1
|
Yang GD, Ma DS, Ma CY, Bai Y. Research Progress on Cardiac Tissue Construction of Mesenchymal Stem Cells for Myocardial Infarction. Curr Stem Cell Res Ther 2024; 19:942-958. [PMID: 37612870 DOI: 10.2174/1574888x18666230823091017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/25/2023]
Abstract
Heart failure is still the main complication affecting the prognosis of acute myocardial infarction (AMI), and mesenchymal stem cells (MSCs) are an effective treatment to replace necrotic myocardium and improve cardiac functioning. However, the transplant survival rate of MSCs still presents challenges. In this review, the biological characteristics of MSCs, the progress of mechanism research in the treatment of myocardial infarction, and the advances in improving the transplant survival rate of MSCs in the replacement of necrotic myocardial infarction are systematically described. From a basic to advanced clinical research, MSC transplants have evolved from a pure injection, an exosome injection, the genetic modification of MSCs prior to injection to the cardiac tissue engineering of MSC patch grafting. This study shows that MSCs have wide clinical applications in the treatment of AMI, suggesting improved myocardial tissue creation. A broader clinical application prospect will be explored and developed to improve the survival rate of MSC transplants and myocardial vascularization.
Collapse
Affiliation(s)
- Guo-Dong Yang
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Da-Shi Ma
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Chun-Ye Ma
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yang Bai
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
2
|
Higuera GA, Ramos T, Gloria A, Ambrosio L, Di Luca A, Pechkov N, de Wijn JR, van Blitterswijk CA, Moroni L. PEOT/PBT Polymeric Pastes to Fabricate Additive Manufactured Scaffolds for Tissue Engineering. Front Bioeng Biotechnol 2021; 9:704185. [PMID: 34595158 PMCID: PMC8476768 DOI: 10.3389/fbioe.2021.704185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/30/2021] [Indexed: 11/13/2022] Open
Abstract
The advantages of additive manufactured scaffolds, as custom-shaped structures with a completely interconnected and accessible pore network from the micro- to the macroscale, are nowadays well established in tissue engineering. Pore volume and architecture can be designed in a controlled fashion, resulting in a modulation of scaffold’s mechanical properties and in an optimal nutrient perfusion determinant for cell survival. However, the success of an engineered tissue architecture is often linked to its surface properties as well. The aim of this study was to create a family of polymeric pastes comprised of poly(ethylene oxide therephthalate)/poly(butylene terephthalate) (PEOT/PBT) microspheres and of a second biocompatible polymeric phase acting as a binder. By combining microspheres with additive manufacturing technologies, we produced 3D scaffolds possessing a tailorable surface roughness, which resulted in improved cell adhesion and increased metabolic activity. Furthermore, these scaffolds may offer the potential to act as drug delivery systems to steer tissue regeneration.
Collapse
Affiliation(s)
- Gustavo A Higuera
- Institute for BioMedical Technology and Technical Medicine (MIRA), Tissue Regeneration Department, University of Twente, Enschede, Netherlands
| | - Tiago Ramos
- Institute of Ophthalmology, University College of London, London, United Kingdom
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Luigi Ambrosio
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Andrea Di Luca
- Institute for BioMedical Technology and Technical Medicine (MIRA), Tissue Regeneration Department, University of Twente, Enschede, Netherlands
| | - Nicholas Pechkov
- Institute for BioMedical Technology and Technical Medicine (MIRA), Tissue Regeneration Department, University of Twente, Enschede, Netherlands
| | - Joost R de Wijn
- Institute for BioMedical Technology and Technical Medicine (MIRA), Tissue Regeneration Department, University of Twente, Enschede, Netherlands
| | - Clemens A van Blitterswijk
- MERLN Institute for Technology-inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, Netherlands
| | - Lorenzo Moroni
- MERLN Institute for Technology-inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
3
|
Abstract
Cell-based therapy is a promising approach in the field of regenerative medicine. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., nano- or microparticles, fibers, and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. Therefore, cell spheroids in synergy with biomaterials have recently emerged for cell-based regenerative therapy. Biomaterials-assisted spheroid engineering has been extensively studied for regeneration of bone or/and cartilage defects, critical limb ischemia, and myocardial infarction. Furthermore, it has been expanded to pancreas islets and hair follicle transplantation. This paper comprehensively reviews biomaterials-assisted spheroid engineering for regenerative therapy.
Collapse
Affiliation(s)
- Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Zhou Zechu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | | |
Collapse
|
4
|
Lee NH, Bayaraa O, Zechu Z, Kim HS. Biomaterials-assisted spheroid engineering for regenerative therapy. BMB Rep 2021; 54:356-367. [PMID: 34154700 PMCID: PMC8328824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/23/2021] [Accepted: 06/15/2021] [Indexed: 04/04/2024] Open
Abstract
Cell-based therapy is a promising approach in the field of regenerative medicine. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., nano- or microparticles, fibers, and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. Therefore, cell spheroids in synergy with biomaterials have recently emerged for cell-based regenerative therapy. Biomaterials-assisted spheroid engineering has been extensively studied for regeneration of bone or/and cartilage defects, critical limb ischemia, and myocardial infarction. Furthermore, it has been expanded to pancreas islets and hair follicle transplantation. This paper comprehensively reviews biomaterials-assisted spheroid engineering for regenerative therapy. [BMB Reports 2021; 54(7): 356-367].
Collapse
Affiliation(s)
- Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Zhou Zechu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
5
|
R A V, Kumari S, Poddar P, Dhara D, Maiti S. Poly(N-isopropylacrylamide)-Based Polymers as Additive for Rapid Generation of Spheroid via Hanging Drop Method. Macromol Biosci 2020; 20:e2000180. [PMID: 32794360 DOI: 10.1002/mabi.202000180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Indexed: 11/09/2022]
Abstract
Multicellular tumor spheroid (MCTS) mimics microenvironment for tumor formation and provides predictive insight for in vivo tests. The hanging drop (HD) method of spheroid generation is cost effective, but it is limited by a long time duration for spheroid development and a low rate of formation of larger spheroids. Toward addressing those limitations, thermoresponsive copolymers with poly(N-isopropylacrylamide) (p(NIPA)) backbone are developed, to be used as additives in the MCTS formation via HD method. Upon investigation it is found that in the presence of the polymer, robust and compact spheroids are formed in a short duration of 48 h. Larger spheroids (350-600 µm) can be formed by increasing the number of cells. Spheroids are characterized for their 3D shape and different cellular layers, and drug uptake study is done to prove the efficacy of the spheroids generated in drug screening.
Collapse
Affiliation(s)
- Vignesh R A
- Chemical and Systems Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, Delhi, 110025, India
| | - Shalini Kumari
- Chemical and Systems Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, Delhi, 110025, India
| | - Puja Poddar
- Department of Chemistry, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Dibakar Dhara
- Department of Chemistry, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Souvik Maiti
- Chemical and Systems Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, Delhi, 110025, India.,Academy of Scientific and Innovative Research, CSIR- Human Resource Development Centre, (CSIR-HRDC) Campus, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
6
|
Selig M, Lauer JC, Hart ML, Rolauffs B. Mechanotransduction and Stiffness-Sensing: Mechanisms and Opportunities to Control Multiple Molecular Aspects of Cell Phenotype as a Design Cornerstone of Cell-Instructive Biomaterials for Articular Cartilage Repair. Int J Mol Sci 2020; 21:E5399. [PMID: 32751354 PMCID: PMC7432012 DOI: 10.3390/ijms21155399] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/23/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-β1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-β1- and interleukin 1 beta (IL-1β)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jasmin C. Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Melanie L. Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| |
Collapse
|
7
|
Ching JY, Huang BJ, Hsu YT, Khung YL. Anti-Adhesion Behavior from Ring-Strain Amine Cyclic Monolayers Grafted on Silicon (111) Surfaces. Sci Rep 2020; 10:8758. [PMID: 32472042 PMCID: PMC7260185 DOI: 10.1038/s41598-020-65710-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/07/2020] [Indexed: 01/09/2023] Open
Abstract
In this manuscript, a series of amine tagged short cyclic molecules (cyclopropylamine, cyclobutylamine, cyclopentylamine and cyclohexylamine) were thermally grafted onto p-type silicon (111) hydride surfaces via nucleophilic addition. The chemistries of these grafting were verified via XPS, AFM and sessile droplet measurements. Confocal microscopy and cell viability assay was performed on these surfaces incubated for 24 hours with triple negative breast cancer cells (MDA-MB 231), gastric adenocarcinoma cells (AGS) endometrial adenocarcinoma (Hec1A). All cell types had shown a significant reduction when incubated on these ring-strain cyclic monolayer surfaces than compared to standard controls. The expression level of focal adhesion proteins (vinculin, paxilin, talin and zyxin) were subsequently quantified for all three cell types via qPCR analysis. Cells incubate on these surface grafting were observed to have reduced levels of adhesion protein expression than compared to positive controls (collagen coating and APTES). A potential application of these anti-adhesive surfaces is the maintenance of the chondrocyte phenotype during in-vitro cell expansion. Articular chondrocytes cultured for 6 days on ring strained cyclopropane-modified surfaces was able to proliferate but had maintained a spheroid/aggregated phenotype with higher COL2A1 and ACAN gene expression. Herein, these findings had help promote grafting of cyclic monolayers as an viable alternative for producing antifouling surfaces.
Collapse
Affiliation(s)
- Jing Yuan Ching
- Department of Biological Science and Technology, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Brian J Huang
- Integrative Stem Cell Center, China Medical University Hospital, Taichung, 40447, Taiwan.,Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Yu-Ting Hsu
- Department of Biological Science and Technology, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Yit Lung Khung
- Department of Biological Science and Technology, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan.
| |
Collapse
|
8
|
Carbon nanotube multilayered nanocomposites as multifunctional substrates for actuating neuronal differentiation and functions of neural stem cells. Biomaterials 2018; 175:93-109. [DOI: 10.1016/j.biomaterials.2018.05.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 12/17/2022]
|
9
|
Wongin S, Waikakul S, Chotiyarnwong P, Siriwatwechakul W, Kino-Oka M, Kim MH, Viravaidya-Pasuwat K. Maintenance of human chondrogenic phenotype on a dendrimer-immobilized surface for an application of cell sheet engineering. BMC Biotechnol 2018. [PMID: 29540167 PMCID: PMC5853058 DOI: 10.1186/s12896-018-0426-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dedifferentiation of chondrocytes during cell expansion is one of the barriers in tissue construction for cartilage repair. To understand chondrocyte behavior and improve cell expansion in monolayer culture, this study investigated the effects of morphological changes and cellular aggregation on the maintenance of chondrogenic capacity by observing the expression patterns of chondrogenic (collagen type II and aggrecan) and dedifferentiation (collagen type I) markers. Primary human chondrocytes were cultured on either a polystyrene surface (PS) or a polyamidoamine dendrimer surface with a fifth-generation (G5) dendron structure to create a one-step process of cell expansion and the maintenance of chondrogenic activities prior to the construction of cell sheets. RESULTS During the first two passages (P0 - P2), the relative mRNA level of collagen type II decreased in all cultures, while that of collagen type I increased. Remarkably, the level of collagen type II was higher and aggrecan was retained in the chondrocytes, forming cell aggregates and showing some round-shaped cells with less production of stress fibers on the G5 surface compared to fibroblast-like chondrocytes with abundant stress fibers on the PS surface. The numbers of P2 chondrocytes on the G5 and PS surfaces were nearly the same and sufficient for construction of chondrocyte sheets using a temperature-responsive plate. Without a supporting material during cell sheet manipulation, chondrocyte sheets spontaneously detached and exhibited a honeycomb-like structure of stress fibers. Unlike the chondrocyte sheets constructed from cells on the PS surface, the chondrocyte sheets from cells on the G5 surface had higher chondrogenic activities, as evidenced by the high expression of chondrogenic markers and the low expression of dedifferentiation markers. CONCLUSIONS The one-step process of cell expansion and maintenance of chondrogenic activity could be obtained using the G5 surface. Human chondrocyte sheets were successfully constructed with high chondrogenic activity. These findings may lead to an alternative cultivation technique for human chondrocytes that offers high clinical potential in autologous chondrocyte implantation.
Collapse
Affiliation(s)
- Sopita Wongin
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
| | - Saranatra Waikakul
- Department of Orthopaedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pojchong Chotiyarnwong
- Department of Orthopaedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Wanwipa Siriwatwechakul
- School of Bio-Chemical Engineering and Technology, Sirindhorn International Institute of Technology, Thammasat University, Pathum Thani, 12121, Thailand
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kwanchanok Viravaidya-Pasuwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand. .,Department of Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand.
| |
Collapse
|
10
|
Wang S, Zan F, Ke Y, Wu G. Cells may feel a hard substrate even on a grafted layer of soft hydrogel. J Mater Chem B 2018; 6:1734-1743. [PMID: 32254245 DOI: 10.1039/c7tb02967e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Introducing or grafting molecules onto biomaterial surfaces to regulate cell destination via biophysical cues is one of the important steps for biomaterial design in tissue engineering. Understanding how cells feel the substrate makes it easier to learn the mechanism behind cell-material interaction. In this study, on a glass substrate, we constructed poly-phenoxyethyl methacrylate (PHEMA) brushes having different lengths via a surface-induced atom transfer radical polymerization (SI-ATRP) method. FTIR-ATR and XPS tests of the formed polymer brushes indicate that these brushes have characteristic chemical structures of PHEMA; the polymer brush length revealed by the AFM tests increases linearly with reaction time. Cell lines of BMSCs, ATDC5, and human chondrocytes (HC) were cultured on these substrates to evaluate proliferation, adhesion, and differentiation. Our results demonstrated that the cells cultured on the substrates with short PHEMA brushes developed a spread morphology and organized actin fibers as compared to the cells cultured on those with long brushes. Different cell lines showed different responses depending on the PHEMA brush length. Cells cultured on long PHEMA brushes displayed a more rounded shape, higher gene expression of FAK and integrin, and lower gene expression of NCAM and N-cadherin as compared to those, especially ATDC5 cells, cultured on short PHEMA brushes. On PHEMA brushes with a long length, the cell lines express higher cartilage-specific genes including Sox9 and Col2 and GAG in ECM. The results suggest that polymer brushes having different lengths may interfere with the behavior of the cells cultured on them.
Collapse
Affiliation(s)
- Shuhao Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
| | | | | | | |
Collapse
|
11
|
Wongin S, Waikakul S, Chotiyarnwong P, Siriwatwechakul W, Viravaidya-Pasuwat K. Effect of Cell Sheet Manipulation Techniques on the Expression of Collagen Type II and Stress Fiber Formation in Human Chondrocyte Sheets. Tissue Eng Part A 2018; 24:469-478. [DOI: 10.1089/ten.tea.2017.0013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sopita Wongin
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Saranatra Waikakul
- Department of Orthopaedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pojchong Chotiyarnwong
- Department of Orthopaedic Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanwipa Siriwatwechakul
- School of Bio-Chemical Engineering and Technology, Sirindhorn International Institute of Technology, Thammasat University, Pathum Thani, Thailand
| | - Kwanchanok Viravaidya-Pasuwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
- Department of Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| |
Collapse
|
12
|
Biocompatibility of hydrogel-based scaffolds for tissue engineering applications. Biotechnol Adv 2017; 35:530-544. [DOI: 10.1016/j.biotechadv.2017.05.006] [Citation(s) in RCA: 521] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 05/08/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
|
13
|
Lin H, Zhou J, Cao L, Wang HR, Dong J, Chen ZR. Tissue-engineered cartilage constructed by a biotin-conjugated anti-CD44 avidin binding technique for the repairing of cartilage defects in the weight-bearing area of knee joints in pigs. Bone Joint Res 2017; 6:284-295. [PMID: 28515058 PMCID: PMC5457648 DOI: 10.1302/2046-3758.65.bjr-2016-0277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/20/2017] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES The lack of effective treatment for cartilage defects has prompted investigations using tissue engineering techniques for their regeneration and repair. The success of tissue-engineered repair of cartilage may depend on the rapid and efficient adhesion of transplanted cells to a scaffold. Our aim in this study was to repair full-thickness defects in articular cartilage in the weight-bearing area of a porcine model, and to investigate whether the CD44 monoclonal antibody biotin-avidin (CBA) binding technique could provide satisfactory tissue-engineered cartilage. METHODS Cartilage defects were created in the load-bearing region of the lateral femoral condyle of mini-type pigs. The defects were repaired with traditional tissue-engineered cartilage, tissue-engineered cartilage constructed with the biotin-avidin (BA) technique, tissue-engineered cartilage constructed with the CBA technique and with autologous cartilage. The biomechanical properties, Western blot assay, histological findings and immunohistochemical staining were explored. RESULTS The CBA group showed similar results to the autologous group in biomechanical properties, Moran's criteria, histological tests and Wakitani histological scoring. CONCLUSIONS These results suggest that tissue-engineered cartilage constructed using the CBA technique could be used effectively to repair cartilage defects in the weight-bearing area of joints.Cite this article: H. Lin, J. Zhou, L. Cao, H. R. Wang, J. Dong, Z. R. Chen. Tissue-engineered cartilage constructed by a biotin-conjugated anti-CD44 avidin binding technique for the repairing of cartilage defects in the weight-bearing area of knee joints in pigs. Bone Joint Res 2017;6:-295. DOI: 10.1302/2046-3758.65.BJR-2016-0277.
Collapse
Affiliation(s)
- H Lin
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - J Zhou
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - L Cao
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - H R Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - J Dong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Z R Chen
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
14
|
Cui X, Hartanto Y, Zhang H. Advances in multicellular spheroids formation. J R Soc Interface 2017; 14:20160877. [PMID: 28202590 PMCID: PMC5332573 DOI: 10.1098/rsif.2016.0877] [Citation(s) in RCA: 328] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/11/2017] [Indexed: 12/20/2022] Open
Abstract
Three-dimensional multicellular spheroids (MCSs) have a complex architectural structure, dynamic cell-cell/cell-matrix interactions and bio-mimicking in vivo microenvironment. As a fundamental building block for tissue reconstruction, MCSs have emerged as a powerful tool to narrow down the gap between the in vitro and in vivo model. In this review paper, we discussed the structure and biology of MCSs and detailed fabricating methods. Among these methods, the approach in microfluidics with hydrogel support for MCS formation is promising because it allows essential cell-cell/cell-matrix interactions in a confined space.
Collapse
Affiliation(s)
- X Cui
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Y Hartanto
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - H Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
15
|
Wang H, Wu G, Zhang J, Zhou K, Yin B, Su X, Qiu G, Yang G, Zhang X, Zhou G, Wu Z. Osteogenic effect of controlled released rhBMP-2 in 3D printed porous hydroxyapatite scaffold. Colloids Surf B Biointerfaces 2016; 141:491-498. [PMID: 26896655 DOI: 10.1016/j.colsurfb.2016.02.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/02/2016] [Accepted: 02/03/2016] [Indexed: 01/13/2023]
Abstract
Recently, 3D printing as effective technology has been highlighted in the biomedical field. Previously, a porous hydroxyapatite (HA) scaffold with the biocompatibility and osteoconductivity has been developed by this method. However, its osteoinductivity is limited. The main purpose of this study was to improve it by the introduction of recombinant human bone morphogenetic protein-2 (rhBMP-2). This scaffold was developed by coating rhBMP-2-delivery microspheres with collagen. These synthesized scaffolds were characterized by Scanning Electron Microscopy (SEM), a delivery test in vitro, cell culture, and the experiments in vivo by a Micro-computed tomography (μCT) scan and histological evaluation of VanGieson staining. SEM results indicated the surface of scaffolds were more fit for the adhesion of hMSCs to coat collagen/rhBMP-2 microspheres. Biphasic release of rhBMP-2 could continue for more than 21 days, and keep its osteoinductivity to induce osteogenic differentiation of hMSCs in vitro. In addition, the experiments in vivo showed that the scaffold had a good bone regeneration capacity. These findings demonstrate that the HA/Collagen/Chitosan Microspheres system can simultaneously achieve localized long-term controlled release of rhBMP-2 and bone regeneration, which provides a promising route for improving the treatment of bone defects.
Collapse
Affiliation(s)
- Hai Wang
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital (PUMCH), Beijing 100730, China
| | - Gui Wu
- Department of Orthopaedics, First Affiliated Hospital, Fujian Medical University, Fujian 350108, China
| | - Jing Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Kui Zhou
- College of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bo Yin
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital (PUMCH), Beijing 100730, China
| | - Xinlin Su
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital (PUMCH), Beijing 100730, China
| | - Guixing Qiu
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital (PUMCH), Beijing 100730, China
| | - Guang Yang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xianglin Zhang
- College of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Gang Zhou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Zhihong Wu
- Central Laboratory, Peking Union Medical College Hospital (PUMCH), Beijing 100730, China; Beijing Key Laboratory for Genetic Research of Bone and Joint Disease, Beijing 100730, China.
| |
Collapse
|
16
|
Kutikov AB, Song J. Biodegradable PEG-Based Amphiphilic Block Copolymers for Tissue Engineering Applications. ACS Biomater Sci Eng 2015; 1:463-480. [PMID: 27175443 PMCID: PMC4860614 DOI: 10.1021/acsbiomaterials.5b00122] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biodegradable tissue engineering scaffolds have great potential for delivering cells/therapeutics and supporting tissue formation. Polyesters, the most extensively investigated biodegradable synthetic polymers, are not ideally suited for diverse tissue engineering applications due to limitations associated with their hydrophobicity. This review discusses the design and applications of amphiphilic block copolymer scaffolds integrating hydrophilic poly(ethylene glycol) (PEG) blocks with hydrophobic polyesters. Specifically, we highlight how the addition of PEG results in striking changes to the physical properties (swelling, degradation, mechanical, handling) and biological performance (protein & cell adhesion) of the degradable synthetic scaffolds in vitro. We then perform a critical review of how these in vitro characteristics translate to the performance of biodegradable amphiphilic block copolymer-based scaffolds in the repair of a variety of tissues in vivo including bone, cartilage, skin, and spinal cord/nerve. We conclude the review with recommendations for future optimizations in amphiphilic block copolymer design and the need for better-controlled in vivo studies to reveal the true benefits of the amphiphilic synthetic tissue scaffolds.
Collapse
Affiliation(s)
- Artem B. Kutikov
- Department of Orthopedics and Physical Rehabilitation. University of Massachusetts Medical School. 55 Lake Ave North, Worcester, MA 01655, USA
| | - Jie Song
- Department of Orthopedics and Physical Rehabilitation. University of Massachusetts Medical School. 55 Lake Ave North, Worcester, MA 01655, USA
- Department of Cell and Developmental Biology. University of Massachusetts Medical School. 55 Lake Ave North, Worcester, MA 01655, USA
| |
Collapse
|
17
|
Rothuizen TC, Damanik FF, Anderson JM, Lavrijsen T, Cox MA, Rabelink TJ, Moroni L, Rotmans JI. Tailoring the Foreign Body Response for In Situ Vascular Tissue Engineering. Tissue Eng Part C Methods 2015; 21:436-46. [DOI: 10.1089/ten.tec.2014.0264] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Tonia C. Rothuizen
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | | - James M. Anderson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | | | | | - Ton J. Rabelink
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lorenzo Moroni
- Department of Tissue Regeneration, University Twente, Enschede, The Netherlands
| | - Joris I. Rotmans
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
18
|
Dwivedi P, Bhat S, Nayak V, Kumar A. Study of Different Delivery Modes of Chondroitin Sulfate Using Microspheres and Cryogel Scaffold for Application in Cartilage Tissue Engineering. INT J POLYM MATER PO 2014. [DOI: 10.1080/00914037.2014.886223] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
19
|
Lin H, Zhou J, Shen L, Ruan Y, Dong J, Guo C, Chen Z. Biotin-conjugated anti-CD44 antibody-avidin binding system for the improvement of chondrocyte adhesion to scaffolds. J Biomed Mater Res A 2013; 102:1140-8. [PMID: 23630032 DOI: 10.1002/jbm.a.34770] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/09/2013] [Accepted: 04/23/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Hong Lin
- Department of Orthopaedic Surgery; Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Jian Zhou
- Department of Orthopaedic Surgery; Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Longxiang Shen
- Department of Orthopaedic Surgery; The 6th Hospital of Shanghai; Shanghai Jiaotong University; Shanghai 200233 China
| | - Yuhui Ruan
- Department of Macromolecular Science; Key Laboratory of Molecular Engineering of Polymers of Ministry of Education, Advanced Materials Laboratory, Fudan University; Shanghai 200433 China
| | - Jian Dong
- Department of Orthopaedic Surgery; Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Changan Guo
- Department of Orthopaedic Surgery; Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Zhengrong Chen
- Department of Orthopaedic Surgery; Zhongshan Hospital, Fudan University; Shanghai 200032 China
| |
Collapse
|
20
|
Buitinga M, Truckenmüller R, Engelse MA, Moroni L, Ten Hoopen HWM, van Blitterswijk CA, de Koning EJP, van Apeldoorn AA, Karperien M. Microwell scaffolds for the extrahepatic transplantation of islets of Langerhans. PLoS One 2013; 8:e64772. [PMID: 23737999 PMCID: PMC3667808 DOI: 10.1371/journal.pone.0064772] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/17/2013] [Indexed: 11/30/2022] Open
Abstract
Allogeneic islet transplantation into the liver has the potential to restore normoglycemia in patients with type 1 diabetes. However, the suboptimal microenvironment for islets in the liver is likely to be involved in the progressive islet dysfunction that is often observed post-transplantation. This study validates a novel microwell scaffold platform to be used for the extrahepatic transplantation of islet of Langerhans. Scaffolds were fabricated from either a thin polymer film or an electrospun mesh of poly(ethylene oxide terephthalate)-poly(butylene terephthalate) (PEOT/PBT) block copolymer (composition: 4000PEOT30PBT70) and were imprinted with microwells, ∼400 µm in diameter and ∼350 µm in depth. The water contact angle and water uptake were 39±2° and 52.1±4.0 wt%, respectively. The glucose flux through electrospun scaffolds was three times higher than for thin film scaffolds, indicating enhanced nutrient diffusion. Human islets cultured in microwell scaffolds for seven days showed insulin release and insulin content comparable to those of free-floating control islets. Islet morphology and insulin and glucagon expression were maintained during culture in the microwell scaffolds. Our results indicate that the microwell scaffold platform prevents islet aggregation by confinement of individual islets in separate microwells, preserves the islet’s native rounded morphology, and provides a protective environment without impairing islet functionality, making it a promising platform for use in extrahepatic islet transplantation.
Collapse
Affiliation(s)
- Mijke Buitinga
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Roman Truckenmüller
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Marten A. Engelse
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Lorenzo Moroni
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Hetty W. M. Ten Hoopen
- Department of BioMedical Chemisty, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | - Eelco JP. de Koning
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Endocrinology, Leiden University Medical Centre, Leiden, The Netherlands
- Hubrecht Institute–Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Aart A. van Apeldoorn
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- * E-mail:
| | - Marcel Karperien
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| |
Collapse
|
21
|
Bhat S, Lidgren L, Kumar A. In Vitro Neo-Cartilage Formation on a Three-Dimensional Composite Polymeric Cryogel Matrix. Macromol Biosci 2013; 13:827-37. [DOI: 10.1002/mabi.201200484] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 02/13/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Sumrita Bhat
- Department of Biological Science and Bioengineering; Indian Institute of Technology Kanpur; 208016-Kanpur India
| | - Lars Lidgren
- Department of Orthopaedics; Lund University Hospital; SE-22185 Lund Sweden
| | - Ashok Kumar
- Department of Biological Science and Bioengineering; Indian Institute of Technology Kanpur; 208016-Kanpur India
| |
Collapse
|
22
|
Smith Callahan LA, Ganios AM, Childers EP, Weiner SD, Becker ML. Primary human chondrocyte extracellular matrix formation and phenotype maintenance using RGD-derivatized PEGDM hydrogels possessing a continuous Young's modulus gradient. Acta Biomater 2013; 9:6095-104. [PMID: 23291491 DOI: 10.1016/j.actbio.2012.12.028] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 12/20/2012] [Accepted: 12/21/2012] [Indexed: 11/26/2022]
Abstract
Efficient ex vivo methods for expanding primary human chondrocytes while maintaining the phenotype is critical to advancing the sourcing of autologous cells for tissue engineering applications. While there has been significant research reported in the literature, systematic approaches are necessary to determine and optimize the chemical and mechanical scaffold properties for hyaline cartilage generation using limited cell numbers. Functionalized hydrogels possessing continuous variations in physico-chemical properties are, therefore, an efficient three-dimensional platform for studying several properties simultaneously. Herein we describe a polyethylene glycol dimethacrylate (PEGDM) hydrogel system with a modulus gradient (~27,000-3800 Pa) containing a uniform concentration of arginine-glycine-aspartic acid (RGD) peptide to enhance cell adhesion in order to correlate primary human osteoarthritic chondrocyte proliferation, phenotype maintenance, and extracellular matrix (ECM) production with hydrogel properties. Cell number and chondrogenic phenotype (CD14:CD90 ratios) were found to decline in regions with a higher storage modulus (>13,100 Pa), while regions with a lower storage modulus maintained their cell number and phenotype. Over 3 weeks culture hydrogel regions possessing a lower Young's modulus experienced an increase in ECM content (~200%) compared with regions with a higher storage modulus. Variations in the amount and organization of the cytoskeletal markers actin and vinculin were observed within the modulus gradient, which are indicative of differences in chondrogenic phenotype maintenance and ECM expression. Thus scaffold mechanical properties have a significant impact in modulating human osteoarthritic chondrocyte behavior and tissue formation.
Collapse
|
23
|
Hendriks JAA, Moroni L, Riesle J, de Wijn JR, van Blitterswijk CA. The effect of scaffold-cell entrapment capacity and physico-chemical properties on cartilage regeneration. Biomaterials 2013; 34:4259-65. [PMID: 23489921 DOI: 10.1016/j.biomaterials.2013.02.060] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 02/20/2013] [Indexed: 11/16/2022]
Abstract
An important tenet in designing scaffolds for regenerative medicine consists in mimicking the dynamic mechanical properties of the tissues to be replaced to facilitate patient rehabilitation and restore daily activities. In addition, it is important to determine the contribution of the forming tissue to the mechanical properties of the scaffold during culture to optimize the pore network architecture. Depending on the biomaterial and scaffold fabrication technology, matching the scaffolds mechanical properties to articular cartilage can compromise the porosity, which hampers tissue formation. Here, we show that scaffolds with controlled and interconnected pore volume and matching articular cartilage dynamic mechanical properties, are indeed effective to support tissue regeneration by co-cultured primary and expanded chondrocyte (1:4). Cells were cultured on scaffolds in vitro for 4 weeks. A higher amount of cartilage specific matrix (ECM) was formed on mechanically matching (M) scaffolds after 28 days. A less protein adhesive composition supported chondrocytes rounded morphology, which contributed to cartilaginous differentiation. Interestingly, the dynamic stiffness of matching constructs remained approximately at the same value after culture, suggesting a comparable kinetics of tissue formation and scaffold degradation. Cartilage regeneration in matching scaffolds was confirmed subcutaneously in vivo. These results imply that mechanically matching scaffolds with appropriate physico-chemical properties support chondrocyte differentiation.
Collapse
Affiliation(s)
- J A A Hendriks
- Institute for BioMedical Technology (MIRA), University of Twente, Enschede, The Netherlands.
| | | | | | | | | |
Collapse
|
24
|
Fennema E, Rivron N, Rouwkema J, van Blitterswijk C, de Boer J. Spheroid culture as a tool for creating 3D complex tissues. Trends Biotechnol 2013; 31:108-15. [PMID: 23336996 DOI: 10.1016/j.tibtech.2012.12.003] [Citation(s) in RCA: 717] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 12/10/2012] [Accepted: 12/13/2012] [Indexed: 01/16/2023]
Abstract
3D cell culture methods confer a high degree of clinical and biological relevance to in vitro models. This is specifically the case with the spheroid culture, where a small aggregate of cells grows free of foreign materials. In spheroid cultures, cells secrete the extracellular matrix (ECM) in which they reside, and they can interact with cells from their original microenvironment. The value of spheroid cultures is increasing quickly due to novel microfabricated platforms amenable to high-throughput screening (HTS) and advances in cell culture. Here, we review new possibilities that combine the strengths of spheroid culture with new microenvironment fabrication methods that allow for the creation of large numbers of highly reproducible, complex tissues.
Collapse
Affiliation(s)
- Eelco Fennema
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | |
Collapse
|
25
|
Chen L, Sun J, Zhu Z, Wu K, Li W, Liu H, Xu S. The adhesion and proliferation of bone marrow-derived mesenchymal stem cells promoted by nanoparticle surface. J Biomater Appl 2011; 27:525-36. [PMID: 21862510 DOI: 10.1177/0885328211414750] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This study's aim consists of evaluating the adhesion and proliferation of mesenchymal stem cells (MSCs) derived from rat bone marrow on nanoparticle Titanium (Ti) surface. Hence, passage 3 MSCs were, respectively, seeded on nanoparticle Ti and pure Ti surfaces and then cultured for 32 h. Cell morphology and viability were separately examined by scanning electron microscopy and 3-(4,5-dimethylthiazsol-2-yl)-2,5-diphenyltetrazolium bromide assay. Moreover, the mitotic rate of the attached MSCs was observed through immunocytochemistry. The real-time polymerase chain reaction was applied to determine the adhesion-associated messenger ribonucleic acid (mRNA), CD44 gene encoding variant isoform 6 (CD44 V6), and the integrinβ1 level. The results showed that MSCs performed better in faster extension on the nanoparticle Ti surface than on the pure Ti surface after culturing for 4 h, and were quicker in fusion patterns after 16 h. Furthermore, cell viability was significantly increased on the nanoparticle Ti surface compared to that of the pure Ti surface 16 h after initial seeding (p < 0.05), and the mitotic rate of attached MSCs on the nanoparticle Ti surface was higher than that on the pure Ti surface after 32 h (p < 0.05). More interestingly, the CD44 V6 and integrinβ1 mRNA in the nanoparticle Ti surface group expressed higher than that in the pure Ti surface group after 4 h (p < 0.05), and positive correlation between CD44 V6 and integrinβ1 was found through statistical analysis (correlation coefficient r (s) = 0.98, p < 0.05). Our study's result indicates that a nanoparticle Ti surface can significantly promote the adhesion and proliferation of MSCs, and also improve the bioactivity of Ti surface.
Collapse
Affiliation(s)
- Lian Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
26
|
Griffon DJ, Abulencia JP, Ragetly GR, Fredericks LP, Chaieb S. A comparative study of seeding techniques and three-dimensional matrices for mesenchymal cell attachment. J Tissue Eng Regen Med 2011; 5:169-79. [DOI: 10.1002/term.302] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
27
|
Ragetly G, Griffon DJ, Chung YS. The effect of type II collagen coating of chitosan fibrous scaffolds on mesenchymal stem cell adhesion and chondrogenesis. Acta Biomater 2010; 6:3988-97. [PMID: 20580951 DOI: 10.1016/j.actbio.2010.05.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 05/12/2010] [Accepted: 05/19/2010] [Indexed: 10/19/2022]
Abstract
The biocompatibility of chitosan and its similarity to glycosaminoglycans (GAG) make it attractive for cartilage tissue engineering. We have previously reported improved chondrogenesis but limited cell adhesion on chitosan scaffolds. Our objectives were to produce chitosan scaffolds coated with different densities of type II collagen and to evaluate the effect of this coating on mesenchymal stem cell (MSC) adhesion and chondrogenesis. Chitosan fibrous scaffolds were obtained by a wet spinning method and coated with type II collagen at two different densities. A polyglycolic acid mesh served as a reference group. The scaffolds were characterized by Fourier-transform infrared spectroscopy, scanning electron microscopy (SEM), transmission electron microscopy (TEM) and type II collagen content. Constructs were analyzed after MSCs seeding via live/dead assay, weight and DNA evaluations, SEM, and TEM. Constructs were cultured in chondrogenic medium for 21 days prior to quantitative analysis (weight, DNA, and GAG), SEM, TEM, histology, immunohistochemistry, and quantitative real time polymerase chain reaction. The cell attachment and distribution after seeding correlated with the density of type II collagen. The cell number, the matrix production, and the expression of genes specific for chondrogenesis were improved after culture in collagen coated chitosan constructs. These findings encourage the use of type II collagen for coating chitosan scaffolds to improve MSCs adhesion and chondrogenesis, and confirm the importance of biomimetic scaffolds for tissue engineering.
Collapse
|
28
|
Jukes JM, van der Aa LJ, Hiemstra C, van Veen T, Dijkstra PJ, Zhong Z, Feijen J, van Blitterswijk CA, de Boer J. A newly developed chemically crosslinked dextran-poly(ethylene glycol) hydrogel for cartilage tissue engineering. Tissue Eng Part A 2010; 16:565-73. [PMID: 19737051 DOI: 10.1089/ten.tea.2009.0173] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cartilage tissue engineering, in which chondrogenic cells are combined with a scaffold, is a cell-based approach to regenerate damaged cartilage. Various scaffold materials have been investigated, among which are hydrogels. Previously, we have developed dextran-based hydrogels that form under physiological conditions via a Michael-type addition reaction. Hydrogels can be formed in situ by mixing a thiol-functionalized dextran with a tetra-acrylated star poly(ethylene glycol) solution. In this article we describe how the degradation time of dextran-poly(ethylene glycol) hydrogels can be varied from 3 to 7 weeks by changing the degree of substitution of thiol groups on dextran. The degradation times increased slightly after encapsulation of chondrocytes in the gels. The effect of the gelation reaction on cell viability and cartilage formation in the hydrogels was investigated. Chondrocytes or embryonic stem cells were mixed in the aqueous dextran solution, and we confirmed that the cells survived gelation. After a 3-week culturing period, chondrocytes and embryonic stem cell-derived embryoid bodies were still viable and both cell types produced cartilaginous tissue. Our data demonstrate the potential of dextran hydrogels for cartilage tissue engineering strategies.
Collapse
Affiliation(s)
- Jojanneke M Jukes
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ragetly GR, Griffon DJ, Lee HB, Chung YS. Effect of collagen II coating on mesenchymal stem cell adhesion on chitosan and on reacetylated chitosan fibrous scaffolds. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2010; 21:2479-2490. [PMID: 20499139 DOI: 10.1007/s10856-010-4096-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 05/05/2010] [Indexed: 05/29/2023]
Abstract
The biocompatibility and biomimetic properties of chitosan make it attractive for tissue engineering but its use is limited by its cell adhesion properties. Our objectives were to produce and characterize chitosan and reacetylated-chitosan fibrous scaffolds coated with type II collagen and to evaluate the effect of these chemical modifications on mesenchymal stem cell (MSC) adhesion. Chitosan and reacetylated-chitosan scaffolds obtained by a wet spinning method were coated with type II collagen. Scaffolds were characterized prior to seeding with MSCs. The constructs were analyzed for cell binding kinetics, numbers, distribution and viability. Cell attachment and distribution were improved on chitosan coated with type II collagen. MSCs adhered less to reacetylated-chitosan and collagen coating did not improve MSCs attachment on those scaffolds. These findings are promising and encourage the evaluation of the differentiation of MSCs in collagen-coated chitosan scaffolds. However, the decreased cell adhesion on reacetylated chitosan scaffold seems difficult to overcome and will limit its use for tissue engineering.
Collapse
Affiliation(s)
- Guillaume R Ragetly
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, IL 61802, USA.
| | | | | | | |
Collapse
|
30
|
Schuh E, Kramer J, Rohwedel J, Notbohm H, Müller R, Gutsmann T, Rotter N. Effect of matrix elasticity on the maintenance of the chondrogenic phenotype. Tissue Eng Part A 2010; 16:1281-90. [PMID: 19903088 DOI: 10.1089/ten.tea.2009.0614] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to examine the influence of matrix elasticity on the maintenance of the chondrogenic phenotype of chondrocytes cultured in monolayer. We used a two-dimensional culturing system in which polyacrylamide gels with different concentrations of bis-acrylamide were coated with collagen type I. Matrices with a Young's modulus of 4, 10, 40, and 100 kPa were produced, as determined by atomic force microscopy. Porcine chondrocytes were cultivated on these matrices at a low density for 7 days. The proliferation of cells was analyzed by 5-Bromo-2'-deoxy-uridine incorporation. Maintenance of the chondrogenic phenotype was analyzed by measuring collagen type I, type II, and aggrecan gene expression, immunofluorescence staining for collagen type II, and phalloidin staining for actin filaments. Cellular proliferation and actin organization were decreased on matrices of 4 kPa compared with stiffer substrates. The differentiated phenotype of the chondrocytes grown on matrices of 4 kPa was stabilized, indicated by higher collagen type II and aggrecan, and lower collagen type I expression. These findings indicate that chondrocytes sense the elasticity of the matrix and might be used for the design of scaffolds with mechanical properties specifically tailored to support the chondrogenic phenotype in tissue engineering applications.
Collapse
Affiliation(s)
- Elena Schuh
- Institute of Virology and Cell Biology, University of Lübeck , Lübeck, Germany
| | | | | | | | | | | | | |
Collapse
|
31
|
Ragetly GR, Slavik GJ, Cunningham BT, Schaeffer DJ, Griffon DJ. Cartilage tissue engineering on fibrous chitosan scaffolds produced by a replica molding technique. J Biomed Mater Res A 2010; 93:46-55. [PMID: 19484774 DOI: 10.1002/jbm.a.32514] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The biocompatibility of chitosan and its similarity with glycosaminoglycans make it attractive as a scaffold for cartilage engineering. Fibrous scaffolds may simulate cartilage extracellular matrix structure and promote chondrocyte functions. Our objectives were to produce chitosan fibers of different size and evaluate their potential for chondrogenesis. A novel replica molding technique was developed to produce chitosan nonwoven scaffolds made of fiber measuring 4, 13, or 22 mum in width. A polyglycolic acid mesh (PGA) served as a reference group. Controls were analyzed 48 h after seeding porcine chondrocytes via scanning electron microscopy (SEM), DNA, and glycosaminoglycan (GAG) quantifications. Constructs were cultured for 21 days prior to confocal microscopy, SEM, histology, and quantitative analysis (weight, water, DNA, GAG and collagen II). Chondrocytes maintained their phenotypic appearance and a viability above 85% on the chitosan scaffolds. Chondrocytes attach preferentially to PGA, resulting in a greater cellularity of these constructs. However, based on the GAG/DNA and Collagen II/DNA ratios, matrix production per chondrocyte was improved in chitosan constructs, especially on smaller fibers. The differences between PGA and chitosan are more likely to result from the chemical composition rather than their structural characteristics. Although chitosan appears to promote matrix formation, further studies should be aimed at improving its cell adhesion properties.
Collapse
Affiliation(s)
- Guillaume R Ragetly
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, IL 61802, USA.
| | | | | | | | | |
Collapse
|
32
|
Fernandes H, Dechering K, Van Someren E, Steeghs I, Apotheker M, Leusink A, Bank R, Janeczek K, Van Blitterswijk C, de Boer J. The role of collagen crosslinking in differentiation of human mesenchymal stem cells and MC3T3-E1 cells. Tissue Eng Part A 2010; 15:3857-67. [PMID: 19694522 DOI: 10.1089/ten.tea.2009.0011] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Collagen is the main protein component of the extracellular matrix of bone, and it has structural and instructive properties. Collagen undergoes many post-translational modifications, including extensive crosslinking. Although defective crosslinking has been implicated in human syndromes (e.g., osteogenesis imperfecta or Ehlers-Danlos syndrome), it is not clear to what extent crosslinking is necessary for collagen's instructive properties during bone formation. Here we report that inhibition of collagen crosslinking in the mouse pre-osteoblast cell line MC3T3-E1 impairs the osteogenic program. Genome-wide expression profiling of beta-aminopropionitrile-treated and control cells revealed that matrix deposition by MC3T3-E1 cells provides a feed back signal, driving cells through the differentiation process, that is strongly impaired when crosslinking is inhibited. Inhibition of crosslinking did not affect osteogenic differentiation of human mesenchymal stem cells (hMSCs), shown by the expression of alkaline phosphatase and genome-wide gene expression analysis, although it enhances matrix mineralization. In conclusion, collagen crosslinking harbors instructive properties in MC3T3-E1 differentiation but plays a more-passive role in differentiation of bone marrow-derived hMSCs.
Collapse
Affiliation(s)
- Hugo Fernandes
- Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Functional Biomaterials for Controlling Stem Cell Differentiation. STUDIES IN MECHANOBIOLOGY, TISSUE ENGINEERING AND BIOMATERIALS 2010. [DOI: 10.1007/8415_2010_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Tonnarelli B, Manferdini C, Piacentini A, Codeluppi K, Zini N, Ghisu S, Facchini A, Lisignoli G. Surface-dependent modulation of proliferation, bone matrix molecules, and inflammatory factors in human osteoblasts. J Biomed Mater Res A 2009; 89:687-96. [DOI: 10.1002/jbm.a.32019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
35
|
Jansen EJP, Pieper J, Gijbels MJJ, Guldemond NA, Riesle J, Van Rhijn LW, Bulstra SK, Kuijer R. PEOT/PBT based scaffolds with low mechanical properties improve cartilage repair tissue formation in osteochondral defects. J Biomed Mater Res A 2009; 89:444-52. [PMID: 18431789 DOI: 10.1002/jbm.a.31986] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The aim of our study was to compare the healing response of biomechanically and biochemically different scaffolds in osteochondral defects created in rabbit medial femoral condyles. A block copolymer comprised of poly(ethylene oxide terephthalate) and poly(butylene terephthalate) was used to prepare porous scaffolds. The 70/30 scaffold (70 wt % poly(ethylene oxide terephthalate)) was compared to the stiffer 55/45 (55 wt % poly(ethylene oxide terephthalate)) scaffold. Nine 6-month-old rabbits were used. Osteochondral defects were filled with 55/45 scaffolds (n = 6); 70/30 scaffolds (n = 6); or left empty (n = 6). Defect sites were allowed to heal for 12 weeks. Condyles were macroscopically evaluated and analysed histologically using the O'Driscoll score for evaluating repair of osteochondral defects. Repair tissue in 70/30 scaffolds consisted of cartilage-like tissue on top of trabecular bone, whereas the tissue within the 55/45 scaffolds consisted predominantly of trabecular bone. O'Driscoll scores for 70/30 scaffolds were significantly better (p = 0.024) in comparison to untreated osteochondral defects and 55/45 scaffolds. This study reveals that the biomechanical and biochemical properties of the scaffold play an important role by themselves, and can affect the healing response of osteochondral defects. Scaffolds with low mechanical properties were superior in cartilage repair tissue formation.
Collapse
Affiliation(s)
- Edwin J P Jansen
- Department of Orthopedic Surgery, University Hospital of Maastricht, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kim MH, Kino-oka M, Morinaga Y, Sawada Y, Kawase M, Yagi K, Taya M. Morphological regulation and aggregate formation of rabbit chondrocytes on dendrimer-immobilized surfaces with d-glucose display. J Biosci Bioeng 2009; 107:196-205. [DOI: 10.1016/j.jbiosc.2008.10.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Accepted: 10/07/2008] [Indexed: 10/20/2022]
|
37
|
Seeding density modulates migration and morphology of rabbit chondrocytes cultured in collagen gels. Biotechnol Bioeng 2009; 102:294-302. [DOI: 10.1002/bit.22029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
38
|
Sohier J, Moroni L, van Blitterswijk C, de Groot K, Bezemer JM. Critical factors in the design of growth factor releasing scaffolds for cartilage tissue engineering. Expert Opin Drug Deliv 2008; 5:543-66. [PMID: 18491981 DOI: 10.1517/17425247.5.5.543] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Trauma or degenerative diseases of the joints are common clinical problems resulting in high morbidity. Although various orthopedic treatments have been developed and evaluated, the low repair capacities of articular cartilage renders functional results unsatisfactory in the long term. Over the last decade, a different approach (tissue engineering) has emerged that aims not only to repair impaired cartilage, but also to fully regenerate it, by combining cells, biomaterials mimicking extracellular matrix (scaffolds) and regulatory signals. The latter is of high importance as growth factors have the potency to induce, support or enhance the growth and differentiation of various cell types towards the chondrogenic lineage. Therefore, the controlled release of different growth factors from scaffolds appears to have great potential to orchestrate tissue repair effectively. OBJECTIVE This review aims to highlight considerations and limitations of the design, materials and processing methods available to create scaffolds, in relation to the suitability to incorporate and release growth factors in a safe and defined manner. Furthermore, the current state of the art of signalling molecules release from scaffolds and the impact on cartilage regeneration in vitro and in vivo is reported and critically discussed. METHODS The strict aspects of biomaterials, scaffolds and growth factor release from scaffolds for cartilage tissue engineering applications are considered. CONCLUSION Engineering defined scaffolds that deliver growth factors in a controlled way is a task seldom attained. If growth factor delivery appears to be beneficial overall, the optimal delivery conditions for cartilage reconstruction should be more thoroughly investigated.
Collapse
Affiliation(s)
- J Sohier
- Laboratory for osteo-articular and dental tissue engineering (LIOAD), Faculté de chirurgie dentaire de Nantes, Inserm U791, 1 Place Alexis Ricordeau, 44042 Nantes Cedex 1, France.
| | | | | | | | | |
Collapse
|
39
|
Moroni L, de Wijn JR, van Blitterswijk CA. Integrating novel technologies to fabricate smart scaffolds. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2008; 19:543-72. [PMID: 18419938 DOI: 10.1163/156856208784089571] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tissue engineering aims at restoring or regenerating a damaged tissue by combining cells, derived from a patient biopsy, with a 3D porous matrix functioning as a scaffold. After isolation and eventual in vitro expansion, cells are seeded on the 3D scaffolds and implanted directly or at a later stage in the patient's body. 3D scaffolds need to satisfy a number of requirements: (i) biocompatibility, (ii) biodegradability and/or bioresorbability, (iii) suitable mechanical properties, (iv) adequate physicochemical properties to direct cell-material interactions matching the tissue to be replaced and (v) ease in regaining the original shape of the damaged tissue and the integration with the surrounding environment. Still, it appears to be a challenge to satisfy all the aforementioned requisites with the biomaterials and the scaffold fabrication technologies nowadays available. 3D scaffolds can be fabricated with various techniques, among which rapid prototyping and electrospinning seem to be the most promising. Rapid prototyping technologies allow manufacturing scaffolds with a controlled, completely accessible pore network--determinant for nutrient supply and diffusion--in a CAD/CAM fashion. Electrospinning (ESP) allows mimicking the extracellular matrix (ECM) environment of the cells and can provide fibrous scaffolds with instructive surface properties to direct cell faith into the proper lineage. Yet, these fabrication methods have some disadvantages if considered alone. This review aims at summarizing conventional and novel scaffold fabrication techniques and the biomaterials used for tissue engineering and drug-delivery applications. A new trend seems to emerge in the field of scaffold design where different scaffolds fabrication technologies and different biomaterials are combined to provide cells with mechanical, physicochemical and biological cues at the macro-, micro- and nano-scale. If merged together, these integrated technologies may lead to the generation of a new set of 3D scaffolds that satisfies all of the scaffolds' requirements for tissue-engineering applications and may contribute to their success in a long-term scenario.
Collapse
Affiliation(s)
- L Moroni
- Institute for BioMedical Technology (BMTI), University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands.
| | | | | |
Collapse
|
40
|
Guo L, Kawazoe N, Fan Y, Ito Y, Tanaka J, Tateishi T, Zhang X, Chen G. Chondrogenic differentiation of human mesenchymal stem cells on photoreactive polymer-modified surfaces. Biomaterials 2008; 29:23-32. [DOI: 10.1016/j.biomaterials.2007.08.043] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 08/27/2007] [Indexed: 11/29/2022]
|
41
|
Moroni L, Hendriks JAA, Schotel R, de Wijn JR, van Blitterswijk CA. Design of biphasic polymeric 3-dimensional fiber deposited scaffolds for cartilage tissue engineering applications. ACTA ACUST UNITED AC 2007; 13:361-71. [PMID: 17504063 DOI: 10.1089/ten.2006.0127] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This report describes a novel system to create rapid prototyped 3-dimensional (3D) fibrous scaffolds with a shell-core fiber architecture in which the core polymer supplies the mechanical properties and the shell polymer acts as a coating providing the desired physicochemical surface properties. Poly[(ethylene oxide) terephthalate-co-poly(butylene) terephthalate] (PEOT/PBT) 3D fiber deposited (3DF) scaffolds were fabricated and examined for articular cartilage tissue regeneration. The shell polymer contained a higher molecular weight of the initial poly(ethylene glycol) (PEG) segments used in the copolymerization and a higher weight percentage of the PEOT domains compared with the core polymer. The 3DF scaffolds entirely produced with the shell or with the core polymers were also considered. After 3 weeks of culture, scaffolds were homogeneously filled with cartilage tissue, as assessed by scanning electron microscopy. Although comparable amounts of entrapped chondrocytes and of extracellular matrix formation were found for all analyzed scaffolds, chondrocytes maintained their rounded shape and aggregated during the culture period on shell-core 3DF scaffolds, suggesting a proper cell differentiation into articular cartilage. This finding was also observed in the 3DF scaffolds fabricated with the shell composition only. In contrast, cells spread and attached on scaffolds made simply with the core polymer, implying a lower degree of differentiation into articular cartilaginous tissue. Furthermore, the shell-core scaffolds displayed an improved dynamic stiffness as a result of a "prestress" action of the shell polymer on the core one. In addition, the dynamic stiffness of the constructs increased compared with the stiffness of the bare scaffolds before culture. These findings suggest that shell-core 3DF PEOT/PBT scaffolds with desired mechanical and surface properties are a promising solution for improved cartilage tissue engineering.
Collapse
Affiliation(s)
- L Moroni
- Institute for BioMedical Technology, University of Twente, Enschede, The Netherlands.
| | | | | | | | | |
Collapse
|
42
|
Dadsetan M, Szatkowski JP, Yaszemski MJ, Lu L. Characterization of photo-cross-linked oligo[poly(ethylene glycol) fumarate] hydrogels for cartilage tissue engineering. Biomacromolecules 2007; 8:1702-9. [PMID: 17419584 DOI: 10.1021/bm070052h] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Photo-cross-linkable oligo[poly(ethylene glycol) fumarate] (OPF) hydrogels have been developed for use in tissue engineering applications. We demonstrated that compressive modulus of these hydrogels increased with increasing polymer concentration, and hydrogels with different mechanical properties were formed by altering the ratio of cross-linker/polymer in precursor solution. Conversely, swelling of hydrogels decreased with increasing polymer concentration and cross-linker/polymer ratio. These hydrogels are degradable and degradation rates vary with the change in cross-linking level. Chondrocyte attachment was quantified as a method for evaluating adhesion of cells to the hydrogels. These data revealed that cross-linking density affects cell behavior on the hydrogel surfaces. Cell attachment was greater on the samples with increased cross-linking density. Chondrocytes on these samples exhibited spread morphology with distinct actin stress fibers, whereas they maintained their rounded morphology on the samples with lower cross-linking density. Moreover, chondrocytes were photoencapsulated within various hydrogel networks. Our results revealed that cells encapsulated within 2-mm thick OPF hydrogel disks remained viable throughout the 3-week culture period, with no difference in viability across the thickness of hydrogels. Photoencapsulated chondrocytes expressed the mRNA of type II collagen and produced cartilaginous matrix within the hydrogel constructs after three weeks. These findings suggest that photo-cross-linkable OPF hydrogels may be useful for cartilage tissue engineering and cell delivery applications.
Collapse
Affiliation(s)
- Mahrokh Dadsetan
- Tissue Engineering and Polymeric Biomaterials Laboratory, Department of Orthopedic Surgery, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
43
|
Kino-oka M, Morinaga Y, Kim MH, Takezawa Y, Kawase M, Yagi K, Taya M. Morphological regulation of rabbit chondrocytes on glucose-displayed surface. Biomaterials 2007; 28:1680-8. [PMID: 17182094 DOI: 10.1016/j.biomaterials.2006.11.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Accepted: 11/29/2006] [Indexed: 11/22/2022]
Abstract
A culture surface was designed to regulate morphology of rabbit chondrocytes by changing the ratio of D- and L-glucose isomers displayed on a glass plate. With increasing ratio of d-glucose displayed on the surfaces, the efficiency of cell attachment improved, meaning that the attachment exclusively occurred via mediation of an affinity between D-glucose displayed and glucose transporter on cell membrane. At 0% and 100% D-glucose display, the round-shaped cells appeared dominantly, and most of cells became stretched in shape at 50% d-glucose display, indicating that the frequency of round-shaped cells depicted a concave profile against the ratio of D-glucose displayed. From the cytoskeletal staining of F-actin and vinculin, the immature stress fibers with fewer focal contacts were recognized in both the round shaped cells and those stretched in shape on 100% D-glucose-displayed surface. The time-lapse observation revealed that the cells on 100% D-glucose-displayed surface conducted active migration and aggregation with formation of collagen type II. These results suggest that 100% D-glucose-displayed surface can offer culture environment to maintain the chondrocytic phenotype of cells, similarly to the conditions achieved in three-dimensional (3-D) culture.
Collapse
Affiliation(s)
- Masahiro Kino-oka
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, Osaka 560-8531, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Mahmood TA, Miot S, Frank O, Martin I, Riesle J, Langer R, van Blitterswijk CA. Modulation of chondrocyte phenotype for tissue engineering by designing the biologic-polymer carrier interface. Biomacromolecules 2007; 7:3012-8. [PMID: 17096526 DOI: 10.1021/bm060489+] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Therapeutic strategies based on cell and tissue engineering can be advanced by developing material substrates that effectively interrogate the biological compartment, with or without the complimentary local release of growth factors. Poly(ether ester) segmented copolymers were engineered as model material systems to elucidate the interfacial molecular events that govern the function of adhered cells. Surface chemistry was modulated by varying poly(ethylene glycol) (PEG) length and mole fraction with poly(butylene terephthalate) (PBT), leading to differential competitive protein adsorption of fibronectin and vitronectin from serum and consequently to different cell attachment modes. Adhesion within the hydrogel-like milieu of longer surface PEG was mediated via binding to the CD44 transmembrane receptor, rather than the RGD-integrin mechanism, whereas greater substrate-bound fibronectin resulted in cell adhesion via integrins. These adhesion modalities differentially impacted morphological cell phenotype (spread or spheroid) and the subsequent expression of mRNA transcripts (collagen types II, I) characteristic of phenotypically differentiated or dedifferentiated chondrocytes, respectively. These results demonstrate that materials can be designed to directly elicit the membrane bound receptor apparatus desired for downstream cellular response, without requiring exogenous biological growth factors to enable differentiated potential.
Collapse
Affiliation(s)
- Tahir A Mahmood
- Institute of Biomedical Technology, University of Twente, Prof. Bronkhorstlaan 10, 3723 MB Bilthoven, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
45
|
Mahmood TA, Shastri VP, van Blitterswijk CA, Langer R, Riesle J. Evaluation of chondrogenesis within PEGT: PBT scaffolds with high PEG content. J Biomed Mater Res A 2006; 79:216-22. [PMID: 16886218 DOI: 10.1002/jbm.a.30830] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Porous poly(ethylene glycol) terephthalate:poly (butylene terephthalate) (PEGT:PBT) scaffolds with high PEG molecular weight (1000 g/mole) and PEGT content (60%) were fabricated using two different processes-paraffin templating and compression molding-for cartilage engineering applications. This polymer composition has previously been shown to enable chondrocyte adhesion and maintain differentiated phenotype in 2D monolayer culture. The influence of 3D polymer scaffold processing on the formation of cartilaginous tissue was studied by seeding primary immature bovine chondrocytes within cylindrical scaffolds in mixed flask reactors for 3 days, followed by cultivation in culture plates for a total of 10 or 24 days. Tissue-polymer constructs were evaluated morphologically by SEM and histology, and quantitatively for cellularity, total collagen, and glycosaminoglycan content, all of which remained statistically equivalent for each time point tested, irrespective of fabrication method. These data demonstrate that the polymers engineered for this study were able to support chondrogenesis independent of scaffold fabrication process, with the influence of pore architecture lessened by the highly hydrated scaffold microenvironments induced by high PEG content.
Collapse
Affiliation(s)
- Tahir A Mahmood
- Institute of Biomedical Technology, University of Twente, Professor Bronkhorstlaan 10, Bilthoven 3723 MB, The Netherlands.
| | | | | | | | | |
Collapse
|
46
|
Moroni L, Licht R, de Boer J, de Wijn JR, van Blitterswijk CA. Fiber diameter and texture of electrospun PEOT/PBT scaffolds influence human mesenchymal stem cell proliferation and morphology, and the release of incorporated compounds. Biomaterials 2006; 27:4911-22. [PMID: 16762409 DOI: 10.1016/j.biomaterials.2006.05.027] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2006] [Accepted: 05/16/2006] [Indexed: 01/20/2023]
Abstract
Electrospinning (ESP) has lately shown a great potential as a novel scaffold fabrication technique for tissue engineering. Scaffolds are produced by spinning a polymeric solution in fibers through a spinneret connected to a high-voltage electric field. The fibers are then collected on a support, where the scaffold is created. Scaffolds can be of different shapes, depending on the collector geometry, and have high porosity and high surface per volume ratio, since the deposited fibers vary from the microscale to the nanoscale range. Such fibers are quite effective in terms of tissue regeneration, as cells can bridge the scaffold pores and fibers, resulting in a fast and homogeneous tissue growth. Furthermore, fibers can display a nanoporous ultrastructure due to solvent evaporation. The aim of this study was to characterize electrospun scaffolds from poly(ethylene oxide terephthalate)-poly(butylene terephthalate) (PEOT/PBT) copolymers and to unravel the mechanism of pore formation on the fibers. The effect of different fiber diameters and of their surface nanotopology on cell seeding, attachment, and proliferation was studied. Smooth fibers with diameter of 10microm were found to support an optimal cell seeding and attachment within the micrometer range analyzed. Moreover, a nanoporous surface significantly enhanced cell proliferation and cells spreading on the fibers. The fabrication of ESP scaffolds with incorporated dyes with different molecular dimensions is also reported and their release measured. These findings contribute to the field of cell-material interaction and lead to the fabrication of "smart" scaffolds which can direct cells morphology and proliferation, and eventually release biological signals to properly conduct tissue formation.
Collapse
Affiliation(s)
- Lorenzo Moroni
- Institute for BioMedical Technology (BMTI), University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands.
| | | | | | | | | |
Collapse
|
47
|
Hayman DM, Blumberg TJ, Scott CC, Athanasiou KA. The Effects of Isolation on Chondrocyte Gene Expression. ACTA ACUST UNITED AC 2006; 12:2573-81. [PMID: 16995790 DOI: 10.1089/ten.2006.12.2573] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Tissue engineering of articular cartilage usually requires the isolation and culture of chondrocytes. Previous studies have suggested that enzymatic isolation may alter the metabolic activity and growth rate of chondrocytes. This study examined the effects of 4 common isolation protocols on chondrocyte gene expression, morphology, and total cell yield immediately following the digest (t = 0) and after 2 culture periods (24 h and 1 week). Cartilage explants were digested using 1 of 4 protocols: (1) 6-h collagenase digest, (2) 22-h collagenase digest, (3) 45-min trypsin digest followed by a 3-h collagenase digest, or (4) 1.5-h pronase digest followed by a 3-h collagenase digest. Gene expression levels for glyceraldehyde-3-phosphate dehydrogenase, type I collagen, type II collagen, aggrecan, superficial zone protein, matrix metalloproteinase- 1, and tissue inhibitor of metalloproteinase-1 were measured at t = 0 h, 24 h, and 1 week using quantitative reverse transcriptase-polymerase chain reaction. In this study, cell yield was greatest for the 22-h collagenase and pronase-collagenase digests. However, the data indicate that a 6-h collagenase digest has the fewest gene expression changes compared to native cells. For tissue engineering, data from this study suggest that when cell yield is critical, a 22-h collagenase digest is preferable, but when obtaining cells closest to native chondrocytes is more desired, the 6-h collagenase digest is more beneficial.
Collapse
Affiliation(s)
- Danika M Hayman
- Department of Bioengineering, Rice University, Houston, Texas 77005, USA
| | | | | | | |
Collapse
|
48
|
Woodfield TBF, Miot S, Martin I, van Blitterswijk CA, Riesle J. The regulation of expanded human nasal chondrocyte re-differentiation capacity by substrate composition and gas plasma surface modification. Biomaterials 2005; 27:1043-53. [PMID: 16125219 DOI: 10.1016/j.biomaterials.2005.07.032] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2005] [Accepted: 07/21/2005] [Indexed: 11/18/2022]
Abstract
Optimizing re-differentiation of clinically relevant cell sources on biomaterial substrates in serum containing (S+) and serum-free (SF) media is a key consideration in scaffold-based articular cartilage repair strategies. We investigated whether the adhesion and post-expansion re-differentiation of human chondrocytes could be regulated by controlled changes in substrate surface chemistry and composition in S+ and SF media following gas plasma (GP) treatment. Expanded human nasal chondrocytes were plated on gas plasma treated (GP+) or untreated (GP-) poly(ethylene glycol)-terephthalate-poly(butylene terephthalate) (PEGT/PBT) block co-polymer films with two compositions (low or high PEG content). Total cellularity, cell morphology and immunofluorescent staining of vitronectin (VN) and fibronectin (FN) integrin receptors were evaluated, while post-expansion chondrogenic phenotype was assessed by collagen types I and II mRNA expression. We observed a direct relationship between cellularity, cell morphology and re-differentiation potential. Substrates supporting high cell adhesion and a spread morphology (i.e. GP+ and low PEG content films), resulted in a significantly greater number of cells expressing alpha5beta1 FN to alpha(V)beta3 VN integrin receptors, concomitant with reduced collagen type II/ImRNA gene expression. Substrates supporting low cell adhesion and a spherical morphology (GP- and high PEG content films) promoted chondrocyte re-differentiation indicated by high collagen type II/I gene expression and a low percentage of alpha5beta1 FN integrin expressing cells. This study demonstrates that cell-substrate interactions via alpha5beta1 FN integrin mediated receptors negatively impacts expanded human nasal chondrocyte re-differentiation capacity. GP treatment promotes cell adhesion in S+ media but reverses the ability of low PEG content PEGT/PBT substrates to maintain chondrocyte phenotype. We suggest alternative cell immobilization techniques to GP are necessary for clinical application in articular cartilage repair.
Collapse
Affiliation(s)
- Tim B F Woodfield
- Institute for Biomedical Technology, University of Twente, Bilthoven Research Group, Prof. Bronkhorstlaan 10-D, 3723 MB, Bilthoven, The Netherlands.
| | | | | | | | | |
Collapse
|
49
|
Moroni L, de Wijn JR, van Blitterswijk CA. 3D fiber-deposited scaffolds for tissue engineering: influence of pores geometry and architecture on dynamic mechanical properties. Biomaterials 2005; 27:974-85. [PMID: 16055183 DOI: 10.1016/j.biomaterials.2005.07.023] [Citation(s) in RCA: 303] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2005] [Accepted: 07/11/2005] [Indexed: 11/15/2022]
Abstract
One of the main issues in tissue engineering is the fabrication of scaffolds that closely mimic the biomechanical properties of the tissues to be regenerated. Conventional fabrication techniques are not sufficiently suitable to control scaffold structure to modulate mechanical properties. Within novel scaffold fabrication processes 3D fiber deposition (3DF) showed great potential for tissue engineering applications because of the precision in making reproducible 3D scaffolds, characterized by 100% interconnected pores with different shapes and sizes. Evidently, these features also affect mechanical properties. Therefore, in this study we considered the influence of different structures on dynamic mechanical properties of 3DF scaffolds. Pores were varied in size and shape, by changing fibre diameter, spacing and orientation, and layer thickness. With increasing porosity, dynamic mechanical analysis (DMA) revealed a decrease in elastic properties such as dynamic stiffness and equilibrium modulus, and an increase of the viscous parameters like damping factor and creep unrecovered strain. Furthermore, the Poisson's ratio was measured, and the shear modulus computed from it. Scaffolds showed an adaptable degree of compressibility between sponges and incompressible materials. As comparison, bovine cartilage was tested and its properties fell in the fabricated scaffolds range. This investigation showed that viscoelastic properties of 3DF scaffolds could be modulated to accomplish mechanical requirements for tailored tissue engineered applications.
Collapse
Affiliation(s)
- L Moroni
- Institute for BioMedical Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands.
| | | | | |
Collapse
|
50
|
Moroni L, de Wijn JR, van Blitterswijk CA. Three-dimensional fiber-deposited PEOT/PBT copolymer scaffolds for tissue engineering: Influence of porosity, molecular network mesh size, and swelling in aqueous media on dynamic mechanical properties. J Biomed Mater Res A 2005; 75:957-65. [PMID: 16118789 DOI: 10.1002/jbm.a.30499] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Among novel scaffold fabrication techniques, 3D fiber deposition (3DF) has recently emerged as a means to fabricate well-defined and custom-made scaffolds for tissue regeneration, with 100% interconnected pores. The mechanical behavior of these constructs is dependent not only on different three-dimensional architectural and geometric features, but also on the intrinsic chemical properties of the material used. These affect the mechanics of the solid material and eventually of 3D porous constructs derived from them. For instance, poly(ethylene oxide terephthalate)-poly(butylene terephthalate) (PEOT/PBT) block copolymers are known to have mechanical properties, depending on the PEOT/PBT weight ratio in block form and on the molecular weight of the initial poly(ethylene glycol) (PEG) blocks. These differences are enhanced even more by their different swelling properties in aqueous media. Therefore, this article examines the influence of copolymer compositions in terms of their swelling on dynamic mechanical properties of solid material and porous 3DF scaffolds. The molecular weight of the starting PEG blocks used in the copolymer synthesis varied from 300 to 1000 g/mol. The PEOT/PBT weight ratio in the blocks used varied from 55/45 to 80/20. This corresponded to an increase of the swelling ratio Q from 1.06 to 2.46, and of the mesh size xi from approximately 9 Angstrom to approximately 47 Angstrom. With increased swelling, dynamic mechanical analysis (DMA) revealed a decrease in elastic response and an increase of viscoelasticity. Thus, by coupling structural and chemical characteristics, the viscoelastic properties of PEOT/PBT 3DF scaffolds may be fine tuned to achieve mechanical requirements for a variety of engineered tissues. Ultimately, the combination of 3DF and DMA may be useful to validate the hypothesis that mimicking the biomechanical behavior of a specific tissue for its optimal replacement is an important issue for at least some tissue-engineering applications.
Collapse
Affiliation(s)
- L Moroni
- Institute for BioMedical Technology (BMTI), University of Twente, P. O. Box 217, 7500 AE Enschede, The Netherlands.
| | | | | |
Collapse
|