1
|
A S S, G MK. In vitro chondrogenic potential of marine biocomposite hydrogel construct for cartilage tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:2845-2866. [PMID: 39431438 DOI: 10.1080/09205063.2024.2391223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/07/2024] [Indexed: 10/22/2024]
Abstract
Cartilage tissue engineering (CTE) is a field of regenerative medicine focused on constructing ideal substitutes for injured cartilage by effectively combining cells, scaffolds, and stimulatory factors. In vitro CTE employing chondrocytes and biopolymer-based hydrogels has the potential to repair damaged cartilage. In this research, primary chondrocytes were extracted from the rib cartilage of rats and seeded on a hydrogel construct named HACF, which is made from hydroxyapatite, alginate, chitosan, and fucoidan. We then evaluated in vitro chondrogenesis on HACF cartilage construct. The results revealed that the primary chondrocytes were successfully isolated from rat rib cartilage by collagenase D digestion and HACF cartilage construct was effectively synthesized. Chondrocyte viability and its differentiation inside the scaffold HACF were determined by MTT assay, NRU assay, live/dead assay, DAPI nuclear staining, flow cytometry analysis (FCA), mRNA expression studies, and quantification of extracellular matrix components in the HACF scaffold. The findings indicated excellent chondrocyte viability within the HACF scaffold, with no noticeable changes in morphology. Apoptosis was not detected in the chondrocytes cultured on these hydrogels, as confirmed by DAPI staining, live/dead assay, and FCA. This demonstrates that the cells were capable of proliferating, dividing, multiplying, and maintaining their integrity on HACF scaffold. The results also showed more collagen deposition and glycosaminoglycan synthesis showing the good health of chondrocytes on the HACF construct. It indicates that HACF is an ideal scaffold supporting stable cartilage matrix production, highlighting its suitability for cartilage tissue engineering.
Collapse
Affiliation(s)
- Sumayya A S
- Assistant Professor, Department of Biochemistry, T.K.M. College of Arts and Science, kollam-5, kerala, India
| | | |
Collapse
|
2
|
Strauß S, Diemer M, Bucan V, Kuhbier JW, Asendorf T, Vogt PM, Schlottmann F. Spider silk enhanced tissue engineering of cartilage tissue: Approach of a novel bioreactor model using adipose derived stromal cells. J Appl Biomater Funct Mater 2024; 22:22808000241226656. [PMID: 38253568 DOI: 10.1177/22808000241226656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024] Open
Abstract
Human cartilage tissue remains a challenge for the development of therapeutic options due to its poor vascularization and reduced regenerative capacities. There are a variety of research approaches dealing with cartilage tissue engineering. In addition to different biomaterials, numerous cell populations have been investigated in bioreactor-supported experimental setups to improve cartilage tissue engineering. The concept of the present study was to investigate spider silk cocoons as scaffold seeded with adipose-derived stromal cells (ASC) in a custom-made bioreactor model using cyclic axial compression to engineer cartilage-like tissue. For chemical induction of differentiation, BMP-7 and TGF-β2 were added and changes in cell morphology and de-novo tissue formation were investigated using histological staining to verify chondrogenic differentiation. By seeding spider silk cocoons with ASC, a high colonization density and cell proliferation could be achieved. Mechanical induction of differentiation using a newly established bioreactor model led to a more roundish cell phenotype and new extracellular matrix formation, indicating a chondrogenic differentiation. The addition of BMP-7 and TGF-β2 enhanced the expression of cartilage specific markers in immunohistochemical staining. Overall, the present study can be seen as pilot study and valuable complementation to the published literature.
Collapse
Affiliation(s)
- Sarah Strauß
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Maximilian Diemer
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Vesna Bucan
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Jörn W Kuhbier
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
- Department of Plastic, Aesthetic and Hand Surgery, Helios Klinikum Hildesheim, Hildesheim, Germany
| | - Tomke Asendorf
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Peter M Vogt
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Frederik Schlottmann
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
3
|
The Fight against Cancer by Microgravity: The Multicellular Spheroid as a Metastasis Model. Int J Mol Sci 2022; 23:ijms23063073. [PMID: 35328492 PMCID: PMC8953941 DOI: 10.3390/ijms23063073] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is a disease exhibiting uncontrollable cell growth and spreading to other parts of the organism. It is a heavy, worldwide burden for mankind with high morbidity and mortality. Therefore, groundbreaking research and innovations are necessary. Research in space under microgravity (µg) conditions is a novel approach with the potential to fight cancer and develop future cancer therapies. Space travel is accompanied by adverse effects on our health, and there is a need to counteract these health problems. On the cellular level, studies have shown that real (r-) and simulated (s-) µg impact survival, apoptosis, proliferation, migration, and adhesion as well as the cytoskeleton, the extracellular matrix, focal adhesion, and growth factors in cancer cells. Moreover, the µg-environment induces in vitro 3D tumor models (multicellular spheroids and organoids) with a high potential for preclinical drug targeting, cancer drug development, and studying the processes of cancer progression and metastasis on a molecular level. This review focuses on the effects of r- and s-µg on different types of cells deriving from thyroid, breast, lung, skin, and prostate cancer, as well as tumors of the gastrointestinal tract. In addition, we summarize the current knowledge of the impact of µg on cancerous stem cells. The information demonstrates that µg has become an important new technology for increasing current knowledge of cancer biology.
Collapse
|
4
|
|
5
|
Theodoridis K, Aggelidou E, Manthou ME, Keklikoglou K, Tsimponis A, Demiri E, Bakopoulou A, Mihailidis A, Kritis A. An effective device and method for enhanced cell growth in 3D scaffolds: Investigation of cell seeding and proliferation under static and dynamic conditions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:111060. [PMID: 32993978 DOI: 10.1016/j.msec.2020.111060] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/26/2020] [Accepted: 05/05/2020] [Indexed: 12/26/2022]
Abstract
Cell adhesion on 3D-scaffolds is a challenging task to succeed high cell densities and even cell distribution. We aimed to design a 3D-cell Culture Device (3D-CD) for static seeding and cultivation, to be used with any kind of scaffold, limiting cell loss and facilitating nutrient supply. 3D printing technology was used for both scaffold and device fabrication. Apart from testing the device, the purpose of this study was to assess and compare static and dynamic seeding and cultivation methods, of wet and dry scaffolds, under normoxic and hypoxic conditions and their effects on parameters such as cell seeding efficiency, cell distribution and cell proliferation. Human adipose tissue was harvested and cultured in 3D-printed poly(epsilon-caprolactone) scaffolds. Micro-CT scans were performed and projection images were reconstructed into cross section images. We created 3D images to visualize cell distribution and orientation inside the scaffolds. The group of prewetted scaffolds was the most favorable to cell attachment. The 3D-cell Culture Device (3D-CD) enhanced cell seeding efficiency with almost no cell loss. We suggest that the most favorable outcome can be produced with static seeding in the device for 24 h, followed either by static cultivation in the same device or by dynamic cultivation.
Collapse
Affiliation(s)
- Konstantinos Theodoridis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece.
| | - Eleni Aggelidou
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Maria-Eleni Manthou
- cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; Laboratory of Histology, Embryology and Anthropology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Kleoniki Keklikoglou
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research (HCMR), Thalassocosmos, 71003 Heraklion, Crete, Greece
| | - Antonios Tsimponis
- Department of Plastic Surgery, Medical School, Papageorgiou Hospital, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Efterpi Demiri
- Department of Plastic Surgery, Medical School, Papageorgiou Hospital, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Athina Bakopoulou
- cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), GR-54124 Thessaloniki, Greece
| | - Athanasios Mihailidis
- Laboratory of Machine Elements and Machine Design, School of Mechanical Engineering, Aristotle University of Thessaloniki (A.U.Th.), GR-54624 Thessaloniki, Greece
| | - Aristeidis Kritis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece; cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| |
Collapse
|
6
|
Rahmani Del Bakhshayesh A, Babaie S, Tayefi Nasrabadi H, Asadi N, Akbarzadeh A, Abedelahi A. An overview of various treatment strategies, especially tissue engineering for damaged articular cartilage. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:1089-1104. [DOI: 10.1080/21691401.2020.1809439] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Azizeh Rahmani Del Bakhshayesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soraya Babaie
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahideh Asadi
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Akbarzadeh
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Paim Á, Cardozo NSM, Pranke P, Tessaro IC. SENSITIVITY ANALYSIS FOR MODEL COMPARISON AND SELECTION IN TISSUE ENGINEERING. BRAZILIAN JOURNAL OF CHEMICAL ENGINEERING 2019. [DOI: 10.1590/0104-6632.20190361s20170268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Ágata Paim
- Universidade Federal do Rio Grande do Sul, Brasil; Universidade Federal do Rio Grande do Sul, Brasil
| | | | - Patricia Pranke
- Universidade Federal do Rio Grande do Sul, Brasil; Stem Cell Research Institute, Brasil
| | | |
Collapse
|
8
|
Lin TH, Wang HC, Cheng WH, Hsu HC, Yeh ML. Osteochondral Tissue Regeneration Using a Tyramine-Modified Bilayered PLGA Scaffold Combined with Articular Chondrocytes in a Porcine Model. Int J Mol Sci 2019; 20:ijms20020326. [PMID: 30650528 PMCID: PMC6359257 DOI: 10.3390/ijms20020326] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 12/28/2022] Open
Abstract
Repairing damaged articular cartilage is challenging due to the limited regenerative capacity of hyaline cartilage. In this study, we fabricated a bilayered poly (lactic-co-glycolic acid) (PLGA) scaffold with small (200–300 μm) and large (200–500 μm) pores by salt leaching to stimulate chondrocyte differentiation, cartilage formation, and endochondral ossification. The scaffold surface was treated with tyramine to promote scaffold integration into native tissue. Porcine chondrocytes retained a round shape during differentiation when grown on the small pore size scaffold, and had a fibroblast-like morphology during transdifferentiation in the large pore size scaffold after five days of culture. Tyramine-treated scaffolds with mixed pore sizes seeded with chondrocytes were pressed into three-mm porcine osteochondral defects; tyramine treatment enhanced the adhesion of the small pore size scaffold to osteochondral tissue and increased glycosaminoglycan and collagen type II (Col II) contents, while reducing collagen type X (Col X) production in the cartilage layer. Col X content was higher for scaffolds with a large pore size, which was accompanied by the enhanced generation of subchondral bone. Thus, chondrocytes seeded in tyramine-treated bilayered scaffolds with small and large pores in the upper and lower parts, respectively, can promote osteochondral regeneration and integration for articular cartilage repair.
Collapse
Affiliation(s)
- Tzu-Hsiang Lin
- Department of Biomedical Engineering, National Cheng Kung University, 1 University Rd., Tainan 701, Taiwan.
| | - Hsueh-Chun Wang
- Department of Biomedical Engineering, National Cheng Kung University, 1 University Rd., Tainan 701, Taiwan.
| | - Wen-Hui Cheng
- Department of Biomedical Engineering, National Cheng Kung University, 1 University Rd., Tainan 701, Taiwan.
| | - Horng-Chaung Hsu
- Department of Orthopedics, China Medical University Hospital, 2 Yude Rd., Taichung 40447, Taiwan.
| | - Ming-Long Yeh
- Department of Biomedical Engineering, National Cheng Kung University, 1 University Rd., Tainan 701, Taiwan.
- Medical Device Innovation Center, National Cheng Kung University, 1 University Rd., Tainan 701, Taiwan.
| |
Collapse
|
9
|
Choi JR, Yong KW, Choi JY. Effects of mechanical loading on human mesenchymal stem cells for cartilage tissue engineering. J Cell Physiol 2018; 233:1913-1928. [PMID: 28542924 DOI: 10.1002/jcp.26018] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/18/2017] [Indexed: 12/22/2022]
Abstract
Today, articular cartilage damage is a major health problem, affecting people of all ages. The existing conventional articular cartilage repair techniques, such as autologous chondrocyte implantation (ACI), microfracture, and mosaicplasty, have many shortcomings which negatively affect their clinical outcomes. Therefore, it is essential to develop an alternative and efficient articular repair technique that can address those shortcomings. Cartilage tissue engineering, which aims to create a tissue-engineered cartilage derived from human mesenchymal stem cells (MSCs), shows great promise for improving articular cartilage defect therapy. However, the use of tissue-engineered cartilage for the clinical therapy of articular cartilage defect still remains challenging. Despite the importance of mechanical loading to create a functional cartilage has been well demonstrated, the specific type of mechanical loading and its optimal loading regime is still under investigation. This review summarizes the most recent advances in the effects of mechanical loading on human MSCs. First, the existing conventional articular repair techniques and their shortcomings are highlighted. The important parameters for the evaluation of the tissue-engineered cartilage, including chondrogenic and hypertrophic differentiation of human MSCs are briefly discussed. The influence of mechanical loading on human MSCs is subsequently reviewed and the possible mechanotransduction signaling is highlighted. The development of non-hypertrophic chondrogenesis in response to the changing mechanical microenvironment will aid in the establishment of a tissue-engineered cartilage for efficient articular cartilage repair.
Collapse
Affiliation(s)
- Jane Ru Choi
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
- UBC Engineering Lab, University of British Columbia, Vancouver, Canada
| | - Kar Wey Yong
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Canada
| | - Jean Yu Choi
- Faculty of Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
10
|
Gokorsch S, Nehring D, Grottke C, Czermak P. Hydrodynamic Stimulation and Long Term Cultivation of Nucleus Pulposus Cells: A New Bioreactor System to Induce Extracellular Matrix Synthesis by Nucleus Pulposus Cells Dependent on Intermittent Hydrostatic Pressure. Int J Artif Organs 2018; 27:962-70. [PMID: 15636054 DOI: 10.1177/039139880402701109] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A novel bioreactor system was constructed to induce extracellular matrix (ECM) synthesis by intervertebral disc (ID) cells due to intermittent hydrostatic pressure. The developed system is completely sterilizable and reusable. It is viable for cultivation, immobilization, and stimulation of various other cell types and tissues especially for cartilage. The custom made lid allows long-run cultivation through semi-continuous operation. Manual interferences and therefore the risk of contamination are reduced. Sampling, medium changing and addition of supplements are easily performed from the connected conditioning vessel, which could be placed in an incubator. For the present investigations nucleus pulposus cells from pigs were taken and immobilized in agarose to obtain three-dimensional cell matrix constructs which were subjected to intermittent hydrostatic pressure. Afterwards the construct was biochemically examined. The proven constituents of ECM were found to be released in dependence of the magnitude and profile of the applied pressure.
Collapse
Affiliation(s)
- S Gokorsch
- University of Applied Sciences Giessen-Friedberg, Department of Biotechnology, Giessen, Germany
| | | | | | | |
Collapse
|
11
|
Tribe HC, McEwan J, Taylor H, Oreffo ROC, Tare RS. Mesenchymal Stem Cells: Potential Role in the Treatment of Osteochondral Lesions of the Ankle. Biotechnol J 2017; 12:1700070. [PMID: 29068173 PMCID: PMC5765412 DOI: 10.1002/biot.201700070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/13/2017] [Indexed: 12/11/2022]
Abstract
Given articular cartilage has a limited repair potential, untreated osteochondral lesions of the ankle can lead to debilitating symptoms and joint deterioration necessitating joint replacement. While a wide range of reparative and restorative surgical techniques have been developed to treat osteochondral lesions of the ankle, there is no consensus in the literature regarding which is the ideal treatment. Tissue engineering strategies, encompassing stem cells, somatic cells, biomaterials, and stimulatory signals (biological and mechanical), have a potentially valuable role in the treatment of osteochondral lesions. Mesenchymal stem cells (MSCs) are an attractive resource for regenerative medicine approaches, given their ability to self-renew and differentiate into multiple stromal cell types, including chondrocytes. Although MSCs have demonstrated significant promise in in vitro and in vivo preclinical studies, their success in treating osteochondral lesions of the ankle is inconsistent, necessitating further clinical trials to validate their application. This review highlights the role of MSCs in cartilage regeneration and how the application of biomaterials and stimulatory signals can enhance chondrogenesis. The current treatments for osteochondral lesions of the ankle using regenerative medicine strategies are reviewed to provide a clinical context. The challenges for cartilage regeneration, along with potential solutions and safety concerns are also discussed.
Collapse
Affiliation(s)
- Howard C. Tribe
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- Foot and Ankle Orthopaedic DepartmentRoyal Bournemouth HospitalBournemouthBH7 7DWUK
| | - Josephine McEwan
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Heath Taylor
- Foot and Ankle Orthopaedic DepartmentRoyal Bournemouth HospitalBournemouthBH7 7DWUK
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Rahul S. Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- Bioengineering Science, Mechanical Engineering DepartmentFaculty of Engineering and the EnvironmentUniversity of SouthamptonSouthamptonSO17 1BJUK
| |
Collapse
|
12
|
Islam A, Romijn EI, Lilledahl MB, Martinez-Zubiaurre I. Non-linear optical microscopy as a novel quantitative and label-free imaging modality to improve the assessment of tissue-engineered cartilage. Osteoarthritis Cartilage 2017; 25:1729-1737. [PMID: 28668541 DOI: 10.1016/j.joca.2017.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 05/22/2017] [Accepted: 06/20/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Current systems to evaluate outcomes from tissue-engineered cartilage (TEC) are sub-optimal. The main purpose of our study was to demonstrate the use of second harmonic generation (SHG) microscopy as a novel quantitative approach to assess collagen deposition in laboratory made cartilage constructs. METHODS Scaffold-free cartilage constructs were obtained by condensation of in vitro expanded Hoffa's fat pad derived stromal cells (HFPSCs), incubated in the presence or absence of chondrogenic growth factors (GF) during a period of 21 d. Cartilage-like features in constructs were assessed by Alcian blue staining, transmission electron microscopy (TEM), SHG and two-photon excited fluorescence microscopy. A new scoring system, using second harmonic generation microscopy (SHGM) index for collagen density and distribution, was adapted to the existing "Bern score" in order to evaluate in vitro TEC. RESULTS Spheroids with GF gave a relative high Bern score value due to appropriate cell morphology, cell density, tissue-like features and proteoglycan content, whereas spheroids without GF did not. However, both TEM and SHGM revealed striking differences between the collagen framework in the spheroids and native cartilage. Spheroids required a four-fold increase in laser power to visualize the collagen matrix by SHGM compared to native cartilage. Additionally, collagen distribution, determined as the area of tissue generating SHG signal, was higher in spheroids with GF than without GF, but lower than in native cartilage. CONCLUSION SHG represents a reliable quantitative approach to assess collagen deposition in laboratory engineered cartilage, and may be applied to improve currently established scoring systems.
Collapse
Affiliation(s)
- A Islam
- Institute of Clinical Medicine, University of Tromsø, Norway.
| | - E I Romijn
- Department of Physics, Norwegian University of Science and Technology, Norway.
| | - M B Lilledahl
- Department of Physics, Norwegian University of Science and Technology, Norway.
| | | |
Collapse
|
13
|
Dickinson SC, Sutton CA, Brady K, Salerno A, Katopodi T, Williams RL, West CC, Evseenko D, Wu L, Pang S, Ferro de Godoy R, Goodship AE, Péault B, Blom AW, Kafienah W, Hollander AP. The Wnt5a Receptor, Receptor Tyrosine Kinase-Like Orphan Receptor 2, Is a Predictive Cell Surface Marker of Human Mesenchymal Stem Cells with an Enhanced Capacity for Chondrogenic Differentiation. Stem Cells 2017; 35:2280-2291. [PMID: 28833807 PMCID: PMC5707440 DOI: 10.1002/stem.2691] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 07/07/2017] [Accepted: 07/23/2017] [Indexed: 12/24/2022]
Abstract
Multipotent mesenchymal stem cells (MSCs) have enormous potential in tissue engineering and regenerative medicine. However, until now, their development for clinical use has been severely limited as they are a mixed population of cells with varying capacities for lineage differentiation and tissue formation. Here, we identify receptor tyrosine kinase‐like orphan receptor 2 (ROR2) as a cell surface marker expressed by those MSCs with an enhanced capacity for cartilage formation. We generated clonal human MSC populations with varying capacities for chondrogenesis. ROR2 was identified through screening for upregulated genes in the most chondrogenic clones. When isolated from uncloned populations, ROR2+ve MSCs were significantly more chondrogenic than either ROR2–ve or unfractionated MSCs. In a sheep cartilage‐repair model, they produced significantly more defect filling with no loss of cartilage quality compared with controls. ROR2+ve MSCs/perivascular cells were present in developing human cartilage, adult bone marrow, and adipose tissue. Their frequency in bone marrow was significantly lower in patients with osteoarthritis (OA) than in controls. However, after isolation of these cells and their initial expansion in vitro, there was greater ROR2 expression in the population derived from OA patients compared with controls. Furthermore, osteoarthritis‐derived MSCs were better able to form cartilage than MSCs from control patients in a tissue engineering assay. We conclude that MSCs expressing high levels of ROR2 provide a defined population capable of predictably enhanced cartilage production. Stem Cells2017;35:2280–2291
Collapse
Affiliation(s)
- Sally C Dickinson
- Institute of Integrative Biology, University of Liverpool, United Kingdom
| | - Catherine A Sutton
- School of Cellular and Molecular Medicine, Faculty of Medical and Veterinary Sciences, University of Bristol, United Kingdom
| | - Kyla Brady
- Institute of Integrative Biology, University of Liverpool, United Kingdom
| | - Anna Salerno
- Institute of Integrative Biology, University of Liverpool, United Kingdom
| | - Theoni Katopodi
- Institute of Integrative Biology, University of Liverpool, United Kingdom
| | - Rhys L Williams
- School of Cellular and Molecular Medicine, Faculty of Medical and Veterinary Sciences, University of Bristol, United Kingdom
| | - Christopher C West
- The University of Edinburgh, MRC Center for Regenerative Medicine, Scotland, United Kingdom
| | - Denis Evseenko
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA.,Department of Stem Cell Research and Regenerative Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Ling Wu
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA.,Department of Stem Cell Research and Regenerative Medicine, University of Southern California (USC), Los Angeles, California, USA
| | - Suzanna Pang
- School of Cellular and Molecular Medicine, Faculty of Medical and Veterinary Sciences, University of Bristol, United Kingdom
| | - Roberta Ferro de Godoy
- Royal National Orthopaedic Hospital, Institute of Orthopaedics and Musculoskeletal Science, University College London, Brockley Hill, Stanmore, United Kingdom
| | - Allen E Goodship
- Royal National Orthopaedic Hospital, Institute of Orthopaedics and Musculoskeletal Science, University College London, Brockley Hill, Stanmore, United Kingdom
| | - Bruno Péault
- The University of Edinburgh, MRC Center for Regenerative Medicine, Scotland, United Kingdom.,The University of Edinburgh, Center for Cardiovascular Science, Scotland, United Kingdom.,David Geffen School of Medicine and Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California, Los Angeles, California, USA
| | - Ashley W Blom
- School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, United Kingdom
| | - Wael Kafienah
- School of Cellular and Molecular Medicine, Faculty of Medical and Veterinary Sciences, University of Bristol, United Kingdom
| | | |
Collapse
|
14
|
Kwon H, Haudenschild AK, Brown WE, Vapniarsky N, Paschos NK, Arzi B, Hu JC, Athanasiou KA. Tissue engineering potential of human dermis-isolated adult stem cells from multiple anatomical locations. PLoS One 2017; 12:e0182531. [PMID: 28767737 PMCID: PMC5540597 DOI: 10.1371/journal.pone.0182531] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023] Open
Abstract
Abundance and accessibility render skin-derived stem cells an attractive cell source for tissue engineering applications. Toward assessing their utility, the variability of constructs engineered from human dermis-isolated adult stem (hDIAS) cells was examined with respect to different anatomical locations (foreskin, breast, and abdominal skin), both in vitro and in a subcutaneous, athymic mouse model. All anatomical locations yielded hDIAS cells with multi-lineage differentiation potentials, though adipogenesis was not seen for foreskin-derived hDIAS cells. Using engineered cartilage as a model, tissue engineered constructs from hDIAS cells were compared. Construct morphology differed by location. The mechanical properties of human foreskin- and abdominal skin-derived constructs were similar at implantation, remaining comparable after 4 additional weeks of culture in vivo. Breast skin-derived constructs were not mechanically testable. For all groups, no signs of abnormality were observed in the host. Addition of aggregate redifferentiation culture prior to construct formation improved chondrogenic differentiation of foreskin-derived hDIAS cells, as evident by increases in glycosaminoglycan and collagen contents. More robust Alcian blue staining and homogeneous cell populations were also observed compared to controls. Human DIAS cells elicited no adverse host responses, reacted positively to chondrogenic regimens, and possessed multi-lineage differentiation potential with the caveat that efficacy may differ by anatomical origin of the skin. Taken together, these results suggest that hDIAS cells hold promise as a potential cell source for a number of tissue engineering applications.
Collapse
Affiliation(s)
- Heenam Kwon
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Anne K. Haudenschild
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Wendy E. Brown
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Natalia Vapniarsky
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Nikolaos K. Paschos
- Department of Orthopaedic Surgery, Penn Sports Medicine, University of Pennsylvania Health System, Philadelphia, PA, United States of America
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Jerry C. Hu
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, United States of America
- * E-mail:
| |
Collapse
|
15
|
Takahashi T, Nieda T, Miyazaki E, Enzan H. Novel Technique for Suspension Culture of Autologous Chondrocytes Improves Cell Proliferation and Tissue Architecture. Cell Transplant 2017; 12:667-76. [PMID: 14579935 DOI: 10.3727/000000003108747145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We have developed a new and simple method of chondrocyte suspension culture using a spinner bottle with rotation of the matrices. We compared the characteristics of chondrocytes cultured by this method with those grown in standard monolayer cultures. We also determined the optimal nutritional medium for suspension cultures. Periosteum explants seeded with chondrocytes were grown in monolayer and suspension cultures under three conditions: in medium with no additive (control), with 10% fetal bovine serum (FBS), or with 10% autologous serum (AS). After culturing, the explants were harvested, processed for histology, and stained with hematoxylin-eosin or TUNEL, or immunostained for type I, II, and III collagen, and Ki-67 antigen. In monolayer cultures, the attachment of the chondrocytes to the periosteum was weak and the superficial layer consisted of fibrotic tissue and few nucleated cells. Collagen type II staining was strong, but types I and III were weak. Among the suspension cultures the AS group produced the thickest layer of chondrocytes with the fewest apoptotic cells. The superficial layer of cartilage in these cultures stained positive for type I and III collagen and Ki-67 antigen. Among the suspension cultures, total chondroitin and chondroitin-4 sulfate (C-4S) concentration was highest in the AS group, while prostaglandin E2 (PGE2) was highest in the FBS group. In summary, our new method of suspension culture of periosteal explants using rotational matrices combined with AS nutritional media was the most effective method for maintaining the bond between the chondrocyte layer and periosteum, as well as the production of type I and III collagen in the superficial layer.
Collapse
Affiliation(s)
- Toshiaki Takahashi
- Department of Orthopaedic Surgery, Kochi Medical School, Oko-cho, Nankoku, Kochi 783-8505, Japan.
| | | | | | | |
Collapse
|
16
|
Ushida T, Furukawa K, Toita K, Tateishi T. Three-Dimensional Seeding of Chondrocytes Encapsulated in Collagen Gel into PLLA Scaffolds. Cell Transplant 2017. [DOI: 10.3727/000000002783985611] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Tissue engineering approaches have been clinically tried to repair damaged articular cartilages. It is an essential step to uniformly seed chondrocytes into 3D scaffolds in order to reconstruct tissue-engineered cartilages in vitro, but the tissue engineering could not have been provided with efficient cell seeding methods. Type I collagen is clinically used and known as a cytocompatible material, having recognition sites for integrins. Collagen gel encapsulating chondrocytes has been tried for making regenerated cartilages, but it is found difficult to have the gel keep its original shape after long-term culture, because of shrinking. On the other hand, 3D scaffolds, either of a nonwoven structure or a sponge-like structure, involve difficulty in that chondrocytes could not be uniformly seeded, although they have adequate initial mechanical properties. In this study, by combining collagen gelation with a nonwoven PLLA scaffold, we achieved uniform cell seeding into the 3D scaffold. Bovine articular chondrocytes were mixed with type I collagen solution, and the solution was poured into the nonwoven PLLA scaffold (1.5 mm thick, f 15 mm). The collagen–chondrocyte mixture was made into gel at 37°C for 1 h. The 0.39% collagen mixture was viscous enough to prevent cells from precipitating during gelation. Almost all chondrocytes were able to be incorporated into the PLLA scaffolds by mixing with collagen solution and subsequently making into gel, while 30–40% of the chondrocytes seeded as a cell suspension were not trapped into the PLLA scaffolds. The method presented, where chondrocytes were mixed with collagen solution, and the mixture was incorporated into a 3D scaffold, then made into gel in the scaffold, could serve as an alternative for in vitro cartilage regeneration, also simultaneously having the advantages of both materials.
Collapse
Affiliation(s)
- Takashi Ushida
- Biomedical Engineering Laboratory, Graduate School of Engineering, The University of Tokyo, 7-3-1 Bunkyo, 113-8656 Tokyo, Japan
| | - Katsuko Furukawa
- Biomedical Engineering Laboratory, Graduate School of Engineering, The University of Tokyo, 7-3-1 Bunkyo, 113-8656 Tokyo, Japan
| | - Kenshi Toita
- Biomedical Engineering Laboratory, Graduate School of Engineering, The University of Tokyo, 7-3-1 Bunkyo, 113-8656 Tokyo, Japan
| | - Tetsuya Tateishi
- Biomedical Engineering Laboratory, Graduate School of Engineering, The University of Tokyo, 7-3-1 Bunkyo, 113-8656 Tokyo, Japan
| |
Collapse
|
17
|
Risbud M, Ringe J, Bhonde R, Sittinger M. In Vitro Expression of Cartilage-Specific Markers by Chondrocytes on a Biocompatible Hydrogel: Implications for Engineering Cartilage Tissue. Cell Transplant 2017. [DOI: 10.3727/000000001783986224] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Makarand Risbud
- Tissue Engineering Laboratory, University Medical Centre, Charité, Humboldt University of Berlin, Tucholskystrasse-2, 10117 Berlin, Germany
- Tissue Engineering and Banking Laboratory, National Centre for Cell Science, Ganeshkhind, Pune 411 007, India
| | - Jochen Ringe
- Tissue Engineering Laboratory, University Medical Centre, Charité, Humboldt University of Berlin, Tucholskystrasse-2, 10117 Berlin, Germany
| | - Ramesh Bhonde
- Tissue Engineering and Banking Laboratory, National Centre for Cell Science, Ganeshkhind, Pune 411 007, India
| | - Michael Sittinger
- Tissue Engineering Laboratory, University Medical Centre, Charité, Humboldt University of Berlin, Tucholskystrasse-2, 10117 Berlin, Germany
| |
Collapse
|
18
|
Osteochondral Tissue Engineering and Regenerative Strategies. REGENERATIVE STRATEGIES FOR THE TREATMENT OF KNEE JOINT DISABILITIES 2017. [DOI: 10.1007/978-3-319-44785-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
19
|
Kar S, Smith DW, Gardiner BS, Grodzinsky AJ. Systems Based Study of the Therapeutic Potential of Small Charged Molecules for the Inhibition of IL-1 Mediated Cartilage Degradation. PLoS One 2016; 11:e0168047. [PMID: 27977731 PMCID: PMC5158201 DOI: 10.1371/journal.pone.0168047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/23/2016] [Indexed: 11/18/2022] Open
Abstract
Inflammatory cytokines are key drivers of cartilage degradation in post-traumatic osteoarthritis. Cartilage degradation mediated by these inflammatory cytokines has been extensively investigated using in vitro experimental systems. Based on one such study, we have developed a computational model to quantitatively assess the impact of charged small molecules intended to inhibit IL-1 mediated cartilage degradation. We primarily focus on the simplest possible computational model of small molecular interaction with the IL-1 system-direct binding of the small molecule to the active site on the IL-1 molecule itself. We first use the model to explore the uptake and release kinetics of the small molecule inhibitor by cartilage tissue. Our results show that negatively charged small molecules are excluded from the negatively charged cartilage tissue and have uptake kinetics in the order of hours. In contrast, the positively charged small molecules are drawn into the cartilage with uptake and release timescales ranging from hours to days. Using our calibrated computational model, we subsequently explore the effect of small molecule charge and binding constant on the rate of cartilage degradation. The results from this analysis indicate that the small molecules are most effective in inhibiting cartilage degradation if they are either positively charged and/or bind strongly to IL-1α, or both. Furthermore, our results showed that the cartilage structural homeostasis can be restored by the small molecule if administered within six days following initial tissue exposure to IL-1α. We finally extended the scope of the computational model by simulating the competitive inhibition of cartilage degradation by the small molecule. Results from this model show that small molecules are more efficient in inhibiting cartilage degradation by binding directly to IL-1α rather than binding to IL-1α receptors. The results from this study can be used as a template for the design and development of more pharmacologically effective osteoarthritis drugs, and to investigate possible therapeutic options.
Collapse
Affiliation(s)
- Saptarshi Kar
- School of Computer Science and Software Engineering, University of Western Australia, Crawley, WA, Australia
| | - David W. Smith
- School of Computer Science and Software Engineering, University of Western Australia, Crawley, WA, Australia
| | - Bruce S. Gardiner
- Department of Physics and Nanotechnology, Murdoch University, Murdoch, WA, Australia
| | - Alan J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| |
Collapse
|
20
|
Li Y, He L, Pan S, Zhang L, Zhang W, Yi H, Niu Y. Three-dimensional simulated microgravity culture improves the proliferation and odontogenic differentiation of dental pulp stem cell in PLGA scaffolds implanted in mice. Mol Med Rep 2016; 15:873-878. [PMID: 28000851 DOI: 10.3892/mmr.2016.6042] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 11/17/2016] [Indexed: 11/06/2022] Open
Abstract
Tooth regeneration through stem cell-based therapy is a promising treatment for tooth decay and loss. Human dental pulp stem cells (hDPSCs) have been widely identified as the stem cells with the most potential for tooth tissue regeneration. However, the culture of hDPSCs in vitro for tissue engineering is challenging, as cells may proliferate slowly or/and differentiate poorly in vivo. Dynamic three‑dimensional (3D) simulated microgravity (SMG) created using the rotary cell culture system is considered to an effective tool, which contributes to several cell functions. Thus, the present study aimed to investigate the effect of dynamic 3D SMG culture on the proliferation and odontogenic differentiation abilities of hDPSCs in poly (lactic‑co‑glycolic acid) (PLGA) scaffolds in nude mice. The hDPSCs on PLGA scaffolds were maintained separately in the 3D SMG culture system and static 3D cultures with osteogenic medium for 7 days in vitro. Subsequently, the cell‑PLGA complexes were implanted subcutaneously on the backs of nude mice for 4 weeks. The results of histological and immunohistochemical examinations of Ki‑67, type I collagen, dentin sialoprotein and DMP‑1 indicated that the proliferation and odontogenic differentiation abilities of the hDPSCs prepared in the 3D SMG culture system were higher, compared with those prepared in the static culture system. These findings suggested that dynamic 3D SMG culture likely contributes to tissue engineering by improving the proliferation and odontogenic differentiation abilities of hDPSCs in vivo.
Collapse
Affiliation(s)
- Yanping Li
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Lina He
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Shuang Pan
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Lin Zhang
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Weiwei Zhang
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hong Yi
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yumei Niu
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
21
|
Soares JS, Sacks MS. A triphasic constrained mixture model of engineered tissue formation under in vitro dynamic mechanical conditioning. Biomech Model Mechanobiol 2016; 15:293-316. [PMID: 26055347 PMCID: PMC4712131 DOI: 10.1007/s10237-015-0687-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/21/2015] [Indexed: 10/23/2022]
Abstract
While it has become axiomatic that mechanical signals promote in vitro engineered tissue formation, the underlying mechanisms remain largely unknown. Moreover, efforts to date to determine parameters for optimal extracellular matrix (ECM) development have been largely empirical. In the present work, we propose a two-pronged approach involving novel theoretical developments coupled with key experimental data to develop better mechanistic understanding of growth and development of dense connective tissue under mechanical stimuli. To describe cellular proliferation and ECM synthesis that occur at rates of days to weeks, we employ mixture theory to model the construct constituents as a nutrient-cell-ECM triphasic system, their transport, and their biochemical reactions. Dynamic conditioning protocols with frequencies around 1 Hz are described with multi-scale methods to couple the dissimilar time scales. Enhancement of nutrient transport due to pore fluid advection is upscaled into the growth model, and the spatially dependent ECM distribution describes the evolving poroelastic characteristics of the scaffold-engineered tissue construct. Simulation results compared favorably to the existing experimental data, and most importantly, distinguish between static and dynamic conditioning regimes. The theoretical framework for mechanically conditioned tissue engineering (TE) permits not only the formulation of novel and better-informed mechanistic hypothesis describing the phenomena underlying TE growth and development, but also the exploration/optimization of conditioning protocols in a rational manner.
Collapse
Affiliation(s)
- Joao S Soares
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences (ICES), Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th Street, Austin, TX, 78712-1129, USA
| | - Michael S Sacks
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences (ICES), Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th Street, Austin, TX, 78712-1129, USA.
| |
Collapse
|
22
|
Kar S, Smith DW, Gardiner BS, Li Y, Wang Y, Grodzinsky AJ. Modeling IL-1 induced degradation of articular cartilage. Arch Biochem Biophys 2016; 594:37-53. [PMID: 26874194 DOI: 10.1016/j.abb.2016.02.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
Abstract
In this study, we develop a computational model to simulate the in vitro biochemical degradation of articular cartilage explants sourced from the femoropatellar grooves of bovine calves. Cartilage explants were incubated in culture medium with and without the inflammatory cytokine IL-1α. The spatio-temporal evolution of the cartilage explant's extracellular matrix components is modelled. Key variables in the model include chondrocytes, aggrecan, collagen, aggrecanase, collagenase and IL-1α. The model is first calibrated for aggrecan homeostasis of cartilage in vivo, then for data on (explant) controls, and finally for data on the IL-1α driven proteolysis of aggrecan and collagen over a 4-week period. The model was found to fit the experimental data best when: (i) chondrocytes continue to synthesize aggrecan during the cytokine challenge, (ii) a one to two day delay is introduced between the addition of IL-1α to the culture medium and subsequent aggrecanolysis, (iii) collagen degradation does not commence until the total concentration of aggrecan (i.e. both intact and degraded aggrecan) at any specific location within the explant becomes ≤ 1.5 mg/ml and (iv) degraded aggrecan formed due to the IL-1α induced proteolysis of intact aggrecan protects the collagen network while collagen degrades in a two-step process which, together, significantly modulate the collagen network degradation. Under simulated in vivo conditions, the model predicts increased aggrecan turnover rates in the presence of synovial IL-1α, consistent with experimental observations. Such models may help to infer the course of events in vivo following traumatic joint injury, and may also prove useful in quantitatively evaluating the efficiency of various therapeutic molecules that could be employed to avoid or modify the course of cartilage disease states.
Collapse
Affiliation(s)
- Saptarshi Kar
- School of Computer Science and Software Engineering, University of Western Australia, Crawley, WA, Australia
| | - David W Smith
- School of Computer Science and Software Engineering, University of Western Australia, Crawley, WA, Australia.
| | - Bruce S Gardiner
- School of Computer Science and Software Engineering, University of Western Australia, Crawley, WA, Australia
| | - Yang Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yang Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
23
|
Biodegradable Materials for Bone Repair and Tissue Engineering Applications. MATERIALS 2015; 8:5744-5794. [PMID: 28793533 PMCID: PMC5512653 DOI: 10.3390/ma8095273] [Citation(s) in RCA: 399] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/09/2015] [Accepted: 08/24/2015] [Indexed: 12/21/2022]
Abstract
This review discusses and summarizes the recent developments and advances in the use of biodegradable materials for bone repair purposes. The choice between using degradable and non-degradable devices for orthopedic and maxillofacial applications must be carefully weighed. Traditional biodegradable devices for osteosynthesis have been successful in low or mild load bearing applications. However, continuing research and recent developments in the field of material science has resulted in development of biomaterials with improved strength and mechanical properties. For this purpose, biodegradable materials, including polymers, ceramics and magnesium alloys have attracted much attention for osteologic repair and applications. The next generation of biodegradable materials would benefit from recent knowledge gained regarding cell material interactions, with better control of interfacing between the material and the surrounding bone tissue. The next generations of biodegradable materials for bone repair and regeneration applications require better control of interfacing between the material and the surrounding bone tissue. Also, the mechanical properties and degradation/resorption profiles of these materials require further improvement to broaden their use and achieve better clinical results.
Collapse
|
24
|
Madry H, Cucchiarini M. Tissue-engineering strategies to repair joint tissue in osteoarthritis: nonviral gene-transfer approaches. Curr Rheumatol Rep 2015; 16:450. [PMID: 25182678 DOI: 10.1007/s11926-014-0450-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Loss of articular cartilage is a common clinical consequence of osteoarthritis (OA). In the past decade, substantial progress in tissue engineering, nonviral gene transfer, and cell transplantation have provided the scientific foundation for generating cartilaginous constructs from genetically modified cells. Combining tissue engineering with overexpression of therapeutic genes enables immediate filling of a cartilage defect with an engineered construct that actively supports chondrogenesis. Several pioneering studies have proved that spatially defined nonviral overexpression of growth-factor genes in constructs of solid biomaterials or hydrogels is advantageous compared with gene transfer or scaffold alone, both in vitro and in vivo. Notably, these investigations were performed in models of focal cartilage defects, because advanced cartilage-repair strategies based on the principles of tissue engineering have not advanced sufficiently to enable resurfacing of extensively degraded cartilage as therapy for OA. These studies serve as prototypes for future technological developments, because they raise the possibility that cartilage constructs engineered from genetically modified chondrocytes providing autocrine and paracrine stimuli could similarly compensate for the loss of articular cartilage in OA. Because cartilage-tissue-engineering strategies are already used in the clinic, combining tissue engineering and nonviral gene transfer could prove a powerful approach to treat OA.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics and Department of Orthopaedic Surgery, Saarland University, 66421, Homburg, Germany,
| | | |
Collapse
|
25
|
Li D, Wang Y, Liu Y, Xie Z, Wang L, Tan H. Preparation and properties of polyurethane-modified epoxy cured in different simulated gravity environments. J Appl Polym Sci 2015. [DOI: 10.1002/app.42063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Defeng Li
- School of Chemical Engineering and Technology, Harbin Institute of Technology; Harbin 150001 China
| | - Youshan Wang
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments; Harbin Institute of Technology; Harbin 150001 China
| | - Yuyan Liu
- School of Chemical Engineering and Technology, Harbin Institute of Technology; Harbin 150001 China
| | - Zhimin Xie
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments; Harbin Institute of Technology; Harbin 150001 China
| | - Lei Wang
- School of Chemical Engineering and Technology, Harbin Institute of Technology; Harbin 150001 China
| | - Huifeng Tan
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments; Harbin Institute of Technology; Harbin 150001 China
| |
Collapse
|
26
|
Li D, Liu Y, Wang Y, Kang H, Wang L, Tan H. Influences of different gravity environments on the curing process and cured products of carbon-nanotube-reinforced epoxy composites. J Appl Polym Sci 2015. [DOI: 10.1002/app.41413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Defeng Li
- School of Chemical Engineering and Technology, Harbin Institute of Technology; Harbin 150001 China
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments, Harbin Institute of Technology; Harbin 150080 China
| | - Yuyan Liu
- School of Chemical Engineering and Technology, Harbin Institute of Technology; Harbin 150001 China
| | - Youshan Wang
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments, Harbin Institute of Technology; Harbin 150080 China
| | - Hongjun Kang
- School of Chemical Engineering and Technology, Harbin Institute of Technology; Harbin 150001 China
| | - Lei Wang
- School of Chemical Engineering and Technology, Harbin Institute of Technology; Harbin 150001 China
| | - Huifeng Tan
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments, Harbin Institute of Technology; Harbin 150080 China
| |
Collapse
|
27
|
Guha Thakurta S, Kraft M, Viljoen HJ, Subramanian A. Enhanced depth-independent chondrocyte proliferation and phenotype maintenance in an ultrasound bioreactor and an assessment of ultrasound dampening in the scaffold. Acta Biomater 2014; 10:4798-4810. [PMID: 25065549 DOI: 10.1016/j.actbio.2014.07.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 01/20/2023]
Abstract
Chondrocyte-seeded scaffolds were cultured in an ultrasound (US)-assisted bioreactor, which supplied the cells with acoustic energy around resonance frequencies (~5.0 MHz). Polyurethane-polycarbonate (BM), chitosan (CS) and chitosan-n-butanol (CSB) based scaffolds with varying porosities were chosen and the following US regimen was employed: 15 kPa and 60 kPa, 5 min per application and 6 applications per day for 21 days. Non-stimulated scaffolds served as control. For BM scaffolds, US stimulation significantly impacted cell proliferation and depth-independent cell population density compared to controls. The highest COL2A1/COL1A1 ratios and ACAN mRNA were noted on US-treated BM scaffolds compared to controls. A similar trend was noted on US-treated cell-seeded CS and CSB scaffolds, though COL2A1/COL1A1 ratios were significantly lower compared to BM scaffolds. Expression of Sox-9 was also elevated under US and paralleled the COL2A1/COL1A1 ratio. As an original contribution, a simplified mathematical model based on Biot theory was developed to understand the propagation of the incident US wave through the scaffolds and the model analysis was connected to cellular responses. Scaffold architecture influenced the distribution of US field, with the US field being the least attenuated in BM scaffolds, thus coupling more mechanical energy into cells, and leading to increased cellular activity.
Collapse
Affiliation(s)
- Sanjukta Guha Thakurta
- Department of Chemical Engineering, 207L Othmer Hall, University of Nebraska-Lincoln, Lincoln, NE 68588-0643, USA
| | - Mikail Kraft
- Department of Chemical Engineering, 207L Othmer Hall, University of Nebraska-Lincoln, Lincoln, NE 68588-0643, USA
| | - Hendrik J Viljoen
- Department of Chemical Engineering, 207L Othmer Hall, University of Nebraska-Lincoln, Lincoln, NE 68588-0643, USA
| | - Anuradha Subramanian
- Department of Chemical Engineering, 207L Othmer Hall, University of Nebraska-Lincoln, Lincoln, NE 68588-0643, USA.
| |
Collapse
|
28
|
Fu AS, Solorio LD, Alsberg E, Saidel GM. Mathematical modelling of glycosaminoglycan production by stem cell aggregates incorporated with growth factor-releasing polymer microspheres. J Tissue Eng Regen Med 2014; 11:481-488. [PMID: 25047254 DOI: 10.1002/term.1940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 05/07/2014] [Accepted: 05/27/2014] [Indexed: 11/12/2022]
Abstract
Systems composed of high density cells incorporated with growth factor-releasing polymer microspheres have recently been shown to promote chondrogenic differentiation and cartilage formation. Within these systems, the effects of spatial and temporal patterning of growth factor release on hyaline cartilage-specific extracellular matrix production have been examined. However, at present, it is unclear which microsphere densities and growth factor delivery profiles are optimal for inducing human mesenchymal stem cell differentiation and glycosaminoglycan production. A mathematical model to describe glycosaminoglycan production as a function of initial microsphere loading and microsphere degradation rate over a period of 3 weeks is presented. Based on predictions generated by this model, it may be feasible to design a bioactive microsphere system with specific spatiotemporal growth factor presentation characteristics to promote glycosaminoglycan production at controllable rates. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Andrew S Fu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Loran D Solorio
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.,Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH, USA
| | - Gerald M Saidel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
29
|
Somoza RA, Welter JF, Correa D, Caplan AI. Chondrogenic differentiation of mesenchymal stem cells: challenges and unfulfilled expectations. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:596-608. [PMID: 24749845 DOI: 10.1089/ten.teb.2013.0771] [Citation(s) in RCA: 252] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Articular cartilage repair and regeneration provides a substantial challenge in Regenerative Medicine because of the high degree of morphological and mechanical complexity intrinsic to hyaline cartilage due, in part, to its extracellular matrix. Cartilage remains one of the most difficult tissues to heal; even state-of-the-art regenerative medicine technology cannot yet provide authentic cartilage resurfacing. Mesenchymal stem cells (MSCs) were once believed to be the panacea for cartilage repair and regeneration, but despite years of research, they have not fulfilled these expectations. It has been observed that MSCs have an intrinsic differentiation program reminiscent of endochondral bone formation, which they follow after exposure to specific reagents as a part of current differentiation protocols. Efforts have been made to avoid the resulting hypertrophic fate of MSCs; however, so far, none of these has recreated a fully functional articular hyaline cartilage without chondrocytes exhibiting a hypertrophic phenotype. We reviewed the current literature in an attempt to understand why MSCs have failed to regenerate articular cartilage. The challenges that must be overcome before MSC-based tissue engineering can become a front-line technology for successful articular cartilage regeneration are highlighted.
Collapse
Affiliation(s)
- Rodrigo A Somoza
- Department of Biology, Skeletal Research Center, Case Western Reserve University , Cleveland, Ohio
| | | | | | | |
Collapse
|
30
|
Evaluation of Diffusive Transport and Cellular Uptake of Nutrients in Tissue Engineered Constructs Using a Hybrid Discrete Mathematical Model. Processes (Basel) 2014. [DOI: 10.3390/pr2020333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
31
|
Lam J, Lu S, Meretoja VV, Tabata Y, Mikos AG, Kasper FK. Generation of osteochondral tissue constructs with chondrogenically and osteogenically predifferentiated mesenchymal stem cells encapsulated in bilayered hydrogels. Acta Biomater 2014; 10:1112-23. [PMID: 24300948 DOI: 10.1016/j.actbio.2013.11.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 10/28/2013] [Accepted: 11/22/2013] [Indexed: 10/25/2022]
Abstract
This study investigated the ability of chondrogenic and osteogenic predifferentiation of mesenchymal stem cells (MSCs) to play a role in the development of osteochondral tissue constructs using injectable bilayered oligo(poly(ethylene glycol) fumarate) (OPF) hydrogel composites. We hypothesized that the combinatorial approach of encapsulating cell populations of both chondrogenic and osteogenic lineages in a spatially controlled manner within bilayered constructs would enable these cells to maintain their respective phenotypes via the exchange of biochemical factors even without the influence of external growth factors. During monolayer expansion prior to hydrogel encapsulation, it was found that 7 (CG7) and 14 (CG14) days of MSC exposure to TGF-β3 allowed for the generation of distinct cell populations with corresponding chondrogenic maturities as indicated by increasing aggrecan and type II collagen/type I collagen expression. Chondrogenic and osteogenic cells were then encapsulated within their respective (chondral/subchondral) layers in bilayered hydrogel composites to include four experimental groups. Encapsulated CG7 cells within the chondral layer exhibited enhanced chondrogenic phenotype when compared to other cell populations based on stronger type II collagen and aggrecan gene expression and higher glycosaminoglycan-to-hydroxyproline ratios. Osteogenic cells that were co-cultured with chondrogenic cells (in the chondral layer) showed higher cellularity over time, suggesting that chondrogenic cells stimulated the proliferation of osteogenic cells. Groups with osteogenic cells displayed mineralization in the subchondral layer, confirming the effect of osteogenic predifferentiation. In summary, it was found that MSCs that underwent 7 days, but not 14 days, of chondrogenic predifferentiation most closely resembled the phenotype of native hyaline cartilage when combined with osteogenic cells in a bilayered OPF hydrogel composite, indicating that the duration of chondrogenic preconditioning is an important factor to control. Furthermore, the respective chondrogenic and osteogenic phenotypes were maintained for 28 days in vitro without the need for external growth factors, demonstrating the exciting potential of this novel strategy for the generation of osteochondral tissue constructs for cartilage engineering applications.
Collapse
|
32
|
Shakeel M, Raza S. Nonlinear Computational Model of Biological Cell Proliferation and Nutrient Delivery in a Bioreactor. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/am.2014.515222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Improvement of PHBV scaffolds with bioglass for cartilage tissue engineering. PLoS One 2013; 8:e71563. [PMID: 23951190 PMCID: PMC3739736 DOI: 10.1371/journal.pone.0071563] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 07/01/2013] [Indexed: 11/19/2022] Open
Abstract
Polymer scaffold systems consisting of poly(hydroxybutyrate-co-hydroxyvalerate) (PHBV) have proven to be possible matrices for the three-dimensional growth of chondrocyte cultures. However, the engineered cartilage grown on these PHBV scaffolds is currently unsatisfactory for clinical applications due to PHBV’s poor hydrophilicity, resulting in inadequate thickness and poor biomechanical properties of the engineered cartilage. It has been reported that the incorporation of Bioglass (BG) into PHBV can improve the hydrophilicity of the composites. In this study, we compared the effects of PHBV scaffolds and PHBV/BG composite scaffolds on the properties of engineered cartilage in vivo. Rabbit articular chondrocytes were seeded into PHBV scaffolds and PHBV/BG scaffolds. Short-term in vitro culture followed by long-term in vivo transplantation was performed to evaluate the difference in cartilage regeneration between the cartilage layers grown on PHBV and PHBV/BG scaffolds. The results show that the incorporation of BG into PHBV efficiently improved both the hydrophilicity of the composites and the percentage of adhered cells and promoted cell migration into the inner part the constructs. With prolonged incubation time in vivo, the chondrocyte-scaffold constructs in the PHBV/BG group formed thicker cartilage-like tissue with better biomechanical properties and a higher cartilage matrix content than the constructs in the PHBV/BG group. These results indicate that PHBV/BG scaffolds can be used to prepare better engineered cartilage than pure PHBV.
Collapse
|
34
|
Simulated microgravity combined with polyglycolic acid scaffold culture conditions improves the function of pancreatic islets. BIOMED RESEARCH INTERNATIONAL 2013; 2013:150739. [PMID: 24024182 PMCID: PMC3758870 DOI: 10.1155/2013/150739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/01/2013] [Accepted: 06/16/2013] [Indexed: 01/13/2023]
Abstract
The in vitro culture of pancreatic islets reduces their immunogenicity and prolongs their availability for transplantation. Both simulated microgravity (sMG) and a polyglycolic acid scaffold (PGA) are believed to confer advantages to cell culture. Here, we evaluated the effects of sMG combined with a PGA on the viability, insulin-producing activity and morphological alterations of pancreatic islets. Under PGA-sMG conditions, the purity of the islets was ≥85%, and the islets had a higher survival rate and an increased ability to secrete insulin compared with islets cultured alone in the static, sMG, or PGA conditions. In addition, morphological analysis under scanning electron microscopy (SEM) revealed that the PGA-sMG treatment preserved the integral structure of the islets and facilitated islet adhesion to the scaffolds. These results suggest that PGA-sMG coculture has the potential to improve the viability and function of islets in vitro and provides a promising method for islet transplantation.
Collapse
|
35
|
Mesallati T, Buckley CT, Kelly DJ. A comparison of self-assembly and hydrogel encapsulation as a means to engineer functional cartilaginous grafts using culture expanded chondrocytes. Tissue Eng Part C Methods 2013; 20:52-63. [PMID: 23672760 DOI: 10.1089/ten.tec.2013.0118] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite an increased interest in the use of hydrogel encapsulation and cellular self-assembly (often termed "self-aggregating" or "scaffold-free" approaches) for tissue-engineering applications, to the best of our knowledge, no study to date has been undertaken to directly compare both approaches for generating functional cartilaginous grafts. The objective of this study was to directly compare self-assembly (SA) and agarose hydrogel encapsulation (AE) as a means to engineer such grafts using passaged chondrocytes. Agarose hydrogels (5 mm diameter × 1.5 mm thick) were seeded with chondrocytes at two cell seeding densities (900,000 cells or 4 million cells in total per hydrogel), while SA constructs were generated by adding the same number of cells to custom-made molds. Constructs were either supplemented with transforming growth factor (TGF)-β3 for 6 weeks, or only supplemented with TGF-β3 for the first 2 weeks of the 6 week culture period. The SA method was only capable of generating geometrically uniform cartilaginous tissues at high seeding densities (4 million cells). At these high seeding densities, we observed that total sulphated glycosaminoglycan (sGAG) and collagen synthesis was greater with AE than SA, with higher sGAG retention also observed in AE constructs. When normalized to wet weight, however, SA constructs exhibited significantly higher levels of collagen accumulation compared with agarose hydrogels. Furthermore, it was possible to engineer such functionality into these tissues in a shorter timeframe using the SA approach compared with AE. Therefore, while large numbers of chondrocytes are required to engineer cartilaginous grafts using the SA approach, it would appear to lead to the faster generation of a more hyaline-like tissue, with a tissue architecture and a ratio of collagen to sGAG content more closely resembling native articular cartilage.
Collapse
Affiliation(s)
- Tariq Mesallati
- 1 Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin , Dublin, Ireland
| | | | | |
Collapse
|
36
|
Ye K, Felimban R, Moulton SE, Wallace GG, Bella CD, Traianedes K, Choong PFM, Myers DE. Bioengineering of articular cartilage: past, present and future. Regen Med 2013; 8:333-49. [DOI: 10.2217/rme.13.28] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The treatment of cartilage defects poses a clinical challenge owing to the lack of intrinsic regenerative capacity of cartilage. The use of tissue engineering techniques to bioengineer articular cartilage is promising and may hold the key to the successful regeneration of cartilage tissue. Natural and synthetic biomaterials have been used to recreate the microarchitecture of articular cartilage through multilayered biomimetic scaffolds. Acellular scaffolds preserve the microarchitecture of articular cartilage through a process of decellularization of biological tissue. Although promising, this technique often results in poor biomechanical strength of the graft. However, biomechanical strength could be improved if biomaterials could be incorporated back into the decellularized tissue to overcome this limitation.
Collapse
Affiliation(s)
- Ken Ye
- Department of Orthopaedics, St Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia.
| | - Raed Felimban
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
- Department of Orthopaedics, St Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
| | - Simon E Moulton
- Intelligent Polymer Research Institute, University of Wollongong, ARC Centre of Excellence for Electromaterials Science (ACES), Squires Way, North Wollongong, New South Wales 2552, Australia
| | - Gordon G Wallace
- Intelligent Polymer Research Institute, University of Wollongong, ARC Centre of Excellence for Electromaterials Science (ACES), Squires Way, North Wollongong, New South Wales 2552, Australia
| | - Claudia Di Bella
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
- Department of Orthopaedics, St Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
| | - Kathy Traianedes
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Peter FM Choong
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
- Department of Orthopaedics, St Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
| | - Damian E Myers
- Department of Surgery, St Vincent’s Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
- Department of Orthopaedics, St Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
| |
Collapse
|
37
|
A temperature-cured dissolvable gelatin microsphere-based cell carrier for chondrocyte delivery in a hydrogel scaffolding system. Acta Biomater 2013; 9:6459-67. [PMID: 23142479 DOI: 10.1016/j.actbio.2012.10.047] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/27/2012] [Accepted: 10/30/2012] [Indexed: 12/27/2022]
Abstract
In this study, a novel therapeutic cell delivery methodology in the form of hydrogel encapsulating cell-laden microspheres was developed and investigated. As a model cell for cartilage tissue engineering, chondrocytes were successfully encapsulated in gelatin-based microspheres (mostly of diameter 50-100 μm, centred at 75-100 μm) with high cell viability during the formation of microspheres via a water-in-oil single emulsion process under a low temperature without any chemical treatment. These cell-laden microspheres were then encapsulated in alginate-based hydrogel constructs. By elevating the temperature to 37°C, the cell-laden microspheres were completely dissolved within 2 days, resulting in the same number of same-sized spherical cavities in hydrogel bulk, along with which the encapsulated cells were released from the microspheres and suspended inside the cavities to be cultivated for further development. In this cell delivery system, the microspheres played a dual role as both removable cell vehicles and porogens for creation of the intra-hydrogel cavities, in which the delivered cells were provided with both free living spaces and a better permeable environment. This temperature-cured dissolvable gelatin microsphere-based cell carrier (tDGMC) associating with cell-laden hydrogel scaffold was attempted and evaluated through WST-1, quantitative polymerase chain reaction, biochemical assays and various immunohistochemistry and histology stains. The results indicate that tDGMC technology can facilitate the delivery of chondrocytes, as a non-anchorage-dependent therapeutic cell, with significantly greater efficiency.
Collapse
|
38
|
Madry H, Kaul G, Zurakowski D, Vunjak-Novakovic G, Cucchiarini M. Cartilage constructs engineered from chondrocytes overexpressing IGF-I improve the repair of osteochondral defects in a rabbit model. Eur Cell Mater 2013; 25:229-47. [PMID: 23588785 PMCID: PMC4476264 DOI: 10.22203/ecm.v025a17] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Tissue engineering combined with gene therapy is a promising approach for promoting articular cartilage repair. Here, we tested the hypothesis that engineered cartilage with chondrocytes overexpressing a human insulin-like growth factor I (IGF-I) gene can enhance the repair of osteochondral defects, in a manner dependent on the duration of cultivation. Genetically modified chondrocytes were cultured on biodegradable polyglycolic acid scaffolds in dynamic flow rotating bioreactors for either 10 or 28 d. The resulting cartilaginous constructs were implanted into osteochondral defects in rabbit knee joints. After 28 weeks of in vivo implantation, immunoreactivity to ß-gal was detectable in the repair tissue of defects that received lacZ constructs. Engineered cartilaginous constructs based on IGF-I-overexpressing chondrocytes markedly improved osteochondral repair compared with control (lacZ) constructs. Moreover, IGF-I constructs cultivated for 28 d in vitro significantly promoted osteochondral repair vis-à-vis similar constructs cultivated for 10 d, leading to significantly decreased osteoarthritic changes in the cartilage adjacent to the defects. Hence, the combination of spatially defined overexpression of human IGF-I within a tissue-engineered construct and prolonged bioreactor cultivation resulted in most enhanced articular cartilage repair and reduction of osteoarthritic changes in the cartilage adjacent to the defect. Such genetically enhanced tissue engineering provides a versatile tool to evaluate potential therapeutic genes in vivo and to improve our comprehension of the development of the repair tissue within articular cartilage defects. Insights gained with additional exploration using this model may lead to more effective treatment options for acute cartilage defects.
Collapse
Affiliation(s)
- Henning Madry
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany,Department of Orthopaedic Surgery, Saarland University, Homburg, Germany,Address for correspondence: Henning Madry Centre of Experimental Orthopaedics Medical Faculty Building 37 Saarland University D-66421 Homburg, Germany Telephone Number: +49-6841-1624515 FAX Number: +49-6841-1624988
| | - Gunter Kaul
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - David Zurakowski
- Departments of Anaesthesia and Surgery, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Magali Cucchiarini
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany
| |
Collapse
|
39
|
Neumann AJ, Alini M, Archer CW, Stoddart MJ. Chondrogenesis of human bone marrow-derived mesenchymal stem cells is modulated by complex mechanical stimulation and adenoviral-mediated overexpression of bone morphogenetic protein 2. Tissue Eng Part A 2013; 19:1285-94. [PMID: 23289669 DOI: 10.1089/ten.tea.2012.0411] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Currently available methods to treat articular cartilage defects still fail to demonstrate satisfactory outcomes for many patients. Functional tissue engineering using human bone marrow-derived mesenchymal stem cells (hMSCs) is a promising alternative approach for the treatment of these defects. This study strived to investigate the combined effect of complex mechanical stimulation and adenoviral-mediated overexpression of bone morphogenetic protein 2 (BMP-2) on hMSC chondrogenesis. hMSCs were encapsulated in a fibrin hydrogel and seeded into biodegradable polyurethane (PU) scaffolds. A novel three-dimensional transduction protocol was used to transduce cells with an adenovirus encoding for BMP-2 (Ad.BMP-2). Control cells were left untransduced. Cells were cultured for 7 or 28 days in a chondropermessive medium, which lacks any exogenous growth factors. Thereby, the in vivo situation is mimicked more precisely. hMSCs in fibrin-PU composite scaffolds were either left as free-swelling controls or mechanically stimulated using a custom-built bioreactor system that is able to generate joint-like forces. Outcome parameters measured were BMP-2 concentration within the culture medium, and biochemical and gene expression analysis. Mechanical stimulation resulted in an upregulation of chondrogenic genes. Further, glycosaminoglycan (GAG)/DNA ratios were elevated in mechanically stimulated groups. Transduction with Ad.BMP-2 led to a pronounced upregulation of the gene aggrecan and an upregulation of Sox9 message after 7 days. Furthermore, a synergistic effect in combination with mechanical stimulation on collagen 2 message was detected after 7 days. This synergistic increase was more than 8-fold if compared to the additive effect of the application of each stimulus on its own. However, BMP-2 overexpression consistently resulted in a trend toward decreased GAG/DNA ratios in both mechanical stimulated and unloaded groups.
Collapse
Affiliation(s)
- Alexander J Neumann
- Musculoskeletal Regeneration Program, AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
| | | | | | | |
Collapse
|
40
|
Abstract
Tissue engineering (TE) has promise as a biological solution and a disease modifying treatment for arthritis. Although cartilage can be generated by TE, substantial inter- and intra-donor variability makes it impossible to guarantee optimal, reproducible results. TE cartilage must be able to perform the functions of native tissue, thus mechanical and biological properties approaching those of native cartilage are likely a pre-requisite for successful implantation. A quality-control assessment of these properties should be part of the implantation release criteria for TE cartilage. Release criteria should certify that selected tissue properties have reached certain target ranges, and should be predictive of the likelihood of success of an implant in vivo. Unfortunately, it is not currently known which properties are needed to establish release criteria, nor how close one has to be to the properties of native cartilage to achieve success. Achieving properties approaching those of native cartilage requires a clear understanding of the target properties and reproducible assessment methodology. Here, we review several main aspects of quality control as it applies to TE cartilage. This includes a look at known mechanical and biological properties of native cartilage, which should be the target in engineered tissues. We also present an overview of the state of the art of tissue assessment, focusing on native articular and TE cartilage. Finally, we review the arguments for developing and validating non-destructive testing methods for assessing TE products.
Collapse
Affiliation(s)
- Joseph M. Mansour
- Skeletal Research Center, Department of Biology Case Western Reserve University Cleveland, OH, 44106
| | - Jean F. Welter
- Skeletal Research Center, Department of Biology Case Western Reserve University Cleveland, OH, 44106
| |
Collapse
|
41
|
Zhang F, Leong W, Su K, Fang Y, Wang DA. A transduced living hyaline cartilage graft releasing transgenic stromal cell-derived factor-1 inducing endogenous stem cell homing in vivo. Tissue Eng Part A 2013; 19:1091-9. [PMID: 23167782 DOI: 10.1089/ten.tea.2012.0441] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Stromal cell-derived factor-1 (SDF-1), also known as a homing factor, is a potent chemokine that activates and directs mobilization, migration, and retention of certain cell species via systemic circulation. The responding homing cells largely consist of activated stem cells, so that, in case of tissue lesions, such SDF-1-induced cell migration may execute recruitment of endogenous stem cells to perform autoreparation and compensatory regeneration in situ. In this study, a recombinant adenoviral vector carrying SDF-1 transgene was constructed and applied to transduce a novel scaffold-free living hyaline cartilage graft (SDF-t-LhCG). As an engineered transgenic living tissue, SDF-t-LhCG is capable of continuously producing and releasing SDF-1 in vitro and in vivo. The in vitro trials were examined with ELISA, while the in vivo trials were subsequently performed via a subcutaneous implantation of SDF-t-LhCG in a nude mouse model, followed by series of biochemical and biological analyses. The results indicate that transgenic SDF-1 enhanced the presence of this chemokine in mouse's circulation system; in consequence, SDF-1-induced activation and recruitment of endogenous stem cells were also augmented in both peripheral blood and SDF-t-LhCG implant per se. These results were obtained via flow cytometry analyses on mouse blood samples and implanted SDF-t-LhCG samples, indicating an upregulation of the CXCR4(+)(SDF-1 receptor) cell population, accompanied by upregulation of the CD34(+), CD44(+), and Sca-1(+) cell populations as well as a downregulation of the CD11b(+) cell population. With the supply of SDF-1-recruited endogenous stem cells, enhanced chondrogenesis was observed in SDF-t-LhCG implants in situ.
Collapse
Affiliation(s)
- Feng Zhang
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Republic of Singapore
| | | | | | | | | |
Collapse
|
42
|
2-D coupled computational model of biological cell proliferation and nutrient delivery in a perfusion bioreactor. Math Biosci 2013; 242:86-94. [PMID: 23291465 DOI: 10.1016/j.mbs.2012.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Revised: 12/09/2012] [Accepted: 12/18/2012] [Indexed: 11/21/2022]
Abstract
Tissue engineering aims to regenerate, repair or replace organs or tissues which have become defective due to trauma, disease or age related degeneration. This engineering may take place within the patient's body or tissue can be regenerated in a bioreactor for later implantation into the patient. Regeneration of soft tissue is one of the most demanding applications of tissue engineering. Producing proper nutrient supply, uniform cell distribution and high cell density are the important challenges. Many experimental models exist for tissue growth in a bioreactor. It is important to put experiments into a theoretical framework. Mathematical modelling in terms of physical and biochemical mechanisms is the best tool to understand experimental results. In this work a mathematical model of convective and diffusive transport of nutrients and cell evolution in a perfusion bioreactor is developed. A cell-seeded porous scaffold is placed in a perfusion bioreactor and fluid delivers the nutrients to the cells for their growth. The model describes the key features of the tissue engineering processes which includes the interaction between the cell growth, variation of material permeability due to cell proliferation, flow of fluid through the material and delivery of nutrients to the cells. The fluid flow through the porous scaffold is modelled by Darcy's law, and the delivery of nutrients to the cells is modelled by the advection-diffusion equation. A non-linear reaction diffusion system is used to model the cell growth. The growth of cells is modelled by logistic growth. COMSOL (a commercial finite element solver) is used to numerically solve the model. The results show that the distribution of cells and total cell number in the scaffold does not depend on the initial cell density but depend on the material permeability.
Collapse
|
43
|
Zhang F, Yao Y, Su K, Fang Y, Citra F, Wang DA. Co-transduction of lentiviral and adenoviral vectors for co-delivery of growth factor and shRNA genes in mesenchymal stem cells-based chondrogenic system. J Tissue Eng Regen Med 2012. [DOI: 10.1002/term.1656] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Feng Zhang
- Division of Bioengineering, School of Chemical and Biomedical Engineering; Nanyang Technological University; Singapore
| | - Yongchang Yao
- Division of Bioengineering, School of Chemical and Biomedical Engineering; Nanyang Technological University; Singapore
- School of Materials Science and Engineering; South China University of Technology; Guangzhou 510641 People's Republic of China
- National Engineering Research Centre for Tissue Restoration and Reconstruction; Guangzhou 510006 People's Republic of China
| | - Kai Su
- Division of Bioengineering, School of Chemical and Biomedical Engineering; Nanyang Technological University; Singapore
| | - Yu Fang
- Division of Bioengineering, School of Chemical and Biomedical Engineering; Nanyang Technological University; Singapore
| | - Fudiman Citra
- Division of Bioengineering, School of Chemical and Biomedical Engineering; Nanyang Technological University; Singapore
| | - Dong-An Wang
- Division of Bioengineering, School of Chemical and Biomedical Engineering; Nanyang Technological University; Singapore
| |
Collapse
|
44
|
Zhang F, Su K, Fang Y, Sandhya S, Wang DA. A mixed co-culture of mesenchymal stem cells and transgenic chondrocytes in alginate hydrogel for cartilage tissue engineering. J Tissue Eng Regen Med 2012; 9:77-84. [PMID: 23166064 DOI: 10.1002/term.1641] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 08/31/2012] [Accepted: 09/27/2012] [Indexed: 01/10/2023]
Abstract
To regenerate articular cartilage tissue from degeneration and trauma, synovial mesenchymal stem cells (SMSCs) were used in this study as therapeutic progenitor cells to induce therapeutic chondrogenesis. To accomplish this, chondrocytes pre-transduced with adenoviral vectors carrying the transforming growth factor (TGF) β3 gene were selected as transgenic companion cells and co-cultured side-by-side with SMSCs in a 3D environment to provide chondrogenic growth factors in situ. We adopted a mixed co-culture strategy for this purpose. Transgenic delivery of TGF-β3 in chondrocytes was performed via recombinant adenoviral vectors. The mixed co-culture of SMSCs and transgenic chondrocytes was produced in alginate gel constructs. Gene expression in both SMSCs and chondrocytes were characterized. Biochemical assays in vitro and in vivo showed that release of TGF-ß3 from transgenic chondrocytes not only induced SMSC differentiation into chondrocytic cells but also preserved the chondrocytic phenotype of chondrocytes from suspected dedifferentiation. As a result, this mixed co-culture strategy in conjunction with TGF-ß3 gene delivery could be a promising approach in cartilage tissue engineering.
Collapse
Affiliation(s)
- Feng Zhang
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Republic of Singapore
| | | | | | | | | |
Collapse
|
45
|
Izadifar Z, Chen X, Kulyk W. Strategic design and fabrication of engineered scaffolds for articular cartilage repair. J Funct Biomater 2012; 3:799-838. [PMID: 24955748 PMCID: PMC4030923 DOI: 10.3390/jfb3040799] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 09/13/2012] [Accepted: 10/17/2012] [Indexed: 01/19/2023] Open
Abstract
Damage to articular cartilage can eventually lead to osteoarthritis (OA), a debilitating, degenerative joint disease that affects millions of people around the world. The limited natural healing ability of cartilage and the limitations of currently available therapies make treatment of cartilage defects a challenging clinical issue. Hopes have been raised for the repair of articular cartilage with the help of supportive structures, called scaffolds, created through tissue engineering (TE). Over the past two decades, different designs and fabrication techniques have been investigated for developing TE scaffolds suitable for the construction of transplantable artificial cartilage tissue substitutes. Advances in fabrication technologies now enable the strategic design of scaffolds with complex, biomimetic structures and properties. In particular, scaffolds with hybrid and/or biomimetic zonal designs have recently been developed for cartilage tissue engineering applications. This paper reviews critical aspects of the design of engineered scaffolds for articular cartilage repair as well as the available advanced fabrication techniques. In addition, recent studies on the design of hybrid and zonal scaffolds for use in cartilage tissue repair are highlighted.
Collapse
Affiliation(s)
- Zohreh Izadifar
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, 57 Campus Dr., Saskatoon SK S7N5A9, Canada.
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, 57 Campus Dr., Saskatoon SK S7N5A9, Canada.
| | - William Kulyk
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, 107 Wiggins Rd., Saskatoon SK S7N 5E5, Canada.
| |
Collapse
|
46
|
Singhatanadgit W, Varodomrujiranon M. Osteogenic potency of a 3-dimensional scaffold-free bonelike sphere of periodontal ligament stem cells in vitro. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 116:e465-72. [PMID: 22901658 DOI: 10.1016/j.oooo.2012.02.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 01/27/2012] [Accepted: 02/21/2012] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The present study aimed to investigate the osteogenic potency of scaffold-free 3-dimensional (3D) spheres of periodontal ligament stem cells (PDLSCs). STUDY DESIGN The osteogenic potency of PDLSC spheres was determined by the ability to form mineralization and to express key osteogenesis-associated genes. The alkaline phosphatase (ALP) activity and the protein content of PDLSC spheres were also measured. RESULTS The 3D sphere developed its osteogenic potency in a time-dependent manner, containing approximately 10-fold higher mineralization, 5-fold higher protein content, and 4-fold greater ALP activity than those in the controls. The expression of key osteogenic genes was also upregulated in the 3D PDLSC spheres. Cellular outgrowth was observed when reintroduced into 2D culture. CONCLUSIONS PDLSCs were able to undergo osteogenic differentiation in a scaffold-free 3D culture, producing bonelike mineralization in vitro. This suggests, at least in vitro, the osteogenic potency of the 3D PDLSC spheres.
Collapse
Affiliation(s)
- Weerachai Singhatanadgit
- Assistant Professor, Faculty of Dentistry, Thammasat University, Klong, Luang, Pathum-Thani, Thailand.
| | | |
Collapse
|
47
|
Kuo SM, Chiang MY, Lan CW, Niu GCC, Chang SJ. Evaluation of nanoarchitectured collagen type II molecules on cartilage engineering. J Biomed Mater Res A 2012; 101:368-77. [DOI: 10.1002/jbm.a.34335] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 03/05/2012] [Accepted: 06/15/2012] [Indexed: 12/29/2022]
|
48
|
Yan X, Bergstrom DJ, Chen XB. Modeling of cell cultures in perfusion bioreactors. IEEE Trans Biomed Eng 2012; 59:2568-75. [PMID: 22772976 DOI: 10.1109/tbme.2012.2206077] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cultivating cells and tissues in bioreactors is a critical step in forming artificial tissues or organs prior to transplantation. Among various bioreactors, the perfusion bioreactor is known for its enhanced convection through the cell-scaffold constructs. Knowledge of mass transfer is essential for controlling the cell culture process; however, obtaining this information remains a challenging task. In this research, a novel mathematical model is developed to represent the nutrient transport and cell growth in a 3-D scaffold cultivated in a perfusion bioreactor. Numerical methods are employed to solve the equations involved, with a focus on identifying the effect of factors such as porosity, culturing time, and flow rate, which are controllable in the scaffold fabrication and culturing process, on cell cultures. To validate the new model, the results from the model simulations were compared to the experimental results extracted from the literature. With the validated model, further simulations were carried out to investigate the glucose and oxygen distribution and the cell growth within the cell-scaffold construct in a perfusion bioreactor, thus providing insight into the cell culture process.
Collapse
Affiliation(s)
- X Yan
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, Canada.
| | | | | |
Collapse
|
49
|
Reuther MS, Wong VW, Briggs KK, Chang AA, Nguyen QT, Schumacher BL, Masuda K, Sah RL, Watson D. Culture of human septal chondrocytes in a rotary bioreactor. Otolaryngol Head Neck Surg 2012; 147:661-7. [PMID: 22597575 DOI: 10.1177/0194599812448049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES (1) To show that extracellular matrix deposition in 3-dimensional culture of human septal chondrocytes cultured in a rotary bioreactor is comparable to the deposition achieved under static culture conditions. (2) To demonstrate that the biomechanical properties of human septal chondrocytes cultured in a bioreactor are enhanced with time and are analogous to beads cultured under static culture. STUDY DESIGN Prospective, basic science. SETTING Research laboratory. METHODS Human septal chondrocytes from 9 donors were expanded in monolayer and seeded in alginate beads. The beads were cultured in a rotary bioreactor for 21 days in media supplemented with growth factors and human serum, using static culture as the control. Biochemical and biomechanical properties of the beads were measured. RESULTS Glycosaminoglycan (GAG) accumulation significantly increased during 2 measured time intervals, 0 to 21 days and 10 to 21 days (P < .01). No significant difference was seen between the static and bioreactor conditions. Substantial type II collagen production was demonstrated in the beads terminated at day 21 of culture in both conditions. In addition, the biomechanical properties of the beads were significantly improved at 21 days in comparison to beads from day 0. CONCLUSION Human septal chondrocytes cultured in alginate beads exhibit significant matrix deposition and improved biomechanical properties after 21 days. Alginate bead diameter and stiffness positively correlated with GAG and type II collagen accretion. Matrix production in beads is supported by the use of a rotary bioreactor.
Collapse
Affiliation(s)
- Marsha S Reuther
- Division of Otolaryngology-Head and Neck Surgery, University of California, San Diego, La Jolla, California, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rogers CM, Woolley TS, Cruwys SC, Buttery LDK, Rose FRAJ, Shakesheff KM. Engineering an in-vitro model of rodent cartilage. J Pharm Pharmacol 2012; 64:821-31. [PMID: 22571260 DOI: 10.1111/j.2042-7158.2012.01491.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVES The purpose of this study was to identify a cell source, scaffold substrate and culture environment suitable for use in engineering an in-vitro model of rodent cartilage. METHODS The chondrogenic activity and stability of cells isolated at Day 18 of gestation was assessed under normoxia and hypoxia using a cytokine stimulation assay and gene expression analysis. The ability of the selected cells seeded in fibrous electrospun scaffolds to form cartilaginous tissue during longterm static and dynamic culture was assessed using immunocytochemistry and biochemical analysis. KEY FINDINGS Rodent fetal chondrocytes appear to have enhanced phenotypic stability compared with other cell sources. Following 16 weeks under static culture, the engineered constructs were found to have greater cellularity and collagen content that native rodent cartilage. CONCLUSIONS A cell source, scaffold and culture environment have been identified that support the generation of in-vitro rodent cartilage. In future work, cytokine treatment of the engineered tissues will take place to generate in-vitro osteoarthritis models.
Collapse
Affiliation(s)
- Catherine M Rogers
- School of Pharmacy, University of Nottingham, Nottingham R&D, AstraZeneca, Leics, Loughborough, UK.
| | | | | | | | | | | |
Collapse
|