1
|
Peters JR, Hoogenboom M, Abinzano F, Callens SJP, Foolen J, Ito K. Tissue growth as a mechanism for collagen fiber alignment in articular cartilage. Sci Rep 2024; 14:31121. [PMID: 39732864 DOI: 10.1038/s41598-024-82441-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Articular cartilage is distinguished by the unique alignment of type II collagen, a feature crucial for its mechanical properties and function. This characteristic organization is established during postnatal development of the tissue, yet the underlying mechanisms remain poorly understood. In this study, a potential mechanism for type II collagen alignment by cartilage-specific growth from within the tissue was investigated. Bovine chondrocyte-derived cartilage organoids were cultured in a transwell system, subjecting the created tissue to transforming growth factor β1 stimulation from either the bottom (bottom-up) or the top (top-down) compartment to induce interstitial growth and appositional growth, respectively. The results demonstrate that interstitial growth within the tissue, stimulated from underneath, successfully produced aligned type II collagen parallel to the direction of this growth. In contrast, appositional growth did not yield such alignment. These findings underscore the critical role of the direction of growth in recreating the characteristic collagen organization of articular cartilage, offering valuable insights for the advancement of creating functional tissue in tissue engineering strategies.
Collapse
Affiliation(s)
- Jet R Peters
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Postbus 513, Eindhoven, 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Marit Hoogenboom
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Postbus 513, Eindhoven, 5600 MB, The Netherlands
| | - Florencia Abinzano
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Postbus 513, Eindhoven, 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Sebastien J P Callens
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Postbus 513, Eindhoven, 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jasper Foolen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Postbus 513, Eindhoven, 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Postbus 513, Eindhoven, 5600 MB, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
2
|
Bačenková D, Trebuňová M, Demeterová J, Živčák J. Human Chondrocytes, Metabolism of Articular Cartilage, and Strategies for Application to Tissue Engineering. Int J Mol Sci 2023; 24:17096. [PMID: 38069417 PMCID: PMC10707713 DOI: 10.3390/ijms242317096] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Hyaline cartilage, which is characterized by the absence of vascularization and innervation, has minimal self-repair potential in case of damage and defect formation in the chondral layer. Chondrocytes are specialized cells that ensure the synthesis of extracellular matrix components, namely type II collagen and aggregen. On their surface, they express integrins CD44, α1β1, α3β1, α5β1, α10β1, αVβ1, αVβ3, and αVβ5, which are also collagen-binding components of the extracellular matrix. This article aims to contribute to solving the problem of the possible repair of chondral defects through unique methods of tissue engineering, as well as the process of pathological events in articular cartilage. In vitro cell culture models used for hyaline cartilage repair could bring about advanced possibilities. Currently, there are several variants of the combination of natural and synthetic polymers and chondrocytes. In a three-dimensional environment, chondrocytes retain their production capacity. In the case of mesenchymal stromal cells, their favorable ability is to differentiate into a chondrogenic lineage in a three-dimensional culture.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, Letná 9, 042 00 Košice, Slovakia; (M.T.); (J.D.); (J.Ž.)
| | | | | | | |
Collapse
|
3
|
Integration of a miniaturized DMMB assay with high-throughput screening for identifying regulators of proteoglycan metabolism. Sci Rep 2022; 12:1083. [PMID: 35058478 PMCID: PMC8776954 DOI: 10.1038/s41598-022-04805-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/21/2021] [Indexed: 11/19/2022] Open
Abstract
Defective biosynthesis or function of proteoglycans causes pathological conditions in a variety of tissue systems. Osteoarthritis (OA) is a prevalent degenerative joint disorder characterized by progressive cartilage destruction caused by imbalanced proteoglycan synthesis and degradation. Identifying agents that regulate proteoglycan metabolism may benefit the development of OA-modifying therapeutics. High-throughput screening (HTS) of chemical libraries has paved the way for achieving this goal. However, the implementation and adaptation of HTS assays based on proteoglycan measurement remain underexploited. Using primary porcine chondrocytes as a model, we report a miniaturized dimethyl-methylene blue (DMMB) assay, which is commonly used to quantitatively evaluate sulfated glycosaminoglycan (GAG) content, with an optimized detection range and reproducibility and its integration with HTS. Treatment with TGF-β1 and IL1-α, known as positive and negative proteoglycan regulators, respectively, supported the assay specificity. A pre-test of chemical screening of 960 compounds identified both stimulators (4.48%) and inhibitors (6.04%) of GAG production. Fluorophore-assisted carbohydrate electrophoresis validated the activity of selected hits on chondroitin sulfate expression in an alginate culture system. Our findings support the implementation of this simple colorimetric assay in HTS to discover modifiers of OA or other diseases related to dysregulated proteoglycan metabolism.
Collapse
|
4
|
Application of Alginate Hydrogels for Next-Generation Articular Cartilage Regeneration. Int J Mol Sci 2022; 23:ijms23031147. [PMID: 35163071 PMCID: PMC8835677 DOI: 10.3390/ijms23031147] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022] Open
Abstract
The articular cartilage has insufficient intrinsic healing abilities, and articular cartilage injuries often progress to osteoarthritis. Alginate-based scaffolds are attractive biomaterials for cartilage repair and regeneration, allowing for the delivery of cells and therapeutic drugs and gene sequences. In light of the heterogeneity of findings reporting the benefits of using alginate for cartilage regeneration, a better understanding of alginate-based systems is needed in order to improve the approaches aiming to enhance cartilage regeneration with this compound. This review provides an in-depth evaluation of the literature, focusing on the manipulation of alginate as a tool to support the processes involved in cartilage healing in order to demonstrate how such a material, used as a direct compound or combined with cell and gene therapy and with scaffold-guided gene transfer procedures, may assist cartilage regeneration in an optimal manner for future applications in patients.
Collapse
|
5
|
Visscher DO, Gleadall A, Buskermolen JK, Burla F, Segal J, Koenderink GH, Helder MN, van Zuijlen PPM. Design and fabrication of a hybrid alginate hydrogel/poly(ε-caprolactone) mold for auricular cartilage reconstruction. J Biomed Mater Res B Appl Biomater 2019; 107:1711-1721. [PMID: 30383916 PMCID: PMC6587956 DOI: 10.1002/jbm.b.34264] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/14/2018] [Accepted: 09/23/2018] [Indexed: 11/11/2022]
Abstract
The aim of this study was to design and manufacture an easily assembled cartilage implant model for auricular reconstruction. First, the printing accuracy and mechanical properties of 3D-printed poly-ε-caprolactone (PCL) scaffolds with varying porosities were determined to assess overall material properties. Next, the applicability of alginate as cell carrier for the cartilage implant model was determined. Using the optimal outcomes of both experiments (in terms of (bio)mechanical properties, cell survival, neocartilage formation, and printing accuracy), a hybrid auricular implant model was developed. PCL scaffolds with 600 μm distances between strands exhibited the best mechanical properties and most optimal printing quality for further exploration. In alginate, chondrocytes displayed high cell survival (~83% after 21 days) and produced cartilage-like matrix in vitro. Alginate beads cultured in proliferation medium exhibited slightly higher compressive moduli (6 kPa) compared to beads cultured in chondrogenic medium (3.5 kPa, p > .05). The final auricular mold could be printed with 300 μm pores and high fidelity, and the injected chondrocytes survived the culture period of 21 days. The presented hybrid auricular mold appears to be an adequate model for cartilage tissue engineering and may provide a novel approach to auricular cartilage regeneration for facial reconstruction. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1711-1721, 2019.
Collapse
Affiliation(s)
- D. O. Visscher
- Department of Plastic, Reconstructive and Hand SurgeryAmsterdam University Medical Center, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - A. Gleadall
- Manufacturing and Process TechnologiesFaculty of Engineering, University of NottinghamNottinghamEnglandUK
- Wolfson School of Mechanical and Manufacturing EngineeringLoughborough UniversityLeicestershireLE11 3TUUK
| | - J. K. Buskermolen
- Department of DermatologyAmsterdam University Medical Center, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - F. Burla
- Department of Living Matter, AMOLFAmsterdamThe Netherlands
| | - J. Segal
- Manufacturing and Process TechnologiesFaculty of Engineering, University of NottinghamNottinghamEnglandUK
| | | | - M. N. Helder
- Department of Oral and Maxillofacial Surgery/Oral PathologyAmsterdam University Medical Center, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - P. P. M. van Zuijlen
- Department of Plastic, Reconstructive and Hand SurgeryAmsterdam University Medical Center, Amsterdam Movement SciencesAmsterdamThe Netherlands
- Department of PlasticReconstructive and Hand Surgery, Red Cross HospitalBeverwijkThe Netherlands
- Association of Dutch Burn CentersBeverwijkThe Netherlands
| |
Collapse
|
6
|
Lehmann J, Nürnberger S, Narcisi R, Stok KS, van der Eerden BCJ, Koevoet WJLM, Kops N, Ten Berge D, van Osch GJ. Recellularization of auricular cartilage via elastase-generated channels. Biofabrication 2019; 11:035012. [PMID: 30921774 DOI: 10.1088/1758-5090/ab1436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Decellularized tissue matrices are promising substrates for tissue generation by stem cells to replace poorly regenerating tissues such as cartilage. However, the dense matrix of decellularized cartilage impedes colonisation by stem cells. Here, we show that digestion of elastin fibre bundles traversing auricular cartilage creates channels through which cells can migrate into the matrix. Human chondrocytes and bone marrow-derived mesenchymal stromal cells efficiently colonise elastin-treated scaffolds through these channels, restoring a glycosaminoglycan-rich matrix and improving mechanical properties while maintaining size and shape of the restored tissue. The scaffolds are also rapidly colonised by endogenous cartilage-forming cells in a subcutaneously implanted osteochondral biopsy model. Creating channels for cells in tissue matrices may be a broadly applicable strategy for recellularization and restoration of tissue function.
Collapse
Affiliation(s)
- J Lehmann
- Department of Otorhinolaryngology and Head and Neck Surgery Erasmus MC, Rotterdam, The Netherlands. Department of Cell Biology Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Lavernia L, Brown WE, Wong BJF, Hu JC, Athanasiou KA. Toward tissue-engineering of nasal cartilages. Acta Biomater 2019; 88:42-56. [PMID: 30794988 DOI: 10.1016/j.actbio.2019.02.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/15/2019] [Accepted: 02/18/2019] [Indexed: 12/25/2022]
Abstract
Nasal cartilage pathologies are common; for example, up to 80% of people are afflicted by deviated nasal septum conditions. Because cartilage provides the supportive framework of the nose, afflicted patients suffer low quality of life. To correct pathologies, graft cartilage is often required. Grafts are currently sourced from the patient's septum, ear, or rib. However, their use yields donor site morbidity and is limited by tissue quantity and quality. Additionally, rhinoplasty revision rates exceed 15%, exacerbating the shortage of graft cartilage. Alternative grafts, such as irradiated allogeneic rib cartilage, are associated with complications. Tissue-engineered neocartilage holds promise to address the limitations of current grafts. The engineering design process may be used to create suitable graft tissues. This process begins by identifying the surgeon's needs. Second, nasal cartilages' properties must be understood to define engineering design criteria. Limited investigations have examined nasal cartilage properties; numerous additional studies need to be performed to examine topographical variations, for example. Third, tissue-engineering processes must be applied to achieve the engineering design criteria. Within the recent past, strategies have frequently utilized human septal chondrocytes. As autologous and allogeneic rib graft cartilage is used, its suitability as a cell source should also be examined. Fourth, quantitative verification of engineered neocartilage is critical to check for successful achievement of the engineering design criteria. Finally, following the FDA paradigm, engineered neocartilage must be orthotopically validated in animals. Together, these steps delineate a path to engineer functional nasal neocartilages that may, ultimately, be used to treat human patients. STATEMENT OF SIGNIFICANCE: Nasal cartilage pathologies are common and lead to greatly diminished quality of life. The ability to correct pathologies is limited by cartilage graft quality and quantity, as well as donor site morbidity and surgical complications, such as infection and resorption. Despite the significance of nasal cartilage pathologies and high rhinoplasty revision rates (15%), little characterization and tissue-engineering work has been performed compared to other cartilages, such as articular cartilage. Furthermore, most work is published in clinical journals, with little in biomedical engineering. Therefore, this review discusses what nasal cartilage properties are known, summarizes the current state of nasal cartilage tissue-engineering, and makes recommendations via the engineering design process toward engineering functional nasal neocartilage to address current limitations.
Collapse
Affiliation(s)
- Laura Lavernia
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA 92697-2715, USA
| | - Wendy E Brown
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA 92697-2715, USA.
| | - Brian J F Wong
- Division of Facial Plastic Surgery, Department of Otolaryngology-Head and Neck Surgery, University of California Irvine, 1002 Health Sciences Road, Irvine, CA 92617, USA; Department of Biomedical Engineering, University of California Irvine, 1002 Health Sciences Road, Irvine, CA 92617, USA.
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA 92697-2715, USA.
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA 92697-2715, USA.
| |
Collapse
|
8
|
Goepfert C, Lutz V, Lünse S, Kittel S, Wiegandt K, Kammal M, Püschel K, Pörtner R. Evaluation of Cartilage Specific Matrix Synthesis of Human Articular Chondrocytes after Extended Propagation on Microcarriers by Image Analysis. Int J Artif Organs 2018. [DOI: 10.1177/039139881003300405] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Cell-based technologies for the repair of cartilage defects usually rely on the expansion of low numbers of chondrocytes isolated from biopsies of healthy cartilage. Proliferating chondrocytes are known to undergo dedifferentiation characterized by downregulation of collagen type II and proteoglycan production, and by upregulation of collagen type I synthesis. Re-expression of cartilage specific matrix components by expanded chondrocytes is therefore critical for successful cartilage repair. Methods Human articular chondrocytes were expanded on microcarriers Cytodex 3. The growth area was increased by adding empty microcarriers. Added microcarriers were colonized by bead-to-bead transfer of the cells. The chondrocytes were harvested from the microcarriers and characterized by their ability to synthesize collagen type II when cultivated in alginate beads using chondrogenic growth factors. A semi-automatic image analysis technique was developed to determine the fractions of collagen type II and type I positive cells. Results The expansion of human articular chondrocytes on microcarriers yielded high cell numbers and propagation rates compared to chondrocytes expanded in flask culture for one passage. The proportion of collagen type II positive cells compared to collagen type I synthesizing cells was increased compared to chondrocytes expanded using conventional methods. The matrix synthesis upon treatment with chondrogenic factors IGF-I and BMP-7 was enhanced whereas TGF-β had an inhibitory effect on microcarrier expanded chondrocytes. Conclusions Expanding human articular chondrocytes on microcarriers omitting subcultivation steps leads to superior ratios of collagen type II to type I forming cells compared to the expansion in conventional monolayer culture.
Collapse
Affiliation(s)
- Christiane Goepfert
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Vivien Lutz
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Svenja Lünse
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Sabrina Kittel
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Katharina Wiegandt
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Michael Kammal
- University Medical Center Hamburg-Eppendorf, Department of Legal Medicine, Hamburg - Germany
| | - Klaus Püschel
- University Medical Center Hamburg-Eppendorf, Department of Legal Medicine, Hamburg - Germany
| | - Ralf Pörtner
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| |
Collapse
|
9
|
Rapid Chondrocyte Isolation for Tissue Engineering Applications: The Effect of Enzyme Concentration and Temporal Exposure on the Matrix Forming Capacity of Nasal Derived Chondrocytes. BIOMED RESEARCH INTERNATIONAL 2017. [PMID: 28337445 DOI: 10.1155/2017/2395138.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Laboratory based processing and expansion to yield adequate cell numbers had been the standard in Autologous Disc Chondrocyte Transplantation (ADCT), Allogeneic Juvenile Chondrocyte Implantation (NuQu®), and Matrix-Induced Autologous Chondrocyte Implantation (MACI). Optimizing cell isolation is a key challenge in terms of obtaining adequate cell numbers while maintaining a vibrant cell population capable of subsequent proliferation and matrix elaboration. However, typical cell yields from a cartilage digest are highly variable between donors and based on user competency. The overall objective of this study was to optimize chondrocyte isolation from cartilaginous nasal tissue through modulation of enzyme concentration exposure (750 and 3000 U/ml) and incubation time (1 and 12 h), combined with physical agitation cycles, and to assess subsequent cell viability and matrix forming capacity. Overall, increasing enzyme exposure time was found to be more detrimental than collagenase concentration for subsequent viability, proliferation, and matrix forming capacity (sGAG and collagen) of these cells resulting in nonuniform cartilaginous matrix deposition. Taken together, consolidating a 3000 U/ml collagenase digest of 1 h at a ratio of 10 ml/g of cartilage tissue with physical agitation cycles can improve efficiency of chondrocyte isolation, yielding robust, more uniform matrix formation.
Collapse
|
10
|
Rapid Chondrocyte Isolation for Tissue Engineering Applications: The Effect of Enzyme Concentration and Temporal Exposure on the Matrix Forming Capacity of Nasal Derived Chondrocytes. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2395138. [PMID: 28337445 PMCID: PMC5350344 DOI: 10.1155/2017/2395138] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/06/2017] [Indexed: 12/22/2022]
Abstract
Laboratory based processing and expansion to yield adequate cell numbers had been the standard in Autologous Disc Chondrocyte Transplantation (ADCT), Allogeneic Juvenile Chondrocyte Implantation (NuQu®), and Matrix-Induced Autologous Chondrocyte Implantation (MACI). Optimizing cell isolation is a key challenge in terms of obtaining adequate cell numbers while maintaining a vibrant cell population capable of subsequent proliferation and matrix elaboration. However, typical cell yields from a cartilage digest are highly variable between donors and based on user competency. The overall objective of this study was to optimize chondrocyte isolation from cartilaginous nasal tissue through modulation of enzyme concentration exposure (750 and 3000 U/ml) and incubation time (1 and 12 h), combined with physical agitation cycles, and to assess subsequent cell viability and matrix forming capacity. Overall, increasing enzyme exposure time was found to be more detrimental than collagenase concentration for subsequent viability, proliferation, and matrix forming capacity (sGAG and collagen) of these cells resulting in nonuniform cartilaginous matrix deposition. Taken together, consolidating a 3000 U/ml collagenase digest of 1 h at a ratio of 10 ml/g of cartilage tissue with physical agitation cycles can improve efficiency of chondrocyte isolation, yielding robust, more uniform matrix formation.
Collapse
|
11
|
Athanasiou KA, Responte DJ, Brown WE, Hu JC. Harnessing biomechanics to develop cartilage regeneration strategies. J Biomech Eng 2015; 137:020901. [PMID: 25322349 DOI: 10.1115/1.4028825] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Indexed: 12/24/2022]
Abstract
As this review was prepared specifically for the American Society of Mechanical Engineers H.R. Lissner Medal, it primarily discusses work toward cartilage regeneration performed in Dr. Kyriacos A. Athanasiou's laboratory over the past 25 years. The prevalence and severity of degeneration of articular cartilage, a tissue whose main function is largely biomechanical, have motivated the development of cartilage tissue engineering approaches informed by biomechanics. This article provides a review of important steps toward regeneration of articular cartilage with suitable biomechanical properties. As a first step, biomechanical and biochemical characterization studies at the tissue level were used to provide design criteria for engineering neotissues. Extending this work to the single cell and subcellular levels has helped to develop biochemical and mechanical stimuli for tissue engineering studies. This strong mechanobiological foundation guided studies on regenerating hyaline articular cartilage, the knee meniscus, and temporomandibular joint (TMJ) fibrocartilage. Initial tissue engineering efforts centered on developing biodegradable scaffolds for cartilage regeneration. After many years of studying scaffold-based cartilage engineering, scaffoldless approaches were developed to address deficiencies of scaffold-based systems, resulting in the self-assembling process. This process was further improved by employing exogenous stimuli, such as hydrostatic pressure, growth factors, and matrix-modifying and catabolic agents, both singly and in synergistic combination to enhance neocartilage functional properties. Due to the high cell needs for tissue engineering and the limited supply of native articular chondrocytes, costochondral cells are emerging as a suitable cell source. Looking forward, additional cell sources are investigated to render these technologies more translatable. For example, dermis isolated adult stem (DIAS) cells show potential as a source of chondrogenic cells. The challenging problem of enhanced integration of engineered cartilage with native cartilage is approached with both familiar and novel methods, such as lysyl oxidase (LOX). These diverse tissue engineering strategies all aim to build upon thorough biomechanical characterizations to produce functional neotissue that ultimately will help combat the pressing problem of cartilage degeneration. As our prior research is reviewed, we look to establish new pathways to comprehensively and effectively address the complex problems of musculoskeletal cartilage regeneration.
Collapse
|
12
|
Pretzel D, Linss S, Ahrem H, Endres M, Kaps C, Klemm D, Kinne RW. A novel in vitro bovine cartilage punch model for assessing the regeneration of focal cartilage defects with biocompatible bacterial nanocellulose. Arthritis Res Ther 2014; 15:R59. [PMID: 23673274 PMCID: PMC4060236 DOI: 10.1186/ar4231] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 02/04/2013] [Accepted: 05/14/2013] [Indexed: 12/21/2022] Open
Abstract
Introduction Current therapies for articular cartilage defects fail to achieve qualitatively sufficient tissue regeneration, possibly because of a mismatch between the speed of cartilage rebuilding and the resorption of degradable implant polymers. The present study focused on the self-healing capacity of resident cartilage cells in conjunction with cell-free and biocompatible (but non-resorbable) bacterial nanocellulose (BNC). This was tested in a novel in vitro bovine cartilage punch model. Methods Standardized bovine cartilage discs with a central defect filled with BNC were cultured for up to eight weeks with/without stimulation with transforming growth factor-β1 (TGF-β1. Cartilage formation and integrity were analyzed by histology, immunohistochemistry and electron microscopy. Content, release and neosynthesis of the matrix molecules proteoglycan/aggrecan, collagen II and collagen I were also quantified. Finally, gene expression of these molecules was profiled in resident chondrocytes and chondrocytes migrated onto the cartilage surface or the implant material. Results Non-stimulated and especially TGF-β1-stimulated cartilage discs displayed a preserved structural and functional integrity of the chondrocytes and surrounding matrix, remained vital in long-term culture (eight weeks) without signs of degeneration and showed substantial synthesis of cartilage-specific molecules at the protein and mRNA level. Whereas mobilization of chondrocytes from the matrix onto the surface of cartilage and implant was pivotal for successful seeding of cell-free BNC, chondrocytes did not immigrate into the central BNC area, possibly due to the relatively small diameter of its pores (2 to 5 μm). Chondrocytes on the BNC surface showed signs of successful redifferentiation over time, including increase of aggrecan/collagen type II mRNA, decrease of collagen type I mRNA and initial deposition of proteoglycan and collagen type II in long-term high-density pellet cultures. Although TGF-β1 stimulation showed protective effects on matrix integrity, effects on other parameters were limited. Conclusions The present bovine cartilage punch model represents a robust, reproducible and highly suitable tool for the long-term culture of cartilage, maintaining matrix integrity and homoeostasis. As an alternative to animal studies, this model may closely reflect early stages of cartilage regeneration, allowing the evaluation of promising biomaterials with/without chondrogenic factors.
Collapse
|
13
|
Madej W, van Caam A, Blaney Davidson EN, van der Kraan PM, Buma P. Physiological and excessive mechanical compression of articular cartilage activates Smad2/3P signaling. Osteoarthritis Cartilage 2014; 22:1018-25. [PMID: 24795273 DOI: 10.1016/j.joca.2014.04.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 04/18/2014] [Accepted: 04/23/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Transforming growth factor beta (TGF-β) in articular cartilage can signal via two routes, the ALK5/Smad2/3P and the ALK1/Smad1/5/8P route, the first being protective and the latter favoring chondrocyte terminal differentiation. Since biomechanical factors are known to play an essential role in osteoarthritis (OA) initiation and progression, we investigated if excessive mechanical compression can alter TGF-β signaling in cartilage shifting it from ALK5/Smad2/3P to ALK1/Smad1/5/8P pathway, favoring terminal differentiation of chondrocytes. DESIGN Articular cartilage explants were harvested from bovine metacarpophalangeal joints. After equilibration, explants were subjected to unconfined dynamic mechanical compression (1 Hz) with 3 MPa (physiological) or 12 MPa (excessive) stress. After different time intervals samples were frozen and mRNA levels of selected genes were examined using real-time polymerase chain reaction. RESULTS In articular cartilage compressed with 3 MPa and also 12 MPa stress the expression of Smad2/3P responsive genes bSerpine1, bSmad7 and bAlk5 was up-regulated, whereas the expression of Smad1/5/8P responsive gene bId1 was down-regulated. Furthermore, the expression of bTgfb1 was significantly up-regulated in both compression groups. When ALK5/Smad2/3P pathway was blocked with a selective ALK4/5/7 inhibitor, the effect of excessive mechanical compression on bSmad7 and bAlk5 expression was prevented. CONCLUSIONS Here we show that excessive mechanical compression alone is not able to shift TGF-β signaling toward the ALK1/Smad1/5/8P pathway. In contrast, we show that mechanical compression not only with physiological but also with excessive stress can activate Smad2/3P signaling, which is known to be protective for articular cartilage and to block chondrocyte terminal differentiation.
Collapse
Affiliation(s)
- W Madej
- Orthopaedic Research Laboratory, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - A van Caam
- Experimental Rheumatology & Advanced Therapeutics, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - E N Blaney Davidson
- Experimental Rheumatology & Advanced Therapeutics, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - P M van der Kraan
- Experimental Rheumatology & Advanced Therapeutics, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - P Buma
- Orthopaedic Research Laboratory, Radboud University Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
14
|
Zamani S, Hashemibeni B, Esfandiari E, Kabiri A, Rabbani H, Abutorabi R. Assessment of TGF-β3 on production of aggrecan by human articular chondrocytes in pellet culture system. Adv Biomed Res 2014; 3:54. [PMID: 24627862 PMCID: PMC3950793 DOI: 10.4103/2277-9175.125799] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 08/23/2012] [Indexed: 11/27/2022] Open
Abstract
Background: The Autologous Chondrocytes Transplantation (ACT) method is being studied for repair of cartilage diseases. As the chondrocytes dedifferentiated during monolayer culture, three-dimensional cultures are suggested to redifferentiate them. The aim of this study was investigation of the effect of TGF-β3 growth factor on chondrocytes in pellet culture system. Materials and Methods: The chondrocytes were isolated from three human articular cartilages by enzymatic digestion. The cells of the second passage were transferred to pellet culture system. We determined the chondrogenic medium with TGF-β3 as the experimental group and without it as the control group. After 2 weeks, the aggrecan production was investigated using histological and immunohistochemical (IHC) methods. Results: The presence of glycosaminoglycans was proved through Toluiden blue staining. Comparison of IHC results using MATLAB software showed that aggrecan in the experimental group was significantly higher than in the control group (P ≤ 0.05). Conclusion: The presence of TGF-β3 in the chondrogenic medium could lead to the production of more aggrecan in chondrocytes cultivated in pellet culture system.
Collapse
Affiliation(s)
- Saeed Zamani
- Department of Anatomical Sciences and Molecular Biology, Medical Faculty, Isfahan University of Medical Sciences, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences and Molecular Biology, Medical Faculty, Isfahan University of Medical Sciences, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences and Molecular Biology, Medical Faculty, Isfahan University of Medical Sciences, Iran
| | - Azadeh Kabiri
- Department of Anatomical Sciences and Molecular Biology, Medical Faculty, Isfahan University of Medical Sciences, Iran
| | - Hossein Rabbani
- Department of Biophysics, Medical Faculty, Isfahan University of Medical Sciences, Iran
| | - Roshanak Abutorabi
- Department of Anatomical Sciences and Molecular Biology, Medical Faculty, Isfahan University of Medical Sciences, Iran
| |
Collapse
|
15
|
Dahlin RL, Ni M, Meretoja VV, Kasper FK, Mikos AG. TGF-β3-induced chondrogenesis in co-cultures of chondrocytes and mesenchymal stem cells on biodegradable scaffolds. Biomaterials 2013; 35:123-32. [PMID: 24125773 DOI: 10.1016/j.biomaterials.2013.09.086] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 09/24/2013] [Indexed: 10/26/2022]
Abstract
In this work, it was hypothesized that co-cultures of articular chondrocytes (ACs) and mesenchymal stem cells (MSCs) would exhibit enhanced sensitivity to chondrogenic stimuli, such as TGF-β3, and would require a reduced concentration of TGF-β3 to achieve an equivalent level of chondrogenesis compared to monocultures of each cell type. Furthermore, it was hypothesized that compared to monocultures, the chondrogenic phenotype of AC/MSC co-cultures would be more stable upon the removal of TGF-β3 from the culture medium. These hypotheses were investigated by culturing ACs and MSCs alone and in a 1:3 ratio on electrospun poly(ε-caprolactone) scaffolds. All cell populations were cultured for two weeks with 0, 1, 3, or 10 ng/ml of TGF-β3. After two weeks growth factor supplementation was removed, and the constructs were cultured for two additional weeks. Cell proliferation, extracellular matrix production, and chondrogenic gene expression were evaluated after two and four weeks. The results demonstrated that co-cultures of ACs and MSCs require a reduced concentration and duration of TGF-β3 exposure to achieve an equivalent level of chondrogenesis compared to AC or MSC monocultures. Thus, the present work implicates that the promise of co-cultures for cartilage engineering is enhanced by their robust phenotype and heightened sensitivity to TGF-β3.
Collapse
|
16
|
Kock LM, Geraedts J, Ito K, van Donkelaar CC. Low agarose concentration and TGF-β3 distribute extracellular matrix in tissue-engineered cartilage. Tissue Eng Part A 2013; 19:1621-31. [PMID: 23469833 DOI: 10.1089/ten.tea.2012.0541] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The mechanical properties of articular cartilage are dominated by the interterritorial matrix, as the matrix in this region is stiffer, greater in volume, and more interconnected compared to that in the pericellular and territorial region. Hence, tissue-engineered constructs in which a newly synthesized matrix accumulates in the pericellular and territorial regions may be of a lower mechanical quality compared to constructs in which the interterritorial region contains abundant matrix. OBJECTIVE In this study, we explored the extent to which matrix distribution may be modulated by altering the agarose concentration and the presence of the transforming growth factor-β (TGF-β) and how this affects the mechanical properties of cultured cartilage constructs. METHODS Cartilage development in constructs with agarose concentrations varying from 1%, 2%, and 3% (study 1) and in constructs with no or very low agarose concentrations of 0.25%, 0.5%, and 1% (study 2) were compared. In both studies, the effect of TGF-β3 was compared to fetal bovine serum. After 21 and 42 days of culture, the matrix content and distribution were analyzed and mechanical properties were assessed at day 42. RESULTS Culture in lower agarose concentrations did not significantly influence the matrix content per wet weight, but did result in a more homogeneous distribution. Constructs cultured with less agarose also showed a higher equilibrium modulus. The presence of TGF-β3 resulted in an increased extracellular matrix (ECM) deposition, a more homogeneous matrix distribution, and an equilibrium modulus. CONCLUSIONS Culturing with no or low agarose concentrations and TGF-β3 is favorable for cartilage tissue-engineering studies, because both stimulate the formation of a more homogeneous ECM and consequently result in improved mechanical properties.
Collapse
Affiliation(s)
- Linda M Kock
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | | | | | | |
Collapse
|
17
|
Chondrocyte-alginate constructs with or without TGF-β1 produces superior extracellular matrix expression than monolayer cultures. Mol Cell Biochem 2012; 376:11-20. [DOI: 10.1007/s11010-012-1543-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 12/06/2012] [Indexed: 10/27/2022]
|
18
|
Liebman J, Goldberg RL. Chondrocyte culture and assay. CURRENT PROTOCOLS IN PHARMACOLOGY 2012; Chapter 12:Unit12.2. [PMID: 21959754 DOI: 10.1002/0471141755.ph1202s12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chondrocytes constitute the sole cell type found within cartilage, and control the formation and composition of cartilage. Cellular, biochemical and pharmacological studies of arthritis and other cartilage disorders have increasingly focused on chondrocyte function. Three methods are presented in this unit for culturing chondrocytes, and two assays are described that characterize proteoglycan synthesis, a key measure of chondrocyte function.Chondrocytes constitute the sole cell type found within cartilage, and control the formation and composition of cartilage.
Collapse
Affiliation(s)
- J Liebman
- Novartis Institute of Biomedical Research, Summit, New Jersey, USA
| | | |
Collapse
|
19
|
Ha CW, Noh MJ, Choi KB, Lee KH. Initial phase I safety of retrovirally transduced human chondrocytes expressing transforming growth factor-beta-1 in degenerative arthritis patients. Cytotherapy 2012; 14:247-56. [PMID: 22242865 PMCID: PMC3793276 DOI: 10.3109/14653249.2011.629645] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background aims. TissueGene-C (TG-C) represents a cell-mediated gene therapy for localized delivery of allogeneic chondrocytes expressing transforming growth factor (TGF)-β1 directly to the damaged knee joint. Untransduced human chondrocytes (hChonJ cells) have also been incorporated into the TG-C product at a 3:1 ratio with TGF-β1-expressing chondrocytes (hChonJb#7) in order to help fill in the defect and as target cells for the actions of the expressed TGF-β1. Methods. A phase I dose-escalating clinical trial was performed to evaluate the safety and biologic activity of TG-C in patients with advanced osteoarthritis of the knee joint (full thickness cartilage defect) that was refractory to existing non-operative therapies. Following a single intra-articular injection into the joint space of the damaged knee, patients were monitored for safety, and an evaluation was performed to assess the pharmacokinetics and biologic activity of TG-C. Results. There were no treatment-related serious adverse events. Swelling, effusion and minor localized reactions such as warming sensation or itching were observed in a dose-dependent manner at the injection site. Knee evaluation scores seemed to indicate a dose-dependent trend toward efficacy; however, patient numbers were not sufficient to determine statistical significance. Conclusions. Overall, there were no significant safety issues related to the administration of TG-C, with only some minor injection site reactions observed. Additionally, knee scoring analyzes indicated a possibility that TG-C may contribute to improvement of arthritic symptoms. More study is warranted to evaluate further the safety and determine the potential efficacy of TG-C.
Collapse
Affiliation(s)
- Chul-Won Ha
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
20
|
Ansar MM, Esfandiariy E, Mardani M, Hashemibeni B, Zarkesh-Esfahani SH, Hatef M, Kabiri A. A comparative study of aggrecan synthesis between natural articular chondrocytes and differentiated chondrocytes from adipose derived stem cells in 3D culture. Adv Biomed Res 2012; 1:24. [PMID: 23210083 PMCID: PMC3507023 DOI: 10.4103/2277-9175.98145] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 03/03/2012] [Indexed: 11/09/2022] Open
Abstract
Introduction: The main obstacle for tissue engineering is to find the most appropriate cell which is able to produce extracellular matrix (ECM) similar or better than natural chondrocytes in vitro. This study compared aggrecan synthesis's potential between differentiated chondrocytes (DCs) from adipose-derived stem cells (ADSCs) and natural articular chondrocytes (NCs) in 3D culture in vitro. Materials and Methods: Human ADSCs were isolated from sub-cutaneous adipose tissue and then the surface markers including CD 14, 45 CD105, CD90, CD44 were analyzed by flow cytometry. Also human articular chondrocytes were yielded of non-weight bearing area of Knee cartilage. Both types of the cells were encapsulated in alginate scaffolds and cultured in chondrogenic medium with and without TGFβ3 for 3 weeks. Then the extent of aggercan (AGC) production was evaluated by ELISA on days 14 and 21. Results: Our findings indicated that differentiated chondrocytes (DCs) with and without TGFβ3 synthesized more AGC than natural chondrocytes (NCs) on day 14. But DCs without TGFβ3 had higher production than other groups on day 21. Application of TGFβ3 resulted in an increase of amount of AGC in DCs on day 14 but a decrease on day 21 than same group. Conclusion: Since, aggrecan is an important chondrogenic marker, it was concluded that ADSCs can be possible reliable alternative cell source for cartilage tissue engineering in future.
Collapse
Affiliation(s)
- Malek Masoud Ansar
- Department of Anatomical Sciences and Molecular Biology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | | | | | | | | | |
Collapse
|
21
|
JONITZ ANIKA, LOCHNER KATRIN, TISCHER THOMAS, HANSMANN DORIS, BADER RAINER. TGF-β1 and IGF-1 influence the re-differentiation capacity of human chondrocytes in 3D pellet cultures in relation to different oxygen concentrations. Int J Mol Med 2012; 30:666-72. [DOI: 10.3892/ijmm.2012.1042] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/05/2012] [Indexed: 11/06/2022] Open
|
22
|
Mounts T, Ginley N, Schluchter M, Dennis J. Optimization of the Expansion and Differentiation of Rabbit Chondrocytes In Vitro. Cartilage 2012; 3:181-7. [PMID: 26069631 PMCID: PMC4297132 DOI: 10.1177/1947603511420999] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE To develop a tissue culture expansion method for rabbit chondrocytes that promotes robust expansion while preserving chondrogenic potential. DESIGN Rabbit chondrocytes isolated from articular or auricular chondrocytes were assessed for chondrogenic differentiation potential versus population doubling using different expansion and differentiation conditions. Expansion conditions included serum alone, serum plus basic fibroblast growth factor 2 (FGF-2), and serum plus insulin-like growth factor 1 (IGF-1) and FGF-2. Differentiation conditions consisted of defined medium with and without bone morphogenetic protein 2 (BMP-2). RESULTS Nonsupplemented chondrocytes showed limited expandability, whereas supplementation with FGF-2 allowed articular chondrocytes to be expanded past 10 population doublings (PDs) and allowed auricular chondrocytes to expand past 15 population doublings. Differentiation, as measured by glycosaminoglycan production in aggregate cultures, was minimal in articular chondrocytes without BMP-2 supplementation and diminished to less than 50% maximal in auricular chondrocytes by PD 20. However, when FGF-2 was used during expansion and BMP-2 used during differentiation, both articular and auricular chondrocytes retained greater than 50% maximal differentiation for more than 25 PDs. The addition of IGF-1 to FGF-2 during expansion decreased chondrogenicity of auricular chondrocytes exposed to BMP-2, whereas for articular chondrocytes, chondrogenic expression increased. CONCLUSION These results demonstrate that FGF-2, for expansion, and BMP-2, for differentiation, dramatically increase the functional expansion of auricular and articular chondrocytes and provide a methodology to expand sufficient numbers of chondrocytes for tissue engineering applications.
Collapse
Affiliation(s)
- T. Mounts
- Case Western Reserve University, Cleveland, OH, USA
| | - N. Ginley
- Case Western Reserve University, Cleveland, OH, USA
| | | | - J.E. Dennis
- Case Western Reserve University, Cleveland, OH, USA,Benaroya Research Institute, Seattle, WA, USA
| |
Collapse
|
23
|
Responte DJ, Arzi B, Natoli RM, Hu JC, Athanasiou KA. Mechanisms underlying the synergistic enhancement of self-assembled neocartilage treated with chondroitinase-ABC and TGF-β1. Biomaterials 2012; 33:3187-94. [PMID: 22284584 DOI: 10.1016/j.biomaterials.2012.01.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/09/2012] [Indexed: 11/26/2022]
Abstract
Developing a platform for in vitro cartilage formation would enhance the study of cartilage development, pathogenesis, and regeneration. To improve neocartilage formation, our group developed a novel self-assembly process for articular chondrocytes, which has been improved in this study using a novel combination of catabolic and anabolic agents. TGF-β1 was applied in conjunction with the enzyme chondroitinase-ABC (C-ABC) to additively increase tensile properties and synergistically enhance collagen content. Additionally, microarray analysis indicated that TGF-β1 up-regulated MAPK signaling in contrast to C-ABC, which did not enrich genetic pathways. The lack of genetic signaling spurred investigation of the biophysical role of C-ABC, which showed that C-ABC treatment increased collagen fibril diameter and density. After four weeks of culture in nude mice, neocartilage exhibited stability and maturation. This study illustrated an innovative strategy for improving in vitro and in vivo articular cartilage formation and elucidated mechanisms underlying TGF-β1 and C-ABC treatment.
Collapse
Affiliation(s)
- Donald J Responte
- Department of Biomedical Engineering, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
24
|
Xu C, Zhang Z, Wu M, Zhu S, Gao J, Zhang J, Yuan Y, Zhang K, Yu Y, Han W. Recombinant human midkine stimulates proliferation and decreases dedifferentiation of auricular chondrocytes in vitro. Exp Biol Med (Maywood) 2011; 236:1254-62. [DOI: 10.1258/ebm.2011.011022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Autologous chondrocyte implantation (ACI) is widely used for the repair of cartilage defects. However, due to the lack of chondrocyte growth factor and dedifferentiation of the cultured primary chondrocytes, cell source has limited the clinical potential of ACI. Auricular cartilage is an attractive potential source of cells for cartilage tissue engineering. Here we demonstrated that recombinant human midkine (rhMK) significantly promoted proliferation of rat primary auricular chondrocytes cultured and passaged in monolayer, which was mediated by the activation of mitogen-activated protein kinase and phosphoinositide 3-kinase pathways. Furthermore, rhMK attenuated the dedifferentiation of cultured chondrocytes by maintaining the expression of chondrocyte-specific matrix proteins during culture expansion and passage. Importantly, rhMK-expanded chondrocytes reserved their full chondrogenic potential and redifferentiated into elastic chondrocytes after being cultured in high density. The results suggest that rhMK may be used for the preparation of chondrocytes in cartilage tissue engineering.
Collapse
Affiliation(s)
- Chuanying Xu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology
| | - Zhonghui Zhang
- Laboratory of Regeneromics, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240
| | - Mingyuan Wu
- Laboratory of Regeneromics, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240
| | - Shunying Zhu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology
| | - Jin Gao
- Laboratory of Regeneromics, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240
| | - Jing Zhang
- Laboratory of Regeneromics, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240
| | - Yunsheng Yuan
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology
| | - Kejian Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yan Yu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology
| | - Wei Han
- Laboratory of Regeneromics, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240
| |
Collapse
|
25
|
Jonitz A, Lochner K, Peters K, Salamon A, Pasold J, Mueller-Hilke B, Hansmann D, Bader R. Differentiation capacity of human chondrocytes embedded in alginate matrix. Connect Tissue Res 2011; 52:503-11. [PMID: 21787134 DOI: 10.3109/03008207.2011.593673] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Healing capacity of cartilage is low. Thus, cartilage defects do not regenerate as hyaline but mostly as fibrous cartilage which is a major drawback since this tissue is not well adapted to the mechanical loading within the joint. During in vitro cultivation in monolayers, chondrocytes proliferate and de-differentiate to fibroblasts. In three-dimensional cell cultures, de-differentiated chondrocytes could re-differentiate toward the chondrogenic lineage and re-express the chondrogenic phenotype. The objective of this study was to characterize the mesenchymal stem cell (MSC) potential of human chondrocytes isolated from articular cartilage. Furthermore, the differentiation capacity of human chondrocytes in three-dimensional cell cultures was analyzed to target differentiation direction into hyaline cartilage. After isolation and cultivation of chondrogenic cells, the expression of the MSC-associated markers: cluster of differentiation (CD)166, CD44, CD105, and CD29 was performed by flow cytometry. The differentiation capacity of human chondrocytes was analyzed in alginate matrix cultured in Dulbecco?s modified eagle medium with (chondrogenic stimulation) and without (control) chondrogenic growth factors. Additionally, the expression of collagen type II, aggrecan, and glycosaminoglycans was determined. Cultivated chondrocytes showed an enhanced expression of the MSC-associated markers with increasing passages. After chondrogenic stimulation in alginate matrix, the chondrocytes revealed a significant increase of cell number compared with unstimulated cells. Further, a higher synthesis rate of glycosaminoglycans and a positive collagen type II and aggrecan immunostaining was detected in stimulated alginate beads. Human chondrocytes showed plasticity whilst cells were encapsulated in alginate and stimulated by growth factors. Stimulated cells demonstrated characteristics of chondrogenic re-differentiation due to collagen type II and aggrecan synthesis.
Collapse
Affiliation(s)
- Anika Jonitz
- Department of Orthopaedics, Biomechanics and Implant Technology Research Laboratory, University of Rostock, Rostock, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Ng KW, O'Conor CJ, Kugler LE, Cook JL, Ateshian GA, Hung CT. Transient supplementation of anabolic growth factors rapidly stimulates matrix synthesis in engineered cartilage. Ann Biomed Eng 2011; 39:2491-500. [PMID: 21833681 DOI: 10.1007/s10439-011-0356-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 07/08/2011] [Indexed: 12/29/2022]
Abstract
The purpose of the presented work is to examine the response of engineered cartilage to a transient, 2-week application of anabolic growth factors compared to continuous exposure in in vitro culture. Immature bovine chondrocytes were suspended in agarose hydrogel and cultured for 28 days (Study 1) or 42 days (Study 2) in chondrogenic media with TGF-β1, TGF-β3, or IGF-I either added for only the first 14 days in culture or added to the media for the entire study period. In both studies, there were no statistical differences in tissue mechanical or biochemical properties between the growth factors on day 14. In Study 1, growth factor removal led to a significant and drastic increase in Young's modulus and glycosaminoglycans content compared to continuously exposed controls on day 28. In Study 2, both TGF-β1 and β3 led to significantly higher mechanical properties and collagen content vs. IGF-I on day 42. These results indicate that the rapid rise in tissue properties (previously observed with TGF-β3 only) is not dependent on the type but rather the temporal application of the anabolic growth factor. These findings shed light on possible techniques to rapidly develop engineered cartilage tissue for the future treatment of osteoarthritis.
Collapse
Affiliation(s)
- Kenneth W Ng
- Research Division, Hospital for Special Surgery, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
27
|
Dickhut A, Dexheimer V, Martin K, Lauinger R, Heisel C, Richter W. Chondrogenesis of human mesenchymal stem cells by local transforming growth factor-beta delivery in a biphasic resorbable carrier. Tissue Eng Part A 2010; 16:453-64. [PMID: 19705961 DOI: 10.1089/ten.tea.2009.0168] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Little is known about the potential of growth factor-augmented biphasic implants composed of a gel and a solid scaffold to enhance chondrogenesis of mesenchymal stem cells (MSCs). We analyzed whether a collagen type I/III carrier and fibrin glue (FG) combined to a biphasic construct support in vitro chondrogenesis of MSCs and allow for local release of bioactive transforming growth factor-beta1 (TGF-beta1). Further, a possible advantage of partial autologous fibrin glue (PAF) over commercial FG was assessed. Collagen carriers seeded with 5 x 10(5) human MSCs with or without FG, PAF, or TGF-beta1-upgraded FG were cultured for 6 weeks in chondrogenic medium with or without TGF-beta1. Pellets with or without FG/PAF served as controls. FG and collagen carriers allowed strong upregulation of COL2A1, AGC, and COL10A1 mRNA, deposition of collagen-type II, and mediated a significantly higher proteoglycan content compared with biomaterial-free pellets. Collagen-carrier groups contained significantly more proteoglycan than FG and PAF pellets, whereas biphasic PAF-carrier constructs were inferior to FG-carrier constructs. Upgrading of biphasic FG-carrier constructs with 50 ng TGF-beta1/construct mediated chondrogenesis as successfully as supply of TGF-beta1 via the medium. In conclusion, the biphasic carrier constructs showed a high biofunctionality by continuous form stability with improved chondrogenesis and long-term local supply of bioactive TGF-beta1 which may be useful to enhance matrix-assisted repair strategies for damaged cartilage.
Collapse
Affiliation(s)
- Andrea Dickhut
- Division of Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Noh MJ, Copeland RO, Yi Y, Choi KB, Meschter C, Hwang S, Lim CL, Yip V, Hyun JP, Lee HY, Lee KH. Pre-clinical studies of retrovirally transduced human chondrocytes expressing transforming growth factor-beta-1 (TG-C). Cytotherapy 2010; 12:384-93. [PMID: 20370350 DOI: 10.3109/14653240903470639] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS The aim was to evaluate cartilage regeneration in animal models involving induced knee joint damage. Through cell-mediated gene therapy methods, a cell mixture comprising a 3:1 ratio of genetically unmodified human chondrocytes and transforming growth factor beta-1 (TGF-beta1)-secreting human chondrocytes (TG-C), generated via retroviral transduction, resulted in successful cartilage proliferation in damaged regions. METHODS Non-clinical toxicology assessments for efficacy, biodistribution and local/systemic toxicity of single intra-articular administration of the cell mixture in mice, rabbits and goats was conducted. RESULTS Administration of the mixture was tolerated well in all of the species. There was evidence of cartilage proliferation in rabbits and goats. As an additional precautionary step, the efficacy of TGF-beta1 secretion in irradiated human chondrocytes was also demonstrated. CONCLUSIONS Four studies in rabbits and goats demonstrated the safety and efficacy of TG-C following direct intra-articular administration in animal models involving induced knee joint damage. Based on these pre-clinical studies authorization has been received from the USA Food and Drug Administration (FDA) to proceed with an initial phase I clinical study of TG-C for degenerative arthritis.
Collapse
|
29
|
Choi KH, Choi BH, Park SR, Kim BJ, Min BH. The chondrogenic differentiation of mesenchymal stem cells on an extracellular matrix scaffold derived from porcine chondrocytes. Biomaterials 2010; 31:5355-65. [DOI: 10.1016/j.biomaterials.2010.03.053] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 03/21/2010] [Indexed: 12/20/2022]
|
30
|
van der Windt AE, Jahr H, Farrell E, Verhaar JAN, Weinans H, van Osch GJVM. Calcineurin inhibitors promote chondrogenic marker expression of dedifferentiated human adult chondrocytes via stimulation of endogenous TGFbeta1 production. Tissue Eng Part A 2010; 16:1-10. [PMID: 19604038 DOI: 10.1089/ten.tea.2009.0082] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In vitro chondrocyte expansion is required for several cell-based approaches for the repair of chondral lesions. During expansion, loss of chondrogenic phenotype takes place (dedifferentiation). The objective of this study was to investigate calcineurin (Cn) as a potential target to improve chondrocyte phenotype for cartilage repair purposes. Cn activity in human articular chondrocytes was significantly increased during dedifferentiation and decreased during redifferentiation in vitro. Inhibition of Cn activity by FK506 increased the expression of chondrogenic markers collagen type 2, aggrecan, and SOX9 in culture-expanded cells. Addition of FK506 increased endogenous transforming growth factor 2 (TGF) beta1 expression on both mRNA and protein level. The effect of FK506 on chondrogenic markers was abolished by addition of anti-TGFbeta1 antibody, indicating that the endogenous TGFbeta1 was necessary to increase chondrogenic marker expression. We also showed that chondrocyte redifferentiation by TGFbeta requires calcium influx and does not depend on changes in Cn activity. In conclusion, inhibition of Cn activity by FK506 increases the expression of chondrogenic markers via endogenous TGFbeta1 production in human articular chondrocytes. Cn inhibitors might be an alternative for the application of (recombinant) TGFbeta, to promote chondrocyte phenotype for cell-based cartilage repair procedures.
Collapse
Affiliation(s)
- Anna E van der Windt
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
31
|
Bernstein P, Dong M, Graupner S, Graupher S, Corbeil D, Gelinsky M, Günther KP, Fickert S. Sox9 expression of alginate-encapsulated chondrocytes is stimulated by low cell density. J Biomed Mater Res A 2010; 91:910-8. [PMID: 19097150 DOI: 10.1002/jbm.a.32308] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recent research in tissue engineering for the treatment of cartilage defects have demonstrated that matrix-biomaterial, cell culture conditions, and cytokine-related factors influence the chondrogenic differentiation pattern, especially for the expression of matrix genes. However, little is known about the impact of cell seeding density in a three-dimensional environment on the key chondrogenic transcription factor Sox9. Here we investigated, whether the cell concentration of alginate encapsulated chondrocytes influences the Sox9 expression. Dedifferentiated passage-4 porcine chondrocytes were encapsulated in alginate beads at two different concentrations (4 x 10(6) versus 7 x 10(7) cells/mL) and cultivated for up to 4 weeks under TGF-ss stimulation. The expression of Sox9, Collagen I, II, and X was assessed via quantitative RT-PCR and compared to those observed in the initial monolayer culture. Cellular viability, cell morphology, and the sulphated glycosaminoglycan-production were monitored. Interestingly Sox9 expression was significantly upregulated in the low-cell-density group, whereas no difference between high-cell-density and monolayer culture group could be observed. Furthermore, the cellular survival and the sulphated glycosaminoglycan production were higher in the low-cell-density group. Collagen I expression was downregulated in the low-cell-density group whereas it was upregulated in the high-cell-density one. Surprisingly, only the high-cell-density group showed the expression of Collagen II, although it appeared not significant. Collagen X expression was upregulated in the low-cell-density group. Taken together our data indicate that a low concentration of cell seeding in a three-dimensional environment is beneficial for the overall chondrogenic development. However, this article reveals discrepancies between Sox9 and the chondrogenic pathway in redifferentiating chondrocytes that should be addressed in further work.
Collapse
Affiliation(s)
- Peter Bernstein
- Department of Orthopaedic Surgery, University Hospital Carl Gustav Carus Dresden, Medical Faculty of Technical University Dresden, Dresden, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Giovannini S, Diaz-Romero J, Aigner T, Mainil-Varlet P, Nesic D. Population doublings and percentage of S100-positive cells as predictors of in vitro chondrogenicity of expanded human articular chondrocytes. J Cell Physiol 2009; 222:411-20. [PMID: 19890919 DOI: 10.1002/jcp.21965] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aim of this study was to investigate the interconnection between the processes of proliferation, dedifferentiation, and intrinsic redifferentiation (chondrogenic) capacities of human articular chondrocyte (HAC), and to identify markers linking HAC dedifferentiation status with their chondrogenic potential. Cumulative population doublings (PD) of HAC expanded in monolayer culture were determined, and a threshold range of 3.57-4.19 PD was identified as indicative of HAC loss of intrinsic chondrogenic capacity in pellets incubated without added chondrogenic factors. While several specific gene and surface markers defined early HAC dedifferentiation process, no clear correlation with the loss of intrinsic chondrogenic potential could be established. CD90 expression during HAC monolayer culture revealed two subpopulations, with sorted CD90-negative cells showing lower proliferative capacity and higher chondrogenic potential compared to CD90-positive cells. Although these data further validated PD as critical for in vitro chondrogenesis, due to the early shift in expression, CD90 could not be considered for predicting chondrogenic potential of HAC expanded for several weeks. In contrast, an excellent mathematically modeled correlation was established between PD and the decline of HAC expressing the intracellular marker S100, providing a direct link between the number of cell divisions and dedifferentiation/loss of intrinsic chondrogenic capacity. Based on the dynamics of S100-positive HAC during expansion, we propose asymmetric cell division as a potential mechanism of HAC dedifferentiation, and S100 as a marker to assess chondrogenicity of HAC during expansion, of potential value for cell-based cartilage repair treatments.
Collapse
|
33
|
Rai MF, Rachakonda PS, Manning K, Palissa C, Sittinger M, Ringe J, Schmidt MFG. Molecular and phenotypic modulations of primary and immortalized canine chondrocytes in different culture systems. Res Vet Sci 2009; 87:399-407. [PMID: 19439332 DOI: 10.1016/j.rvsc.2009.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 03/24/2009] [Accepted: 04/14/2009] [Indexed: 11/24/2022]
Abstract
This study was conducted to determine physiological and functional features of primary and immortalized canine chondrocytes. Chondrocytes were immortalized by introducing the catalytic component of human telomerase namely human telomerase reverse transcriptase (hTERT). Primary chondrocytes lost their characteristic phenotype and growth properties whereas the immortalized cells remained polygonal with rapid growth rate. The expression of chondrocyte-specific markers decreased many-fold whereas that of chondrocyte-non-specific gene increased in primary chondrocytes. The immortalized cells did not express chondrocyte-specific genes in monolayers. Both primary and immortalized cells were encapsulated in alginate microspheres to construct three-dimensional (3D) culture system. As the primary chondrocytes, also the telomerase-transfected cells adopted a chondrocyte-specific gene expression pattern in alginate culture. Thus, the expression of telomerase represents possibility to expand chondrocytes without limitation while maintaining the chondrocyte-specific phenotype in 3D cultures. Use of such cells provides a standardized tool for testing different tissue engineering applications in canine model.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Institute of Immunology and Molecular Biology, Faculty of Veterinary Medicine, Freie Universität, House 18, Philippstrasse 13, D-10115 Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Bastiaansen-Jenniskens YM, Koevoet W, De Bart ACW, Zuurmond AM, Bank RA, Verhaar JAN, DeGroot J, van Osch GJVM. TGFbeta affects collagen cross-linking independent of chondrocyte phenotype but strongly depending on physical environment. Tissue Eng Part A 2009. [PMID: 19230128 DOI: 10.1089/tea.2007.0345] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transforming growth factor beta (TGFbeta) is often used in cartilage tissue engineering to increase matrix formation by cells with various phenotypes. However, adverse effects of TGFbeta, such as extensive crosslinking in cultured fibroblasts, have also been reported. Our goal was to study effects of TGFbeta on collagen cross-linking and evaluating the role of cellular phenotype and physical environment. We therefore used four different cell populations in two very different physical environments: primary and expanded chondrocytes and fibroblasts embedded in alginate gel and attached to tissue culture plastic. Matrix production, collagen cross-linking, and alpha-smooth muscle actin (alphaSMA) were analyzed during 4 weeks with or without 2.5 ng/ mL TGFbeta2. TGFbeta2 did not affect collagen deposition by primary cells. In expanded cells, TGFbeta2 increased collagen deposition. Chondrocytes and fibroblasts in monolayer produced more collagen cross-links with TGFbeta2. In alginate, primary and expanded cells displayed an unexpected decrease in collagen cross-linking with TGFbeta2. alphaSMA was not present in alginate cultures and barely upregulated by TGFbeta2. Organized alphaSMA fibers were present in all monolayer cultures and became more pronounced with TGFbeta2. This study demonstrates that the physical environment determined by the substrate used co-determines the response of cells to TGFbeta. The presence of mechanical stress, determined with alphaSMA-staining, is probably responsible for the increase in collagen cross-linking upon addition of TGFbeta.
Collapse
|
35
|
Bastiaansen-Jenniskens YM, Koevoet W, de Bart AC, Zuurmond AM, Bank RA, Verhaar JA, DeGroot J, van Osch GJ. TGFβ Affects Collagen Cross-Linking Independent of Chondrocyte Phenotype but Strongly Depending on Physical Environment. Tissue Eng Part A 2008; 14:1059-66. [DOI: 10.1089/ten.tea.2007.0345] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yvonne Maria Bastiaansen-Jenniskens
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- TNO Quality of Life, Business Unit BioSciences, Leiden, The Netherlands
| | - Wendy Koevoet
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | - Ruud A. Bank
- TNO Quality of Life, Business Unit BioSciences, Leiden, The Netherlands
- Department of Oral Cell Biology, Academic Center of Dentistry, Amsterdam, The Netherlands
| | - Jan A.N. Verhaar
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeroen DeGroot
- TNO Quality of Life, Business Unit BioSciences, Leiden, The Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
36
|
Tai TF, Chan CP, Lin CC, Chen LI, Jeng JH, Chang MC. Transforming Growth Factor β2 Regulates Growth and Differentiation of Pulp Cells via ALK5/Smad2/3. J Endod 2008; 34:427-32. [DOI: 10.1016/j.joen.2008.02.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2007] [Revised: 01/29/2008] [Accepted: 02/01/2008] [Indexed: 11/26/2022]
|
37
|
Lima EG, Bian L, Ng KW, Mauck RL, Byers BA, Tuan RS, Ateshian GA, Hung CT. The beneficial effect of delayed compressive loading on tissue-engineered cartilage constructs cultured with TGF-beta3. Osteoarthritis Cartilage 2007; 15:1025-33. [PMID: 17498976 PMCID: PMC2724596 DOI: 10.1016/j.joca.2007.03.008] [Citation(s) in RCA: 185] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2006] [Accepted: 03/11/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine whether the functional properties of tissue-engineered constructs cultured in a chemically-defined medium supplemented briefly with TGF-beta3 can be enhanced with the application of dynamic deformational loading. METHODS Primary immature bovine cells (2-3 months old) were encapsulated in agarose hydrogel (2%, 30 x 10(6)cells/ml) and cultured in chemically-defined medium supplemented for the first 2 weeks with transforming growth factor beta 3 (TGF-beta3) (10 microg/ml). Physiologic deformational loading (1 Hz, 3 h/day, 10% unconfined deformation initially and tapering to 2% peak-to-peak deformation by day 42) was applied either concurrent with or after the period of TGF-beta3 supplementation. Mechanical and biochemical properties were evaluated up to day 56. RESULTS Dynamic deformational loading applied concurrently with TGF-beta3 supplementation yielded significantly lower (-90%) overall mechanical properties when compared to free-swelling controls. In contrast, the same loading protocol applied after the discontinuation of the growth factor resulted in significantly increased (+10%) overall mechanical properties relative to free-swelling controls. Equilibrium modulus values reach 1306+/-79 kPa and glycosaminoglycan levels reach 8.7+/-1.6% w.w. during this 8-week period and are similar to host cartilage properties (994+/-280 kPa, 6.3+/-0.9% w.w.). CONCLUSIONS An optimal strategy for the functional tissue engineering of articular cartilage, particularly to accelerate construct development, may incorporate sequential application of different growth factors and applied deformational loading.
Collapse
Affiliation(s)
- E G Lima
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Xu X, Urban JPG, Browning JA, Tirlapur U, Wilkins RJ, Wu MH, Cui Z, Cui Z. Influences of buffer systems on chondrocyte growth during long-term culture in alginate. Osteoarthritis Cartilage 2007; 15:396-402. [PMID: 17070714 DOI: 10.1016/j.joca.2006.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Accepted: 09/17/2006] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Chondrocyte behavior is very sensitive to culture environment such as physical and biochemical conditions. As extracellular pH (pHo) and the existence of bicarbonate could affect the chondrocyte fate, hence, the purpose of this study is to investigate the buffer system effect on chondrocyte fate during relatively long-term culture. METHODS In order to examine whether effects seen were due to bicarbonate or to pHo, we had to devise a system which could differentiate between the two effects. Culture media buffered by N-2-hydroxyethyl piperazine-N'-2-ethanesulfonic acid (HEPES) only and the combination of HEPES and bicarbonate were used. Bovine articular chondrocytes were cultured in alginate beads for up to 12 days. pHo was kept constant by culture of 3 beads in 2 ml culture medium. Cell density, intracellular pH (pHi) and glycosaminoglycan (GAG) were measured at day 5 and day 12. Cell morphology, distribution and viability in alginate beads were monitored over 12 days of culture. RESULTS Compared to culture in the absence of bicarbonate, a higher proliferation rate of chondrocytes was observed in the presence of bicarbonate. pHi was more alkaline, about 0.2 pH unit, in the presence of bicarbonate than that in the absence of bicarbonate. About 50% more GAG was deposited in alginate beads when chondrocytes were cultured in the combination of HEPES and bicarbonate, compared to chondrocytes cultured in the absence of NaHCO3 at the end of 12 days of culture. CONCLUSION The presence of bicarbonate results in more alkaline in the pHi of bovine chondrocytes after long-term culture. The combination of bicarbonate and HEPES in culture medium improves cell growth, matrix production in three-dimensional alginate beads.
Collapse
Affiliation(s)
- X Xu
- Department of Engineering Science, University of Oxford, Parks Road, Oxford OX1 3PJ, UK
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Han F, Gilbert JR, Harrison G, Adams CS, Freeman T, Tao Z, Zaka R, Liang H, Williams C, Tuan RS, Norton PA, Hickok NJ. Transforming growth factor-beta1 regulates fibronectin isoform expression and splicing factor SRp40 expression during ATDC5 chondrogenic maturation. Exp Cell Res 2007; 313:1518-32. [PMID: 17391668 PMCID: PMC1920702 DOI: 10.1016/j.yexcr.2007.01.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2005] [Revised: 01/09/2007] [Accepted: 01/17/2007] [Indexed: 11/28/2022]
Abstract
Fibronectin (FN) isoform expression is altered during chondrocyte commitment and maturation, with cartilage favoring expression of FN isoforms that includes the type II repeat extra domain B (EDB) but excludes extra domain A (EDA). We and others have hypothesized that the regulated splicing of FN mRNAs is necessary for the progression of chondrogenesis. To test this, we treated the pre-chondrogenic cell line ATDC5 with transforming growth factor-beta1, which has been shown to modulate expression of the EDA and EDB exons, as well as the late markers of chondrocyte maturation; it also slightly accelerates the early acquisition of a sulfated proteoglycan matrix without affecting cell proliferation. When chondrocytes are treated with TGF-beta1, the EDA exon is preferentially excluded at all times whereas the EDB exon is relatively depleted at early times. This regulated alternative splicing of FN correlates with the regulation of alternative splicing of SRp40, a splicing factor facilitating inclusion of the EDA exon. To determine if overexpression of the SRp40 isoforms altered FN and FN EDA organization, cDNAs encoding these isoforms were overexpressed in ATDC5 cells. Overexpression of the long-form of SRp40 yielded an FN organization similar to TGF-beta1 treatment; whereas overexpression of the short form of SRp40 (which facilitates EDA inclusion) increased formation of long-thick FN fibrils. Therefore, we conclude that the effects of TGF-beta1 on FN splicing during chondrogenesis may be largely dependent on its effect on SRp40 isoform expression.
Collapse
Affiliation(s)
- Fei Han
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - James R. Gilbert
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Gerald Harrison
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Christopher S. Adams
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Theresa Freeman
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Zhuliang Tao
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Raihana Zaka
- Division of Rheumatology, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Hongyan Liang
- Department of Biochemistry & Molecular Pharmacology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Charlene Williams
- Department of Biochemistry & Molecular Pharmacology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Division of Rheumatology, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rocky S. Tuan
- Cartilage Biology and Orthopaedics Branch, NIAMS, NIH, Bethesda, MD
| | - Pamela A. Norton
- Department of Biochemistry & Molecular Pharmacology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Jefferson Center for Biomedical Research, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Noreen J. Hickok
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- ‡ To Whom Correspondence Should be Addressed: Noreen J. Hickok, Ph.D., Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut St., Suite 501, Philadelphia, PA 19107, Tel: 215-955-6979, Fax: 215-955-4317, e-mail:
| |
Collapse
|
40
|
Jenniskens YM, Koevoet W, de Bart ACW, Weinans H, Jahr H, Verhaar JAN, DeGroot J, van Osch GJVM. Biochemical and functional modulation of the cartilage collagen network by IGF1, TGFbeta2 and FGF2. Osteoarthritis Cartilage 2006; 14:1136-46. [PMID: 16730198 DOI: 10.1016/j.joca.2006.04.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Accepted: 04/04/2006] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Examine effects of insulin-like growth factor 1 (IGF1), transforming growth factor beta2 (TGFbeta2) and fibroblast growth factor 2 (FGF2) on proteoglycan and collagen network and biomechanical properties of the newly formed cartilage matrix. METHODS Bovine articular chondrocytes were cultured in alginate beads for 3 weeks with or without FGF2, TGFbeta2 or IGF1 in the presence of 10% FCS. Proteoglycan content, collagen content, hydroxylysylpyridinoline cross-links and overall matrix metalloproteinase (MMP) activity in the culture medium were measured. Alginate disks cultured for 5 weeks were used to evaluate the effect of growth factors on mechanical properties of the construct by determining the equilibrium aggregate modulus and secant modulus. RESULTS IGF1 increased collagen and proteoglycan deposition. FGF2 mainly decreased collagen deposition and TGFbeta2 proteoglycan deposition. A decrease in cross-links was observed in matrix produced by chondrocytes cultured in the presence of TGFbeta2. IGF1 and FGF2 had no influence on the number of cross-links per collagen molecule. Overall MMP activity was significantly higher in culture medium of cells cultured with FGF2. TGFbeta2 and IGF1 had no effect on MMP activity. After 35 days of culture, the matrix produced under influence of IGF1 had a lower permeability and a trend to increase stiffness. FGF2 showed a trend to lower both properties. TGFbeta2 had no effect on these parameters. CONCLUSION IGF1, TGFbeta2 and FGF2 had differential effects on collagen network formation. Of the three growth factors tested, IGF1 seems to be best in promoting the formation of a functional collagen network since it increased proteoglycan and collagen deposition and improved the mechanical properties.
Collapse
Affiliation(s)
- Y M Jenniskens
- Erasmus MC, University Medical Centre Rotterdam, Department of Orthopaedics, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Shakibaei M, Seifarth C, John T, Rahmanzadeh M, Mobasheri A. Igf-I extends the chondrogenic potential of human articular chondrocytes in vitro: Molecular association between Sox9 and Erk1/2. Biochem Pharmacol 2006; 72:1382-95. [PMID: 17010943 DOI: 10.1016/j.bcp.2006.08.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 08/21/2006] [Accepted: 08/22/2006] [Indexed: 10/24/2022]
Abstract
Expansion of articular chondrocytes in monolayer culture leads to loss of the unique chondrocyte phenotype and the cells' redifferentiation capacity. Dedifferentiation of chondrocytes in monolayer culture is a challenging problem for autologous chondrocyte transplantation (ACT). It is well established that Igf-I exerts positive anabolic effects on chondrocytes in vivo and in vitro. Accordingly, in this study, we examined whether the anabolic insulin-like growth factor-I (Igf-I) is capable of extending the chondrogenic potential of dedifferentiated chondrocytes in vitro. Chondrocyte monolayers were cultured up to 10 passages. At each passage chondrocytes were stimulated with Igf-I (10ng/ml) and introduced to high-density cultures for up to 7 days. Expression of collagen type II, cartilage-specific proteoglycans, activated caspase-3, integrin beta1, extracellular signal-regulated kinase (Erk) and Sox9 was examined by Western blotting, immunoprecipitation and immunomorphological techniques. Monolayer chondrocytes rapidly lost their differentiated phenotype. When introduced to high-density cultures, only chondrocytes from P1-P4 redifferentiated. In contrast, Igf-I treated cells from P1 up to P7 redifferentiated and formed cartilage-like tissue in high-density culture. P8-P10 cells exhibited apoptotic alterations and produced significantly less matrix. Igf-I markedly increased expression of integrin beta1, Erk and Sox9. Immunoprecipitation revealed that phosphorylated Erk1/2 physically interacts with Sox9 in chondrocyte nuclei, suggesting a previously unreported functional association which was markedly enhanced by Igf-I. Treatment of chondrocyte cultures with Igf-I stabilizes chondrogenic potential, stimulates Sox9 and promotes molecular interactions between Erk and Sox9. These effects appear to be regulated by the integrin/MAPK signaling pathways.
Collapse
Affiliation(s)
- Mehdi Shakibaei
- Institute of Anatomy, Ludwig-Maximilians-University, Pettenkoferstrasse 11, 80336 Munich, Germany.
| | | | | | | | | |
Collapse
|
42
|
Terada S, Fuchs JR, Yoshimoto H, Fauza DO, Vacanti JP. In Vitro Cartilage Regeneration From Proliferated Adult Elastic Chondrocytes. Ann Plast Surg 2005; 55:196-201. [PMID: 16034253 DOI: 10.1097/01.sap.0000164388.33965.4e] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The purpose of this study was to investigate cellular feasibility in the proliferation and differentiation status of adult chondrocytes for cartilage regeneration in comparison to fetal chondrocytes. Primary cells were isolated from adult (n = 6) and fetal (n = 6) sheep ear cartilages and expanded in 10% fetal bovine serum (FBS) containing Ham's F12 medium, in which adult and fetal cell proliferation rates were compared using a WST-1 assay kit. Approximately 4 million cells were seeded onto each 1 x 1 x 0.2-cm (200 microL) nonwoven fabric scaffold made from polyglycolic acid. Cell/polymer constructs were cultured in serum-free DMEM/F12 medium supplemented with 5 ng/mL TGF-beta2 and 5 ng/mL des(1-3)IGF-I (adult chondrocytes, group A) or in 10% FBS containing Ham's F12 medium (adult chondrocytes, group B, and fetal chondrocytes, group C) as controls in a rotating bioreactor for 6 weeks. The proliferation assay showed that fetal cells had a significantly better growth potential than did adult cells. Histology and extracellular matrix analyses revealed that groups A and C qualitatively displayed better matrix deposition than did group B. In conclusion, although adult sheep elastic chondrocytes had less growth potential than did fetal cells, the serum-free medium supplemented with growth factors significantly enhanced the production of cartilage matrix secreted from proliferated adult sheep elastic chondrocytes.
Collapse
Affiliation(s)
- Shinichi Terada
- Laboratory for Tissue Engineering and Organ Fabrication, Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
43
|
Li WJWJ, Tuli R, Okafor C, Derfoul A, Danielson KGKG, Hall DJDJ, Tuan RSRS. A three-dimensional nanofibrous scaffold for cartilage tissue engineering using human mesenchymal stem cells. Biomaterials 2005; 26:599-609. [PMID: 15282138 DOI: 10.1016/j.biomaterials.2004.03.005] [Citation(s) in RCA: 601] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Accepted: 03/13/2004] [Indexed: 12/16/2022]
Abstract
The utilization of adult stem cells in tissue engineering is a promising solution to the problem of tissue or organ shortage. Adult bone marrow derived mesenchymal stem cells (MSCs) are undifferentiated, multipotential cells which are capable of giving rise to chondrocytes when maintained in a three-dimensional culture and treated with members of the transforming growth factor-beta (TGF-beta) family of growth factors. In this study, we fabricated a nanofibrous scaffold (NFS) made of a synthetic biodegradable polymer, poly(-caprolactone) (PCL), and examined its ability to support in vitro chondrogenesis of MSCs. The electrospun PCL porous scaffold was constructed of uniform, randomly oriented nanofibers with a diameter of 700 nm, and structural integrity of this scaffold was maintained over a 21-day culture period. MSCs cultured in NFSs in the presence of TGF-beta1 differentiated to a chondrocytic phenotype, as evidenced by chondrocyte-specific gene expression and synthesis of cartilage-associated extracellular matrix (ECM) proteins. The level of chondrogenesis observed in MSCs seeded within NFSs was comparable to that observed for MSCs maintained as cell aggregates or pellets, a widely used culture protocol for studying chondrogenesis of MSCs in vitro. Due to the physical nature and improved mechanical properties of NFSs, particularly in comparison to cell pellets, the findings reported here suggest that the PCL NFS is a practical carrier for MSC transplantation, and represents a candidate scaffold for cell-based tissue engineering approaches to cartilage repair.
Collapse
Affiliation(s)
- W-J Wan-Ju Li
- Department of Health and Human Services, Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institute of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Tew SR, Li Y, Pothacharoen P, Tweats LM, Hawkins RE, Hardingham TE. Retroviral transduction with SOX9 enhances re-expression of the chondrocyte phenotype in passaged osteoarthritic human articular chondrocytes. Osteoarthritis Cartilage 2005; 13:80-9. [PMID: 15639641 DOI: 10.1016/j.joca.2004.10.011] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Accepted: 10/05/2004] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Articular chondrocytes proliferate in monolayer culture, but the expression of the transcription factor SOX9 falls and the ability of the cells to reform cartilage tissue declines. We have investigated whether retroviral SOX9 expression in extensively passaged human articular chondrocytes from osteoarthritic (OA) joints enables the cells to regain a cartilage matrix forming phenotype in pellet culture. DESIGN Chondrocytes from normal and OA joints were retrovirally transduced with SOX9 and grown to passages 7-10 before being cultured as pellets of 500,000 cells for 14 days. Pellets were analysed by real time polymerase chain reaction, histology, immunohistochemistry and 1,9-dimethylmethylene blue assay. RESULTS Chondrocytes from OA joints displayed higher expression of COL2A1 gene when transduced with SOX9 and cultured as pellets with 10% serum, but glycosaminoglycan (GAG) synthesis was low. Addition of transforming growth factor beta-3 and insulin like growth factor-1 increased collagen II expression and GAG synthesis in these SOX9 transduced cell pellets. The cells adopted a rounded morphology and there was increased deposition of collagen II protein compared to control green fluorescent protein transduced cell pellets. Similar results were seen with transduced chondrocytes from OA or healthy cartilage. SOX9 transduced human dermal fibroblasts did not show any chondrogenic response. DISCUSSION Transduction with SOX9 primed the passaged articular chondrocytes to regain a chondrocytic phenotype in pellet culture and to form a cartilaginous matrix, which was enhanced by growth factors. Following transduction, chondrocytes from OA joints showed a similar capacity for chondrogenic recovery as those from healthy joints, which suggested that OA does not permanently compromise the chondrocyte phenotype.
Collapse
Affiliation(s)
- Simon R Tew
- UK Centre for Tissue Engineering at The Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
An increasing number of patients are treated by autologous chondrocyte implantation (ACI). This study tests the hypothesis that culture within a defined chondrogenic medium containing TGF-β enhances the reexpression of a chondrocytic phenotype and the subsequent production of cartilaginous extracellular matrix by human chondrocytes used in ACI. Chondrocytes surplus to clinical requirements for ACI from 24 patients were pelleted and cultured in either DMEM (Dulbecco’s modified eagles medium)/ITS+Premix/TGF-β1 or DMEM/10%FCS (fetal calf serum) and were subsequently analysed biochemically and morphologically. Pellets cultured in DMEM/ITS+/TGF-β1 stained positively for type-II collagen, while those maintained in DMEM/10%FCS expressed type-I collagen. The pellets cultured in DMEM/ITS+/TGF-β1 were larger and contained significantly greater amounts of DNA and glycosaminoglycans. This study suggests that the use of a defined medium containing TGF-β is necessary to induce the re-expression of a differentiated chondrocytic phenotype and the subsequent stimulation of glycosaminoglycan and type-II collagen production by human monolayer expanded chondrocytes.
Collapse
Affiliation(s)
- A. J. Goldberg
- Interdisciplinary Research Centre, Institute of Orthopaedics, Royal Free & University College Medical School, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK
| | - D. A. Lee
- Medical Engineering Division and IRC in Biomedical Materials, Department of Engineering, Queen Mary, University of London, Mile End Road, London E1 4NS, UK
| | - D. L. Bader
- Medical Engineering Division and IRC in Biomedical Materials, Department of Engineering, Queen Mary, University of London, Mile End Road, London E1 4NS, UK
| | - G. Bentley
- Interdisciplinary Research Centre, Institute of Orthopaedics, Royal Free & University College Medical School, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK
| |
Collapse
|
46
|
Chowdhury TT, Salter DM, Bader DL, Lee DA. Integrin-mediated mechanotransduction processes in TGFbeta-stimulated monolayer-expanded chondrocytes. Biochem Biophys Res Commun 2004; 318:873-81. [PMID: 15147953 DOI: 10.1016/j.bbrc.2004.04.107] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Indexed: 11/17/2022]
Abstract
Previous studies have demonstrated that passage in monolayer detrimentally affects the response of articular chondrocytes to the application of dynamic compression. Transforming growth factor beta (TGFbeta) is known to regulate metabolic processes in articular cartilage and can enhance the re-expression of a chondrocytic phenotype following monolayer expansion. The current study tests the hypothesis that TGFbeta also modulates the response of monolayer-expanded human chondrocytes to the application of dynamic compression, via an integrin-mediated mechanotransduction process. The data presented demonstrate that TGFbeta3 enhanced 35SO4 and [3H]thymidine incorporation and inhibited nitrite release after 48 h of culture when compared to unsupplemented constructs. Dynamic compression also enhanced 35SO4 and [3H]thymidine incorporation and inhibited nitrite release in the presence of TGFbeta3. By contrast, dynamic compression did not alter these parameters in the absence of the growth factor. The addition of the peptide, GRGDSP, which acts as a competitive ligand for the alpha5beta1 integrin, reversed the compression-induced stimulation of 35SO4 incorporation, [3H]thymidine incorporation, and suppression of nitrite release. No effect was observed when the control peptide, GRADSP, was used. The current data clearly demonstrate that the dynamic compression-induced changes observed in cell metabolism for human monolayer-expanded chondrocytes were dependent on the presence of TGFbeta3 and are integrin-mediated.
Collapse
Affiliation(s)
- T T Chowdhury
- Medical Engineering Division and IRC in Biomedical Materials, Department of Engineering, Queen Mary, University of London, Mile End Road, London E1 4NS, UK.
| | | | | | | |
Collapse
|
47
|
Awad HA, Halvorsen YDC, Gimble JM, Guilak F. Effects of transforming growth factor beta1 and dexamethasone on the growth and chondrogenic differentiation of adipose-derived stromal cells. ACTA ACUST UNITED AC 2004; 9:1301-12. [PMID: 14670117 DOI: 10.1089/10763270360728215] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The effects of soluble mediators and medium supplements commonly used to induce chondrogenic differentiation in different cell culture systems were investigated to define their dose-response profiles and potentially synergistic effects on the chondrogenic differentiation of adipose-derived adult stromal (ADAS) cells. Human ADAS cells were suspended within alginate beads and cultured in basal medium with insulin, transferrin, and selenious acid (ITS+) or fetal bovine serum (FBS) and treated with different doses and combinations of TGF-beta1 (0, 1, and 10 ng/mL) and dexamethasone (0, 10, and 100 nM). Cell growth and chondrogenic differentiation were assessed by measuring DNA content, protein and proteoglycan synthesis rates, and proteoglycan accumulation. The combination of ITS+ and TGF-beta1 significantly increased cell proliferation. Protein synthesis rates were increased by TGF-beta1 and dexamethasone in the presence of ITS+ or FBS. While TGF-beta1 significantly increased proteoglycan synthesis and accumulation by 1.5- to 2-fold in the presence of FBS, such effects were suppressed by dexamethasone. In summary, the combination of TGF-beta1 and ITS+ stimulated cell growth and synthesis of proteins and proteoglycans by human ADAS cells. The addition of dexamethasone appeared to amplify protein synthesis but had suppressive effects on proteoglycan synthesis and accumulation.
Collapse
Affiliation(s)
- Hani A Awad
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
48
|
Abstract
Transforming growth factor-beta (TGF-beta) has multiple functions including increasing extracellular matrix deposition in fibrosis. It functions through a complex family of cell surface receptors that mediate downstream signaling. We report here that a transmembrane heparan sulfate proteoglycan, syndecan-2 (S2), can regulate TGF-beta signaling. S2 protein increased in the renal interstitium in diabetes and regulated TGF-beta-mediated increased matrix deposition in vitro. Transfection of renal papillary fibroblasts with S2 or a S2 construct that has a truncated cytoplasmic domain (S2DeltaS) promoted TGF-beta binding and S2 core protein ectodomain directly bound TGF-beta. Transfection with S2 increased the amounts of type I and type II TGF-beta receptors (TbetaRI and TbetaRII), whereas S2DeltaS was much less effective. In contrast, S2DeltaS dramatically increased the level of type III TGF-beta receptor (TbetaRIII), betaglycan, whereas S2 resulted in a decrease. Syndecan-2 specifically co-immunoprecipitated with betaglycan but not with TbetaRI or TbetaRII. This is a novel mechanism of control of TGF-beta action that may be important in fibrosis.
Collapse
Affiliation(s)
- Ligong Chen
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | |
Collapse
|
49
|
Malda J, Rouwkema J, Martens DE, Le Comte EP, Kooy FK, Tramper J, van Blitterswijk CA, Riesle J. Oxygen gradients in tissue-engineered Pegt/Pbt cartilaginous constructs: Measurement and modeling. Biotechnol Bioeng 2004; 86:9-18. [PMID: 15007836 DOI: 10.1002/bit.20038] [Citation(s) in RCA: 245] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The supply of oxygen within three-dimensional tissue-engineered (TE) cartilage polymer constructs is mainly by diffusion. Oxygen consumption by cells results in gradients in the oxygen concentration. The aims of this study were, firstly, to identify the gradients within TE cartilage polymer constructs and, secondly, to predict the profiles during in vitro culture. A glass microelectrode system was adapted and used to penetrate cartilage and TE cartilaginous constructs, yielding reproducible measurements with high spatial resolution. Cartilage polymer constructs were cultured for up to 41 days in vitro. Oxygen concentrations, as low as 2-5%, were measured within the center of these constructs. At the beginning of in vitro culture, the oxygen gradients were steeper in TE constructs in comparison to native tissue. Nevertheless, during the course of culture, oxygen concentrations approached the values measured in native tissue. A mathematical model was developed which yields oxygen profiles within cartilage explants and TE constructs. Model input parameters were assessed, including the diffusion coefficient of cartilage (2.2 x 10(-9)) + (0.4 x 10(-9) m(2) s(-1)), 70% of the diffusion coefficient of water and the diffusion coefficient of constructs (3.8 x 10(-10) m(2) s(-1)). The model confirmed that chondrocytes in polymer constructs cultured for 27 days have low oxygen requirements (0.8 x 10(-19) mol m(-3) s(-1)), even lower than chondrocytes in native cartilage. The ability to measure and predict local oxygen tensions offers new opportunities to obtain more insight in the relation between oxygen tension and chondrogenesis.
Collapse
Affiliation(s)
- J Malda
- IsoTis S.A., P.O. Box 98, 3720 AB Bilthoven, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Graff RD, Kelley SS, Lee GM. Role of pericellular matrix in development of a mechanically functional neocartilage. Biotechnol Bioeng 2003; 82:457-64. [PMID: 12632402 DOI: 10.1002/bit.10593] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The role of the chondrocyte pericellular matrix (PCM) was examined in a three-dimensional chondrocyte culture system to determine whether retention of the native pericellular matrix could stimulate collagen and proteoglycan accumulation and also promote the formation of a mechanically functional hyaline-like neocartilage. Porcine chondrocytes and chondrons, consisting of the chondrocyte with its intact pericellular matrix, were maintained in pellet culture for up to 12 weeks. Sulfated glycosaminoclycans and type II collagen were measured biochemically. Immunocytochemistry was used to examine collagen localization as well as cell distribution within the pellets. In addition, the equilibrium compressive moduli of developing pellets were measured to determine whether matrix deposition contributed to the mechanical stiffness of the cartilage constructs. Pellets increased in size and weight over a 6-week period without apparent cell proliferation. Although chondrocytes quickly rebuilt a PCM rich in type VI collagen, chondron pellets accumulated significantly more proteoglycan and type II collagen than did chondrocyte pellets, indicating a greater positive effect of the native PCM. After 5 weeks in chondron pellets, matrix remodeling was evident by microscopy. Cells that had been uniformly distributed throughout the pellets began to cluster between large areas of interterritorial matrix rich in type II collagen. After 12 weeks, clusters were stacked in columns. A rapid increase in compressive strength was observed between 1 and 3 weeks in culture for both chondron and chondrocyte pellets and, by 6 weeks, both had achieved 25% of the equilibrium compressive stiffness of cartilage explants. Retention of the in vivo PCM during chondrocyte isolation promotes the formation of a mechanically functional neocartilage construct, suitable for modeling the responses of articular cartilage to chemical stimuli or mechanical compression.
Collapse
Affiliation(s)
- Ronald D Graff
- Department of Orthopaedics and Thurston Arthritis Research Center, CB #7280, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599-7280, USA.
| | | | | |
Collapse
|