1
|
Godivier J, Lawrence EA, Wang M, Hammond CL, Nowlan NC. Compressive stress gradients direct mechanoregulation of anisotropic growth in the zebrafish jaw joint. PLoS Comput Biol 2024; 20:e1010940. [PMID: 38330044 PMCID: PMC10880962 DOI: 10.1371/journal.pcbi.1010940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/21/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Mechanical stimuli arising from fetal movements are critical factors underlying joint growth. Abnormal fetal movements negatively affect joint shape features with important implications for joint health, but the mechanisms by which mechanical forces from fetal movements influence joint growth are still unclear. In this research, we quantify zebrafish jaw joint growth in 3D in free-to-move and immobilised fish larvae between four and five days post fertilisation. We found that the main changes in size and shape in normally moving fish were in the ventrodorsal axis, while growth anisotropy was lost in the immobilised larvae. We next sought to determine the cell level activities underlying mechanoregulated growth anisotropy by tracking individual cells in the presence or absence of jaw movements, finding that the most dramatic changes in growth rates due to jaw immobility were in the ventrodorsal axis. Finally, we implemented mechanobiological simulations of joint growth with which we tested hypotheses relating specific mechanical stimuli to mechanoregulated growth anisotropy. Different types of mechanical stimulation were incorporated into the simulation to provide the mechanoregulated component of growth, in addition to the baseline (non-mechanoregulated) growth which occurs in the immobilised animals. We found that when average tissue stress over the opening and closing cycle of the joint was used as the stimulus for mechanoregulated growth, joint morphogenesis was not accurately predicted. Predictions were improved when using the stress gradients along the rudiment axes (i.e., the variation in magnitude of compression to magnitude of tension between local regions). However, the most accurate predictions were obtained when using the compressive stress gradients (i.e., the variation in compressive stress magnitude) along the rudiment axes. We conclude therefore that the dominant biophysical stimulus contributing to growth anisotropy during early joint development is the gradient of compressive stress experienced along the growth axes under cyclical loading.
Collapse
Affiliation(s)
- Josepha Godivier
- Department of Bioengineering, Imperial College London, London, United Kingdom
- School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
| | - Elizabeth A. Lawrence
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Mengdi Wang
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Chrissy L. Hammond
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Niamh C. Nowlan
- Department of Bioengineering, Imperial College London, London, United Kingdom
- School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Abusharkh HA, Robertson T, Mendenhall J, Gozen BA, Tingstad EM, Abu-Lail NI, Thiessen DB, Van Wie BJ. Impact of interstitial flow on cartilage matrix synthesis and NF-kB transcription factor mRNA expression in a novel perfusion bioreactor. Biotechnol Prog 2024; 40:e3404. [PMID: 37985202 PMCID: PMC10922130 DOI: 10.1002/btpr.3404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 09/15/2023] [Accepted: 10/23/2023] [Indexed: 11/22/2023]
Abstract
This work is focused on designing an easy-to-use novel perfusion system for articular cartilage (AC) tissue engineering and using it to elucidate the mechanism by which interstitial shear upregulates matrix synthesis by articular chondrocytes (AChs). Porous chitosan-agarose (CHAG) scaffolds were synthesized and compared to bulk agarose (AG) scaffolds. Both scaffolds were seeded with osteoarthritic human AChs and cultured in a novel perfusion system with a medium flow velocity of 0.33 mm/s corresponding to 0.4 mPa surfice shear and 40 mPa CHAG interstitial shear. While there were no statistical differences in cell viability for perfusion versus static cultures for either scaffold type, CHAG scaffolds exhibited a 3.3-fold higher (p < 0.005) cell viability compared to AG scaffold cultures. Effects of combined superficial and interstitial perfusion for CHAG showed 150- and 45-fold (p < 0.0001) increases in total collagen (COL) and 13- and 2.2-fold (p < 0.001) increases in glycosaminoglycans (GAGs) over AG non-perfusion and perfusion cultures, respectively, and a 1.5-fold and 3.6-fold (p < 0.005) increase over non-perfusion CHAG cultures. Contrasting CHAG perfusion and static cultures, chondrogenic gene comparisons showed a 3.5-fold increase in collagen type II/type I (COL2A1/COL1A1) mRNA ratio (p < 0.05), and a 1.3-fold increase in aggrecan mRNA. Observed effects are linked to NF-κB signal transduction pathway inhibition as confirmed by a 3.2-fold (p < 0.05) reduction of NF-κB mRNA expression upon exposure to perfusion. Our results demonstrate that pores play a critical role in improving cell viability and that interstitial flow caused by medium perfusion through the porous scaffolds enhances the expression of chondrogenic genes and extracellular matrix through downregulating NF-κB1.
Collapse
Affiliation(s)
- Haneen A Abusharkh
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164-6515
| | - Terreill Robertson
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164-6515
| | | | - Bulent A Gozen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920
| | - Edwin M Tingstad
- Inland Orthopedic Surgery and Sports Medicine Clinic, 825 SE Bishop Blvd, Suite 120, Pullman, WA 99163
| | - Nehal I Abu-Lail
- Department of Biomedical Engineering and Chemical Engineering, the University of Texas at San Antonio, San Antonio, TX 78249
| | - David B Thiessen
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164-6515
| | - Bernard J Van Wie
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164-6515
| |
Collapse
|
3
|
Ciulla MG, Marchini A, Gazzola J, Sambrotta M, Gelain F. Low-power microwaves: a cell-compatible physical treatment to enhance the mechanical properties of self-assembling peptides. NANOSCALE 2023; 15:15840-15854. [PMID: 37747054 DOI: 10.1039/d3nr02738d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Biomaterials designed for tissue engineering applications should, among other requirements, mimic the native extracellular matrix (ECM) of the tissues to be regenerated, both in terms of biomimetic and mechanical properties. Ideally, the scaffold stiffness and stress resistance should be tuned for each specific implantation therapy. Self-assembling peptides (SAPs) are promising synthetic bionanomaterials prone to easy multi-functionalization, bestowing biomimetic properties. However, they usually yield soft and fragile hydrogels unsuited for the regeneration of medium-to-hard tissues. For this purpose, chemical cross-linking of SAPs is an option, but it often requires a moderately toxic and expensive chemical compound and/or the presence of specific residues/reactive sites, posing issues for its feasibility and translational potential. In this work, we introduced, characterized by rheology, atomic force microscopy (AFM), Thioflavin-T assay (ThT), and Fourier transform infrared (FT-IR) tests, and optimized (by tuning the power, temperature and treatment time) a novel fast, green and affordable methodology using mild microwave (MW) irradiation to increase the mechanical properties of diverse classes of SAPs. Low-power MWs increase stiffness, resilience, and β-structuration, while high-power MW treatments partially denature the tested SAPs. Our pure-physical methodology does not alter the SAP biomimetic properties (verified via in vitro tests of viability and differentiation of human neural stem cells), is compatible with already seeded cells, and is also synergic with genipin-based cross-linking of SAPs; therefore, it may become the next standard for SAP preparation in tissue engineering applications at hand of all research labs and in clinics.
Collapse
Affiliation(s)
- Maria Gessica Ciulla
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy.
| | - Amanda Marchini
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy.
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Jacopo Gazzola
- Department of Biotechnology and Bioscience, University of Milan - Bicocca, 20125 Milan, Italy
| | - Manuel Sambrotta
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, 20133 Milan, Italy
| | - Fabrizio Gelain
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy.
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| |
Collapse
|
4
|
Azizi P, Drobek C, Budday S, Seitz H. Simulating the mechanical stimulation of cells on a porous hydrogel scaffold using an FSI model to predict cell differentiation. Front Bioeng Biotechnol 2023; 11:1249867. [PMID: 37799813 PMCID: PMC10549991 DOI: 10.3389/fbioe.2023.1249867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
3D-structured hydrogel scaffolds are frequently used in tissue engineering applications as they can provide a supportive and biocompatible environment for the growth and regeneration of new tissue. Hydrogel scaffolds seeded with human mesenchymal stem cells (MSCs) can be mechanically stimulated in bioreactors to promote the formation of cartilage or bone tissue. Although in vitro and in vivo experiments are necessary to understand the biological response of cells and tissues to mechanical stimulation, in silico methods are cost-effective and powerful approaches that can support these experimental investigations. In this study, we simulated the fluid-structure interaction (FSI) to predict cell differentiation on the entire surface of a 3D-structured hydrogel scaffold seeded with cells due to dynamic compressive load stimulation. The computational FSI model made it possible to simultaneously investigate the influence of both mechanical deformation and flow of the culture medium on the cells on the scaffold surface during stimulation. The transient one-way FSI model thus opens up significantly more possibilities for predicting cell differentiation in mechanically stimulated scaffolds than previous static microscale computational approaches used in mechanobiology. In a first parameter study, the impact of the amplitude of a sinusoidal compression ranging from 1% to 10% on the phenotype of cells seeded on a porous hydrogel scaffold was analyzed. The simulation results show that the number of cells differentiating into bone tissue gradually decreases with increasing compression amplitude, while differentiation into cartilage cells initially multiplied with increasing compression amplitude in the range of 2% up to 7% and then decreased. Fibrous cell differentiation was predicted from a compression of 5% and increased moderately up to a compression of 10%. At high compression amplitudes of 9% and 10%, negligible areas on the scaffold surface experienced high stimuli where no cell differentiation could occur. In summary, this study shows that simulation of the FSI system is a versatile approach in computational mechanobiology that can be used to study the effects of, for example, different scaffold designs and stimulation parameters on cell differentiation in mechanically stimulated 3D-structured scaffolds.
Collapse
Affiliation(s)
- Pedram Azizi
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany
| | - Christoph Drobek
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany
| | - Silvia Budday
- Department of Mechanical Engineering, Institute of Applied Mechanics, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Hermann Seitz
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany
| |
Collapse
|
5
|
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
|
6
|
Binaymotlagh R, Chronopoulou L, Palocci C. Peptide-Based Hydrogels: Template Materials for Tissue Engineering. J Funct Biomater 2023; 14:jfb14040233. [PMID: 37103323 PMCID: PMC10145623 DOI: 10.3390/jfb14040233] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Tissue and organ regeneration are challenging issues, yet they represent the frontier of current research in the biomedical field. Currently, a major problem is the lack of ideal scaffold materials' definition. As well known, peptide hydrogels have attracted increasing attention in recent years thanks to significant properties such as biocompatibility, biodegradability, good mechanical stability, and tissue-like elasticity. Such properties make them excellent candidates for 3D scaffold materials. In this review, the first aim is to describe the main features of a peptide hydrogel in order to be considered as a 3D scaffold, focusing in particular on mechanical properties, as well as on biodegradability and bioactivity. Then, some recent applications of peptide hydrogels in tissue engineering, including soft and hard tissues, will be discussed to analyze the most relevant research trends in this field.
Collapse
Affiliation(s)
- Roya Binaymotlagh
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Laura Chronopoulou
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
- Research Center for Applied Sciences to the Safeguard of Environment and Cultural Heritage (CIABC), Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Cleofe Palocci
- Department of Chemistry, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
- Research Center for Applied Sciences to the Safeguard of Environment and Cultural Heritage (CIABC), Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
7
|
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
|
8
|
Plaas AHK, Moran MM, Sandy JD, Hascall VC. Aggrecan and Hyaluronan: The Infamous Cartilage Polyelectrolytes - Then and Now. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:3-29. [PMID: 37052843 DOI: 10.1007/978-3-031-25588-5_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Cartilages are unique in the family of connective tissues in that they contain a high concentration of the glycosaminoglycans, chondroitin sulfate and keratan sulfate attached to the core protein of the proteoglycan, aggrecan. Multiple aggrecan molecules are organized in the extracellular matrix via a domain-specific molecular interaction with hyaluronan and a link protein, and these high molecular weight aggregates are immobilized within the collagen and glycoprotein network. The high negative charge density of glycosaminoglycans provides hydrophilicity, high osmotic swelling pressure and conformational flexibility, which together function to absorb fluctuations in biomechanical stresses on cartilage during movement of an articular joint. We have summarized information on the history and current knowledge obtained by biochemical and genetic approaches, on cell-mediated regulation of aggrecan metabolism and its role in skeletal development, growth as well as during the development of joint disease. In addition, we describe the pathways for hyaluronan metabolism, with particular focus on the role as a "metabolic rheostat" during chondrocyte responses in cartilage remodeling in growth and disease.Future advances in effective therapeutic targeting of cartilage loss during osteoarthritic diseases of the joint as an organ as well as in cartilage tissue engineering would benefit from 'big data' approaches and bioinformatics, to uncover novel feed-forward and feed-back mechanisms for regulating transcription and translation of genes and their integration into cell-specific pathways.
Collapse
Affiliation(s)
- Anna H K Plaas
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, Chicago, IL, USA
| | - Meghan M Moran
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - John D Sandy
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, The Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
9
|
Kim B, Bonassar LJ. Understanding the Influence of Local Physical Stimuli on Chondrocyte Behavior. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:31-44. [PMID: 37052844 DOI: 10.1007/978-3-031-25588-5_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Investigating the mechanobiology of chondrocytes is challenging due to the complex micromechanical environment of cartilage tissue. The innate zonal differences and poroelastic properties of the tissue combined with its heterogeneous composition create spatial- and temporal-dependent cell behavior, which further complicates the investigation. Despite the numerous challenges, understanding the mechanobiology of chondrocytes is crucial for developing strategies for treating cartilage related diseases as chondrocytes are the only cell type within the tissue. The effort to understand chondrocyte behavior under various mechanical stimuli has been ongoing over the last 50 years. Early studies examined global biosynthetic behavior under unidirectional mechanical stimulus. With the technological development in high-speed confocal imaging techniques, recent studies have focused on investigating real-time individual and collective cell responses to multiple / combined modes of mechanical stimuli. Such efforts have led to tremendous advances in understanding the influence of local physical stimuli on chondrocyte behavior. In addition, we highlight the wide variety of experimental techniques, spanning from static to impact loading, and analysis techniques, from biochemical assays to machine learning, that have been utilized to study chondrocyte behavior. Finally, we review the progression of hypotheses about chondrocyte mechanobiology and provide a perspective on the future outlook of chondrocyte mechanobiology.
Collapse
Affiliation(s)
- Byumsu Kim
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Lawrence J Bonassar
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
10
|
Xu W, Zhu J, Hu J, Xiao L. Engineering the biomechanical microenvironment of chondrocytes towards articular cartilage tissue engineering. Life Sci 2022; 309:121043. [DOI: 10.1016/j.lfs.2022.121043] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/24/2022] [Accepted: 10/02/2022] [Indexed: 11/28/2022]
|
11
|
Cacciamali A, Villa R, Dotti S. 3D Cell Cultures: Evolution of an Ancient Tool for New Applications. Front Physiol 2022; 13:836480. [PMID: 35936888 PMCID: PMC9353320 DOI: 10.3389/fphys.2022.836480] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Recently, research is undergoing a drastic change in the application of the animal model as a unique investigation strategy, considering an alternative approach for the development of science for the future. Although conventional monolayer cell cultures represent an established and widely used in vitro method, the lack of tissue architecture and the complexity of such a model fails to inform true biological processes in vivo. Recent advances in cell culture techniques have revolutionized in vitro culture tools for biomedical research by creating powerful three-dimensional (3D) models to recapitulate cell heterogeneity, structure and functions of primary tissues. These models also bridge the gap between traditional two-dimensional (2D) single-layer cultures and animal models. 3D culture systems allow researchers to recreate human organs and diseases in one dish and thus holds great promise for many applications such as regenerative medicine, drug discovery, precision medicine, and cancer research, and gene expression studies. Bioengineering has made an important contribution in the context of 3D systems using scaffolds that help mimic the microenvironments in which cells naturally reside, supporting the mechanical, physical and biochemical requirements for cellular growth and function. We therefore speak of models based on organoids, bioreactors, organ-on-a-chip up to bioprinting and each of these systems provides its own advantages and applications. All of these techniques prove to be excellent candidates for the development of alternative methods for animal testing, as well as revolutionizing cell culture technology. 3D systems will therefore be able to provide new ideas for the study of cellular interactions both in basic and more specialized research, in compliance with the 3R principle. In this review, we provide a comparison of 2D cell culture with 3D cell culture, provide details of some of the different 3D culture techniques currently available by discussing their strengths as well as their potential applications.
Collapse
Affiliation(s)
| | | | - Silvia Dotti
- *Correspondence: Andrea Cacciamali, ; Silvia Dotti,
| |
Collapse
|
12
|
Alves-Simões M. Rodent models of knee osteoarthritis for pain research. Osteoarthritis Cartilage 2022; 30:802-814. [PMID: 35139423 DOI: 10.1016/j.joca.2022.01.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease and a leading cause of disability worldwide. Pain is the main symptom, yet no current treatment can halt disease progression or effectively provide symptomatic relief. Numerous animal models have been described for studying OA and some for the associated OA pain. This review aims to update on current models used for studying OA pain, focusing on mice and rats. These models include surgical, chemical, mechanical, and spontaneous OA models. The impact of sex and age will also be addressed in the context of OA modelling. Although no single animal model has been shown ideal for studying OA pain, increased efforts to phenotype OA will likely impact the choice of models for pre-clinical and basic research studies.
Collapse
Affiliation(s)
- M Alves-Simões
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK.
| |
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
|
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
|
15
|
Mechanical Cues: Bidirectional Reciprocity in the Extracellular Matrix Drives Mechano-Signalling in Articular Cartilage. Int J Mol Sci 2021; 22:ijms222413595. [PMID: 34948394 PMCID: PMC8707858 DOI: 10.3390/ijms222413595] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 12/29/2022] Open
Abstract
The composition and organisation of the extracellular matrix (ECM), particularly the pericellular matrix (PCM), in articular cartilage is critical to its biomechanical functionality; the presence of proteoglycans such as aggrecan, entrapped within a type II collagen fibrillar network, confers mechanical resilience underweight-bearing. Furthermore, components of the PCM including type VI collagen, perlecan, small leucine-rich proteoglycans—decorin and biglycan—and fibronectin facilitate the transduction of both biomechanical and biochemical signals to the residing chondrocytes, thereby regulating the process of mechanotransduction in cartilage. In this review, we summarise the literature reporting on the bidirectional reciprocity of the ECM in chondrocyte mechano-signalling and articular cartilage homeostasis. Specifically, we discuss studies that have characterised the response of articular cartilage to mechanical perturbations in the local tissue environment and how the magnitude or type of loading applied elicits cellular behaviours to effect change. In vivo, including transgenic approaches, and in vitro studies have illustrated how physiological loading maintains a homeostatic balance of anabolic and catabolic activities, involving the direct engagement of many PCM molecules in orchestrating this slow but consistent turnover of the cartilage matrix. Furthermore, we document studies characterising how abnormal, non-physiological loading including excessive loading or joint trauma negatively impacts matrix molecule biosynthesis and/or organisation, affecting PCM mechanical properties and reducing the tissue’s ability to withstand load. We present compelling evidence showing that reciprocal engagement of the cells with this altered ECM environment can thus impact tissue homeostasis and, if sustained, can result in cartilage degradation and onset of osteoarthritis pathology. Enhanced dysregulation of PCM/ECM turnover is partially driven by mechanically mediated proteolytic degradation of cartilage ECM components. This generates bioactive breakdown fragments such as fibronectin, biglycan and lumican fragments, which can subsequently activate or inhibit additional signalling pathways including those involved in inflammation. Finally, we discuss how bidirectionality within the ECM is critically important in enabling the chondrocytes to synthesise and release PCM/ECM molecules, growth factors, pro-inflammatory cytokines and proteolytic enzymes, under a specified load, to influence PCM/ECM composition and mechanical properties in cartilage health and disease.
Collapse
|
16
|
Abusharkh HA, Reynolds OM, Mendenhall J, Gozen BA, Tingstad E, Idone V, Abu-Lail NI, Van Wie BJ. Combining stretching and gallic acid to decrease inflammation indices and promote extracellular matrix production in osteoarthritic human articular chondrocytes. Exp Cell Res 2021; 408:112841. [PMID: 34563516 DOI: 10.1016/j.yexcr.2021.112841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/21/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
Osteoarthritis (OA) patients undergo cartilage degradation and experience painful joint swelling. OA symptoms are caused by inflammatory molecules and the upregulation of catabolic genes leading to the breakdown of cartilage extracellular matrix (ECM). Here, we investigate the effects of gallic acid (GA) and mechanical stretching on the expression of anabolic and catabolic genes and restoring ECM production by osteoarthritic human articular chondrocytes (hAChs) cultured in monolayers. hAChs were seeded onto conventional plates or silicone chambers with or without 100 μM GA. A 5% cyclic tensile strain (CTS) was applied to the silicone chambers and the deposition of collagen and glycosaminoglycan, and gene expressions of collagen types II (COL2A1), XI (COL11A2), I (COL1A1), and X (COL10A1), and matrix metalloproteinases (MMP-1 and MMP-13) as inflammation markers, were quantified. CTS and GA acted synergistically to promote the deposition of collagen and glycosaminoglycan in the ECM by 14- and 7-fold, respectively. Furthermore, the synergistic stimuli selectively upregulated the expression of cartilage-specific proteins, COL11A2 by 7-fold, and COL2A1 by 47-fold, and, in contrast, downregulated the expression of MMP-1 by 2.5-fold and MMP-13 by 125-fold. GA supplementation with CTS is a promising approach for restoring osteoarthritic hAChs ECM production ability making them suitable for complex tissue engineering applications.
Collapse
Affiliation(s)
- Haneen A Abusharkh
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164-6515, USA.
| | - Olivia M Reynolds
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164-6515, USA.
| | - Juana Mendenhall
- Department of Chemistry, Morehouse College, Atlanta, GA, 30314, USA.
| | - Bulent A Gozen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164-2920, USA.
| | - Edwin Tingstad
- Inland Orthopedic Surgery and Sports Medicine Clinic, Pullman, WA, 99163, USA.
| | - Vincent Idone
- Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA.
| | - Nehal I Abu-Lail
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, 78249-3209, USA.
| | - Bernard J Van Wie
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, 99164-6515, USA.
| |
Collapse
|
17
|
Elahi SA, Tanska P, Mukherjee S, Korhonen RK, Geris L, Jonkers I, Famaey N. Guide to mechanical characterization of articular cartilage and hydrogel constructs based on a systematic in silico parameter sensitivity analysis. J Mech Behav Biomed Mater 2021; 124:104795. [PMID: 34488174 DOI: 10.1016/j.jmbbm.2021.104795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/07/2021] [Accepted: 08/21/2021] [Indexed: 10/20/2022]
Abstract
Osteoarthritis is a whole joint disease with cartilage degeneration being an important manifestation. Tissue engineering treatment is a solution for repairing cartilage defects by implantation of chondrocyte-laden hydrogel constructs within the defect. In silico models have recently been introduced to simulate and optimize the design of these constructs. These models require accurate knowledge on the mechanical properties of the hydrogel constructs and cartilage explants, which are challenging to obtain due to their anisotropic structure and time-dependent behaviour. We performed a systematic in silico parameter sensitivity analysis to find the most efficient unconfined compression testing protocols for mechanical characterization of hydrogel constructs and cartilage explants, with a minimum number of tests but maximum identifiability of the material parameters. The construct and explant were thereby modelled as porohyperelastic and fibril-reinforced poroelastic materials, respectively. Three commonly used loading regimes were simulated in Abaqus (ramp, relaxation and dynamic loading) with varying compressive strain magnitudes and rates. From these virtual experiments, the resulting material parameters were obtained for each combination using a numerical inverse identification scheme. For hydrogels, maximum sensitivity to the different material parameters was found when using a single step ramp loading (20% compression with 10%/s rate) followed by 15 min relaxation. For cartilage explants, a two-stepped ramp loading (10% compression with 10%/s rate and 10% compression with 1%/s rate), each step followed by 15 min relaxation, yielded the maximum sensitivity to the different material parameters. With these protocols, the material parameters could be retrieved with the lowest amount of uncertainty (hydrogel: < 2% and cartilage: < 6%). These specific results and the overall methodology can be used to optimize mechanical testing protocols to yield reliable material parameters for in silico models of cartilage and hydrogel constructs.
Collapse
Affiliation(s)
- Seyed Ali Elahi
- Human Movement Biomechanics Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium; Soft Tissue Biomechanics Group, Biomechanics Division, Mechanical Engineering Department, KU Leuven, Leuven, Belgium.
| | - Petri Tanska
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Satanik Mukherjee
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium; Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Rami K Korhonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium; Biomechanics Section, KU Leuven, Leuven, Belgium; GIGA in Silico Medicine, University of Liège, Liège, Belgium
| | - Ilse Jonkers
- Human Movement Biomechanics Research Group, Department of Movement Sciences, KU Leuven, Leuven, Belgium
| | - Nele Famaey
- Soft Tissue Biomechanics Group, Biomechanics Division, Mechanical Engineering Department, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Pedrini F, Hausen MA, Duek EAR. Optimized Method to Improve Cell Activity in 3D Scaffolds Under a Dual Real-Time Dynamic Bioreactor System. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2436:127-134. [PMID: 34081312 DOI: 10.1007/7651_2021_410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Bioreactor systems that allow the simulation of in vivo variables in a controlled in vitro environment, were a great advance in the field of tissue engineering. Due to the dynamic-mechanical features that some tissues present, 3D-engineered constructs often do not exhibit the biomechanical properties of these native tissues. Thus, a successful approach must not only achieve tissue repair but also restore its function after injury. Here, we describe a method to improve cell activity in 3D scaffolds in a dynamic bioreactor system through the application of mechanical compression and fluid flow for tissue engineering approaches.
Collapse
Affiliation(s)
- Flavia Pedrini
- Postgraduate Program in Biotechnology and Environmental Monitoring, Federal University of São Carlos (UFSCar), Sorocaba, Brazil.
| | - Moema A Hausen
- Surgery Department, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC/SP), Sorocaba, Brazil
| | - Eliana A R Duek
- Postgraduate Program in Biotechnology and Environmental Monitoring, Federal University of São Carlos (UFSCar), Sorocaba, Brazil.,Surgery Department, Faculty of Medical Sciences and Health, Pontifical Catholic University of São Paulo (PUC/SP), Sorocaba, Brazil
| |
Collapse
|
19
|
Statham P, Jones E, Jennings LM, Fermor HL. Reproducing the Biomechanical Environment of the Chondrocyte for Cartilage Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:405-420. [PMID: 33726527 DOI: 10.1089/ten.teb.2020.0373] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
It is well known that the biomechanical and tribological performance of articular cartilage is inextricably linked to its extracellular matrix (ECM) structure and zonal heterogeneity. Furthermore, it is understood that the presence of native ECM components, such as collagen II and aggrecan, promote healthy homeostasis in the resident chondrocytes. What is less frequently discussed is how chondrocyte metabolism is related to the extracellular mechanical environment, at both the macro and microscale. The chondrocyte is in immediate contact with the pericellular matrix of the chondron, which acts as a mechanocoupler, transmitting external applied loads from the ECM to the chondrocyte. Therefore, components of the pericellular matrix also play essential roles in chondrocyte mechanotransduction and metabolism. Recreating the biomechanical environment through tuning material properties of a scaffold and/or the use of external cyclic loading can induce biosynthetic responses in chondrocytes. Decellularized scaffolds, which retain the native tissue macro- and microstructure also represent an effective means of recapitulating such an environment. The use of such techniques in tissue engineering applications can ensure the regeneration of skeletally mature articular cartilage with appropriate biomechanical and tribological properties to restore joint function. Despite the pivotal role in graft maturation and performance, biomechanical and tribological properties of such interventions is often underrepresented. This review outlines the role of biomechanics in relation to native cartilage performance and chondrocyte metabolism, and how application of this theory can enhance the future development and successful translation of biomechanically relevant tissue engineering interventions. Impact statement Physiological cartilage function is a key criterion in the success of a cartilage tissue engineering solution. The in situ performance is dependent on the initial scaffold design as well as extracellular matrix deposition by endogenous or exogenous cells. Both biological and biomechanical stimuli serve as key regulators of cartilage homeostasis and maturation of the resulting tissue-engineered graft. An improved understanding of the influence of biomechanics on cellular function and consideration of the final biomechanical and tribological performance will help in the successful development and translation of tissue-engineered grafts to restore natural joint function postcartilage trauma or osteoarthritic degeneration, delaying the requirement for prosthetic intervention.
Collapse
Affiliation(s)
- Patrick Statham
- Institute of Medical and Biological Engineering, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds, United Kingdom
| | - Louise M Jennings
- Institute of Medical and Biological Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, United Kingdom
| | - Hazel L Fermor
- Institute of Medical and Biological Engineering, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
20
|
Mills DK, Luo Y, Elumalai A, Esteve S, Karnik S, Yao S. Creating Structured Hydrogel Microenvironments for Regulating Stem Cell Differentiation. Gels 2020; 6:gels6040047. [PMID: 33276682 PMCID: PMC7768466 DOI: 10.3390/gels6040047] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
The development of distinct biomimetic microenvironments for regulating stem cell behavior and bioengineering human tissues and disease models requires a solid understanding of cell-substrate interactions, adhesion, and its role in directing cell behavior, and other physico-chemical cues that drive cell behavior. In the past decade, innovative developments in chemistry, materials science, microfabrication, and associated technologies have given us the ability to manipulate the stem cell microenvironment with greater precision and, further, to monitor effector impacts on stem cells, both spatially and temporally. The influence of biomaterials and the 3D microenvironment's physical and biochemical properties on mesenchymal stem cell proliferation, differentiation, and matrix production are the focus of this review chapter. Mechanisms and materials, principally hydrogel and hydrogel composites for bone and cartilage repair that create "cell-supportive" and "instructive" biomaterials, are emphasized. We begin by providing an overview of stem cells, their unique properties, and their challenges in regenerative medicine. An overview of current fabrication strategies for creating instructive substrates is then reviewed with a focused discussion of selected fabrication methods with an emphasis on bioprinting as a critical tool in creating novel stem cell-based biomaterials. We conclude with a critical assessment of the current state of the field and offer our view on the promises and potential pitfalls of the approaches discussed.
Collapse
Affiliation(s)
- David K. Mills
- School of Biological Sciences, Louisiana Tech University, Ruston, LA 71270, USA;
- Center for Biomedical Engineering and Rehabilitation Science, Louisiana Tech University, Ruston, LA 71270, USA;
- Correspondence:
| | - Yangyang Luo
- Molecular Sciences and Nanotechnology, Louisiana Tech University, Ruston, LA 71270, USA;
| | - Anusha Elumalai
- School of Biological Sciences, Louisiana Tech University, Ruston, LA 71270, USA;
- Center for Biomedical Engineering and Rehabilitation Science, Louisiana Tech University, Ruston, LA 71270, USA;
| | - Savannah Esteve
- Center for Biomedical Engineering and Rehabilitation Science, Louisiana Tech University, Ruston, LA 71270, USA;
| | - Sonali Karnik
- Department of Mechanical and Energy Engineering, IUPUI, Indianapolis, IN 46202, USA;
| | - Shaomian Yao
- Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803, USA;
| |
Collapse
|
21
|
Ding X, Zhao H, Li Y, Lee AL, Li Z, Fu M, Li C, Yang YY, Yuan P. Synthetic peptide hydrogels as 3D scaffolds for tissue engineering. Adv Drug Deliv Rev 2020; 160:78-104. [PMID: 33091503 DOI: 10.1016/j.addr.2020.10.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
The regeneration of tissues and organs poses an immense challenge due to the extreme complexity in the research work involved. Despite the tissue engineering approach being considered as a promising strategy for more than two decades, a key issue impeding its progress is the lack of ideal scaffold materials. Nature-inspired synthetic peptide hydrogels are inherently biocompatible, and its high resemblance to extracellular matrix makes peptide hydrogels suitable 3D scaffold materials. This review covers the important aspects of peptide hydrogels as 3D scaffolds, including mechanical properties, biodegradability and bioactivity, and the current approaches in creating matrices with optimized features. Many of these scaffolds contain peptide sequences that are widely reported for tissue repair and regeneration and these peptide sequences will also be discussed. Furthermore, 3D biofabrication strategies of synthetic peptide hydrogels and the recent advances of peptide hydrogels in tissue engineering will also be described to reflect the current trend in the field. In the final section, we will present the future outlook in the design and development of peptide-based hydrogels for translational tissue engineering applications.
Collapse
Affiliation(s)
- Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Huimin Zhao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Ashlynn Lingzhi Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Zongshao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Mengjing Fu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Chengnan Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
22
|
Di Federico E, Bader DL, Shelton JC. 3D models of chondrocytes within biomimetic scaffolds: Effects of cell deformation from loading regimens. Clin Biomech (Bristol, Avon) 2020; 79:104972. [PMID: 32093973 DOI: 10.1016/j.clinbiomech.2020.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 11/26/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mechanical conditioning has been widely used to attempt to enhance chondrocyte metabolism for the evolution of functionally competent cartilage. However, although upregulation of proteoglycans have been reported through the application of uniaxial compression, minimal collagen has been produced. The study is designed to examine whether alternative loading regimens, equivalent to physiological conditions, involving shear in addition to compression can enhance collagen production. METHODS Finite element models were developed to determine how the local chondrocyte environments within agarose constructs were influenced by a range of static and dynamic loading regimens. 3-D poro-viscoelastic models were validated against experimental data. In particular, these models were used to characterise chondrocyte deformation in compression with and without shear superimposed, with special reference to the formation of pericellular matrix around the cells. FINDINGS The models of the hydrogel constructs under stress relaxation and dynamic cyclic compression conditions were highly correlated with the experimental data. The cell deformation (y/z) in the constructs was greatest in the centre of the constructs, increasing with magnitude of compression up to 25%. The superposition of shear however did not produce significant additional changes in deformation, with the presence of PCM reducing the chondrocyte deformation. INTERPRETATION The use of FE models can prove important in the definition of appropriate, optimised mechanical conditioning regimens for the synthesis and organisation of mature extra cellular matrix by chondrocyte-seeded constructs. They will also provide insight into the mechanisms relating cell deformation to mechanotransduction pathways, thereby progressing the development of functionally competent tissue engineered cartilage.
Collapse
Affiliation(s)
- Erica Di Federico
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Dan L Bader
- Faculty of Health Sciences, University of Southampton, UK
| | - Julia C Shelton
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK.
| |
Collapse
|
23
|
Komeili A, Otoo BS, Abusara Z, Sibole S, Federico S, Herzog W. Chondrocyte Deformations Under Mild Dynamic Loading Conditions. Ann Biomed Eng 2020; 49:846-857. [PMID: 32959133 DOI: 10.1007/s10439-020-02615-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
Dynamic deformation of chondrocytes are associated with cell mechanotransduction and thus may offer a new understanding of the mechanobiology of articular cartilage. Despite extensive research on chondrocyte deformations for static conditions, work for dynamic conditions remains rare. However, it is these dynamic conditions that articular cartilage in joints are exposed to everyday, and that seem to promote biological signaling in chondrocytes. Therefore, the objective of this study was to develop an experimental technique to determine the in situ deformations of chondrocytes when the cartilage is dynamically compressed. We hypothesized that dynamic deformations of chondrocytes vastly differ from those observed under steady-state static strain conditions. Real-time chondrocyte geometry was reconstructed at 10, 15, and 20% compression during ramp compressions with 20% ultimate strain, applied at a strain rate of 0.2% s-1, followed by stress relaxation. Dynamic compressive chondrocyte deformations were non-linear as a function of nominal strain, with large deformations in the early and small deformations in the late part of compression. Early compression (up to about 10%) was associated with chondrocyte volume loss, while late compression (> ~ 10%) was associated with cell deformation but minimal volume loss. Force continued to decrease for 5 min in the stress-relaxation phase, while chondrocyte shape/volume remained unaltered after the first minute of stress-relaxation.
Collapse
Affiliation(s)
- Amin Komeili
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.,School of Engineering, University of Guelph, 50 Stone Rd E, Guelph, N1G 2W1, ON, Canada
| | - Baaba Sekyiwaa Otoo
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Ziad Abusara
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.,Advanced Imaging and Histopathology Core, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, P.O. Box 34110, Doha, Qatar
| | - Scott Sibole
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Salvatore Federico
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.,Department of Mechanical and Manufacturing Engineering, The University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada. .,Biomechanics Laboratory, School of Sports, Federal University of Santa Catarina, Florianopolis, SC, Brazil.
| |
Collapse
|
24
|
Schneider MC, Lalitha Sridhar S, Vernerey FJ, Bryant SJ. Spatiotemporal neocartilage growth in matrix-metalloproteinase-sensitive poly(ethylene glycol) hydrogels under dynamic compressive loading: an experimental and computational approach. J Mater Chem B 2020; 8:2775-2791. [PMID: 32155233 PMCID: PMC7695218 DOI: 10.1039/c9tb02963j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Enzyme-sensitive hydrogels containing encapsulated chondrocytes are a promising platform for cartilage tissue engineering. However, the growth of neotissue is closely coupled to the degradation of the hydrogel and is further complicated due to the encapsulated cells serving as the enzyme source for hydrogel degradation. To better understand these coupled processes, this study combined experimental and computational methods to analyze the transition from hydrogel to neotissue in a biomimetic MMP-sensitive poly(ethylene glycol) (PEG) hydrogel with encapsulated chondrocytes. A physics-based computational model that describes spatial heterogeneities in cell distribution was used. Experimentally, cell-laden hydrogels were cultured for six weeks under free swelling or subjected daily to one-hour of dynamic compressive loading. Extracellular matrix (ECM) synthesis rates were used as model inputs, and the model was fit to the experimentally determined construct modulus over time for the free swelling condition. Experimentally, ECM accumulation comprising collagen II and aggrecan increased over time concomitant with hydrogel degradation observed by a loss in PEG. Simulations demonstrated rapid degradation in regions of high cell density (i.e., cell clusters) reaching complete degradation by day 13, which facilitated localized ECM growth. Regions of low cell density degraded more slowly, had limited ECM, and led to the decrease in construct modulus during the first two weeks. The primary difference between the two culture environments was greater ECM accumulation in the clusters under free swelling, which facilitated a faster recovery in construct modulus. By 6 weeks the compressive modulus increased 2.5-fold to 107 kPa under free swelling, but dropped 1.6-fold to 26 kPa under loading. In summary, this biomimetic MMP-sensitive hydrogel supports neocartilage growth by facilitating rapid ECM growth within cell clusters, which was followed by slower growth in the rest of the hydrogel. Subtle temporal differences in hydrogel degradation and ECM accumulation, however, had a significant impact on the evolving mechanical properties.
Collapse
Affiliation(s)
- Margaret C Schneider
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave., Boulder, Colorado 80309-0596, USA.
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado, 1111 Engineering Dr., Boulder, Colorado 80309-0596, USA.
| | - Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado, 1111 Engineering Dr., Boulder, Colorado 80309-0596, USA. and Materials Science and Engineering Program, University of Colorado, 3415 Colorado Ave., Boulder, Colorado 80309-0596, USA
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave., Boulder, Colorado 80309-0596, USA. and Materials Science and Engineering Program, University of Colorado, 3415 Colorado Ave., Boulder, Colorado 80309-0596, USA and Biofrontiers Institute, University of Colorado, 3415 Colorado Ave., Boulder, Colorado 80309-0596, USA
| |
Collapse
|
25
|
Lee S, Trinh TH, Yoo M, Shin J, Lee H, Kim J, Hwang E, Lim YB, Ryou C. Self-Assembling Peptides and Their Application in the Treatment of Diseases. Int J Mol Sci 2019; 20:E5850. [PMID: 31766475 PMCID: PMC6928719 DOI: 10.3390/ijms20235850] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
Self-assembling peptides are biomedical materials with unique structures that are formed in response to various environmental conditions. Governed by their physicochemical characteristics, the peptides can form a variety of structures with greater reactivity than conventional non-biological materials. The structural divergence of self-assembling peptides allows for various functional possibilities; when assembled, they can be used as scaffolds for cell and tissue regeneration, and vehicles for drug delivery, conferring controlled release, stability, and targeting, and avoiding side effects of drugs. These peptides can also be used as drugs themselves. In this review, we describe the basic structure and characteristics of self-assembling peptides and the various factors that affect the formation of peptide-based structures. We also summarize the applications of self-assembling peptides in the treatment of various diseases, including cancer. Furthermore, the in-cell self-assembly of peptides, termed reverse self-assembly, is discussed as a novel paradigm for self-assembling peptide-based nanovehicles and nanomedicines.
Collapse
Affiliation(s)
- Sungeun Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Trang H.T. Trinh
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Miryeong Yoo
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Junwu Shin
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Hakmin Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Jaehyeon Kim
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Euimin Hwang
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Korea; (E.H.); (Y.-b.L.)
| | - Yong-beom Lim
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Korea; (E.H.); (Y.-b.L.)
| | - Chongsuk Ryou
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| |
Collapse
|
26
|
Effect of strain rate on transient local strain variations in articular cartilage. J Mech Behav Biomed Mater 2019; 95:60-66. [DOI: 10.1016/j.jmbbm.2019.03.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/06/2019] [Accepted: 03/20/2019] [Indexed: 11/18/2022]
|
27
|
Vaca-González JJ, Guevara JM, Moncayo MA, Castro-Abril H, Hata Y, Garzón-Alvarado DA. Biophysical Stimuli: A Review of Electrical and Mechanical Stimulation in Hyaline Cartilage. Cartilage 2019; 10:157-172. [PMID: 28933195 PMCID: PMC6425540 DOI: 10.1177/1947603517730637] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Hyaline cartilage degenerative pathologies induce morphologic and biomechanical changes resulting in cartilage tissue damage. In pursuit of therapeutic options, electrical and mechanical stimulation have been proposed for improving tissue engineering approaches for cartilage repair. The purpose of this review was to highlight the effect of electrical stimulation and mechanical stimuli in chondrocyte behavior. DESIGN Different information sources and the MEDLINE database were systematically revised to summarize the different contributions for the past 40 years. RESULTS It has been shown that electric stimulation may increase cell proliferation and stimulate the synthesis of molecules associated with the extracellular matrix of the articular cartilage, such as collagen type II, aggrecan and glycosaminoglycans, while mechanical loads trigger anabolic and catabolic responses in chondrocytes. CONCLUSION The biophysical stimuli can increase cell proliferation and stimulate molecules associated with hyaline cartilage extracellular matrix maintenance.
Collapse
Affiliation(s)
- Juan J. Vaca-González
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Johana M. Guevara
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogota, Colombia
| | - Miguel A. Moncayo
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Hector Castro-Abril
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Yoshie Hata
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
| | - Diego A. Garzón-Alvarado
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| |
Collapse
|
28
|
Dufour A, Buffier M, Vertu-Ciolino D, Disant F, Mallein-Gerin F, Perrier-Groult E. Combination of bioactive factors and IEIK13 self-assembling peptide hydrogel promotes cartilage matrix production by human nasal chondrocytes. J Biomed Mater Res A 2019; 107:893-903. [PMID: 30650239 DOI: 10.1002/jbm.a.36612] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/17/2018] [Accepted: 01/09/2019] [Indexed: 02/01/2023]
Abstract
Nasal reconstruction remains a challenge for every reconstructive surgeon. Alloplastic implants are proposed to repair nasal cartilaginous defects but they are often associated with high rates of extrusion and infection and poor biocompatibility. In this context, a porous polymeric scaffold filled with an autologous cartilage gel would be advantageous. In this study, we evaluated the capacity of IEIK13 self-assembling peptide (SAP) to serve as support to form such cartilage gel. Human nasal chondrocytes (HNC) were first amplified with FGF-2 and insulin, and then redifferentiated in IEIK13 with BMP-2, insulin, and T3 (BIT). Our results demonstrate that IEIK13 fosters HNC growth and survival. HNC phenotype was assessed by RT-PCR analysis and neo-synthesized extracellular matrix was characterized by western blotting and immunohistochemistry analysis. BIT-treated cells embedded in IEIK13 displayed round morphology and expressed cartilage-specific markers such as type II and type IX collagens and aggrecan. In addition, we did not detect significant production of type I and type X collagens and gene products of dedifferentiated and hypertrophic chondrocytes that are unwanted in hyaline cartilage. The whole of these results indicates that the SAP IEIK13 represents a suitable support for hydrogel-based tissue engineering of nasal cartilage. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 893-903, 2019.
Collapse
Affiliation(s)
- Alexandre Dufour
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS UMR 5305, Institute for Biology and Chemistry of Proteins, Lyon, France
| | | | - Delphine Vertu-Ciolino
- Department of otolaryngology-head and neck surgery, Édouard-Herriot hospital, Lyon, France
| | - François Disant
- Department of otolaryngology-head and neck surgery, Édouard-Herriot hospital, Lyon, France
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS UMR 5305, Institute for Biology and Chemistry of Proteins, Lyon, France
| | - Emeline Perrier-Groult
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS UMR 5305, Institute for Biology and Chemistry of Proteins, Lyon, France
| |
Collapse
|
29
|
Graceffa V, Vinatier C, Guicheux J, Stoddart M, Alini M, Zeugolis DI. Chasing Chimeras - The elusive stable chondrogenic phenotype. Biomaterials 2018; 192:199-225. [PMID: 30453216 DOI: 10.1016/j.biomaterials.2018.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/27/2022]
Abstract
The choice of the best-suited cell population for the regeneration of damaged or diseased cartilage depends on the effectiveness of culture conditions (e.g. media supplements, three-dimensional scaffolds, mechanical stimulation, oxygen tension, co-culture systems) to induce stable chondrogenic phenotype. Herein, advances and shortfalls in in vitro, preclinical and clinical setting of various in vitro microenvironment modulators on maintaining chondrocyte phenotype or directing stem cells towards chondrogenic lineage are critically discussed. Chondrocytes possess low isolation efficiency, limited proliferative potential and rapid phenotypic drift in culture. Mesenchymal stem cells are relatively readily available, possess high proliferation potential, exhibit great chondrogenic differentiation capacity, but they tend to acquire a hypertrophic phenotype when exposed to chondrogenic stimuli. Embryonic and induced pluripotent stem cells, despite their promising in vitro and preclinical data, are still under-investigated. Although a stable chondrogenic phenotype remains elusive, recent advances in in vitro microenvironment modulators are likely to develop clinically- and commercially-relevant therapies in the years to come.
Collapse
Affiliation(s)
- Valeria Graceffa
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Claire Vinatier
- INSERMU1229, Regenerative Medicine and Skeleton (RMeS), University of Nantes, UFR Odontologie & CHU Nantes, PHU 4 OTONN, 44042 Nantes, France
| | - Jerome Guicheux
- INSERMU1229, Regenerative Medicine and Skeleton (RMeS), University of Nantes, UFR Odontologie & CHU Nantes, PHU 4 OTONN, 44042 Nantes, France
| | - Martin Stoddart
- AO Research Institute, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Mauro Alini
- AO Research Institute, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
30
|
Edd SN, Omoumi P, Andriacchi TP, Jolles BM, Favre J. Modeling knee osteoarthritis pathophysiology using an integrated joint system (IJS): a systematic review of relationships among cartilage thickness, gait mechanics, and subchondral bone mineral density. Osteoarthritis Cartilage 2018; 26:1425-1437. [PMID: 30056214 DOI: 10.1016/j.joca.2018.06.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/08/2018] [Accepted: 06/26/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To introduce an integrated joint system (IJS) model of joint health and osteoarthritis (OA) pathophysiology through a systematic review of the cross-sectional relationships among three knee properties (cartilage thickness, gait mechanics, and subchondral bone mineral density). METHODS Searches using keywords associated with the three knee properties of interest were performed in PubMed, Scopus, and Ovid databases. English-language articles reporting cross-sectional correlations between at least two knee properties in healthy or tibiofemoral OA human knees were included. A narrative synthesis of the data was conducted. RESULTS Of the 5600 retrieved articles, 13 were included, eight of which reported relationships between cartilage thickness and gait mechanics. The 744 tested knees were separated into three categories based on knee health: 199 healthy, 340 at-risk/early OA, and 205 late OA knees. Correlations between knee adduction moment and medial-to-lateral cartilage thickness ratios were generally positive in healthy, inconclusive in at-risk/early OA, and negative in late OA knees. Knee adduction moment was positively correlated with medial-to-lateral tibial subchondral bone mineral density ratios in knees of all health categories. One study reported a positive correlation between lateral tibial subchondral bone mineral density and femoral cartilage thickness in at-risk/early OA knees. CONCLUSIONS The correlations identified between knee properties in this review agreed with the proposed relationship-based IJS model of OA pathophysiology. Accordingly, the IJS model could provide insights into overcoming current barriers to developing disease-modifying treatments by considering multiple aspects of OA disease, aspects that could be assessed simultaneously at an in vivo system level.
Collapse
Affiliation(s)
- S N Edd
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), Department of Musculoskeletal Medicine (DAL), Swiss BioMotion Lab, Lausanne, Switzerland.
| | - P Omoumi
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), Department of Diagnostic and Interventional Radiology, Lausanne, Switzerland.
| | - T P Andriacchi
- Department of Mechanical Engineering, Stanford, CA, USA; Palo Alto Veterans Affairs, Palo Alto, CA, USA; Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, CA, USA.
| | - B M Jolles
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), Department of Musculoskeletal Medicine (DAL), Swiss BioMotion Lab, Lausanne, Switzerland; Ecole Polytechnique Fédérale de Lausanne (EPFL), Institute of Microengineering, Lausanne, Switzerland.
| | - J Favre
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), Department of Musculoskeletal Medicine (DAL), Swiss BioMotion Lab, Lausanne, Switzerland.
| |
Collapse
|
31
|
Komeili A, Abusara Z, Federico S, Herzog W. A compression system for studying depth-dependent mechanical properties of articular cartilage under dynamic loading conditions. Med Eng Phys 2018; 60:103-108. [PMID: 30061065 DOI: 10.1016/j.medengphy.2018.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 06/30/2018] [Accepted: 07/15/2018] [Indexed: 10/28/2022]
Abstract
The biological activities of chondrocytes are influenced by the mechanical characteristics of their environment. The overall real-time mechanical response of cartilage has been investigated earlier. However, the instantaneous local mechano-biology of cartilage has not been investigated in detail under dynamic loading conditions. In order to address this gap in the literature, we designed a compression testing device and implemented a dual photon microscopy technique with the goal of measuring local mechanical and biological responses of articular cartilage under dynamic loading conditions. The details of the compression system and results of a pilot study are presented here. A 15% ramp compression at a rate of 0.003/s with a subsequent stress relaxation phase was applied to the cartilage explant samples. The extra cellular matrix was imaged throughout the entire thickness of the cartilage sample, and local tissue strains were measured during the compression and relaxation phase. The axial compressive strains in the middle and superficial zones of cartilage were observed to increase during the relaxation phase: this was a new finding, suggesting the importance of further investigations on the real-time local behavior of cartilage. The compression system showed promising results for investigating the dynamic, real-time mechanical response of articular cartilage, and can now be used to reveal the instantaneous mechanical and biological responses of chondrocytes in response to dynamic loading conditions.
Collapse
Affiliation(s)
- Amin Komeili
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, Calgary, Canada 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Ziad Abusara
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, Calgary, Canada 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Salvatore Federico
- Department of Mechanical and Manufacturing Engineering, The University of Calgary, Calgary, Canada 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, Calgary, Canada 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, The University of Calgary, Calgary, Canada 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada.
| |
Collapse
|
32
|
Lee D, Erickson A, You T, Dudley AT, Ryu S. Pneumatic microfluidic cell compression device for high-throughput study of chondrocyte mechanobiology. LAB ON A CHIP 2018; 18:2077-2086. [PMID: 29897088 PMCID: PMC6467204 DOI: 10.1039/c8lc00320c] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Hyaline cartilage is a specialized type of connective tissue that lines many moveable joints (articular cartilage) and contributes to bone growth (growth plate cartilage). Hyaline cartilage is composed of a single cell type, the chondrocyte, which produces a unique hydrated matrix to resist compressive stress. Although compressive stress has profound effects on transcriptional networks and matrix biosynthesis in chondrocytes, mechanistic relationships between strain, signal transduction, cell metabolism, and matrix production remain superficial. Here, we describe development and validation of a polydimethylsiloxane (PDMS)-based pneumatic microfluidic cell compression device which generates multiple compression conditions in a single platform. The device contained an array of PDMS balloons of different sizes which were actuated by pressurized air, and the balloons compressed chondrocytes cells in alginate hydrogel constructs. Our characterization and testing of the device showed that the developed platform could compress chondrocytes with various magnitudes simultaneously with negligible effect on cell viability. Also, the device is compatible with live cell imaging to probe early effects of compressive stress, and it can be rapidly dismantled to facilitate molecular studies of compressive stress on transcriptional networks. Therefore, the proposed device will enhance the productivity of chondrocyte mechanobiology studies, and it can be applied to study mechanobiology of other cell types.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA.
| | | | | | | | | |
Collapse
|
33
|
Salinas EY, Hu JC, Athanasiou K. A Guide for Using Mechanical Stimulation to Enhance Tissue-Engineered Articular Cartilage Properties. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:345-358. [PMID: 29562835 DOI: 10.1089/ten.teb.2018.0006] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The use of tissue-engineered articular cartilage (TEAC) constructs has the potential to become a powerful treatment option for cartilage lesions resulting from trauma or early stages of pathology. Although fundamental tissue-engineering strategies based on the use of scaffolds, cells, and signals have been developed, techniques that lead to biomimetic AC constructs that can be translated to in vivo use are yet to be fully confirmed. Mechanical stimulation during tissue culture can be an effective strategy to enhance the mechanical, structural, and cellular properties of tissue-engineered constructs toward mimicking those of native AC. This review focuses on the use of mechanical stimulation to attain and enhance the properties of AC constructs needed to translate these implants to the clinic. In vivo, mechanical loading at maximal and supramaximal physiological levels has been shown to be detrimental to AC through the development of degenerative changes. In contrast, multiple studies have revealed that during culture, mechanical stimulation within narrow ranges of magnitude and duration can produce anisotropic, mechanically robust AC constructs with high cellular viability. Significant progress has been made in evaluating a variety of mechanical stimulation techniques on TEAC, either alone or in combination with other stimuli. These advancements include determining and optimizing efficacious loading parameters (e.g., duration and frequency) to yield improvements in construct design criteria, such as collagen II content, compressive stiffness, cell viability, and fiber organization. With the advancement of mechanical stimulation as a potent strategy in AC tissue engineering, a compendium detailing the results achievable by various stimulus regimens would be of great use for researchers in academia and industry. The objective is to list the qualitative and quantitative effects that can be attained when direct compression, hydrostatic pressure, shear, and tensile loading are used to tissue-engineer AC. Our goal is to provide a practical guide to their use and optimization of loading parameters. For each loading condition, we will also present and discuss benefits and limitations of bioreactor configurations that have been used. The intent is for this review to serve as a reference for including mechanical stimulation strategies as part of AC construct culture regimens.
Collapse
Affiliation(s)
- Evelia Y Salinas
- Biomedical Engineering Department, University of California , Irvine, California
| | - Jerry C Hu
- Biomedical Engineering Department, University of California , Irvine, California
| | - Kyriacos Athanasiou
- Biomedical Engineering Department, University of California , Irvine, California
| |
Collapse
|
34
|
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
|
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
|
Rothdiener M, Uynuk-Ool T, Südkamp N, Aurich M, Grodzinsky AJ, Kurz B, Rolauffs B. Human osteoarthritic chondrons outnumber patient- and joint-matched chondrocytes in hydrogel culture-Future application in autologous cell-based OA cartilage repair? J Tissue Eng Regen Med 2017; 12:e1206-e1220. [PMID: 28714570 DOI: 10.1002/term.2516] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 06/09/2017] [Accepted: 07/11/2017] [Indexed: 12/22/2022]
Abstract
Autologous chondrocyte implantation (ACI) is used in 34-60% for osteoarthritic (OA) cartilage defects, although ACI is neither recommended nor designed for OA. Envisioning a hydrogel-based ACI for OA that uses chondrons instead of classically used chondrocytes, we hypothesized that human OA chondrons may outperform OA chondrocytes. We compared patient- and joint surface-matched human OA chondrons with OA chondrocytes cultured for the first time in a hydrogel, using a self-assembling peptide system. We determined yield, viability, cell numbers, mRNA expression, GAPDH mRNA enzyme activity, Collagen II synthesis (CPII) and degradation (C2C), and sulfated glycosaminoglycan. Ex vivo, mRNA expression was comparable. Over time, significant differences in survival led to 3.4-fold higher OA chondron numbers in hydrogels after 2 weeks (p = .002). Significantly, more enzymatically active GAPDH protein indicated higher metabolic activity. The number of cultures that expressed mRNA for Collagen Types I and VI, COMP, aggrecan, VEGF, TGF-β1, and FGF-2 (but not Collagen Types II and X) was different, resulting in a 3.5-fold higher number of expression-positive OA chondron cultures (p < .05). Measuring CPII and C2C per hydrogel, OA chondron hydrogels synthesized more than they degraded Collagen Type II, the opposite was true for OA chondrocytes. Per cell, OA chondrons but not OA chondrocytes displayed more synthesis than degradation. Thus, OA chondrons displayed superior biosynthesis and mRNA expression of tissue engineering and phenotype-relevant genes. Moreover, human OA chondrons displayed a significant survival advantage in hydrogel culture, whose presence, drastic extent, and timescale was novel and is clinically significant. Collectively, these data highlight the high potential of human OA chondrons for OA ACI, as they would outnumber and, thus, surpass OA chondrocytes.
Collapse
Affiliation(s)
- Miriam Rothdiener
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic, Eberhard Karls University, Tuebingen, Germany
| | - Tatiana Uynuk-Ool
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic, Eberhard Karls University, Tuebingen, Germany
| | - Norbert Südkamp
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, University Medical Center Freiburg, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Matthias Aurich
- Department of Orthopaedic and Trauma Surgery, Sana Kliniken Leipziger Land, Borna, Germany.,Department of Trauma, Hand and Reconstructive Surgery, Universitätsklinikum Jena, Jena, Germany.,Department of Biochemistry, Rush Medical College, Chicago, IL, USA
| | - Alan J Grodzinsky
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bodo Kurz
- Department of Anatomy, Christian Albrechts University, Kiel, Germany
| | - Bernd Rolauffs
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, University Medical Center Freiburg, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany.,Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
38
|
Zhang Y, Guo W, Wang M, Hao C, Lu L, Gao S, Zhang X, Li X, Chen M, Li P, Jiang P, Lu S, Liu S, Guo Q. Co-culture systems-based strategies for articular cartilage tissue engineering. J Cell Physiol 2017; 233:1940-1951. [PMID: 28548713 DOI: 10.1002/jcp.26020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 01/01/2023]
Abstract
Cartilage engineering facilitates repair and regeneration of damaged cartilage using engineered tissue that restores the functional properties of the impaired joint. The seed cells used most frequently in tissue engineering, are chondrocytes and mesenchymal stem cells. Seed cells activity plays a key role in the regeneration of functional cartilage tissue. However, seed cells undergo undesirable changes after in vitro processing procedures, such as degeneration of cartilage cells and induced hypertrophy of mesenchymal stem cells, which hinder cartilage tissue engineering. Compared to monoculture, which does not mimic the in vivo cellular environment, co-culture technology provides a more realistic microenvironment in terms of various physical, chemical, and biological factors. Co-culture technology is used in cartilage tissue engineering to overcome obstacles related to the degeneration of seed cells, and shows promise for cartilage regeneration and repair. In this review, we focus first on existing co-culture systems for cartilage tissue engineering and related fields, and discuss the conditions and mechanisms thereof. This is followed by methods for optimizing seed cell co-culture conditions to generate functional neo-cartilage tissue, which will lead to a new era in cartilage tissue engineering.
Collapse
Affiliation(s)
- Yu Zhang
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, People's Republic of China
| | - Weimin Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, People's Republic of China
| | - Mingjie Wang
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, People's Republic of China
| | - Chunxiang Hao
- Institute of Anesthesia, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Liang Lu
- Anhui Provincial Hospital, Hefei, People's Republic of China
| | - Shuang Gao
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, People's Republic of China
| | - Xueliang Zhang
- Shanxi Traditional Chinese, Taiyuan, People's Republic of China
| | - Xu Li
- School of Medicine, Naikai University, Tianjin, People's Republic of China
| | - Mingxue Chen
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, People's Republic of China
| | - Penghao Li
- School of Medicine, Naikai University, Tianjin, People's Republic of China
| | - Peng Jiang
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, People's Republic of China
| | - Shibi Lu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, People's Republic of China
| | - Shuyun Liu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, People's Republic of China
| | - Quanyi Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, Beijing, People's Republic of China
| |
Collapse
|
39
|
Schneider MC, Barnes CA, Bryant SJ. Characterization of the chondrocyte secretome in photoclickable poly(ethylene glycol) hydrogels. Biotechnol Bioeng 2017; 114:2096-2108. [PMID: 28436002 DOI: 10.1002/bit.26320] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/28/2016] [Accepted: 04/17/2017] [Indexed: 12/30/2022]
Abstract
Poly(ethylene glycol) (PEG) hydrogels are highly tunable platforms that are promising cell delivery vehicles for chondrocytes and cartilage tissue engineering. In addition to characterizing the type of extracellular matrix (ECM) that forms, understanding the types of proteins that are secreted by encapsulated cells may be important. Thus, the objectives for this study were to characterize the secretome of chondrocytes encapsulated in PEG hydrogels and determine whether the secretome varies as a function of hydrogel stiffness and culture condition. Bovine chondrocytes were encapsulated in photoclickable PEG hydrogels with a compressive modulus of 8 and 46 kPa and cultured under free swelling or dynamic compressive loading conditions. Cartilage ECM deposition was assessed by biochemical assays and immunohistochemistry. The conditioned medium was analyzed by liquid chromatography-tandem mass spectrometry. Chondrocytes maintained their phenotype within the hydrogels and deposited cartilage-specific ECM that increased over time and included aggrecan and collagens II and VI. Analysis of the secretome revealed a total of 64 proteins, which were largely similar among all experimental conditions. The identified proteins have diverse functions such as biological regulation, response to stress, and collagen fibril organization. Notably, many of the proteins important to the assembly of a collagen-rich cartilage ECM were identified and included collagen types II(α1), VI (α1, α2, and α3), IX (α1), XI (α1 and α2), and biglycan. In addition, many of the other identified proteins have been reported to be present within cell-secreted exosomes. In summary, chondrocytes encapsulated within photoclickable PEG hydrogels secrete many types of proteins that diffuse out of the hydrogel and which have diverse functions, but which are largely preserved across different hydrogel culture environments. Biotechnol. Bioeng. 2017;114: 2096-2108. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Margaret C Schneider
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Campus Box 596, Boulder 80309, Colorado.,Biofrontiers Institute, University of Colorado, Boulder, Colorado
| | | | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Campus Box 596, Boulder 80309, Colorado.,Biofrontiers Institute, University of Colorado, Boulder, Colorado.,Material Science and Engineering Program, University of Colorado, Boulder, Colorado
| |
Collapse
|
40
|
Abraham SJK, Yoshioka H. Insights into in vitro environments for human cartilage tissue engineering. Indian J Med Res 2017; 144:796-798. [PMID: 28474614 PMCID: PMC5433270 DOI: 10.4103/ijmr.ijmr_1237_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Samuel J K Abraham
- Department of Surgery II, Yamanashi University-Faculty of Medicine, Chuo, Yamanashi, Japan; The Mary-Yoshio Translational Hexagon, Nichi-In Centre for Regenerative Medicine (NCRM), Chennai 600 034, Tamil Nadu, India
| | | |
Collapse
|
41
|
DiFederico E, Shelton JC, Bader DL. Complex mechanical conditioning of cell-seeded agarose constructs can influence chondrocyte biosynthetic activity. Biotechnol Bioeng 2017; 114:1614-1625. [PMID: 28240346 DOI: 10.1002/bit.26273] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/16/2017] [Accepted: 02/07/2017] [Indexed: 11/10/2022]
Abstract
Articular cartilage with its inherently poor capacity for self-regeneration represents a primary target for tissue engineering strategies, with approaches focusing on the in vitro generation of neo-cartilage using chondrocyte-seeded 3D scaffolds subjected to mechanical conditioning. Although uniaxial compression regimens have significantly up-regulated proteoglycan synthesis, their effects on the synthesis of collagen have been modest. Articular cartilage is subjected to shear forces during joint motion. Accordingly, this study utilized an apparatus to apply biaxial loading to chondrocytes seeded within agarose constructs with endplates. The chondrocytes yielded a monotonic increase in proteoglycan synthesis both in free swelling culture up to day 8 and when the constructs were subjected to dynamic compression alone (15% amplitude at a frequency of 1 Hz for 48 h). However, when dynamic shear (10% amplitude at 1 Hz) was superimposed on dynamic compression, total collagen synthesis was also up-regulated, within 3 days of culture, without compromising proteoglycan synthesis. Histological analysis revealed marked collagen deposition around individual chondrocytes. A significant proportion (50%) of collagen was released into the culture medium, suggesting that it had only been partially synthesized in its mature state. The overall biosynthetic activity was enhanced more when the biaxial stimulation was applied in a continuous mode as opposed to intermittent loading. Results of the present study strongly suggest that proteoglycan and collagen synthesis may be triggered by uncoupled mechanosensitive cellular responses. The proposed in vitro model and the prescribed conditioning protocols demonstrated that a short pre-culture period is preferable to long free swelling culture condition as it enables a significantly higher up-regulation of collagen. Biotechnol. Bioeng. 2017;114: 1614-1625. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Erica DiFederico
- Medical Engineering Division, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK.,Department of Bioengineering, Imperial College, London, UK
| | - Julia C Shelton
- Medical Engineering Division, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Daniel L Bader
- Medical Engineering Division, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK.,Faculty of Health Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
42
|
Bandeiras C, Completo A. A mathematical model of tissue-engineered cartilage development under cyclic compressive loading. Biomech Model Mechanobiol 2016; 16:651-666. [PMID: 27817048 DOI: 10.1007/s10237-016-0843-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/11/2016] [Indexed: 12/23/2022]
Abstract
In this work a coupled model of solute transport and uptake, cell proliferation, extracellular matrix synthesis and remodeling of mechanical properties accounting for the impact of mechanical loading is presented as an advancement of a previously validated coupled model for free-swelling tissue-engineered cartilage cultures. Tissue-engineering constructs were modeled as biphasic with a linear elastic solid, and relevant intrinsic mechanical stimuli in the constructs were determined by numerical simulation for use as inputs of the coupled model. The mechanical dependent formulations were derived from a calibration and parametrization dataset and validated by comparison of normalized ratios of cell counts, total glycosaminoglycans and collagen after 24-h continuous cyclic unconfined compression from another dataset. The model successfully fit the calibration dataset and predicted the results from the validation dataset with good agreement, with average relative errors up to 3.1 and 4.3 %, respectively. Temporal and spatial patterns determined for other model outputs were consistent with reported studies. The results suggest that the model describes the interaction between the simultaneous factors involved in in vitro tissue-engineered cartilage culture under dynamic loading. This approach could also be attractive for optimization of culture protocols, namely through the application to longer culture times and other types of mechanical stimuli.
Collapse
Affiliation(s)
- Cátia Bandeiras
- Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - António Completo
- Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
43
|
Liebesny PH, Byun S, Hung HH, Pancoast JR, Mroszczyk KA, Young WT, Lee RT, Frisbie DD, Kisiday JD, Grodzinsky AJ. Growth Factor-Mediated Migration of Bone Marrow Progenitor Cells for Accelerated Scaffold Recruitment. Tissue Eng Part A 2016; 22:917-27. [PMID: 27268956 DOI: 10.1089/ten.tea.2015.0524] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Tissue engineering approaches using growth factor-functionalized acellular scaffolds to support and guide repair driven by endogenous cells are thought to require a careful balance between cell recruitment and growth factor release kinetics. The objective of this study was to identify a growth factor combination that accelerates progenitor cell migration into self-assembling peptide hydrogels in the context of cartilage defect repair. A novel 3D gel-to-gel migration assay enabled quantification of the chemotactic impact of platelet-derived growth factor-BB (PDGF-BB), heparin-binding insulin-like growth factor-1 (HB-IGF-1), and transforming growth factor-β1 (TGF-β1) on progenitor cells derived from subchondral bovine trabecular bone (bone-marrow progenitor cells, BM-PCs) encapsulated in the peptide hydrogel [KLDL]3. Only the combination of PDGF-BB and TGF-β1 stimulated significant migration of BM-PCs over a 4-day period, measured by confocal microscopy. Both PDGF-BB and TGF-β1 were slowly released from the gel, as measured using their (125)I-labeled forms, and they remained significantly present in the gel at 4 days. In the context of augmenting microfracture surgery for cartilage repair, our strategy of delivering chemotactic and proanabolic growth factors in KLD may provide the necessary local stimulus to help increase defect cellularity, providing more cells to generate repair tissue.
Collapse
Affiliation(s)
- Paul H Liebesny
- 1 Department of Biological Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | - Sangwon Byun
- 1 Department of Biological Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | - Han-Hwa Hung
- 1 Department of Biological Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | | | - Keri A Mroszczyk
- 3 Department of Mechanical Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | - Whitney T Young
- 3 Department of Mechanical Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | - Richard T Lee
- 2 Brigham and Women's Hospital , Boston, Massachusetts
| | - David D Frisbie
- 4 Colorado State University , Orthopaedic Research Center, Fort Collins, Colorado
| | - John D Kisiday
- 4 Colorado State University , Orthopaedic Research Center, Fort Collins, Colorado
| | - Alan J Grodzinsky
- 1 Department of Biological Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts.,3 Department of Mechanical Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts.,5 Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology , Cambridge, Massachusetts
| |
Collapse
|
44
|
Aisenbrey EA, Bryant SJ. Mechanical loading inhibits hypertrophy in chondrogenically differentiating hMSCs within a biomimetic hydrogel. J Mater Chem B 2016; 4:3562-3574. [PMID: 27499854 PMCID: PMC4972607 DOI: 10.1039/c6tb00006a] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Three dimensional hydrogels are a promising vehicle for delivery of adult human bone-marrow derived mesenchymal stem cells (hMSCs) for cartilage tissue engineering. One of the challenges with using this cell type is the default pathway is terminal differentiation, a hypertrophic phenotype and precursor to endochondral ossification. We hypothesized that a synthetic hydrogel consisting of extracellular matrix (ECM) analogs derived from cartilage when combined with dynamic loading provides physiochemical cues for achieving a stable chondrogenic phenotype. Hydrogels were formed from crosslinked poly(ethylyene glycol) as the base chemistry and to which (meth)acrylate functionalized ECM analogs of RGD (cell adhesion peptide) and chondroitin sulfate (ChS, a negatively charged glycosaminoglycan) were introduced. Bone-marrow derived hMSCs from three donors were encapsulated in the hydrogels and cultured under free swelling conditions or under dynamic com pressive loading with 2.5 ng/ml TGF-β3. hMSC differentiation was assessed by quantitative PCR and immunohistochemistry. Nine hydrogel formulations were initially screened containing 0, 0.1 or 1mM RGD and 0, 1 or 2wt% ChS. After 21 days, the 1% ChS and 0.1 mM RGD hydrogel had the highest collagen II gene expression, but this was accompanied by high collagen X gene expression. At the protein level, collagen II was detected in all formulations with ECM analogs, but minimally detectable in the hydrogel without ECM analogs. Collagen X protein was present in all formulations. The 0.1 mM RGD and 1% ChS formulation was selected and subjected to five loading regimes: no loading, 5% strain 0.3Hz (1.5%/s), 10% strain 0.3 Hz (3%/s), 5% strain 1 Hz (5%/s), and 10% strain 1Hz (10%/s). After 21 days, ~70-90% of cells stained positive for collagen II protein regardless of the culture condition. On the contrary, only ~20-30% of cells stained positive for collagen X protein under 3 and 5%/s loading conditions, which was accompanied by minimal staining for RunX2. The other culture conditions had more cells staining positive for collagen X (40-60%) and was accompanied by positive staining for RunX2. In summary, a cartilage-like biomimetic hydrogel supports chondrogenesis of hMSCs, but dynamic loading only under select strain rates is able to inhibit hypertrophy.
Collapse
Affiliation(s)
- E A Aisenbrey
- University of Colorado,Boulder. Chemical and Biological Engineering, UCB 596. Boulder, CO. 80309
| | - S J Bryant
- University of Colorado,Boulder. Chemical and Biological Engineering, UCB 596. Boulder, CO. 80309
| |
Collapse
|
45
|
Finlay S, Seedhom BB, Carey DO, Bulpitt AJ, Treanor DE, Kirkham J. In Vitro Engineering of High Modulus Cartilage-Like Constructs. Tissue Eng Part C Methods 2016; 22:382-97. [PMID: 26850081 PMCID: PMC4827287 DOI: 10.1089/ten.tec.2015.0351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To date, the outcomes of cartilage repair have been inconsistent and have frequently yielded mechanically inferior fibrocartilage, thereby increasing the chances of damage recurrence. Implantation of constructs with biochemical composition and mechanical properties comparable to natural cartilage could be advantageous for long-term repair. This study attempted to create such constructs, in vitro, using tissue engineering principles. Bovine synoviocytes were seeded on nonwoven polyethylene terephthalate fiber scaffolds and cultured in chondrogenic medium for 4 weeks, after which uniaxial compressive loading was applied using an in-house bioreactor for 1 h per day, at a frequency of 1 Hz, for a further 84 days. The initial loading conditions, determined from the mechanical properties of the immature constructs after 4 weeks in chondrogenic culture, were strains ranging between 13% and 23%. After 56 days (sustained at 84 days) of loading, the constructs were stained homogenously with Alcian blue and for type-II collagen. Dynamic compressive moduli were comparable to the high end values for native cartilage and proportional to Alcian blue staining intensity. We suggest that these high moduli values were attributable to the bioreactor setup, which caused the loading regime to change as the constructs developed, that is, the applied stress and strain increased with construct thickness and stiffness, providing continued sufficient cell stimulation as further matrix was deposited. Constructs containing cartilage-like matrix with response to load similar to that of native cartilage could produce long-term effective cartilage repair when implanted.
Collapse
Affiliation(s)
- Scott Finlay
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| | - Bahaa B Seedhom
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| | - Duane O Carey
- 2 School of Computing, University of Leeds , Leeds, United Kingdom
| | - Andy J Bulpitt
- 2 School of Computing, University of Leeds , Leeds, United Kingdom
| | - Darren E Treanor
- 3 Department of Pathology, Leeds Institute of Cancer and Pathology, University of Leeds , Leeds, United Kingdom .,4 Leeds Teaching Hospitals NHS Trust , Leeds, United Kingdom
| | - Jennifer Kirkham
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| |
Collapse
|
46
|
Koutsopoulos S. Self-assembling peptide nanofiber hydrogels in tissue engineering and regenerative medicine: Progress, design guidelines, and applications. J Biomed Mater Res A 2016; 104:1002-16. [DOI: 10.1002/jbm.a.35638] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/30/2015] [Accepted: 12/22/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Sotirios Koutsopoulos
- Center for Biomedical Engineering; Massachusetts Institute of Technology; Cambridge Massachusetts 02139
| |
Collapse
|
47
|
Izadifar Z, Chang T, Kulyk W, Chen X, Eames BF. Analyzing Biological Performance of 3D-Printed, Cell-Impregnated Hybrid Constructs for Cartilage Tissue Engineering. Tissue Eng Part C Methods 2016; 22:173-88. [PMID: 26592915 DOI: 10.1089/ten.tec.2015.0307] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Three-dimensional (3D) bioprinting of hybrid constructs is a promising biofabrication method for cartilage tissue engineering because a synthetic polymer framework and cell-impregnated hydrogel provide structural and biological features of cartilage, respectively. During bioprinting, impregnated cells may be subjected to high temperatures (caused by the adjacent melted polymer) and process-induced mechanical forces, potentially compromising cell function. This study addresses these biofabrication issues, evaluating the heat distribution of printed polycaprolactone (PCL) strands and the rheological property and structural stability of alginate hydrogels at various temperatures and concentrations. The biocompatibility of parameters from these studies was tested by culturing 3D hybrid constructs bioprinted with primary cells from embryonic chick cartilage. During initial two-dimensional culture expansion of these primary cells, two morphologically and molecularly distinct cell populations ("rounded" and "fibroblastic") were isolated. The biological performance of each population was evaluated in 3D hybrid constructs separately. The cell viability, proliferation, and cartilage differentiation were observed at high levels in hybrid constructs of both cell populations, confirming the validity of these 3D bioprinting parameters for effective cartilage tissue engineering. Statistically significant performance variations were observed, however, between the rounded and fibroblastic cell populations. Molecular and morphological data support the notion that such performance differences may be attributed to the relative differentiation state of rounded versus fibroblastic cells (i.e., differentiated chondrocytes vs. chondroprogenitors, respectively), which is a relevant issue for cell-based tissue engineering strategies. Taken together, our study demonstrates that bioprinting 3D hybrid constructs of PCL and cell-impregnated alginate hydrogel is a promising approach for cartilage tissue engineering.
Collapse
Affiliation(s)
- Zohreh Izadifar
- 1 Division of Biomedical Engineering, University of Saskatchewan , Saskatoon, Canada
| | - Tuanjie Chang
- 2 Department of Anatomy and Cell Biology, University of Saskatchewan , Saskatoon, Canada
| | - William Kulyk
- 2 Department of Anatomy and Cell Biology, University of Saskatchewan , Saskatoon, Canada
| | - Xiongbiao Chen
- 1 Division of Biomedical Engineering, University of Saskatchewan , Saskatoon, Canada .,3 Department of Mechanical Engineering, University of Saskatchewan , Saskatoon, Canada
| | - B Frank Eames
- 1 Division of Biomedical Engineering, University of Saskatchewan , Saskatoon, Canada .,2 Department of Anatomy and Cell Biology, University of Saskatchewan , Saskatoon, Canada
| |
Collapse
|
48
|
Ortved KF, Nixon AJ. Cell-based cartilage repair strategies in the horse. Vet J 2015; 208:1-12. [PMID: 26702950 DOI: 10.1016/j.tvjl.2015.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/06/2015] [Accepted: 10/08/2015] [Indexed: 12/12/2022]
Abstract
Damage to the articular cartilage surface is common in the equine athlete and, due to the poor intrinsic healing capabilities of cartilage, can lead to osteoarthritis (OA). Joint disease and OA are the leading cause of retirement in equine athletes and currently there are no effective treatments to stop the progression of OA. Several different cell-based strategies have been investigated to bolster the weak regenerative response of chondrocytes. Such techniques aim to restore the articular surface and prevent further joint degradation. Cell-based cartilage repair strategies include enhancement of endogenous repair mechanisms by recruitment of stem cells from the bone marrow following perforation of the subchondral bone plate; osteochondral implantation; implantation of chondrocytes that are maintained in defects by either a membrane cover or scaffold, and transplantation of mesenchymal stem cells into cartilage lesions. More recently, bioengineered cartilage and scaffoldless cartilage have been investigated for enhancing repair. This review article focuses on the multitude of cell-based repair techniques for cartilage repair across several species, with special attention paid to the horse.
Collapse
Affiliation(s)
- Kyla F Ortved
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA.
| | - Alan J Nixon
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| |
Collapse
|
49
|
Morishita A, Kumabe S, Nakatsuka M, Iwai Y. A histological study of mineralised tissue formation around implants with 3D culture of HMS0014 cells in Cellmatrix Type I-A collagen gel scaffold in vitro. Okajimas Folia Anat Jpn 2015; 91:57-71. [PMID: 25797459 DOI: 10.2535/ofaj.91.57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We cultured HMS0014 Yub621b cells within a 3D collagen gel scaffold (Cellmatrix Type I-A) and aimed to study the fate and contribution of human bone-derived mesenchymal stem cells (MSCs) in the guided bone regeneration(GBR)-engineered tissue which has developed around the titanium (Ti) test dental implant (IP) in vitro. The light microscopy (LM) and transmission electron microscopy (TEM) results of the peri-IP tissue indicated that collagen fibrils of the Cellmatrix Type I-A gel were accumulated and fabricated to provide a 3D meshwork for proliferation and differentiation of the HMS0014 cells in the top (cell) layer; mineralisation of the GBR tissue had commenced since day 1 and became markedly deposited between days 7 and 14 of the experiment. TEM observation revealed sedimentation of cement line at the periphery of the interwoven Cellmatrix fibres and fibrils in the ECM scaffold of the GBR tissue; matrix vesicle-mediated and appositional collagen-mediated mineralisation were identified in the peri-IP ECM scaffold. The fine structure study of the plurimorphic osteoblast(Ob)-like osteogeneic cells demonstrated numerous membranous organelles related with vesicular trafficking, secretion and endocytosis in the cytoplasm; well-developed cytoskeleton networks and intercellular junctional complexes were also observed. The specimens on fluorescence immunohistochemistry (IHC) by confocal laser-scanning microscopy (LSM) showed the expression of LC3 and Cx43 associated with autophagic-lysosomal degeneration pathway and signal conduction mediated with gap junctions (GJS) in maintaining tissue homeostasis of the Ob-like cells which grew and degenerated in the 3D scaffold. Results from this in vitro study suggest that Ob-like HMS0014 cells actively regulate turnover of the peri-IP ECM to recapitulate the development and formation of osteoid tissue-engineered material which might contribute to augment osseointegration around the dental implant.
Collapse
|
50
|
Hudson KD, Mozia RI, Bonassar LJ. Dose-dependent response of tissue-engineered intervertebral discs to dynamic unconfined compressive loading. Tissue Eng Part A 2015; 21:564-72. [PMID: 25277703 DOI: 10.1089/ten.tea.2014.0174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Because of the limitations of current surgical methods in the treatment of degenerative disc disease, tissue-engineered intervertebral discs (TE-IVDs) have become an important target. This study investigated the biochemical and mechanical responses of composite TE-IVDs to dynamic unconfined compression. TE-IVDs were manufactured by floating an injection molded alginate nucleus pulposus (NP) in a type I collagen annulus fibrosus (AF) that was allowed to contract for 2 weeks before loading. The discs were mechanically stimulated at a range of strain amplitude (1-10%) for 2 weeks with a duty cycle of 1 h on-1 h off-1 h on before being evaluated for their biochemical and mechanical properties. Mechanical loading increased all properties in a dose-dependent manner. Glycosaminoglycans (GAGs) increased between 2.8 and 2.2 fold in the AF and NP regions, respectively, whereas the hydroxyproline content increased between 1.2 and 1.8 fold. The discs also experienced a 2-fold increase in the equilibrium modulus and a 4.3-fold increase in the instantaneous modulus. Full effects for all properties were seen by 5% strain amplitude. These data suggest that dynamic loading increases the functionality of our TE-IVDs with region-dependent responses using a method that may be scaled up to larger disc models to expedite maturation for implantation.
Collapse
Affiliation(s)
- Katherine D Hudson
- 1 Department of Biomedical Engineering, Cornell University , Ithaca, New York
| | | | | |
Collapse
|