantified cell responses at two, 24, 48, and 72 h via immunohistochemical labeling of Ki67, Sox9, and pEGFR antibodies. We compared strain, stress, and impact energy density as predictors for mechanotransductive responses from cells, and fit significant correlations using linear regressions. Our study demonstrates that low-energy mechanical impacts (1.5-3.2mJ/mm3) generally stimulate time-dependent anabolic responses in the superficial zone of articular cartilage and catabolic responses in the middle and deep zones. We also found that impact energy density is the most consistent predictor of cell responses to low-energy impact loading. These spatial and temporal changes in chondrocyte behavior result directly from low-energy mechanical impacts, revealing a new level of mechanotransductive sensitivity in chondrocytes not previously appreciated.
Collapse
Affiliation(s)
- Stephany Santos
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States of America
| | - Kelsey Richard
- Department of Global Health, University of Connecticut, Storrs, CT, United States of America
| | - Melanie C Fisher
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Services, University of Connecticut Health Center, Farmington, CT, United States of America
| | - Caroline N Dealy
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Services, University of Connecticut Health Center, Farmington, CT, United States of America; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT, United States of America
| | - David M Pierce
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States of America; Department of Mechanical Engineering, University of Connecticut, Storrs, CT, United States of America.
| |
Collapse
|
22
|
Tekari A, Egli RJ, Schmid V, Justiz J, Luginbuehl R. A Novel Bioreactor System Capable of Simulating the In Vivo Conditions of Synovial Joints. Tissue Eng Part C Methods 2020; 26:617-627. [PMID: 33267725 PMCID: PMC7759289 DOI: 10.1089/ten.tec.2020.0161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Any significant in vitro evaluation of cartilage tissue engineering and cartilage repair strategies has to be performed under the harsh conditions encountered in vivo within synovial joints. To this end, we have developed a novel automated physiological robot reactor system (PRRS) that is capable of recapitulating complex physiological motions and load patterns within an environment similar to that found in the human knee. The PRRS consists of a mechanical stimulation unit (MSU) and an automatic sample changer (ASC) within an environment control box in which the humidity, temperature, and gas composition are tightly regulated. The MSU has three linear (orthogonal) axes and one rotational degree of freedom (around the z-axis). The ASC provides space for up to 24 samples, which can be allocated to individual stimulation patterns. Cell-seeded scaffolds and ex vivo tissue culture systems were established to demonstrate the applicability of the PRRS to the investigation of the effect of load and environmental conditions on engineering and maintenance of articular cartilage in vitro. The bioreactor is a flexible system that has the potential to be applied for culturing connective tissues other than cartilage, such as bone and intervertebral disc tissue, even though the mechanical and environmental parameters are very different.
Collapse
Affiliation(s)
- Adel Tekari
- Group for Bone Biology and Orthopaedic Research, Department for Biomedical Research, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.,Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Rainer J Egli
- Group for Bone Biology and Orthopaedic Research, Department for Biomedical Research, University of Bern, Bern, Switzerland.,RMS Foundation, Bettlach, Switzerland.,Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Veit Schmid
- Institute for Human-Centered Engineering (HuCE) BME Lab, Bern University of Applied Sciences, Biel, Switzerland
| | - Joern Justiz
- Institute for Human-Centered Engineering (HuCE) BME Lab, Bern University of Applied Sciences, Biel, Switzerland
| | - Reto Luginbuehl
- Group for Bone Biology and Orthopaedic Research, Department for Biomedical Research, University of Bern, Bern, Switzerland.,RMS Foundation, Bettlach, Switzerland.,Blaser Swisslube AG, Hasle-Ruegsau, Switzerland
| |
Collapse
|
23
|
Chong PP, Panjavarnam P, Ahmad WNHW, Chan CK, Abbas AA, Merican AM, Pingguan-Murphy B, Kamarul T. Mechanical compression controls the biosynthesis of human osteoarthritic chondrocytes in vitro. Clin Biomech (Bristol, Avon) 2020; 79:105178. [PMID: 32988676 DOI: 10.1016/j.clinbiomech.2020.105178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 05/29/2020] [Accepted: 09/11/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cartilage damage, which can potentially lead to osteoarthritis, is a leading cause of morbidity in the elderly population. Chondrocytes are sensitive to mechanical stimuli and their matrix-protein synthesis may be altered when chondrocytes experience a variety of in vivo loadings. Therefore, a study was conducted to evaluate the biosynthesis of isolated osteoarthritic chondrocytes which subjected to compression with varying dynamic compressive strains and loading durations. METHODS The proximal tibia was resected as a single osteochondral unit during total knee replacement from patients (N = 10). The osteoarthritic chondrocytes were isolated from the osteochondral units, and characterized using reverse transcriptase-polymerase chain reaction. The isolated osteoarthritic chondrocytes were cultured and embedded in agarose, and then subjected to 10% and 20% uniaxial dynamic compression up to 8-days using a bioreactor. The morphological features and changes in the osteoarthritic chondrocytes upon compression were evaluated using scanning electron microscopy. Safranin O was used to detect the presence of cartilage matrix proteoglycan expression while quantitative analysis was conducted by measuring type VI collagen using an immunohistochemistry and fluorescence intensity assay. FINDINGS Gene expression analysis indicated that the isolated osteoarthritic chondrocytes expressed chondrocyte-specific markers, including BGN, CD90 and HSPG-2. Moreover, the compressed osteoarthritic chondrocytes showed a more intense and broader deposition of proteoglycan and type VI collagen than control. The expression of type VI collagen was directly proportional to the duration of compression in which 8-days compression was significantly higher than 4-days compression. The 20% compression showed significantly higher intensity compared to 10% compression in 4- and 8-days. INTERPRETATION The biosynthetic activity of human chondrocytes from osteoarthritic joints can be enhanced using selected compression regimes.
Collapse
Affiliation(s)
- Pan Pan Chong
- National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Ponnurajah Panjavarnam
- National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Wan Nor Hanis Wan Ahmad
- National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Chee Ken Chan
- Mahkota Medical Centre, No 3, Mahkota Melaka, Jalan Merdeka, 75000 Melaka, Malaysia
| | - Azlina A Abbas
- National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Azhar M Merican
- National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Tunku Kamarul
- National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
24
|
Walsh SK, Schneider SE, Amundson LA, Neu CP, Henak CR. Maturity-dependent cartilage cell plasticity and sensitivity to external perturbation. J Mech Behav Biomed Mater 2020; 106:103732. [PMID: 32321631 DOI: 10.1016/j.jmbbm.2020.103732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/04/2020] [Accepted: 03/13/2020] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Articular cartilage undergoes biological and morphological changes throughout maturation. The prevalence of osteoarthritis in the aged population suggests that maturation predisposes cartilage to degradation and/or impaired regeneration, but this process is not fully understood. Therefore, the objective of this study was to characterize the cellular and genetic profile of cartilage, as well as biological plasticity in response to mechanical and culture time stimuli, as a function of animal maturity. METHODS/DESIGN Porcine articular cartilage explants were harvested from stifle joints of immature (2-4 weeks), adolescent (5-6 months), and mature (1-5 years) animals. Half of all samples were subjected to a single compressive mechanical load. Loaded samples were paired with unloaded controls for downstream analyses. Expression of cartilage progenitor cell markers CD105, CD44, and CD29 were determined via flow cytometry. Expression of matrix synthesis genes Col1, Col2, Col10, ACAN, and SOX9 were determined via qPCR. Tissue morphology and matrix content were examined histologically. Post-loading assays were performed immediately and following 7 days in culture. RESULTS CD105 and CD29 expression decreased with maturity, while CD44 expression was upregulated in cartilage from mature animals. Expression of matrix synthesis genes were generally upregulated in cartilage from mature animals, and adolescent animals showed the lowest expression of several matrix synthesizing genes. Culture time and mechanical loading analyses revealed greater plasticity to mechanical loading and culture time in cartilage from younger animals. Histology confirmed distinct structural and biochemical profiles across maturity. CONCLUSION This study demonstrates differential, nonlinear expression of chondroprogenitor markers and matrix synthesis genes as a function of cartilage maturity, as well as loss of biological plasticity in aged tissue. These findings have likely implications for age-related loss of regeneration and osteoarthritis progression.
Collapse
Affiliation(s)
- Shannon K Walsh
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, USA.
| | - Stephanie E Schneider
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA.
| | - Laura A Amundson
- Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| | - Corey P Neu
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA.
| | - Corinne R Henak
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
25
|
Gamez C, Schneider-Wald B, Schuette A, Mack M, Hauk L, Khan AUM, Gretz N, Stoffel M, Bieback K, Schwarz ML. Bioreactor for mobilization of mesenchymal stem/stromal cells into scaffolds under mechanical stimulation: Preliminary results. PLoS One 2020; 15:e0227553. [PMID: 31923210 PMCID: PMC6953860 DOI: 10.1371/journal.pone.0227553] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 12/20/2019] [Indexed: 11/18/2022] Open
Abstract
Introduction Articular cartilage (AC) is a viscoelastic tissue with a limited regenerative capability because of the lack of vasculature. Mechanical stimulation contributes to the homeostasis of functional AC since it promotes the delivery of nutrients, cytokines and growth factors between the distant chondrocytes. We hypothesized that biomechanical stimulation might enhance mobilization of endogenous mesenchymal stem/stromal cells (MSCs) from neighboring niches as the bone marrow. Aim This study aimed to introduce a bioreactor for inducing mobilization of MSCs from one compartment to another above by mechanical stimulation in vitro. Methods A novel mechanical system for evaluating mobilization of cells in a 3D context in vitro is presented. The system consists of a compression bioreactor able to induce loading on hydrogel-based scaffolds, custom-made software for settings management and data recording, and image based biological evaluation. Intermittent load was applied under a periodic regime with frequency of 0.3 Hz and unload phases of 10 seconds each 180 cycles over 24 hours. The mechanical stimulation acted on an alginate scaffold and a cell reservoir containing MSCs below it. The dynamic compression exerted amplitude of 200 μm as 10% strain regarding the original height of the scaffold. Results The bioreactor was able to stimulate the scaffolds and the cells for 24.4 (±1.7) hours, exerting compression with vertical displacements of 185.8 (±17.8) μm and a force-amplitude of 1.87 (±1.37; min 0.31, max 4.42) N. Our results suggest that continuous mechanical stimulation hampered the viability of the cells located at the cell reservoir when comparing to intermittent mechanical stimulation (34.4 ± 2.0% vs. 66.8 ± 5.9%, respectively). Functionalizing alginate scaffolds with laminin-521 (LN521) seemed to enhance the mobilization of cells from 48 (±21) to 194 (±39) cells/mm3 after applying intermittent mechanical loading. Conclusion The bioreactor presented here was able to provide mechanical stimulation that seemed to induce the mobilization of MSCs into LN521-alginate scaffolds under an intermittent loading regime.
Collapse
Affiliation(s)
- Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Barbara Schneider-Wald
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Andy Schuette
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Michael Mack
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Luisa Hauk
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Arif ul Maula Khan
- Medical Research Centre (ZMF), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Norbert Gretz
- Medical Research Centre (ZMF), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Marcus Stoffel
- Institute of General Mechanics, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, FlowCore Mannheim, German Red Cross Blood Service of Baden Württemberg-Hessen, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Markus L. Schwarz
- Department for Experimental Orthopaedics and Trauma Surgery, Orthopaedics and Trauma Surgery Centre (OUZ), Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Baden Württemberg, Germany
- * E-mail:
| |
Collapse
|
26
|
Krueger S, Achilles S, Zimmermann J, Tischer T, Bader R, Jonitz-Heincke A. Re-Differentiation Capacity of Human Chondrocytes in Vitro Following Electrical Stimulation with Capacitively Coupled Fields. J Clin Med 2019; 8:E1771. [PMID: 31652962 PMCID: PMC6912508 DOI: 10.3390/jcm8111771] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 12/27/2022] Open
Abstract
Treatment of cartilage lesions remains a clinical challenge. Therefore, biophysical stimuli like electric fields seem to be a promising tool for chondrocytic differentiation and treatment of cartilage lesions. In this in vitro study, we evaluated the effects of low intensity capacitively coupled electric fields with an alternating voltage of 100 mVRMS (corresponds to 5.2 × 10-5 mV/cm) or 1 VRMS (corresponds to 5.2 × 10-4 mV/cm) with 1 kHz, on human chondrocytes derived from osteoarthritic (OA) and non-degenerative hyaline cartilage. A reduction of metabolic activity after electrical stimulation was more pronounced in non-degenerative cells. In contrast, DNA contents in OA cells were significantly decreased after electrical stimulation. A difference between 100 mVRMS and 1 VRMS was not detected. However, a voltage-dependent influence on gene and protein expression was observed. Both cell types showed increased synthesis rates of collagen (Col) II, glycosaminoglycans (GAG), and Col I protein following stimulation with 100 mVRMS, whereas this increase was clearly higher in OA cells. Our results demonstrated the sensitization of chondrocytes by alternating electric fields, especially at 100 mVRMS, which has an impact on chondrocytic differentiation capacity. However, analysis of further electrical stimulation parameters should be done to induce optimal hyaline characteristics of ex vivo expanded human chondrocytes.
Collapse
Affiliation(s)
- Simone Krueger
- Department of Orthopedics, Rostock University Medical Centre, 18057 Rostock, Germany.
| | - Sophie Achilles
- Department of Orthopedics, Rostock University Medical Centre, 18057 Rostock, Germany.
| | - Julius Zimmermann
- Institute of General Electrical Engineering, University of Rostock, 18059 Rostock, Germany.
| | - Thomas Tischer
- Department of Orthopedics, Rostock University Medical Centre, 18057 Rostock, Germany.
| | - Rainer Bader
- Department of Orthopedics, Rostock University Medical Centre, 18057 Rostock, Germany.
| | - Anika Jonitz-Heincke
- Department of Orthopedics, Rostock University Medical Centre, 18057 Rostock, Germany.
| |
Collapse
|
27
|
Dolzani P, Assirelli E, Pulsatelli L, Meliconi R, Mariani E, Neri S. Ex vivo physiological compression of human osteoarthritis cartilage modulates cellular and matrix components. PLoS One 2019; 14:e0222947. [PMID: 31550275 PMCID: PMC6759151 DOI: 10.1371/journal.pone.0222947] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/10/2019] [Indexed: 01/05/2023] Open
Abstract
Mechanical stimulation appears to play a key role in cartilage homeostasis maintenance, but it can also contribute to osteoarthritis (OA) pathogenesis. Accumulating evidence suggests that cartilage loading in the physiological range contributes to tissue integrity maintenance, whereas excessive or reduced loading have catabolic effects. However, how mechanical stimuli can regulate joint homeostasis is still not completely elucidated and few data are available on human cartilage. We aimed at investigating human OA cartilage response to ex vivo loading at physiological intensity. Cartilage explants from ten OA patients were subjected to ex vivo controlled compression, then recovered and used for gene and protein expression analysis of cartilage homeostasis markers. Compressed samples were compared to uncompressed ones in presence or without interleukin 1β (IL-1β) or interleukin 4 (IL-4). Cartilage explants compressed in combination with IL-4 treatment showed the best histological scores. Mechanical stimulation was able to significantly modify the expression of collagen type II (collagen 2), aggrecan, SOX9 transcription factor, cartilage oligomeric matrix protein (COMP), collagen degradation marker C2C and vascular endothelial growth factor (VEGF). Conversely, ADAMTS4 metallopeptidase, interleukin 4 receptor alpha (IL4Rα), chondroitin sulfate 846 epitope (CS846), procollagen type 2 C-propeptide (CPII) and glycosaminoglycans (GAG) appeared not modulated. Our data suggest that physiological compression of OA human cartilage modulates the inflammatory milieu by differently affecting the expression of components and homeostasis regulators of the cartilage extracellular matrix.
Collapse
Affiliation(s)
- Paolo Dolzani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Assirelli
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Lia Pulsatelli
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Riccardo Meliconi
- Unità di Medicina e Reumatologia, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Erminia Mariani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Simona Neri
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- * E-mail:
| |
Collapse
|
28
|
Qu P, Qi J, Han Y, Zhou L, Xie D, Song H, Geng C, Zhang K, Wang G. Effects of Rolling-Sliding Mechanical Stimulation on Cartilage Preserved In Vitro. Cell Mol Bioeng 2019; 12:301-310. [PMID: 31719916 DOI: 10.1007/s12195-019-00584-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 06/26/2019] [Indexed: 12/18/2022] Open
Abstract
Introduction Mechanical stimulation is important for maintaining cartilage function. We used a loading device to exert rolling-sliding mechanical stimulation on cartilage preserved in vitro to investigate cartilage viability and the involved mechanisms. Methods Osteochondral grafts from pig knees were randomly classified into loading and control groups. The loading group cartilage was subjected to cycles of mechanical stimulation with specified frequency/time/pressure combinations every 3 days; Then the DMEM was refreshed, and the cartilage was preserved in vitro. The control group cartilage was preserved in DMEM throughout the process and was changed every 3 days. On days 14 and 28, the chondrocyte survival rate, histology, and Young's modulus of the cartilage were measured. Western blots were performed after 2 h of loading to evaluate the protein expression. Results The loading group showed a significantly higher chondrocyte survival rate, proteoglycan and type II collagen content, and Young's modulus than did the control group on day 14, but no statistically significant differences were found on day 28. After two hours of the loading, the phosphorylation levels of MEK and ERK1/2 increased, and the expression of caspase-3, cleaved caspase-3 and bax decreased. Conclusion These results suggest that periodic rolling-sliding mechanical stimulation can increase cartilage vitality in 2 weeks; a possible mechanism is that mechanical stimulation activates the MEK/ERK signalling pathway, thus inhibiting apoptotic protein expression. This loading preservation scheme could be used by cartilage tissue banks to improve cartilage preservation in vitro and enhance the quality of cartilage repair.
Collapse
Affiliation(s)
- Pengwei Qu
- Institute of Sports Medicine, Shandong First Medical University&Shandong Academy of Medical Science, 619 Changcheng Road, Taian, 271016 Shandong China
| | - Jianhong Qi
- Institute of Sports Medicine, Shandong First Medical University&Shandong Academy of Medical Science, 619 Changcheng Road, Taian, 271016 Shandong China
| | - Yunning Han
- Institute of Sports Medicine, Shandong First Medical University&Shandong Academy of Medical Science, 619 Changcheng Road, Taian, 271016 Shandong China
| | - Lu Zhou
- Institute of Sports Medicine, Shandong First Medical University&Shandong Academy of Medical Science, 619 Changcheng Road, Taian, 271016 Shandong China
| | - Di Xie
- Institute of Sports Medicine, Shandong First Medical University&Shandong Academy of Medical Science, 619 Changcheng Road, Taian, 271016 Shandong China
| | - Hongqiang Song
- Institute of Sports Medicine, Shandong First Medical University&Shandong Academy of Medical Science, 619 Changcheng Road, Taian, 271016 Shandong China
| | - Caiyun Geng
- Institute of Sports Medicine, Shandong First Medical University&Shandong Academy of Medical Science, 619 Changcheng Road, Taian, 271016 Shandong China
| | - Kaihong Zhang
- Institute of Sports Medicine, Shandong First Medical University&Shandong Academy of Medical Science, 619 Changcheng Road, Taian, 271016 Shandong China
| | - Guozhu Wang
- College of Radiology, Shandong First Medical University&Shandong Academy of Medical Science, Taian, 271016 Shandong China
| |
Collapse
|
29
|
Chen Z, Mai Z, Tu S, Lu H, Chen L, Ai H. Expression of lubricin in rat posterior mandibular condylar cartilage following functional mandibular forward repositioning. J Orofac Orthop 2019; 80:128-135. [PMID: 30953086 DOI: 10.1007/s00056-019-00173-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 02/20/2019] [Indexed: 11/30/2022]
Abstract
PURPOSE The aim of the present study was to investigate the effects of mandibular forward repositioning on expression of lubricin in rat posterior condylar cartilage. METHODS In total, fifty 5‑week-old female Sprague Dawley rats were divided randomly into experimental groups and control groups. The animals in the experimental groups were fitted with modified acrylic inclined planes to advance the mandible, whereas rats in the normal control groups were left intact. Rats were sacrificed on days 3, 7, 14, 21, and 30, and temporomandibular joint (TMJ) samples were collected. The expression of lubricin of the posterior mandibular condylar cartilage was evaluated by immunohistochemistry. RESULTS In the control groups, higher expression of lubricin was observed in the proliferative zone of the posterior mandibular condylar cartilage compared with the hypertrophic zone during the experimental period. Compared with the control group, the positive signals for lubricin of the posterior mandibular condylar cartilage in the experimental animals were significantly higher on days 7, 14, and 21; however, no statistical difference was found on day 3 or 30. CONCLUSIONS Data analyses suggest that the bite jumping appliance temporarily enhanced lubricin expression, providing a good mechanical environment for the physiologic growth of the condyle and mandible, and contributes to TMJ remodeling by the regulation of condylar chondrocyte proliferation.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Zhihui Mai
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Shaoqin Tu
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongfei Lu
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Lin Chen
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Hong Ai
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou, China.
| |
Collapse
|
30
|
Huang X, Das R, Patel A, Nguyen TD. Physical Stimulations for Bone and Cartilage Regeneration. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 4:216-237. [PMID: 30740512 PMCID: PMC6366645 DOI: 10.1007/s40883-018-0064-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/07/2018] [Indexed: 12/26/2022]
Abstract
A wide range of techniques and methods are actively invented by clinicians and scientists who are dedicated to the field of musculoskeletal tissue regeneration. Biological, chemical, and physiological factors, which play key roles in musculoskeletal tissue development, have been extensively explored. However, physical stimulation is increasingly showing extreme importance in the processes of osteogenic and chondrogenic differentiation, proliferation and maturation through defined dose parameters including mode, frequency, magnitude, and duration of stimuli. Studies have shown manipulation of physical microenvironment is an indispensable strategy for the repair and regeneration of bone and cartilage, and biophysical cues could profoundly promote their regeneration. In this article, we review recent literature on utilization of physical stimulation, such as mechanical forces (cyclic strain, fluid shear stress, etc.), electrical and magnetic fields, ultrasound, shock waves, substrate stimuli, etc., to promote the repair and regeneration of bone and cartilage tissue. Emphasis is placed on the mechanism of cellular response and the potential clinical usage of these stimulations for bone and cartilage regeneration.
Collapse
|
31
|
Van Ginckel A, Hall M, Dobson F, Calders P. Effects of long-term exercise therapy on knee joint structure in people with knee osteoarthritis: A systematic review and meta-analysis. Semin Arthritis Rheum 2018; 48:941-949. [PMID: 30392703 DOI: 10.1016/j.semarthrit.2018.10.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/19/2018] [Accepted: 10/11/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To investigate effects of long-term exercise therapy for people with knee osteoarthritis (OA) on radiographic disease severity and cartilage integrity (primary outcomes) as well as severity of bone marrow lesions (BMLs), synovitis and/or effusion (secondary outcomes). METHODS We sourced randomized controlled trials in people with clinical and/or radiographic OA investigating the effect of land-based exercise therapy of > 6 months on radiographic disease severity and magnetic resonance imaging outcomes of cartilage integrity (morphology or composition) as well as BML, synovitis and/or effusion severity, when compared to no exercise. Two independent reviewers extracted data and assessed risk of bias. Random-effects meta-analysis was used to pool standardised mean differences (SMD) (95% confidence intervals (CI)) or odds ratios (OR) (95% CI) and estimate heterogeneity (I2, %). Quality of the pooled body of evidence was rated implementing the GRADE approach. Studies unsuitable for meta-analysis were summarized in a best-evidence synthesis. RESULTS Meta-analysis showed moderate quality evidence of no treatment effect on tibiofemoral radiographic disease severity ((SMD) 95% (CI): 0.06 (-0.07, 0.20), I2 = 0%) and low-quality evidence of no effect on tibiofemoral cartilage morphology (SMD (95%): 0.06 (-0.20, 0.36), I2 = 0%). Low quality evidence revealed no treatment effect on the odds of change in synovitis ((OR) (95% CI): 0.90 (0.51,1.60), I2 = 0%) and effusion ((OR (95% CI): 0.88 (0.64, 1.20), I2 = 0%), but greater odds of tibiofemoral BMLs worsening (OR (95% CI): 1.90 (1.11, 3.26), I2 = 0%). In best-evidence synthesis, limited evidence was found for changes in patellar cartilage composition following exercise in women with mild knee OA compared to no exercise, but not for tibiofemoral cartilage. CONCLUSION Long-term exercise therapy did not change tibiofemoral radiographic disease severity, cartilage morphology or synovitis/effusion, but may slightly increase the likelihood for increased BML severity. Overall, meta-analysis findings were limited in directness and precision and restricted to relatively imprecise effect estimates in people who were obese on average. Limited evidence suggested some protective effects on patellar cartilage composition.
Collapse
Affiliation(s)
- Ans Van Ginckel
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Corneel Heymanslaan 10, Building B3, 9000 Ghent, Belgium.
| | - Michelle Hall
- Centre for Health, Exercise and Sports Medicine, Department of Physiotherapy, The University of Melbourne, Alan Gilbert Building, 161 Barry Street, Carlton, VIC 3053, Australia.
| | - Fiona Dobson
- Centre for Health, Exercise and Sports Medicine, Department of Physiotherapy, The University of Melbourne, Alan Gilbert Building, 161 Barry Street, Carlton, VIC 3053, Australia.
| | - Patrick Calders
- Department of Rehabilitation Sciences and Physiotherapy, Ghent University, Corneel Heymanslaan 10, Building B3, 9000 Ghent, Belgium.
| |
Collapse
|
32
|
Adipose-Derived Mesenchymal Stem Cells in the Use of Cartilage Tissue Engineering: The Need for a Rapid Isolation Procedure. Stem Cells Int 2018; 2018:8947548. [PMID: 29765427 PMCID: PMC5903192 DOI: 10.1155/2018/8947548] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/01/2018] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have shown much promise with respect to their use in cartilage tissue engineering. MSCs can be obtained from many different tissue sources. Among these, adipose tissue can provide an abundant source of adipose-derived mesenchymal stem cells (ADMSCs). The infrapatellar fat pad (IFP) is a promising source of ADMSCs with respect to producing a cartilage lineage. Cell isolation protocols to date are time-consuming and follow conservative approaches that rely on a long incubation period of 24–48 hours. The different types of ADMSC isolation techniques used for cartilage repair will be reviewed and compared with the view of developing a rapid one-step isolation protocol that can be applied in the context of a surgical procedure.
Collapse
|
33
|
Sawatjui N, Limpaiboon T, Schrobback K, Klein T. Biomimetic scaffolds and dynamic compression enhance the properties of chondrocyte‐ and
MSC
‐based tissue‐engineered cartilage. J Tissue Eng Regen Med 2018; 12:1220-1229. [DOI: 10.1002/term.2653] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 12/21/2017] [Accepted: 02/17/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Nopporn Sawatjui
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences Khon Kaen University Khon Kaen Thailand
| | - Temduang Limpaiboon
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences Khon Kaen University Khon Kaen Thailand
| | - Karsten Schrobback
- Cartilage Regeneration Laboratory, Institute of Health and Biomedical Innovation Queensland University of Technology Brisbane Queensland Australia
| | - Travis Klein
- Cartilage Regeneration Laboratory, Institute of Health and Biomedical Innovation Queensland University of Technology Brisbane Queensland Australia
| |
Collapse
|
34
|
Biochemical alterations in inflammatory reactive chondrocytes: evidence for intercellular network communication. Heliyon 2018; 4:e00525. [PMID: 29560438 PMCID: PMC5857518 DOI: 10.1016/j.heliyon.2018.e00525] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/04/2018] [Accepted: 01/23/2018] [Indexed: 12/24/2022] Open
Abstract
Chondrocytes are effectively involved in the pathophysiological processes of inflammation in joints. They form cellular processes in the superficial layer of the articular cartilage and form gap junction coupled syncytium to facilitate cell-to-cell communication. However, very little is known about their physiological cellular identity and communication. The aim with the present work is to evaluate the physiological behavior after stimulation with the inflammatory inducers interleukin-1β and lipopolysaccharide. The cytoskeleton integrity and intracellular Ca2+ release were assessed as indicators of inflammatory state. Cytoskeleton integrity was analyzed through cartilage oligomeric matrix protein and actin labeling with an Alexa 488-conjugated phalloidin probe. Ca2+ responses were assessed through the Ca2+ sensitive fluorophore Fura-2/AM. Western blot analyses of several inflammatory markers were performed. The results show reorganization of the actin filaments. Glutamate, 5-hydoxytryptamine, and ATP evoked intracellular Ca2+ release changed from single peaks to oscillations after inflammatory induction in the chondrocytes. The expression of toll-like receptor 4, the glutamate transporters GLAST and GLT-1, and the matrix metalloproteinase-13 increased. This work demonstrates that chondrocytes are a key part in conditions that lead to inflammation in the cartilage. The inflammatory inducers modulate the cytoskeleton, the Ca2+ signaling, and several inflammatory parameters. In conclusion, our data show that the cellular responses to inflammatory insults from healthy and inflammatory chondrocytes resemble those previously observed in astrocyte and cardiac fibroblasts networks.
Collapse
|
35
|
Chen CH, Kuo CY, Chen JP. Effect of Cyclic Dynamic Compressive Loading on Chondrocytes and Adipose-Derived Stem Cells Co-Cultured in Highly Elastic Cryogel Scaffolds. Int J Mol Sci 2018; 19:370. [PMID: 29373507 PMCID: PMC5855592 DOI: 10.3390/ijms19020370] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/23/2022] Open
Abstract
In this study, we first used gelatin/chondroitin-6-sulfate/hyaluronan/chitosan highly elastic cryogels, which showed total recovery from large strains during repeated compression cycles, as 3D scaffolds to study the effects of cyclic dynamic compressive loading on chondrocyte gene expression and extracellular matrix (ECM) production. Dynamic culture of porcine chondrocytes was studied at 1 Hz, 10% to 40% strain and 1 to 9 h/day stimulation duration, in a mechanical-driven multi-chamber bioreactor for 14 days. From the experimental results, we could identify the optimum dynamic culture condition (20% and 3 h/day) to enhance the chondrocytic phenotype of chondrocytes from the expression of marker (Col I, Col II, Col X, TNF-α, TGF-β1 and IGF-1) genes by quantitative real-time polymerase chain reactions (qRT-PCR) and production of ECM (GAGs and Col II) by biochemical analysis and immunofluorescence staining. With up-regulated growth factor (TGF-β1 and IGF-1) genes, co-culture of chondrocytes with porcine adipose-derived stem cells (ASCs) was employed to facilitate chondrogenic differentiation of ASCs during dynamic culture in cryogel scaffolds. By replacing half of the chondrocytes with ASCs during co-culture, we could obtain similar production of ECM (GAGs and Col II) and expression of Col II, but reduced expression of Col I, Col X and TNF-α. Subcutaneous implantation of cells/scaffold constructs in nude mice after mono-culture (chondrocytes or ASCs) or co-culture (chondrocytes + ASCs) and subject to static or dynamic culture condition in vitro for 14 days was tested for tissue-engineering applications. The constructs were retrieved 8 weeks post-implantation for histological analysis by Alcian blue, Safranin O and Col II immunohistochemical staining. The most abundant ectopic cartilage tissue was found for the chondrocytes and chondrocytes + ASCs groups using dynamic culture, which showed similar neo-cartilage formation capability with half of the chondrocytes replaced by ASCs for co-culture. This combined co-culture/dynamic culture strategy is expected to cut down the amount of donor chondrocytes needed for cartilage-tissue engineering.
Collapse
Affiliation(s)
- Chih-Hao Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan.
| | - Chang-Yi Kuo
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan.
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan.
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
36
|
Anderson DE, Johnstone B. Dynamic Mechanical Compression of Chondrocytes for Tissue Engineering: A Critical Review. Front Bioeng Biotechnol 2017; 5:76. [PMID: 29322043 PMCID: PMC5732133 DOI: 10.3389/fbioe.2017.00076] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/23/2017] [Indexed: 01/19/2023] Open
Abstract
Articular cartilage functions to transmit and translate loads. In a classical structure-function relationship, the tissue resides in a dynamic mechanical environment that drives the formation of a highly organized tissue architecture suited to its biomechanical role. The dynamic mechanical environment includes multiaxial compressive and shear strains as well as hydrostatic and osmotic pressures. As the mechanical environment is known to modulate cell fate and influence tissue development toward a defined architecture in situ, dynamic mechanical loading has been hypothesized to induce the structure-function relationship during attempts at in vitro regeneration of articular cartilage. Researchers have designed increasingly sophisticated bioreactors with dynamic mechanical regimes, but the response of chondrocytes to dynamic compression and shear loading remains poorly characterized due to wide variation in study design, system variables, and outcome measurements. We assessed the literature pertaining to the use of dynamic compressive bioreactors for in vitro generation of cartilaginous tissue from primary and expanded chondrocytes. We used specific search terms to identify relevant publications from the PubMed database and manually sorted the data. It was very challenging to find consensus between studies because of species, age, cell source, and culture differences, coupled with the many loading regimes and the types of analyses used. Early studies that evaluated the response of primary bovine chondrocytes within hydrogels, and that employed dynamic single-axis compression with physiologic loading parameters, reported consistently favorable responses at the tissue level, with upregulation of biochemical synthesis and biomechanical properties. However, they rarely assessed the cellular response with gene expression or mechanotransduction pathway analyses. Later studies that employed increasingly sophisticated biomaterial-based systems, cells derived from different species, and complex loading regimes, did not necessarily corroborate prior positive results. These studies report positive results with respect to very specific conditions for cellular responses to dynamic load but fail to consistently achieve significant positive changes in relevant tissue engineering parameters, particularly collagen content and stiffness. There is a need for standardized methods and analyses of dynamic mechanical loading systems to guide the field of tissue engineering toward building cartilaginous implants that meet the goal of regenerating articular cartilage.
Collapse
Affiliation(s)
- Devon E Anderson
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, OR, United States
| | - Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
37
|
A novel bioreactor system for biaxial mechanical loading enhances the properties of tissue-engineered human cartilage. Sci Rep 2017; 7:16997. [PMID: 29208903 PMCID: PMC5717235 DOI: 10.1038/s41598-017-16523-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/13/2017] [Indexed: 11/15/2022] Open
Abstract
The ex vivo engineering of autologous cartilage tissues has the potential to revolutionize the clinical management of joint disorders. Yet, high manufacturing costs and variable outcomes associated with tissue-engineered implants are still limiting their application. To improve clinical outcomes and facilitate a wider use of engineered tissues, automated bioreactor systems capable of enhancing and monitoring neotissues are required. Here, we developed an innovative system capable of applying precise uni- or biaxial mechanical stimulation to developing cartilage neotissues in a tightly controlled and automated fashion. The bioreactor allows for simple control over the loading parameters with a user-friendly graphical interface and is equipped with a load cell for monitoring tissue maturation. Applying our bioreactor, we demonstrate that human articular chondrocytes encapsulated in hydrogels composed of gelatin methacryloyl (GelMA) and hyaluronic acid methacrylate (HAMA) respond to uni- and biaxial mechanical stimulation by upregulation of hyaline cartilage-specific marker genes. We further demonstrate that intermittent biaxial mechanostimulation enhances accumulation of hyaline cartilage-specific extracellular matrix. Our study underlines the stimulatory effects of mechanical loading on the biosynthetic activity of human chondrocytes in engineered constructs and the need for easy-to-use, automated bioreactor systems in cartilage tissue engineering.
Collapse
|
38
|
Zhu D, Tong X, Trinh P, Yang F. Mimicking Cartilage Tissue Zonal Organization by Engineering Tissue-Scale Gradient Hydrogels as 3D Cell Niche. Tissue Eng Part A 2017; 24:1-10. [PMID: 28385124 DOI: 10.1089/ten.tea.2016.0453] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Zonal organization plays an important role in cartilage structure and function, whereas most tissue-engineering strategies developed to date have only allowed the regeneration of cartilage with homogeneous biochemical and mechanical cues. To better restore tissue structure and function, there is a strong need to engineer materials with biomimetic gradient niche cues that recapitulate native tissue organization. To address this critical unmet need, in this study, we report a method for rapid formation of tissue-scale gradient hydrogels as a three-dimensional (3D) cell niche with tunable biochemical and physical properties. When encapsulated in stiffness gradient hydrogels, both chondrocytes and mesenchymal stem cells demonstrated zone-specific response and extracellular deposition that mimics zonal organization of articular cartilage. Blocking cell mechanosensing using blebbistatin abolished the zonal response of chondrocytes in 3D hydrogels with a stiffness gradient. Such tissue-scale gradient hydrogels can provide a 3D artificial cell niche to enable tissue engineering of various tissue types with zonal organizations or tissue interfaces.
Collapse
Affiliation(s)
- Danqing Zhu
- 1 Department of Bioengineering, Stanford University , Stanford, California
| | - Xinming Tong
- 2 Department of Orthopaedic Surgery, Stanford University , Stanford, California
| | - Pavin Trinh
- 3 Department of Biology, Stanford University , Stanford, California
| | - Fan Yang
- 1 Department of Bioengineering, Stanford University , Stanford, California.,2 Department of Orthopaedic Surgery, Stanford University , Stanford, California
| |
Collapse
|
39
|
Dong P, Tang X, Wang J, Jiang Y, Yao W, Gui J. [Short-term effectiveness of joint distraction by Ilizarov combined with arthroscopic debridement in treatment of knee osteoarthritis]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:794-798. [PMID: 29798521 PMCID: PMC8498158 DOI: 10.7507/1002-1892.201701099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 05/20/2017] [Indexed: 01/24/2023]
Abstract
Objective To investigate the short-term effectiveness of joint distraction by Ilizarov combined with arthroscopic debridement in the treatment of knee osteoarthritis (KOA). Methods Between January 2014 and January 2015, 15 patients (15 knees) with KOA were treated using arthroscopic debridement assisting with the Ilizarov distraction technology. There were 7 males and 8 females, aged from 45 to 64 years (mean, 55 years). The left knee and the right knee were involved in 6 and 9 cases respectively. The disease duration was 2.0-9.5 years (median, 6 years). They all had received conservative treatment for 6 months and got poor clinical improvement. The preoperative visual analogue scale (VAS) score, the Western Ontario and McMaster University Osteoarthritis Index (WOMAC) score, the knee injury and osteoarthritis outcome score (KOOS), the range of motion (ROM) for knee, and the radiographic joint space width were 76.2±8.8, 59.3±5.7, 44.3±7.2, (75±21)°, and (2.5±0.4) mm respectively. According to Kellgren-Lawrence grade system, 11 cases were rated as grade III and 4 cases as grade IV. Results There was no poor incision healing, infection, and deep vein thrombosis. All the 15 patients were followed up 12-18 months (mean, 15.5 months). Patients achieved pain relief. The knee activity was obviously improved. The postoperative VAS score, WOMAC score, KOOS score, and ROM at 12 months were 20.9±7.8, 38.2±5.5, 92.1±6.9, and (118±14)° respectively, showing significant difference when compared with preoperative ones ( t=18.213, P=0.000; t=10.317, P=0.000; t=18.564, P=0.000; t=6.599, P=0.000). Postoperative X-ray film showed that joint space width at 12 months was (3.8±0.3) mm, showing significant difference when compared with preoperative one ( t=10.070, P=0.000). Conclusion Joint distraction by Ilizarov combined with arthroscopic debridement can effectively relieve pain, improve the function and quality of life. It was beneficial to cartilaginous tissue repair and delaying the degenerative process of KOA. The short-term effectiveness is satisfactory.
Collapse
Affiliation(s)
- Peilong Dong
- Department of Orthopedics, Nanjing Hospital Affiliated to Nanjing Medical University, Nanjing Jiangsu, 210006, P.R.China;Department of Orthopedics, Jianhu Hospital Affiliated to Nantong University, Yancheng Jiangsu, 224700, P.R.China
| | - Xiaobo Tang
- Department of Orthopedics, Jianhu Hospital Affiliated to Nantong University, Yancheng Jiangsu, 224700, P.R.China
| | - Jian Wang
- Department of Orthopedics, Jianhu Hospital Affiliated to Nantong University, Yancheng Jiangsu, 224700, P.R.China
| | - Yiqiu Jiang
- Department of Orthopedics, Nanjing Hospital Affiliated to Nanjing Medical University, Nanjing Jiangsu, 210006, P.R.China
| | - Wangxiang Yao
- Department of Orthopedics, Nanjing Hospital Affiliated to Nanjing Medical University, Nanjing Jiangsu, 210006, P.R.China
| | - Jianchao Gui
- Department of Orthopedics, Nanjing Hospital Affiliated to Nanjing Medical University, Nanjing Jiangsu, 210006,
| |
Collapse
|
40
|
Zeineddine HA, Frush TJ, Saleh ZM, El-Othmani MM, Saleh KJ. Applications of Tissue Engineering in Joint Arthroplasty: Current Concepts Update. Orthop Clin North Am 2017; 48:275-288. [PMID: 28577777 DOI: 10.1016/j.ocl.2017.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Research in tissue engineering has undoubtedly achieved significant milestones in recent years. Although it is being applied in several disciplines, tissue engineering's application is particularly advanced in orthopedic surgery and in degenerative joint diseases. The literature is full of remarkable findings and trials using tissue engineering in articular cartilage disease. With the vast and expanding knowledge, and with the variety of techniques available at hand, the authors aimed to review the current concepts and advances in the use of cell sources in articular cartilage tissue engineering.
Collapse
Affiliation(s)
- Hussein A Zeineddine
- Department of Surgery, University of Chicago Medical Center, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | - Todd J Frush
- Department of Orthopaedics and Sports Medicine, Detroit Medical Center, University Health Center (UHC) 9B, 4201 Saint Antoine Street, Detroit, MI 48201-2153, USA
| | - Zeina M Saleh
- Department of Surgery, American University of Beirut Medical Center, Bliss Street, Riad El-Solh, Beirut 11072020, Lebanon
| | - Mouhanad M El-Othmani
- Department of Orthopaedics and Sports Medicine, Musculoskeletal Institute of Excellence, Detroit Medical Center, University Health Center (UHC) 9B, 4201 Saint Antoine Street, Detroit, MI 48201-2153, USA
| | - Khaled J Saleh
- Department of Orthopaedics and Sports Medicine, Detroit Medical Center, University Health Center (UHC) 9B, 4201 Saint Antoine Street, Detroit, MI 48201-2153, USA.
| |
Collapse
|
41
|
Silverstein AM, Stoker AM, Ateshian GA, Bulinski JC, Cook JL, Hung CT. Transient expression of the diseased phenotype of osteoarthritic chondrocytes in engineered cartilage. J Orthop Res 2017; 35:829-836. [PMID: 27183499 PMCID: PMC5383531 DOI: 10.1002/jor.23301] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 05/10/2016] [Indexed: 02/04/2023]
Abstract
Due to the degradation of osteoarthritic (OA) cartilage in post-traumatic OA (PTOA), these tissues are challenging to study and manipulate in vitro. In this study, chondrocytes isolated from either PTOA (meniscal-release (MR) model) or normal (contralateral limb) cartilage of canine knee joints were used to form micropellets to assess the maintenance of the OA chondrocyte phenotype in vitro. Media samples from the micropellet cultures were used to measure matrix metalloproteinase (MMP), chemokine, and cytokine concentrations. Significant differences in matrix synthesis were observed as a function of disease with OA chondrocytes generally synthesizing more extracellular matrix with increasing time in culture. No donor dependent differences were detected. Luminex multiplex analysis of pellet culture media showed disease and time-dependent differences in interleukin (IL)-8, keratinocyte chemoattractant (KC)-like protein, MMP-1, MMP-2, and MMP-3, which are differentially expressed in OA. This memory of their diseased phenotype persists for the first 2 weeks of culture. These results demonstrate the potential to use chondrocytes from an animal model of OA to study phenotype alterations during the progression and treatment of OA. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:829-836, 2017.
Collapse
Affiliation(s)
- Amy M. Silverstein
- Department of Biomedical Engineering, Columbia University, New York, 1210 Amsterdam Avenue, 351 Engineering Terrace, New York 10027
| | - Aaron M. Stoker
- Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, 1210 Amsterdam Avenue, 351 Engineering Terrace, New York 10027,Department of Mechanical Engineering, Columbia University, New York, New York
| | - J. Chloe Bulinski
- Department of Biological Sciences, Columbia University, New York, New York
| | - James L. Cook
- Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, 1210 Amsterdam Avenue, 351 Engineering Terrace, New York 10027
| |
Collapse
|
42
|
Diao HJ, Fung HS, Yeung P, Lam K, Yan CH, Chan BP. Dynamic cyclic compression modulates the chondrogenic phenotype in human chondrocytes from late stage osteoarthritis. Biochem Biophys Res Commun 2017; 486:14-21. [DOI: 10.1016/j.bbrc.2017.02.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 11/24/2022]
|
43
|
Completo A, Bandeiras C, Fonseca F. Comparative assessment of intrinsic mechanical stimuli on knee cartilage and compressed agarose constructs. Med Eng Phys 2017; 44:87-93. [PMID: 28318948 DOI: 10.1016/j.medengphy.2017.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 02/15/2017] [Accepted: 02/20/2017] [Indexed: 11/28/2022]
Abstract
A well-established cue for improving the properties of tissue-engineered cartilage is mechanical stimulation. However, the explicit ranges of mechanical stimuli that correspond to favorable metabolic outcomes are elusive. Usually, these outcomes have only been associated with the applied strain and frequency, an oversimplification that can hide the fundamental relationship between the intrinsic mechanical stimuli and the metabolic outcomes. This highlights two important key issues: the firstly is related to the evaluation of the intrinsic mechanical stimuli of native cartilage; the second, assuming that the intrinsic mechanical stimuli will be important, deals with the ability to replicate them on the tissue-engineered constructs. This study quantifies and compares the volume of cartilage and agarose subjected to a given magnitude range of each intrinsic mechanical stimulus, through a numerical simulation of a patient-specific knee model coupled with experimental data of contact during the stance phase of gait, and agarose constructs under direct-dynamic compression. The results suggest that direct compression loading needs to be parameterized with time-dependence during the initial culture period in order to better reproduce each one of the intrinsic mechanical stimuli developed in the patient-specific cartilage. A loading regime which combines time periods of low compressive strain (5%) and frequency (0.5Hz), in order to approach the maximal principal strain and fluid velocity stimulus of the patient-specific cartilage, with time periods of high compressive strain (20%) and frequency (3Hz), in order to approach the pore pressure values, may be advantageous relatively to a single loading regime throughout the full culture period.
Collapse
Affiliation(s)
- A Completo
- Mechanical Engineering Department, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal .
| | - C Bandeiras
- Mechanical Engineering Department, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - F Fonseca
- Coimbra Hospital and University Centre, Av. Bissaya Barreto, 3000-075 Coimbra, Portugal
| |
Collapse
|
44
|
Bar Oz M, Kumar A, Elayyan J, Reich E, Binyamin M, Kandel L, Liebergall M, Steinmeyer J, Lefebvre V, Dvir‐Ginzberg M. Acetylation reduces SOX9 nuclear entry and ACAN gene transactivation in human chondrocytes. Aging Cell 2016; 15:499-508. [PMID: 26910618 PMCID: PMC4854920 DOI: 10.1111/acel.12456] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2016] [Indexed: 12/13/2022] Open
Abstract
Changes in the content of aggrecan, an essential proteoglycan of articular cartilage, have been implicated in the pathophysiology of osteoarthritis (OA), a prevalent age-related, degenerative joint disease. Here, we examined the effect of SOX9 acetylation on ACAN transactivation in the context of osteoarthritis. Primary chondrocytes freshly isolated from degenerated OA cartilage displayed lower levels of ACAN mRNA and higher levels of acetylated SOX9 compared with cells from intact regions of OA cartilage. Degenerated OA cartilage presented chondrocyte clusters bearing diffused immunostaining for SOX9 compared with intact cartilage regions. Primary human chondrocytes freshly isolated from OA knee joints were cultured in monolayer or in three-dimensional alginate microbeads (3D). SOX9 was hypo-acetylated in 3D cultures and displayed enhanced binding to a -10 kb ACAN enhancer, a result consistent with higher ACAN mRNA levels than in monolayer cultures. It also co-immunoprecipitated with SIRT1, a major deacetylase responsible for SOX9 deacetylation. Finally, immunofluorescence assays revealed increased nuclear localization of SOX9 in primary chondrocytes treated with the NAD SIRT1 cofactor, than in cells treated with a SIRT1 inhibitor. Inhibition of importin β by importazole maintained SOX9 in the cytoplasm, even in the presence of NAD. Based on these data, we conclude that deacetylation promotes SOX9 nuclear translocation and hence its ability to activate ACAN.
Collapse
Affiliation(s)
- Michal Bar Oz
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Ashok Kumar
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Jinan Elayyan
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Eli Reich
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Milana Binyamin
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| | - Leonid Kandel
- Joint Replacement and Reconstructive Surgery UnitOrthopaedic Surgery ComplexHadassah Mount Scopus HospitalJerusalemIsrael
| | - Meir Liebergall
- Joint Replacement and Reconstructive Surgery UnitOrthopaedic Surgery ComplexHadassah Mount Scopus HospitalJerusalemIsrael
| | - Juergen Steinmeyer
- Laboratory for Experimental OrthopaedicsDepartment of Orthopaedic SurgeryUniversity Hospital Giessen & Marburg GmbHGießenGermany
| | | | - Mona Dvir‐Ginzberg
- Laboratory of Cartilage BiologyInstitute of Dental SciencesHebrew University of JerusalemJerusalemIsrael
| |
Collapse
|
45
|
Abstract
One of the most important issues facing cartilage tissue engineering is the inability to move technologies into the clinic. Despite the multitude of current research in the field, it is known that 90% of new drugs that advance past animal studies fail clinical trials. The objective of this review is to provide readers with an understanding of the scientific details of tissue engineered cartilage products that have demonstrated a certain level of efficacy in humans, so that newer technologies may be developed upon this foundation. Compared to existing treatments, such as microfracture or autologous chondrocyte implantation, a tissue engineered product can potentially provide more consistent clinical results in forming hyaline repair tissue and in filling the entirety of the defect. The various tissue engineering strategies (e.g., cell expansion, scaffold material, media formulations, biomimetic stimuli, etc.) used in forming these products, as collected from published literature, company websites, and relevant patents, are critically discussed. The authors note that many details about these products remain proprietary, not all information is made public, and that advancements to the products are continuously made. Nevertheless, by understanding the design and production processes of these emerging technologies, one can gain tremendous insight into how to best use them and also how to design the next generation of tissue engineered cartilage products.
Collapse
|
46
|
Flouzat-Lachaniette CH, Roubineau F, Heyberger C, Bouthors C. Distraction to treat knee osteoarthritis. Joint Bone Spine 2016; 84:141-144. [PMID: 27178780 DOI: 10.1016/j.jbspin.2016.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2016] [Indexed: 01/27/2023]
Abstract
The objective of this article is to review data on joint distraction used to treat knee osteoarthritis. Joint distraction is a surgical procedure in which the two bony ends of the joint are gradually pulled apart then kept separated for 2 months in an external fixation frame. Weight bearing is continued to ensure variations in hydrostatic pressure within the joint. In published studies, joint distraction provided substantial clinical and structural improvements in patients with knee osteoarthritis, delaying joint replacement surgery for at least 2 years. Animal studies showed that joint distraction was associated with decrease in the secondary inflammatory response, cartilage breakdown, and subchondral bone remodeling. In vitro, the intermittent application of hydrostatic pressure stimulated the production of extracellular matrix, particularly in joints with osteoarthritis. Nevertheless, several considerations invite caution when considering the more widespread use of joint distraction. Published studies have short follow-ups and small sample sizes. In addition, the high frequency of pin tract infection is of concern, since most patients eventually require knee replacement surgery. These two considerations indicate a need for longer-term prospective studies of patient cohorts.
Collapse
Affiliation(s)
- Charles-Henri Flouzat-Lachaniette
- Service de chirurgie orthopédique et traumatologique, hôpital Henri-Mondor, AP-HP-UPEC, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil cedex, France.
| | - François Roubineau
- Service de chirurgie orthopédique et traumatologique, hôpital Henri-Mondor, AP-HP-UPEC, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil cedex, France
| | - Clémence Heyberger
- Service de chirurgie orthopédique et traumatologique, hôpital Henri-Mondor, AP-HP-UPEC, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil cedex, France
| | - Charlie Bouthors
- Service de chirurgie orthopédique et traumatologique, hôpital Henri-Mondor, AP-HP-UPEC, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil cedex, France
| |
Collapse
|
47
|
Huang BJ, Hu JC, Athanasiou KA. Cell-based tissue engineering strategies used in the clinical repair of articular cartilage. Biomaterials 2016; 98:1-22. [PMID: 27177218 DOI: 10.1016/j.biomaterials.2016.04.018] [Citation(s) in RCA: 282] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/15/2016] [Accepted: 04/20/2016] [Indexed: 12/12/2022]
Abstract
One of the most important issues facing cartilage tissue engineering is the inability to move technologies into the clinic. Despite the multitude of current research in the field, it is known that 90% of new drugs that advance past animal studies fail clinical trials. The objective of this review is to provide readers with an understanding of the scientific details of tissue engineered cartilage products that have demonstrated a certain level of efficacy in humans, so that newer technologies may be developed upon this foundation. Compared to existing treatments, such as microfracture or autologous chondrocyte implantation, a tissue engineered product can potentially provide more consistent clinical results in forming hyaline repair tissue and in filling the entirety of the defect. The various tissue engineering strategies (e.g., cell expansion, scaffold material, media formulations, biomimetic stimuli, etc.) used in forming these products, as collected from published literature, company websites, and relevant patents, are critically discussed. The authors note that many details about these products remain proprietary, not all information is made public, and that advancements to the products are continuously made. Nevertheless, by understanding the design and production processes of these emerging technologies, one can gain tremendous insight into how to best use them and also how to design the next generation of tissue engineered cartilage products.
Collapse
Affiliation(s)
- Brian J Huang
- Department of Biomedical Engineering, University of California Davis, USA.
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Davis, USA.
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Davis, USA; Department of Orthopedic Surgery, University of California Davis, USA.
| |
Collapse
|
48
|
Chen C, Wei X, Wang S, Jiao Q, Zhang Y, Du G, Wang X, Wei F, Zhang J, Wei L. Compression regulates gene expression of chondrocytes through HDAC4 nuclear relocation via PP2A-dependent HDAC4 dephosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1633-42. [PMID: 27106144 DOI: 10.1016/j.bbamcr.2016.04.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 04/13/2016] [Accepted: 04/17/2016] [Indexed: 11/15/2022]
Abstract
Biomechanics plays a critical role in the modulation of chondrocyte function. The mechanisms by which mechanical loading is transduced into intracellular signals that regulate chondrocyte gene expression remain largely unknown. Histone deacetylase 4 (HDAC4) is specifically expressed in chondrocytes. Mice lacking HDAC4 display chondrocyte hypertrophy, ectopic and premature ossification, and die early during the perinatal period. HDAC4 has a remarkable ability to translocate between the cell's cytoplasm and nucleus. It has been established that subcellular relocation of HDAC4 plays a critical role in chondrocyte differentiation and proliferation. However, it remains unclear whether subcellular relocation of HDAC4 in chondrocytes can be induced by mechanical loading. In this study, we first report that compressive loading induces HDAC4 relocation from the cytoplasm to the nucleus of chondrocytes via stimulation of Ser/Thr-phosphoprotein phosphatases 2A (PP2A) activity, which results in dephosphorylation of HDAC4. Dephosphorylated HDAC4 relocates to the nucleus to achieve transcriptional repression of Runx2 and regulates chondrocyte gene expression in response to compression. Our results elucidate the mechanism by which mechanical compression regulates chondrocyte gene expression through HDAC4 relocation from the cell's cytoplasm to the nucleus via PP2A-dependent HDAC4 dephosphorylation.
Collapse
Affiliation(s)
- Chongwei Chen
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Xiaochun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Shaowei Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Qiang Jiao
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Yang Zhang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Guoqing Du
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Xiaohu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Fangyuan Wei
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Jianzhong Zhang
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Lei Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA.
| |
Collapse
|
49
|
Bartnikowski M, Akkineni AR, Gelinsky M, Woodruff MA, Klein TJ. A Hydrogel Model Incorporating 3D-Plotted Hydroxyapatite for Osteochondral Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2016; 9:E285. [PMID: 28773410 PMCID: PMC5502978 DOI: 10.3390/ma9040285] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/24/2016] [Accepted: 04/06/2016] [Indexed: 12/15/2022]
Abstract
The concept of biphasic or multi-layered compound scaffolds has been explored within numerous studies in the context of cartilage and osteochondral regeneration. To date, no system has been identified that stands out in terms of superior chondrogenesis, osteogenesis or the formation of a zone of calcified cartilage (ZCC). Herein we present a 3D plotted scaffold, comprising an alginate and hydroxyapatite paste, cast within a photocrosslinkable hydrogel made of gelatin methacrylamide (GelMA), or GelMA with hyaluronic acid methacrylate (HAMA). We hypothesized that this combination of 3D plotting and hydrogel crosslinking would form a high fidelity, cell supporting structure that would allow localization of hydroxyapatite to the deepest regions of the structure whilst taking advantage of hydrogel photocrosslinking. We assessed this preliminary design in terms of chondrogenesis in culture with human articular chondrocytes, and verified whether the inclusion of hydroxyapatite in the form presented had any influence on the formation of the ZCC. Whilst the inclusion of HAMA resulted in a better chondrogenic outcome, the effect of HAP was limited. We overall demonstrated that formation of such compound structures is possible, providing a foundation for future work. The development of cohesive biphasic systems is highly relevant for current and future cartilage tissue engineering.
Collapse
Affiliation(s)
- Michal Bartnikowski
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia.
| | - Ashwini Rahul Akkineni
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden D-01307, Germany.
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, Dresden D-01307, Germany.
| | - Maria A Woodruff
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia.
| | - Travis J Klein
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Queensland 4059, Australia.
| |
Collapse
|
50
|
Hyaluronic Acid Suppresses the Expression of Metalloproteinases in Osteoarthritic Cartilage Stimulated Simultaneously by Interleukin 1β and Mechanical Load. PLoS One 2016; 11:e0150020. [PMID: 26934732 PMCID: PMC4774918 DOI: 10.1371/journal.pone.0150020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/08/2016] [Indexed: 12/05/2022] Open
Abstract
Purpose In patients with osteoarthritis (OA), intraarticular injection of hyaluronic acid (HA) frequently results in reduced pain and improved function for prolonged periods of time, i.e. more than 6 months. However, the mechanisms underlying these effects are not fully understood. Our underlying hypothesis is that HA modifies the enzymatic breakdown of joint tissues. Methods To test this hypothesis, we examined osteochondral cylinders from 12 OA patients. In a bioreactor, these samples were stimulated by interleukin 1β (Il1ß) (2 ng/ml) plus mechanical load (2.0 Mpa at 0.5 Hz horizontal and 0.1 Hz vertical rotation), thus the experimental setup recapitulated both catabolic and anabolic clues of the OA joint. Results Upon addition of HA at either 1 or 3 mg/ml, we observed a significant suppression of expression of metalloproteinase (MMP)-13. A more detailed analysis based on the Kellgren and Lawrence (K&L) OA grade, showed a much greater degree of suppression of MMP-13 expression in grade IV as compared to grade II OA. In contrast to the observed MMP-13 suppression, treatment with HA resulted in a suppression of MMP-1 expression only at 1 mg/ml HA, while MMP-2 expression was not significantly affected by either HA concentration. Conclusion Together, these data suggest that under concurrent catabolic and anabolic stimulation, HA exhibits a pronounced suppressive effect on MMP-13. In the long-run these findings may benefit the development of treatment strategies aimed at blocking tissue degradation in OA patients.
Collapse
|