1
|
Karami P, Laurent A, Philippe V, Applegate LA, Pioletti DP, Martin R. Cartilage Repair: Promise of Adhesive Orthopedic Hydrogels. Int J Mol Sci 2024; 25:9984. [PMID: 39337473 PMCID: PMC11432485 DOI: 10.3390/ijms25189984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Cartilage repair remains a major challenge in human orthopedic medicine, necessitating the application of innovative strategies to overcome existing technical and clinical limitations. Adhesive hydrogels have emerged as promising candidates for cartilage repair promotion and tissue engineering, offering key advantages such as enhanced tissue integration and therapeutic potential. This comprehensive review navigates the landscape of adhesive hydrogels in cartilage repair, discussing identified challenges, shortcomings of current treatment options, and unique advantages of adhesive hydrogel products and scaffolds. While emphasizing the critical need for in situ lateral integration with surrounding tissues, we dissect current limitations and outline future perspectives for hydrogel scaffolds in cartilage repair. Moreover, we examine the clinical translation pathway and regulatory considerations specific to adhesive hydrogels. Overall, this review synthesizes the existing insights and knowledge gaps and highlights directions for future research regarding adhesive hydrogel-based devices in advancing cartilage tissue engineering.
Collapse
Affiliation(s)
- Peyman Karami
- Department of Orthopedic Surgery and Traumatology, University Hospital of Lausanne, CH-1011 Lausanne, Switzerland
- Laboratory of Biomechanical Orthopaedics, Institute of Bioengineering, School of Engineering, EPFL, CH-1015 Lausanne, Switzerland
| | - Alexis Laurent
- Manufacturing Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland
- Regenerative Therapy Unit, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Virginie Philippe
- Department of Orthopedic Surgery and Traumatology, University Hospital of Lausanne, CH-1011 Lausanne, Switzerland
- Regenerative Therapy Unit, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopaedics, Institute of Bioengineering, School of Engineering, EPFL, CH-1015 Lausanne, Switzerland
| | - Robin Martin
- Department of Orthopedic Surgery and Traumatology, University Hospital of Lausanne, CH-1011 Lausanne, Switzerland
| |
Collapse
|
2
|
Pan Y, Li B, Sun X, Tu P, Guo Y, Zhao Z, Wu M, Wang Y, Wang Z, Ma Y. Composite Hydrogel Containing Collagen-Modified Polylactic Acid-Hydroxylactic Acid Copolymer Microspheres Loaded with Tetramethylpyrazine Promotes Articular Cartilage Repair. Macromol Biosci 2024; 24:e2400003. [PMID: 38597147 DOI: 10.1002/mabi.202400003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/22/2024] [Indexed: 04/11/2024]
Abstract
Articular cartilage defects pose a significant challenge due to the limited self-healing ability of cartilage. However, traditional techniques face limitations including autologous chondrocyte expansion issues. This study aims to investigate the effects of the polylactic acid-glycolic acid (PLGA) and collagen-surface modified polylactic acid-glycolic acid (CPLGA) microspheres loaded with tetramethylpyrazine (TMP) on two cell types and the regeneration potential of articular cartilage. CPLGA microspheres are prepared by Steglich reaction and characterized. They evaluated the effect of TMP-loaded microspheres on HUVECs (Human Umbilical Vein Endothelial Cells) and examined the compatibility of blank microspheres with BMSCs (Bone marrow mesenchymal stromal cells) and their potential to promote cartilage differentiation. Subcutaneous implant immune tests and cartilage defect treatment are conducted to assess biocompatibility and cartilage repair potential. The results highlight the efficacy of CPLGA microspheres in promoting tissue regeneration, attributed to improved hydrophilicity and collagen-induced mitigation of degradation. Under hypoxic conditions, both CPLGA and PLGA TMP-loaded microspheres exhibit inhibitory effects on HUVEC proliferation, migration, and angiogenesis. Notably, CPLGA microspheres show enhanced compatibility with BMSCs, facilitating chondrogenic differentiation. Moreover, the CPLGA microsphere-composite hydrogel exhibits potential for cartilage repair by modulating angiogenesis and promoting BMSC differentiation.
Collapse
Affiliation(s)
- Yalan Pan
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Bin Li
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Xiaoxian Sun
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Pengcheng Tu
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Yang Guo
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Zitong Zhao
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Mao Wu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214001, P. R. China
| | - Yun Wang
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Soochow, 215600, P. R. China
| | - Zhifang Wang
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Soochow, 215600, P. R. China
| | - Yong Ma
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214001, P. R. China
| |
Collapse
|
3
|
Hammad M, Veyssiere A, Leclercq S, Patron V, Baugé C, Boumédiene K. Hypoxia Differentially Affects Chondrogenic Differentiation of Progenitor Cells from Different Origins. Int J Stem Cells 2023; 16:304-314. [PMID: 37105555 PMCID: PMC10465331 DOI: 10.15283/ijsc21242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 04/29/2023] Open
Abstract
Background and Objectives Ear cartilage malformations are commonly encountered problems in reconstructive surgery, since cartilage has low self-regenerating capacity. Malformations that impose psychological and social burden on one's life are currently treated using ear prosthesis, synthetic implants or autologous flaps from rib cartilage. These approaches are challenging because not only they request high surgical expertise, but also they lack flexibility and induce severe donor-site morbidity. Through the last decade, tissue engineering gained attention where it aims at regenerating human tissues or organs in order to restore normal functions. This technique consists of three main elements, cells, growth factors, and above all, a scaffold that supports cells and guides their behavior. Several studies have investigated different scaffolds prepared from both synthetic or natural materials and their effects on cellular differentiation and behavior. Methods and Results In this study, we investigated a natural scaffold (alginate) as tridimensional hydrogel seeded with progenitors from different origins such as bone marrow, perichondrium and dental pulp. In contact with the scaffold, these cells remained viable and were able to differentiate into chondrocytes when cultured in vitro. Quantitative and qualitative results show the presence of different chondrogenic markers as well as elastic ones for the purpose of ear cartilage, upon different culture conditions. Conclusions We confirmed that auricular perichondrial cells outperform other cells to produce chondrogenic tissue in normal oxygen levels and we report for the first time the effect of hypoxia on these cells. Our results provide updates for cartilage engineering for future clinical applications.
Collapse
Affiliation(s)
- Mira Hammad
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| | - Alexis Veyssiere
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
- Service de chirurgie Maxillo-faciale, CHU de Caen, Caen, France
| | - Sylvain Leclercq
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Clinique Saint Martin, Service de Chirurgie Orthopédique, Caen, France
| | - Vincent Patron
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Service ORL et chirurgie cervico-faciale, CHU de Caen, Caen, France
| | - Catherine Baugé
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| | - Karim Boumédiene
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| |
Collapse
|
4
|
Shigley C, Trivedi J, Meghani O, Owens BD, Jayasuriya CT. Suppressing Chondrocyte Hypertrophy to Build Better Cartilage. Bioengineering (Basel) 2023; 10:741. [PMID: 37370672 DOI: 10.3390/bioengineering10060741] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Current clinical strategies for restoring cartilage defects do not adequately consider taking the necessary steps to prevent the formation of hypertrophic tissue at injury sites. Chondrocyte hypertrophy inevitably causes both macroscopic and microscopic level changes in cartilage, resulting in adverse long-term outcomes following attempted restoration. Repairing/restoring articular cartilage while minimizing the risk of hypertrophic neo tissue formation represents an unmet clinical challenge. Previous investigations have extensively identified and characterized the biological mechanisms that regulate cartilage hypertrophy with preclinical studies now beginning to leverage this knowledge to help build better cartilage. In this comprehensive article, we will provide a summary of these biological mechanisms and systematically review the most cutting-edge strategies for circumventing this pathological hallmark of osteoarthritis.
Collapse
Affiliation(s)
- Christian Shigley
- The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Jay Trivedi
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Ozair Meghani
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Brett D Owens
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Sports Surgery, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
5
|
Menezes R, Vincent R, Osorno L, Hu P, Arinzeh TL. Biomaterials and tissue engineering approaches using glycosaminoglycans for tissue repair: Lessons learned from the native extracellular matrix. Acta Biomater 2023; 163:210-227. [PMID: 36182056 PMCID: PMC10043054 DOI: 10.1016/j.actbio.2022.09.064] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 01/30/2023]
Abstract
Glycosaminoglycans (GAGs) are an important component of the extracellular matrix as they influence cell behavior and have been sought for tissue regeneration, biomaterials, and drug delivery applications. GAGs are known to interact with growth factors and other bioactive molecules and impact tissue mechanics. This review provides an overview of native GAGs, their structure, and properties, specifically their interaction with proteins, their effect on cell behavior, and their mechanical role in the ECM. GAGs' function in the extracellular environment is still being understood however, promising studies have led to the development of medical devices and therapies. Native GAGs, including hyaluronic acid, chondroitin sulfate, and heparin, have been widely explored in tissue engineering and biomaterial approaches for tissue repair or replacement. This review focuses on orthopaedic and wound healing applications. The use of GAGs in these applications have had significant advances leading to clinical use. Promising studies using GAG mimetics and future directions are also discussed. STATEMENT OF SIGNIFICANCE: Glycosaminoglycans (GAGs) are an important component of the native extracellular matrix and have shown promise in medical devices and therapies. This review emphasizes the structure and properties of native GAGs, their role in the ECM providing biochemical and mechanical cues that influence cell behavior, and their use in tissue regeneration and biomaterial approaches for orthopaedic and wound healing applications.
Collapse
Affiliation(s)
- Roseline Menezes
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Richard Vincent
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Laura Osorno
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Phillip Hu
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Treena Livingston Arinzeh
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States; Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States.
| |
Collapse
|
6
|
Taheri S, Ghazali ZS, Montazeri L, Ebrahim FA, Javadpour J, Kamguyan K, Thormann E, Renaud P, Bonakdar S. Engineered substrates incapable of induction of chondrogenic differentiation compared to the chondrocyte imprinted substrates. Biomed Mater 2023; 18. [PMID: 36693281 DOI: 10.1088/1748-605x/acb5d7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
It is well established that surface topography can affect cell functions. However, finding a reproducible and reliable method for regulating stem cell behavior is still under investigation. It has been shown that cell imprinted substrates contain micro- and nanoscale structures of the cell membrane that serve as hierarchical substrates, can successfully alter stem cell fate. This study investigated the effect of the overall cell shape by fabricating silicon wafers containing pit structure in the average size of spherical-like chondrocytes using photolithography technique. We also used chondrocyte cell line (C28/I2) with spindle-like shape to produce cell imprinted substrates. The effect of all substrates on the differentiation of adipose-derived mesenchymal stem cells (ADSCs) has been studied. The AFM and scanning electron microscopy images of the prepared substrates demonstrated that the desired shapes were successfully transferred to the substrates. Differentiation of ADSCs was investigated by immunostaining for mature chondrocyte marker, collagen II, and gene expression of collagen II, Sox9, and aggrecan markers. C28/I2 imprinted substrate could effectively enhanced chondrogenic differentiation compared to regular pit patterns on the wafer. It can be concluded that cell imprinted substrates can induce differentiation signals better than engineered lithographic substrates. The nanostructures on the cell-imprinted patterns play a crucial role in harnessing cell fate. Therefore, the patterns must include the nano-topographies to have reliable and reproducible engineered substrates.
Collapse
Affiliation(s)
- Shiva Taheri
- National Cell Bank Department, Iran Pasteur Institute, Tehran, Iran.,School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Zahra Sadat Ghazali
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Jafar Javadpour
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Khorshid Kamguyan
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Esben Thormann
- Department of Chemistry, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Philippe Renaud
- STI-IMT-LMIS4, École Polytechnique Fédérale de Lausanne, Station 17, 1015 Lausanne, Switzerland
| | - Shahin Bonakdar
- National Cell Bank Department, Iran Pasteur Institute, Tehran, Iran
| |
Collapse
|
7
|
Hou M, Tian B, Bai B, Ci Z, Liu Y, Zhang Y, Zhou G, Cao Y. Dominant role of in situ native cartilage niche for determining the cartilage type regenerated by BMSCs. Bioact Mater 2022; 13:149-160. [PMID: 35224298 PMCID: PMC8843973 DOI: 10.1016/j.bioactmat.2021.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/27/2022] Open
Affiliation(s)
- Mengjie Hou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
- National Tissue Engineering Center of China, Shanghai, PR China
| | - Baoxing Tian
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, PR China
| | - Baoshuai Bai
- National Tissue Engineering Center of China, Shanghai, PR China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, PR China
| | - Zheng Ci
- National Tissue Engineering Center of China, Shanghai, PR China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, PR China
| | - Yu Liu
- National Tissue Engineering Center of China, Shanghai, PR China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, PR China
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
- National Tissue Engineering Center of China, Shanghai, PR China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, PR China
- Corresponding author. Shanghai Key Lab of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
- National Tissue Engineering Center of China, Shanghai, PR China
- Corresponding author. Shanghai Key Lab of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011, PR China.
| |
Collapse
|
8
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
9
|
Cun X, Hosta-Rigau L. Topography: A Biophysical Approach to Direct the Fate of Mesenchymal Stem Cells in Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2070. [PMID: 33092104 PMCID: PMC7590059 DOI: 10.3390/nano10102070] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022]
Abstract
Tissue engineering is a promising strategy to treat tissue and organ loss or damage caused by injury or disease. During the past two decades, mesenchymal stem cells (MSCs) have attracted a tremendous amount of interest in tissue engineering due to their multipotency and self-renewal ability. MSCs are also the most multipotent stem cells in the human adult body. However, the application of MSCs in tissue engineering is relatively limited because it is difficult to guide their differentiation toward a specific cell lineage by using traditional biochemical factors. Besides biochemical factors, the differentiation of MSCs also influenced by biophysical cues. To this end, much effort has been devoted to directing the cell lineage decisions of MSCs through adjusting the biophysical properties of biomaterials. The surface topography of the biomaterial-based scaffold can modulate the proliferation and differentiation of MSCs. Presently, the development of micro- and nano-fabrication techniques has made it possible to control the surface topography of the scaffold precisely. In this review, we highlight and discuss how the main topographical features (i.e., roughness, patterns, and porosity) are an efficient approach to control the fate of MSCs and the application of topography in tissue engineering.
Collapse
Affiliation(s)
| | - Leticia Hosta-Rigau
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Nils Koppels Allé, Building 423, 2800 Kgs. Lyngby, Denmark;
| |
Collapse
|
10
|
Wu Y, Yang Z, Denslin V, Ren X, Lee CS, Yap FL, Lee EH. Repair of Osteochondral Defects With Predifferentiated Mesenchymal Stem Cells of Distinct Phenotypic Character Derived From a Nanotopographic Platform. Am J Sports Med 2020; 48:1735-1747. [PMID: 32191492 DOI: 10.1177/0363546520907137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Articular cartilage has a zonal architecture and biphasic mechanical properties. The recapitulation of surface lubrication properties with high compressibility of the deeper layers of articular cartilage during regeneration is essential in achieving long-term cartilage integrity. Current clinical approaches for cartilage repair, especially with the use of mesenchymal stem cells (MSCs), have yet to restore the hierarchically organized architecture of articular cartilage. HYPOTHESIS MSCs predifferentiated on surfaces with specific nanotopographic patterns can provide phenotypically stable and defined chondrogenic cells and, when delivered as a bilayered stratified construct at the cartilage defect site, will facilitate the formation of functionally superior cartilage tissue in vivo. STUDY DESIGN Controlled laboratory study. METHODS MSCs were subjected to chondrogenic differentiation on specific nanopatterned surfaces. The phenotype of the differentiated cells was assessed by the expression of cartilage markers. The ability of the 2-dimensional nanopattern-generated chondrogenic cells to retain their phenotypic characteristics after removal from the patterned surface was tested by subjecting the enzymatically harvested cells to 3-dimensional fibrin hydrogel culture. The in vivo efficacy in cartilage repair was demonstrated in an osteochondral rabbit defect model. Repair by bilayered construct with specific nanopattern predifferentiated cells was compared with implantation with cell-free fibrin hydrogel, undifferentiated MSCs, and mixed-phenotype nanopattern predifferentiated MSCs. Cartilage repair was evaluated at 12 weeks after implantation. RESULTS Three weeks of predifferentiation on 2-dimensional nanotopographic patterns was able to generate phenotypically stable chondrogenic cells. Implantation of nanopatterned differentiated MSCs as stratified bilayered hydrogel constructs improved the repair quality of cartilage defects, as indicated by histological scoring, mechanical properties, and polarized microscopy analysis. CONCLUSION Our results indicate that with an appropriate period of differentiation, 2-dimensional nanotopographic patterns can be employed to generate phenotypically stable chondrogenic cells, which, when implanted as stratified bilayered hydrogel constructs, were able to form functionally superior cartilage tissue. CLINICAL RELEVANCE Our approach provides a relatively straightforward method of obtaining large quantities of zone-specific chondrocytes from MSCs to engineer a stratified cartilage construct that could recapitulate the zonal architecture of hyaline cartilage, and it represents a significant improvement in current MSC-based cartilage regeneration.
Collapse
Affiliation(s)
- Yingnan Wu
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zheng Yang
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vinitha Denslin
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - XiaFei Ren
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chang Sheng Lee
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Fung Ling Yap
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Eng Hin Lee
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
11
|
Lee S, Choi JH, Park A, Rim M, Youn J, Lee W, Song JE, Khang G. Advanced gellan gum-based glycol chitosan hydrogel for cartilage tissue engineering biomaterial. Int J Biol Macromol 2020; 158:452-460. [PMID: 32335106 DOI: 10.1016/j.ijbiomac.2020.04.135] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/24/2020] [Accepted: 04/18/2020] [Indexed: 01/01/2023]
Abstract
Gellan gum (GG), a nature-derived polysaccharide, is one of the materials widely used in cartilage tissue engineering (TE). Glycol chitosan (GC), a derivative of chitosan, is a water-soluble natural polymer that has excellent biocompatibility and biodegradability as well as cell adhesion. Herein, GG was physically blended with GC to enhance the mechanical properties and microenvironment of the GG to apply in cartilage TE. The study was conducted with a hydrogel model which is similar to the extracellular matrix (ECM) of cartilage tissue. The physicochemical studies were carried out with morphological study, swelling ratio, weight loss, and sol fraction. The mechanical characterization was conducted with compression test and rheological study to confirm availability in cartilage TE material. Furthermore, in vitro studies such as morphology investigation, viability assay, GAG content, qRT-PCR, and histological study were performed to verify biocompatibility and chondrogenesis of the material. The mechanical and biological properties improved with a proper amount of GC. Overall results verify the potential of the material and can be further used for the cartilage TE.
Collapse
Affiliation(s)
- Sumi Lee
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea.
| | - Joo Hee Choi
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea.
| | - Ain Park
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea.
| | - Mina Rim
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea.
| | - Jina Youn
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea.
| | - Wonchan Lee
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea.
| | - Jeong Eun Song
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea.
| | - Gilson Khang
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, Republic of Korea.
| |
Collapse
|
12
|
Zhan X, Cai P, Lei D, Yang Y, Wang Z, Lu Z, Zheng L, Zhao J. Comparative profiling of chondrogenic differentiation of mesenchymal stem cells (MSCs) driven by two different growth factors. Cell Biochem Funct 2019; 37:359-367. [PMID: 31066473 DOI: 10.1002/cbf.3404] [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: 01/18/2019] [Revised: 03/22/2019] [Accepted: 04/05/2019] [Indexed: 12/14/2022]
Abstract
This study aimed to investigate the mechanism of nerve growth factor (NGF) from cobra venom and human transforming growth factor-β1 (TGF-β1) on the chondrogenic induction of mesenchymal stem cells (MSCs). NGF and TGF-β1 were used to induce chondrogenesis of MSCs from rabbits for 7 days. Total RNA was extracted for mRNA sequencing. Differentially expressed genes (DEGs), gene ontology (GO), KEGG pathway enrichment, and PPI network analysis were conducted to screen the specific signalling pathways and target genes. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to further confirm the relative target genes. The results showed that NGF could significantly promote the expression of hyaline cartilage specific genes (collagen type II alpha 1 chain, COL2A1) compared with TGF-β1. PI3K-AKT signalling pathway is commonly involved in the chondrogenesis of MSCs induced by NGF and TGF-β1. However, the expression levels of the genes in the PI3K-AKT signalling pathway were significantly higher in NGF group than that in the TGF-β1 group. In the process of chondrogenesis of MSCs induced by NGF and TGF-β1, integrin (ITGAs) were the targeted hub genes to activate the PI3K-AKT signalling pathway. NGF could activate more proliferation and differentiation genes in the process of chondrogenesis of MSCs than TGF-β1. TGF-β1 promoted angiogenesis by targeting the thrombospondin (THBS1) and THBS2 which might contribute to the osteophyte formation. PI3K-AKT was the crucial signalling pathway for chondrogenic differentiation. NGF could activate the PI3K-AKT signalling pathway to a higher level, and NGF had more specificity for promoting expression of specific genes of chondrocyte compared with TGF-β1. SIGNIFICANCE OF THE STUDY: In our study, we compared two different growth factors in promoting cartilage differentiation of MSCs and found some similarities and differences. We revealed that both NGF and TGF-β1 could activate the PI3K-AKT signalling pathway (the expression of it in NGF was higher) by targeting the ITGAs in the process of chondrogenesis from MSCs. However, NGF could activate more proliferation and differentiation genes in the process of chondrogenesis of MSCs, whereas TGF-β1 caused osteophyte formation by activating THBS1 and THBS2. These might be the reason why NGF could promote cartilage differentiation more specifically.
Collapse
Affiliation(s)
- Xintang Zhan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Peian Cai
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Danqing Lei
- Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Yifeng Yang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zetao Wang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Pharmaceutical college, Guangxi Medical University, Nanning, China
| | - Zhenhui Lu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
13
|
Lehmann J, Nürnberger S, Narcisi R, Stok KS, van der Eerden BCJ, Koevoet WJLM, Kops N, Ten Berge D, van Osch GJ. Recellularization of auricular cartilage via elastase-generated channels. Biofabrication 2019; 11:035012. [PMID: 30921774 DOI: 10.1088/1758-5090/ab1436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Decellularized tissue matrices are promising substrates for tissue generation by stem cells to replace poorly regenerating tissues such as cartilage. However, the dense matrix of decellularized cartilage impedes colonisation by stem cells. Here, we show that digestion of elastin fibre bundles traversing auricular cartilage creates channels through which cells can migrate into the matrix. Human chondrocytes and bone marrow-derived mesenchymal stromal cells efficiently colonise elastin-treated scaffolds through these channels, restoring a glycosaminoglycan-rich matrix and improving mechanical properties while maintaining size and shape of the restored tissue. The scaffolds are also rapidly colonised by endogenous cartilage-forming cells in a subcutaneously implanted osteochondral biopsy model. Creating channels for cells in tissue matrices may be a broadly applicable strategy for recellularization and restoration of tissue function.
Collapse
Affiliation(s)
- J Lehmann
- Department of Otorhinolaryngology and Head and Neck Surgery Erasmus MC, Rotterdam, The Netherlands. Department of Cell Biology Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
|
15
|
Murine and Chinese cobra venom‑derived nerve growth factor stimulate chondrogenic differentiation of BMSCs in vitro: A comparative study. Mol Med Rep 2018; 18:3341-3349. [PMID: 30066875 PMCID: PMC6102669 DOI: 10.3892/mmr.2018.9307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/22/2018] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based therapy has been commonly used in cartilage reconstruction, due to its self-renewing ability and multi-differentiation potential. Nerve growth factor (NGF) from cobra venom has been reported to regulate chondrogenesis of bone-derived MSCs (BMSCs) and chondrocyte metabolism. Therefore, the present study aimed to determine whether other sources of NGF behave in the same manner as NGF from natural venom. The present study compared the effects of NGF from two sources, the commercially purchased recombinant murine β-NGF (mNGF) and cobra venom-derived NGF (cvNGF), on chondrogenesis of BMSCs by performing hematoxylin and eosin and fluorescein diacetate/propidium iodide staining, DNA and glycosaminoglycan quantization and reverse transcription-quantitative polymerase chain reaction to investigate cell morphology, viability, proliferation, glycosaminoglycan synthesis and cartilage-specific gene expression. The results demonstrated that cvNGF significantly accelerated cell proliferation and upregulated the expression of cartilage-specific genes, including aggrecan, SRY-box 9 and collagen type II α1 chain. Conversely, cvNGF reduced the expression levels of collagen type I α1 chain (a fibrocartilage marker), runt-related transcription factor 2 and enolase 2 compared with in the mNGF and control groups. In addition, Chinese cobra venom, which is the main resource of cvNGF, is abundant and inexpensive, thus greatly decreasing the cost. In conclusion, the present study demonstrated that cvNGF may be considered a potential growth factor for inducing chondrogenic differentiation of BMSCs.
Collapse
|
16
|
Li A, Wei Y, Hung C, Vunjak-Novakovic G. Chondrogenic properties of collagen type XI, a component of cartilage extracellular matrix. Biomaterials 2018; 173:47-57. [PMID: 29758546 DOI: 10.1016/j.biomaterials.2018.05.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/30/2018] [Accepted: 05/03/2018] [Indexed: 12/17/2022]
Abstract
Cartilage extracellular matrix (ECM) has been used for promoting tissue engineering. However, the exact effects of ECM on chondrogenesis and the acting mechanisms are not well understood. In this study, we investigated the chondrogenic effects of cartilage ECM on human mesenchymal stem cells (MSCs) and identified the contributing molecular components. To this end, a preparation of articular cartilage ECM was supplemented to pellets of chondrogenically differentiating MSCs, pellets of human chondrocytes, and bovine articular cartilage explants to evaluate the effects on cell proliferation and the production of cartilaginous matrix. Selective enzymatic digestion and screening of ECM components were conducted to identify matrix molecules with chondrogenic properties. Cartilage ECM promoted MSC proliferation, production of cartilaginous matrix, and maturity of chondrogenic differentiation, and inhibited the hypertrophic differentiation of MSC-derived chondrocytes. Selective digestion of ECM components revealed a contributory role of collagens in promoting chondrogenesis. The screening of various collagen subtypes revealed strong chondrogenic effect of collagen type XI. Finally, collagen XI was found to promote production and inhibit degradation of cartilage matrix in human articular chondrocyte pellets and bovine articular cartilage explants. Our results indicate that cartilage ECM promotes chondrogenesis and inhibits hypertrophic differentiation in MSCs. Collagen type XI is the ECM component that has the strongest effects on enhancing the production and inhibiting the degradation of cartilage matrix.
Collapse
Affiliation(s)
- Ang Li
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yiyong Wei
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Clark Hung
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
17
|
Li D, Zhu L, Liu Y, Yin Z, Liu Y, Liu F, He A, Feng S, Zhang Y, Zhang Z, Zhang W, Liu W, Cao Y, Zhou G. Stable subcutaneous cartilage regeneration of bone marrow stromal cells directed by chondrocyte sheet. Acta Biomater 2017; 54:321-332. [PMID: 28342879 DOI: 10.1016/j.actbio.2017.03.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 02/16/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022]
Abstract
In vivo niche plays an important role in regulating differentiation fate of stem cells. Due to lack of proper chondrogenic niche, stable cartilage regeneration of bone marrow stromal cells (BMSCs) in subcutaneous environments is always a great challenge. This study explored the feasibility that chondrocyte sheet created chondrogenic niche retained chondrogenic phenotype of BMSC engineered cartilage (BEC) in subcutaneous environments. Porcine BMSCs were seeded into biodegradable scaffolds followed by 4weeks of chondrogenic induction in vitro to form BEC, which were wrapped with chondrocyte sheets (Sheet group), acellular small intestinal submucosa (SIS, SIS group), or nothing (Blank group) respectively and then implanted subcutaneously into nude mice to trace the maintenance of chondrogenic phenotype. The results showed that all the constructs in Sheet group displayed typical cartilaginous features with abundant lacunae and cartilage specific matrices deposition. These samples became more mature with prolonged in vivo implantation, and few signs of ossification were observed at all time points except for one sample that had not been wrapped completely. Cell labeling results in Sheet group further revealed that the implanted BEC directly participated in cartilage formation. Samples in both SIS and Blank groups mainly showed ossified tissue at all time points with partial fibrogenesis in a few samples. These results suggested that chondrocyte sheet could create a chondrogenic niche for retaining chondrogenic phenotype of BEC in subcutaneous environment and thus provide a novel research model for stable ectopic cartilage regeneration based on stem cells. STATEMENT OF SIGNIFICANCE In vivo niche plays an important role in directing differentiation fate of stem cells. Due to lack of proper chondrogenic niche, stable cartilage regeneration of bone marrow stromal cells (BMSCs) in subcutaneous environments is always a great challenge. The current study demonstrated that chondrocyte sheet generated by high-density culture of chondrocytes in vitro could cearte a chondrogenic niche in subcutaneous environment and efficiently retain the chondrogenic phenotype of in vitro BMSC engineered cartilage (vitro-BEC). Furthermore, cell tracing results revealed that the regenerated cartilage mainly derived from the implanted vitro-BEC. The current study not only proposes a novel research model for microenvironment simulation but also provides a useful strategy for stable ectopic cartilage regeneration of stem cells.
Collapse
Affiliation(s)
- Dan Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China; National Tissue Engineering Center of China, Shanghai, PR China
| | - Lian Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Yu Liu
- National Tissue Engineering Center of China, Shanghai, PR China
| | - Zongqi Yin
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China; National Tissue Engineering Center of China, Shanghai, PR China
| | - Yi Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, PR China
| | - Fangjun Liu
- Research Institute of Plastic Surgery, Plastic Surgery Hospital, Wei Fang Medical College, Weifang, Shandong, PR China
| | - Aijuan He
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Shaoqing Feng
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Zhiyong Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China; National Tissue Engineering Center of China, Shanghai, PR China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China; National Tissue Engineering Center of China, Shanghai, PR China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China; National Tissue Engineering Center of China, Shanghai, PR China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China; National Tissue Engineering Center of China, Shanghai, PR China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China; National Tissue Engineering Center of China, Shanghai, PR China; Research Institute of Plastic Surgery, Plastic Surgery Hospital, Wei Fang Medical College, Weifang, Shandong, PR China.
| |
Collapse
|
18
|
Moradi L, Vasei M, Dehghan MM, Majidi M, Farzad Mohajeri S, Bonakdar S. Regeneration of meniscus tissue using adipose mesenchymal stem cells-chondrocytes co-culture on a hybrid scaffold: In vivo study. Biomaterials 2017; 126:18-30. [PMID: 28242519 DOI: 10.1016/j.biomaterials.2017.02.022] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 02/07/2017] [Accepted: 02/16/2017] [Indexed: 01/01/2023]
Abstract
The meniscus has poor intrinsic regenerative capacity and its damage inevitably leads to articular cartilage degeneration. We focused on evaluating the effects of Polyvinyl alcohol/Chitosan (PVA/Ch) scaffold seeded by adipose-derived mesenchymal stem cell (ASC) and articular chondrocytes (AC) in meniscus regeneration. The PVA/Ch scaffolds with different molar contents of Ch (Ch1, Ch2, Ch4 and Ch8) were cross-linked by pre-polyurethane chains. By increasing amount of Ch tensile modulus was increased from 83.51 MPa for Ch1 to 110 MPa for Ch8 while toughness showed decrease from 0.33 mJ/mm3 in Ch1 to 0.11 mJ/mm3 in Ch8 constructs. Moreover, swelling ratio and degradation rate increased with an increase in Ch amount. Scanning electron microscopy imaging was performed for pore size measurement and cell attachment. At day 21, Ch4 construct seeded by AC showed the highest expression with 24.3 and 22.64 folds increase in collagen II and aggrecan (p ≤ 0.05), respectively. Since, the mechanical properties, water uptake and degradation rate of Ch4 and Ch8 compositions had no statistically significant differences, Ch4 was selected for in vivo study. New Zealand rabbits were underwent unilateral total medial meniscectomy and AC/scaffold, ASC/scaffold, AC-ASC (co-culture)/scaffold and cell-free scaffold were engrafted. At 7 months post-implantation, macroscopic, histologic, and immunofluorescent studies for regenerated meniscus revealed better results in AC/scaffold group followed by AC-ASC/scaffold and ASC/scaffold groups. In the cell-free scaffold group, there was no obvious meniscus regeneration. Articular cartilages were best preserved in AC/scaffold group. The best histological score was observed in AC/scaffold group. Our results support that Ch4 scaffold seeded by AC alone can successfully regenerate meniscus in tearing injury and ASC has no significant contribution in the healing process.
Collapse
Affiliation(s)
- Lida Moradi
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Vasei
- Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran; Molecular and Cell Biology Laboratory, Department of Pathology, Digestive Disease Research Institute (DDRI), Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Mohammad M Dehghan
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohammad Majidi
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Saeed Farzad Mohajeri
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
19
|
Wu Y, Yang Z, Law JBK, He AY, Abbas AA, Denslin V, Kamarul T, Hui JH, Lee EH. The Combined Effect of Substrate Stiffness and Surface Topography on Chondrogenic Differentiation of Mesenchymal Stem Cells. Tissue Eng Part A 2017; 23:43-54. [DOI: 10.1089/ten.tea.2016.0123] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Yingnan Wu
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - Jaslyn Bee Khuan Law
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Ai Yu He
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Azlina A. Abbas
- Tissue Engineering Group (TEG), National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Pantai Valley, Kuala Lumpur, Malaysia
| | - Vinitha Denslin
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tunku Kamarul
- Tissue Engineering Group (TEG), National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Pantai Valley, Kuala Lumpur, Malaysia
| | - James H.P Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore
| |
Collapse
|
20
|
Cellular Response to Surface Topography and Substrate Stiffness. STEM CELL BIOLOGY AND REGENERATIVE MEDICINE 2017. [DOI: 10.1007/978-3-319-51617-2_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
21
|
Liu J, Yu C, Chen Y, Cai H, Lin H, Sun Y, Liang J, Wang Q, Fan Y, Zhang X. Fast fabrication of stable cartilage-like tissue using collagen hydrogel microsphere culture. J Mater Chem B 2017; 5:9130-9140. [DOI: 10.1039/c7tb02535a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Fabrication of cartilage-like tissue by mimicking chondrogenesis of MSCs in collagen hydrogel microsphere (CHM) culture.
Collapse
Affiliation(s)
- Jun Liu
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Cheng Yu
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yafang Chen
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Hanxu Cai
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Hai Lin
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yong Sun
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Jie Liang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| |
Collapse
|
22
|
Higa K, Kitamura N, Kurokawa T, Goto K, Wada S, Nonoyama T, Kanaya F, Sugahara K, Gong JP, Yasuda K. Fundamental biomaterial properties of tough glycosaminoglycan-containing double network hydrogels newly developed using the molecular stent method. Acta Biomater 2016; 43:38-49. [PMID: 27427226 DOI: 10.1016/j.actbio.2016.07.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/01/2016] [Accepted: 07/13/2016] [Indexed: 12/18/2022]
Abstract
UNLABELLED The purpose of this study was to clarify fundamental mechanical properties and biological responses of the sodium hyaluronate-containing double network (HA-DN) gel and chondroitin sulfate-containing double network (CS-DN) gel, which were newly developed using the molecular stent method. This study discovered the following facts. First, these hydrogels had high mechanical performance comparable to the native cartilage tissue, and the mechanical properties were not affected by immersion in the saline solution for 12weeks. Secondly, the mechanical properties of the CS-DN gel were not significantly reduced at 12weeks in vivo, while the mechanical properties of the HA-DN gel were significantly deteriorated at 6weeks. Thirdly, the degree of inflammation around the HA-DN gel was the same as that around the negative control. The CS-DN gel showed a mild but significant foreign body reaction, which was significantly greater than the negative control and less than the positive control at 1week, while the inflammation was reduced to the same level as the negative control at 4 and 6weeks. Fourthly, these gels induced differentiation of the ATDC5 cells into chondrocytes in the culture with the insulin-free maintenance medium. These findings suggest that these tough hydrogels are potential biomaterials for future application to therapeutic implants such as artificial cartilage. STATEMENT OF SIGNIFICANCE The present study reported fundamental biomaterial properties of the sodium hyaluronate-containing double network (HA-DN) gel and chondroitin sulfate-containing double network (CS-DN) gel, which were newly developed using the molecular stent method. Both the HA- and CS-DN gels had high mechanical properties comparable to the cartilage tissue and showed the ability to induce chondrogenic differentiation of ATDC5 cells in vitro. They are potential biomaterials that may meet the requirements of artificial cartilage concerning the material properties. Further, these DN gels can be also applied to the implantable inducer for cell-free cartilage regeneration therapy.
Collapse
|
23
|
Daley EL, Coleman RM, Stegemann JP. Biomimetic microbeads containing a chondroitin sulfate/chitosan polyelectrolyte complex for cell-based cartilage therapy. J Mater Chem B 2015; 3:7920-7929. [PMID: 26693016 DOI: 10.1039/c5tb00934k] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Articular cartilage has a limited healing capacity that complicates the treatment of joint injuries and osteoarthritis. Newer repair strategies have focused on the use of cells and biomaterials to promote cartilage regeneration. In the present study, we developed and characterized bioinspired materials designed to mimic the composition of the cartilage extracellular matrix. Chondroitin sulfate (CS) and chitosan (CH) were used to form physically cross-linked macromolecular polyelectrolyte complexes (PEC) without the use of additional crosslinkers. A single-step water-in-oil emulsification process was used to either directly embed mesenchymal stem cells (MSC) in PEC particles created with a various concentrations of CS and CH, or to co-embed MSC with PEC in agarose-based microbeads. Direct embedding of MSC in PEC resulted in high cell viability but irregular and large particles. Co-embedding of PEC particles with MSC in agarose (Ag) resulted in uniform microbeads 80-90 μm in diameter that maintained high cell viability over three weeks in culture. Increased serum content resulted in more uniform PEC distribution within the microbead matrix, and both high and low CS:CH ratios resulted in more homogeneous microbeads than 1:1 formulations. Under chondrogenic conditions, expression of sulfated GAG and collagen type II was increased in 10:1 CS:CH PEC-Ag microbeads compared to pure Ag beads, indicating a chondrogenic influence of the PEC component. Such PEC-Ag microbeads may have utility in the directed differentiation and delivery of progenitor cell populations for cartilage repair.
Collapse
Affiliation(s)
- Ethan Lh Daley
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Rhima M Coleman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Zamani M, Prabhakaran MP, Thian ES, Ramakrishna S. Controlled delivery of stromal derived factor-1α from poly lactic-co-glycolic acid core-shell particles to recruit mesenchymal stem cells for cardiac regeneration. J Colloid Interface Sci 2015; 451:144-52. [PMID: 25897850 DOI: 10.1016/j.jcis.2015.04.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 02/08/2023]
Abstract
Stromal derived factor-1α (SDF-1α) has shown promising results in treatment of myocardial infarction (MI), via recruitment of endogenous stem cells into the injured myocardium. However, the bioactivity of this susceptible signalling chemokine is reduced significantly during the common fabrication processes of drug delivery systems, due to the exposure to organic-aqueous interfaces or elevated temperature. In this study, we developed a novel SDF-1α delivery system using coaxial electrospraying, the technique which enables fabrication of core-shell particles with minimized contact of organic-aqueous phases. The SDF-1α incorporated PLGA particles exhibited distinct core-shell structure, confirmed by transmission electron microscopy (TEM). Controlled release of SDF-1α was obtained for at least 40days, and the release rate was tailored by co-encapsulation of bovine serum albumin (BSA) into the core of the particles. The SDF-1α released from PLGA/SDF-1α and PLGA/BSA-SDF-1α particles retained its chemotactic activity, and enhanced the number of migrated mesenchymal stem cells (MSCs) by 38% and 54%, respectively, compared to basal medium used as the control. Moreover, both SDF-1α and BSA supported the proliferation of MSCs within 3days of cell culture. The SDF-1α incorporated core-shell particles developed by electrospraying technique, can be effectively employed as injectable drug delivery system for in situ cardiac regeneration.
Collapse
Affiliation(s)
- Maedeh Zamani
- Department of Mechanical Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore 117576; Center for Nanofibers and Nanotechnology, E3-05-14, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore 117576
| | - Molamma P Prabhakaran
- Center for Nanofibers and Nanotechnology, E3-05-14, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore 117576.
| | - Eng San Thian
- Department of Mechanical Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore 117576
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore 117576; Center for Nanofibers and Nanotechnology, E3-05-14, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore 117576
| |
Collapse
|
25
|
Abstract
Due to a blood supply shortage, articular cartilage has a limited capacity for self-healing once damaged. Articular chondrocytes, cartilage progenitor cells, embryonic stem cells, and mesenchymal stem cells are candidate cells for cartilage regeneration. Significant current attention is paid to improving chondrogenic differentiation capacity; unfortunately, the potential chondrogenic hypertrophy of differentiated cells is largely overlooked. Consequently, the engineered tissue is actually a transient cartilage rather than a permanent one. The development of hypertrophic cartilage ends with the onset of endochondral bone formation which has inferior mechanical properties. In this review, current strategies for inhibition of chondrogenic hypertrophy are comprehensively summarized; the impact of cell source options is discussed; and potential mechanisms underlying these strategies are also categorized. This paper aims to provide guidelines for the prevention of hypertrophy in the regeneration of cartilage tissue. This knowledge may also facilitate the retardation of osteophytes in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Song Chen
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- Department of Joint Surgery, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, China
| | - Peiliang Fu
- Department of Joint Surgery, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, China
| | - Ruijun Cong
- Department of Orthopaedics, The 10th People's Hospital of Shanghai, Affiliated with Tongji University, Shanghai 200072, China
| | - HaiShan Wu
- Department of Joint Surgery, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV 26506, USA
- Corresponding author. Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, PO Box 9196, One Medical Center Drive, Morgantown, WV 26506-9196, USA. Tel.: +1 304 293 1072; fax: +1 304 293 7070.
| |
Collapse
|
26
|
|
27
|
Comparative study of collagen hydrogels modified in two ways using the model of ectopic cartilage construction with diffusion-chamber in immunocompetent host. J Appl Biomater Funct Mater 2014; 12:41-7. [PMID: 22865571 DOI: 10.5301/jabfm.2012.9340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2012] [Indexed: 11/20/2022] Open
Abstract
PURPOSE For scaffolds in cartilage tissue engineering, it is the principle to design the materials with both favorable mechanical and biological property. METHODS In this article, collagen hydrogels modified by two ways to improve mechanical strength were applied for in vivo cartilage reconstruction: one is collagen-alginate hydrogel (CAH) representative of mixture, the other is collagen hydrogel crosslinked by genipin (CGH). To investigate the biological activities of the two materials, it was designed as: scaffolds loaded with allogenous chondrocytes were encased in diffusion chamber, and then implanted subcutaneously in SD rats for 8 weeks. RESULTS Histologic, immunohistochemical, and RT-PCR results showed that collagen type Ⅱ and GAG, indicator of cartilage extracellular matrix (ECM) was highly expressed in constructs of chondrocyte-CAH. Significantly lower cell density and expression of cartilage specific protein were shown in constructs of chondrocyte-CGH than that in chondrocyte-CAH. This demonstrated that CAH may provide a more favorable environment for cartilage reconstruction. In addition, the model with diffusion chamber technique was viable for evaluation of scaffolds in vivo cartilage engineering in immunocompetent host. Instead, directly reconstruction of ectopic cartilage without diffusion chamber suffered from damaged tissue and less neo-cartilage matrix formed. CONCLUSIONS In conclusion, CAH is realistic as scaffold for in vivo cartilage tissue engineering with both satisfactory mechanical properties and biomimetic activity. And the model with diffusion chamber to reconstruct ectopic cartilage in immunocompetent animals is promising for evaluation of scaffolds. This study provided a new insight for in vivo cartilage tissue engineering.
Collapse
|
28
|
Fan H, Zeng X, Wang X, Zhu R, Pei G. Efficacy of prevascularization for segmental bone defect repair using β-tricalcium phosphate scaffold in rhesus monkey. Biomaterials 2014; 35:7407-15. [PMID: 24909103 DOI: 10.1016/j.biomaterials.2014.05.035] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/14/2014] [Indexed: 10/25/2022]
Abstract
Although small animal model (rabbit) showed successful bone defect repair using prevascularized tissue-engineered bone grafts (TEBG), large animal (rhesus monkey) studies are still needed to extrapolate the findings from animal data to humans. In current study, we investigated the efficacy of prevascularized TEBG for segmental bone defect repair in rhesus monkey. The segmental diaphyseal defects were created in both tibias. In group A, the defect was filled with prevascularized MSCs/scaffold prepared by inserting saphenous vascular bundle into the side groove and a fascia flap coverage; In group B, the defect was filled with MSCs/scaffold with a fascia flap coverage; In group C, the defect was filled with MSCs/scaffold; In group D, the defect was filled with only scaffold. The angiogenesis and new bone formation were compared among groups at 4, 8, and 12 weeks postoperatively. The results showed the prevascularized TEBG in group A could augment new bone formation and capillary vessel in-growth. It had significantly higher values of vascularization and radiographic grading score compared with other groups. In conclusion, the in vivo experiment data of prevascularized TEBG was further enriched from small to large animal model. It implies that prevascularized TEBG has great potentials in clinical applications.
Collapse
Affiliation(s)
- Hongbin Fan
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, 17 West Changle Road, Xi'an, China
| | - Xianli Zeng
- Department of Orthopaedics & Traumatology, Nanfang Hospital, Nanfang Medical University, Guangzhou, China
| | - Xueming Wang
- Department of Orthopaedics & Traumatology, Nanfang Hospital, Nanfang Medical University, Guangzhou, China
| | - Rui Zhu
- Collage of Science, Engineering University of Air Force, Xi'an, China
| | - Guoxian Pei
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, 17 West Changle Road, Xi'an, China.
| |
Collapse
|
29
|
Wu YN, Law JBK, He AY, Low HY, Hui JHP, Lim CT, Yang Z, Lee EH. Substrate topography determines the fate of chondrogenesis from human mesenchymal stem cells resulting in specific cartilage phenotype formation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1507-16. [PMID: 24768908 DOI: 10.1016/j.nano.2014.04.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 03/21/2014] [Accepted: 04/05/2014] [Indexed: 12/14/2022]
Abstract
To reproduce a complex and functional tissue, it is crucial to provide a biomimetic cellular microenvironment that not only incorporates biochemical cues, but also physical features including the nano-topographical patterning, for cell/matrix interaction. We developed spatially-controlled nano-topography in the form of nano-pillar, nano-hole and nano-grill on polycaprolactone surface via thermal nanoimprinting. The effects of chondroitin sulfate-coated nano-topographies on cell characteristics and chondrogenic differentiation of human mesenchymal stem cell (MSC) were investigated. Our results show that various nano-topographical patterns triggered changes in MSC morphology and cytoskeletal structure, affecting cell aggregation and differentiation. Compared to non-patterned surface, nano-pillar and nano-hole topography enhanced MSC chondrogenesis and facilitated hyaline cartilage formation. MSCs experienced delayed chondrogenesis on nano-grill topography and were induced to fibro/superficial zone cartilage formation. This study demonstrates the sensitivity of MSC differentiation to surface nano-topography and highlights the importance of incorporating topographical design in scaffolds for cartilage tissue engineering. From the clinical editor: These authors have developed spatially-controlled nano-topography in the form of nano-pillar, nano-hole and nano-grill on polycaprolactone surface via thermal nanoimprinting, and the effects of chondroitin sulfate-coated nano-topographies on cell characteristics and chondrogenic differentiation of human mesenchymal stem cells (MSC) were investigated. It has been concluded that MSC differentiation is sensitive to surface nano-topography, and certain nano-imprinted surfaces are more useful than others for cartilage tissue engineering.
Collapse
Affiliation(s)
- Ying-Nan Wu
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Mechanobiology Institute, National University of Singapore, T-Lab, Singapore
| | - Jaslyn Bee Khuan Law
- Institute of Materials Research Engineering, A*STAR (Agency for Science, Technology and Research), 3 Research Link, Singapore
| | - Ai Yu He
- Institute of Materials Research Engineering, A*STAR (Agency for Science, Technology and Research), 3 Research Link, Singapore
| | - Hong Yee Low
- Institute of Materials Research Engineering, A*STAR (Agency for Science, Technology and Research), 3 Research Link, Singapore; Singapore University of Technology and Design, Engineering Product Development 20, Singapore
| | - James H P Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, T-Lab, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Mechanobiology Institute, National University of Singapore, T-Lab, Singapore; Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.
| |
Collapse
|
30
|
Wise JK, Alford AI, Goldstein SA, Stegemann JP. Comparison of uncultured marrow mononuclear cells and culture-expanded mesenchymal stem cells in 3D collagen-chitosan microbeads for orthopedic tissue engineering. Tissue Eng Part A 2013; 20:210-24. [PMID: 23879621 DOI: 10.1089/ten.tea.2013.0151] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stem cell-based therapies have shown promise in enhancing repair of bone and cartilage. Marrow-derived mesenchymal stem cells (MSC) are typically expanded in vitro to increase cell number, but this process is lengthy, costly, and there is a risk of contamination and altered cellular properties. Potential advantages of using fresh uncultured bone marrow mononuclear cells (BMMC) include heterotypic cell and paracrine interactions between MSC and other marrow-derived cells including hematopoietic, endothelial, and other progenitor cells. In the present study, we compared the osteogenic and chondrogenic potential of freshly isolated BMMC to that of cultured-expanded MSC, when encapsulated in three-dimensional (3D) collagen-chitosan microbeads. The effect of low and high oxygen tension on cell function and differentiation into orthopedic lineages was also examined. Freshly isolated rat BMMC (25 × 10(6) cells/mL, containing an estimated 5 × 10(4) MSC/mL) or purified and culture-expanded rat bone marrow-derived MSC (2 × 10(5) cells/mL) were added to a 65-35 wt% collagen-chitosan hydrogel mixture and fabricated into 3D microbeads by emulsification and thermal gelation. Microbeads were cultured in control MSC growth media in either 20% O2 (normoxia) or 5% O2 (hypoxia) for an initial 3 days, and then in control, osteogenic, or chondrogenic media for an additional 21 days. Microbead preparations were evaluated for viability, total DNA content, calcium deposition, and osteocalcin and sulfated glycosaminoglycan expression, and they were examined histologically. Hypoxia enhanced initial progenitor cell survival in fresh BMMC-microbeads, but it did not enhance osteogenic potential. Fresh uncultured BMMC-microbeads showed a similar degree of osteogenesis as culture-expanded MSC-microbeads, even though they initially contained only 1/10th the number of MSC. Chondrogenic differentiation was not strongly supported in any of the microbead formulations. This study demonstrates the microbead-based approach to culturing and delivering cells for tissue regeneration, and suggests that fresh BMMC may be an alternative to using culture-expanded MSC for bone tissue engineering.
Collapse
Affiliation(s)
- Joel K Wise
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | | | | | | |
Collapse
|
31
|
Bhat A, Hoch AI, Decaris ML, Leach JK. Alginate hydrogels containing cell‐interactive beads for bone formation. FASEB J 2013; 27:4844-52. [DOI: 10.1096/fj.12-213611] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Archana Bhat
- Department of Biomedical EngineeringUniversity of California–DavisDavisCaliforniaUSA
| | - Allison I. Hoch
- Department of Biomedical EngineeringUniversity of California–DavisDavisCaliforniaUSA
| | - Martin L. Decaris
- Department of Biomedical EngineeringUniversity of California–DavisDavisCaliforniaUSA
| | - J. Kent Leach
- Department of Biomedical EngineeringUniversity of California–DavisDavisCaliforniaUSA
- Department of Orthopaedic SurgeryUniversity of California–Davis School of MedicineSacramentoCaliforniaUSA¼
| |
Collapse
|
32
|
Jiang Y, Chen L, Zhang S, Tong T, Zhang W, Liu W, Xu G, Tuan RS, Heng BC, Crawford R, Xiao Y, Ouyang HW. Incorporation of bioactive polyvinylpyrrolidone-iodine within bilayered collagen scaffolds enhances the differentiation and subchondral osteogenesis of mesenchymal stem cells. Acta Biomater 2013; 9:8089-98. [PMID: 23707501 DOI: 10.1016/j.actbio.2013.05.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 04/22/2013] [Accepted: 05/14/2013] [Indexed: 10/26/2022]
Abstract
Polyvinylpyrrolidone-iodine (Povidone-iodine, PVP-I) is widely used as an antiseptic agent for lavation during joint surgery; however, the biological effects of PVP-I on cells from joint tissue are unknown. This study examined the biocompatibility and biological effects of PVP-I on cells from joint tissue, with the aim of optimizing cell-scaffold based joint repair. Cells from joint tissue, including cartilage derived progenitor cells (CPC), subchondral bone derived osteoblast and bone marrow derived mesenchymal stem cells (BM-MSC) were isolated. The concentration-dependent effects of PVP-I on cell proliferation, migration and differentiation were evaluated. Additionally, the efficacy and mechanism of a PVP-I loaded bilayer collagen scaffold for osteochondral defect repair was investigated in a rabbit model. A micromolar concentration of PVP-I was found not to affect cell proliferation, CPC migration or extracellular matrix production. Interestingly, micromolar concentrations of PVP-I promote osteogenic differentiation of BM-MSC, as evidenced by up-regulation of RUNX2 and Osteocalcin gene expression, as well as increased mineralization on the three-dimensional scaffold. PVP-I treatment of collagen scaffolds significantly increased fibronectin binding onto the scaffold surface and collagen type I protein synthesis of cultured BM-MSC. Implantation of PVP-I treated collagen scaffolds into rabbit osteochondral defect significantly enhanced subchondral bone regeneration at 6 weeks post-surgery compared with the scaffold alone (subchondral bone histological score of 8.80±1.64 vs. 3.8±2.19, p<0.05). The biocompatibility and pro-osteogenic activity of PVP-I on the cells from joint tissue and the enhanced subchondral bone formation in PVP-I treated scaffolds would thus indicate the potential of PVP-I for osteochondral defect repair.
Collapse
|
33
|
Wu X, Cai ZD, Lou LM, Chen ZR. The effects of inhibiting hedgehog signaling pathways by using specific antagonist cyclopamine on the chondrogenic differentiation of mesenchymal stem cells. Int J Mol Sci 2013; 14:5966-77. [PMID: 23493060 PMCID: PMC3634433 DOI: 10.3390/ijms14035966] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/05/2013] [Accepted: 03/07/2013] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate the effects of cyclopamine, a specific inhibitor of Hedgehog signaling pathways, on the chondrogenic differentiation of mesenchymal stem cells (MSCs). During culture, the experimental groups were treated with cyclopamine and their cell proliferation status was assessed using the MTT test. The extra-bone cellular matrix (ECM) and Collagen II (Col II) was detected by toluidine blue staining and immunohistochemistry of cells. The concentrations of Col II and aggrecan in the culture solution and cytosol were detected using ELISA on the 7th, 14th, and 21st days of cyclopamine induction. Gene and protein expression of Col II and aggrecan were analyzed on the 14th day of cyclopamine induction using real-time PCR and western blot analyses. No significant differences in proliferation of mesenchymal stem cells were found between the control group and the group treated with cyclopamine. Compared to the blank control group, the ECM level was low and the protein and mRNA concentrations of Collagen II (Col II) and aggrecan in the culture solution and cytosol, respectively, were significantly reduced in the experimental group. The Smo acted as a key point in the regulations of Hedgehog signaling pathway on the chondrogenic differentiation of rabbit MSCs.
Collapse
Affiliation(s)
- Xing Wu
- Department of Orthopaedics, Shanghai tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China; E-Mails: (Z.-D.C.); (L.-M.L.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-21-6630-0588; Fax: +86-21-6630-1051
| | - Zheng-Dong Cai
- Department of Orthopaedics, Shanghai tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China; E-Mails: (Z.-D.C.); (L.-M.L.)
| | - Lei-Ming Lou
- Department of Orthopaedics, Shanghai tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China; E-Mails: (Z.-D.C.); (L.-M.L.)
| | - Zheng-Rong Chen
- Department of Orthopaedics, Shanghai Zhongshan Hospital, Fudan University School of Medicine, Shanghai 200032, China; E-Mail:
| |
Collapse
|
34
|
Chen K, Ng KS, Ravi S, Goh JCH, Toh SL. In vitro generation of whole osteochondral constructs using rabbit bone marrow stromal cells, employing a two-chambered co-culture well design. J Tissue Eng Regen Med 2013; 10:294-304. [PMID: 23495238 DOI: 10.1002/term.1716] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 09/18/2012] [Accepted: 01/05/2013] [Indexed: 12/22/2022]
Abstract
The regeneration of whole osteochondral constructs with a physiological structure has been a significant issue, both clinically and academically. In this study, we present a method using rabbit bone marrow stromal cells (BMSCs) cultured on a silk-RADA peptide scaffold in a specially designed two-chambered co-culture well for the generation of multilayered osteochondral constructs in vitro. This specially designed two-chambered well can simultaneously provide osteogenic and chondrogenic stimulation to cells located in different regions of the scaffold. We demonstrated that this co-culture approach could successfully provide specific chemical stimulation to BMSCs located on different layers within a single scaffold, resulting in the formation of multilayered osteochondral constructs containing cartilage-like and subchondral bone-like tissue, as well as the intermediate osteochondral interface. The cells in the intermediate region were found to be hypertrophic chondrocytes, embedded in a calcified extracellular matrix containing glycosaminoglycans and collagen types I, II and X. In conclusion, this study provides a single-step approach that highlights the feasibility of rabbit BMSCs as a single-cell source for multilayered osteochondral construct generation in vitro.
Collapse
Affiliation(s)
- Kelei Chen
- Department of Bioengineering, National University of Singapore
| | - Kian Siang Ng
- Department of Bioengineering, National University of Singapore
| | - Sujata Ravi
- Department of Bioengineering, National University of Singapore
| | - James C H Goh
- Department of Bioengineering, National University of Singapore.,Department of Orthopaedic Surgery, National University of Singapore
| | - Siew Lok Toh
- Department of Bioengineering, National University of Singapore.,Department of Mechanical Engineering, National University of Singapore
| |
Collapse
|
35
|
Pravdyuk AI, Petrenko YA, Fuller BJ, Petrenko AY. Cryopreservation of alginate encapsulated mesenchymal stromal cells. Cryobiology 2013; 66:215-22. [PMID: 23419981 DOI: 10.1016/j.cryobiol.2013.02.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 12/21/2012] [Accepted: 02/06/2013] [Indexed: 01/28/2023]
Abstract
Human mesenchymal stromal cells (MSCs) can differentiate into various cell types, which makes them attractive for regenerative medicine and tissue engineering. Encapsulation of MSCs in alginate microspheres (AMS) is a novel and promising approach of tissue engineering. Application and research of such cell-hydrogel systems require selection of adequate cryopreservation protocols. In this study we investigated the response of MSCs encapsulated in AMS to different cryopreservation protocols. Bone marrow MSCs either encapsulated in AMS and or as cells in suspension, were cryopreserved with 5% and 10% of dimethyl sulfoxide (ME₂SO) using conventional 2-step slow cooling (protocol 1). The viability and metabolism of MSCs in AMS following cryopreservation with 5% Me₂SO were lower than in the group cryopreserved with 10% Me₂SO. MSCs in suspension were more resistant to cryopreservation than cells in AMS when cryopreserved with 5% Me₂SO, although when using a concentration of 10% Me₂SO, no differences were detected. Comparisons of the viability and metabolic activity of MSC cryopreserved either in AMS or as cell suspensions with 10% ME₂SO using protocol 1 (2-step cooling), protocol 2 (3-step slow cooling with induced ice nucleation) or protocol 3 (rapid 1-step freezing), showed that the highest viabilities and metabolic rates were obtained following cryopreservation of MSCs in AMS by protocol 2 (with controlled ice nucleation). Cryopreservation with protocol 3 resulted in critical damage of the encapsulated MSCs. After cryopreservation by protocol 2, AMS encapsulated MSCs were capable of achieving multilineage differentiation directed towards osteogenic, adipogenic and chondrogenic lineages. The data obtained indicate that cryo-banking of AMS encapsulated MSCs is feasible for future regenerative medicine projects.
Collapse
Affiliation(s)
- Alexey I Pravdyuk
- Institute for Problems of Cryobiology and Cryomedicine of NASU, Kharkov, Ukraine
| | | | | | | |
Collapse
|
36
|
Chen K, Shi P, Teh TKH, Toh SL, Goh JC. In vitro generation of a multilayered osteochondral construct with an osteochondral interface using rabbit bone marrow stromal cells and a silk peptide-based scaffold. J Tissue Eng Regen Med 2013; 10:284-93. [PMID: 23413023 DOI: 10.1002/term.1708] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 11/27/2012] [Accepted: 12/20/2012] [Indexed: 01/25/2023]
Abstract
Tissue engineering of a biological osteochondral multilayered construct with a cartilage-interface subchondral bone layer is a key challenge. This study presented a rabbit bone marrow stromal cell (BMSC)/silk fibroin scaffold-based co-culture approach to generate tissue-engineered osteochondral grafts with an interface. BMSC-seeded scaffolds were first cultured separately in osteogenic and chondrogenic stimulation media. The two differentiated pieces were then combined using an RADA self-assembling peptide and subsequently co-cultured. Gene expression, histological and biochemical analyses were used to evaluate the multilayered structure of the osteochondral graft. A complete osteochondral construct with a cartilage-subchondral bone interface was regenerated and BMSCs were used as the only cell source for the osteochondral construct and interface regeneration. Furthermore, in the intermediate region of co-cultured samples, hypertrophic chondrogenic gene markers type X collagen and MMP-13 were found on both chondrogenic and osteogenic section edges after co-culture. However, significant differences gene expression profile were found in distinct zones of the construct during co-culture and the section in the intermediate region had significantly higher hypertrophic chondrocyte gene expression. Biochemical analyses and histology results further supported this observation. This study showed that specific stimulation from osteogenic and chondrogenic BMSCs affected each other in this co-culture system and induced the formation of an osteochondral interface. Moreover, this system provided a possible approach for generating multilayered osteochondral constructs.
Collapse
Affiliation(s)
- Kelei Chen
- National University of Singapore, Department of Bioengineering, Singapore, Singapore
| | - Pujiang Shi
- National University of Singapore, Department of Orthopaedic Surgery, Singapore, Singapore
| | - Thomas Kok Hiong Teh
- National University of Singapore, Department of Bioengineering, Singapore, Singapore
| | - Siew Lok Toh
- National University of Singapore, Department of Bioengineering, Singapore, Singapore.,National University of Singapore, Department of Mechanical Engineering, Singapore, Singapore
| | - James Ch Goh
- National University of Singapore, Department of Bioengineering, Singapore, Singapore.,National University of Singapore, Department of Orthopaedic Surgery, Singapore, Singapore
| |
Collapse
|
37
|
Solorio LD, Vieregge EL, Dhami CD, Alsberg E. High-density cell systems incorporating polymer microspheres as microenvironmental regulators in engineered cartilage tissues. TISSUE ENGINEERING PART B-REVIEWS 2012; 19:209-20. [PMID: 23126333 DOI: 10.1089/ten.teb.2012.0252] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
To address the significant clinical need for tissue-engineered therapies for the repair and regeneration of articular cartilage, many systems have recently been developed using bioactive polymer microspheres as regulators of the chondrogenic microenvironment within high-density cell cultures. In this review, we highlight various densely cellular systems utilizing polymer microspheres as three-dimensional (3D) structural elements within developing engineered cartilage tissue, carriers for cell expansion and delivery, vehicles for spatiotemporally controlled growth factor delivery, and directors of cell behavior via regulation of cell-biomaterial interactions. The diverse systems described herein represent a shift from the more traditional tissue engineering approach of combining cells and growth factors within a biomaterial scaffold, to the design of modular systems that rely on the assembly of cells and bioactive polymer microspheres as building blocks to guide the creation of articular cartilage. Cell-based assembly of 3D microsphere-incorporated structures represents a promising avenue for the future of tissue engineering.
Collapse
Affiliation(s)
- Loran D Solorio
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
38
|
Hwang JH, Kim OY, Kim AR, Bae JY, Jeong SM, Shim JB, Yoon KH, Lee D, Khang G. EFFECT OF PURIFIED ALGINATE MICROCAPSULES ON THE REGENERATION OF CHONDROCYTES. BIOMEDICAL ENGINEERING-APPLICATIONS BASIS COMMUNICATIONS 2012. [DOI: 10.4015/s1016237212500019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Adult articular cartilage tissue has poor capability of self-repair. Therefore, a variety of tissue engineering approaches are motivated by the clinical need for articular repair. Alginate has been used as a biomaterial for cartilage regeneration. The alginate is a natural polymer that is extracted from seaweeds and purification. However, the main drawback is the immune rejection in vivo. To overcome this problem, we have developed the biocompability of alginate using modified Korbutt method. After alginate was purified, purified alginate microcapsules were used in cartilage regeneration. Chondrocytes were seeded in purified and nonpurified alginate microcapsules, and then cell viability, proliferation and phenotype were analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide (MTT) assay. Reverse transcriptase-polymerase chain reaction (RT-PCR) was conducted to confirm mRNA expression on collagen type I and collagen type II for chondrocytes phenotype. Hematoxylin and eosin (H&E) and Safranin-O histological staining showed tissue growth at the interface during the first 10 days. In this study, chondrocytes in purified alginate microcapsules had higher cell viability, proliferation and more phenotype expression than those in nonpurified alginate microcapsules. The results suggest that the purified alginate microcapsule is useful for cartilage regeneration.
Collapse
Affiliation(s)
- Ji Hye Hwang
- Department of BIN Fusion Technology, Polymer Fusion Research, Center and Department of PolymerNano Science Technology, Chonbuk National University, 567 Baekje-Daero, Jeonju 561-756, Korea
| | - On You Kim
- Department of BIN Fusion Technology, Polymer Fusion Research, Center and Department of PolymerNano Science Technology, Chonbuk National University, 567 Baekje-Daero, Jeonju 561-756, Korea
| | - A Ram Kim
- Department of BIN Fusion Technology, Polymer Fusion Research, Center and Department of PolymerNano Science Technology, Chonbuk National University, 567 Baekje-Daero, Jeonju 561-756, Korea
| | - Ji Yeon Bae
- Department of BIN Fusion Technology, Polymer Fusion Research, Center and Department of PolymerNano Science Technology, Chonbuk National University, 567 Baekje-Daero, Jeonju 561-756, Korea
| | - Su Mi Jeong
- Department of BIN Fusion Technology, Polymer Fusion Research, Center and Department of PolymerNano Science Technology, Chonbuk National University, 567 Baekje-Daero, Jeonju 561-756, Korea
| | - Jung Bo Shim
- Department of BIN Fusion Technology, Polymer Fusion Research, Center and Department of PolymerNano Science Technology, Chonbuk National University, 567 Baekje-Daero, Jeonju 561-756, Korea
| | - Kun Ho Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 505 Banpodong, Seochogu Seoul 137-701, Korea
| | - Dongwon Lee
- Department of BIN Fusion Technology, Polymer Fusion Research, Center and Department of PolymerNano Science Technology, Chonbuk National University, 567 Baekje-Daero, Jeonju 561-756, Korea
| | - Gilson Khang
- Department of BIN Fusion Technology, Polymer Fusion Research, Center and Department of PolymerNano Science Technology, Chonbuk National University, 567 Baekje-Daero, Jeonju 561-756, Korea
| |
Collapse
|
39
|
Fong CY, Subramanian A, Gauthaman K, Venugopal J, Biswas A, Ramakrishna S, Bongso A. Human umbilical cord Wharton's jelly stem cells undergo enhanced chondrogenic differentiation when grown on nanofibrous scaffolds and in a sequential two-stage culture medium environment. Stem Cell Rev Rep 2012; 8:195-209. [PMID: 21671058 DOI: 10.1007/s12015-011-9289-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current treatments used for osteoarthritis from cartilage damage have their disadvantages of donor site morbidity, complicated surgical interventions and risks of infection and graft rejection. Recent advances in tissue engineering have offered much promise in cartilage repair but the best cell source and in vitro system have not as yet been optimised. Human bone marrow mesenchymal stem cells (hBMSCs) have thus far been the cell of choice. However, we derived a unique stem cell from the human umbilical cord Wharton's jelly (hWJSC) that has properties superior to hBMSCs in terms of ready availability, prolonged stemness characteristics in vitro, high proliferation rates, wide multipotency, non-tumorigenicity and tolerance in allogeneic transplantation. We observed enhanced cell attachment, cell proliferation and chondrogenesis of hWJSCs over hBMSCs when grown on PCL/Collagen nanoscaffolds in the presence of a two-stage sequential complex/chondrogenic medium for 21 days. Improvement of these three parameters were confirmed via inverted optics, field emission scanning electron microscopy (FESEM), MTT assay, pellet diameters, Alcian blue histology and staining, glycosaminglycans (GAG) and hyaluronic acid production and expression of key chondrogenic genes (SOX9, Collagen type II, COMP, FMOD) using immunohistochemistry and real-time polymerase chain reaction (qRT-PCR). In separate experiments we demonstrated that the 16 ng/ml of basic fibroblast growth factor (bFGF) present in the complex medium may have contributed to driving chondrogenesis. We conclude that hWJSCs are an attractive stem cell source for inducing chondrogenesis in vitro when grown on nanoscaffolds and exposed sequentially first to complex medium and then followed by chondrogenic medium.
Collapse
Affiliation(s)
- Chui-Yee Fong
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | | | | | | | | | | | | |
Collapse
|
40
|
Xue JX, Gong YY, Zhou GD, Liu W, Cao Y, Zhang WJ. Chondrogenic differentiation of bone marrow-derived mesenchymal stem cells induced by acellular cartilage sheets. Biomaterials 2012; 33:5832-40. [PMID: 22608213 DOI: 10.1016/j.biomaterials.2012.04.054] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 04/23/2012] [Indexed: 10/28/2022]
Abstract
Acellular cartilage sheets (ACSs) have been used as scaffolds for engineering cartilage with mature chondrocytes. In this study we investigated whether ACSs possess a chondrogenic induction activity that may benefit cartilage engineering with multipotent stem cells. Bone marrow-derived mesenchymal stem cells (BMSCs) isolated from newborn pigs were expanded in vitro and seeded on ACSs that were then stacked layer-by-layer to form BMSC-ACS constructs. Cells seeded on polyglycolic acid/polylactic acid (PGA/PLA) scaffolds served as a control. After 4 weeks of culture with or without additional chondrogenic factors, constructs were subcutaneously implanted into nude mice for another 4 weeks. Cartilage-like tissues were formed after 4 weeks of culture. However, formation of cartilage with a typical lacunar structure was only observed in induced groups. RT-PCR showed that aggrecan, COMP, type II collagen and Sox9 were expressed in all groups except the non-induced BMSC-PGA/PLA group. At 4 weeks post-implantation, cartilage formation was achieved in the induced BMSC-ACS group and partial cartilage formation was achieved in the non-induced BMSC-ACS group, confirmed by safranin O staining, toluidine blue staining and type II collagen immunostaining. In addition, enzyme-linked immunosorbent assay demonstrated the presence of transforming growth factor-β1, insulin-like growth factor-1 and bone morphogenic protein-2 in ACSs. These results indicate that ACSs possess a chondrogenic induction activity that promotes BMSC differentiation.
Collapse
Affiliation(s)
- Ji Xin Xue
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 Zhi Zao Ju Road, Shanghai 200011, China
| | | | | | | | | | | |
Collapse
|
41
|
Ge Z, Li C, Heng BC, Cao G, Yang Z. Functional biomaterials for cartilage regeneration. J Biomed Mater Res A 2012; 100:2526-36. [PMID: 22492677 DOI: 10.1002/jbm.a.34147] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 01/30/2012] [Accepted: 02/19/2012] [Indexed: 12/22/2022]
Abstract
The injury and degeneration of articular cartilage and associated arthritis are leading causes of disability worldwide. Cartilage tissue engineering as a treatment modality for cartilage defects has been investigated for over 20 years. Various scaffold materials have been developed for this purpose, but has yet to achieve feasibility and effectiveness for widespread clinical use. Currently, the regeneration of articular cartilage remains a formidable challenge, due to the complex physiology of cartilage tissue and its poor healing capacity. Although intensive research has been focused on the developmental biology and regeneration of cartilage tissue and a diverse plethora of biomaterials have been developed for this purpose, cartilage regeneration is still suboptimal, such as lacking a layered structure, mechanical mismatch with native cartilage and inadequate integration between native tissue and implanted scaffold. The ideal scaffold material should have versatile properties that actively contribute to cartilage regeneration. Functional scaffold materials may overcome the various challenges faced in cartilage tissue engineering by providing essential biological, mechanical, and physical/chemical signaling cues through innovative design. This review thus focuses on the complex structure of native articular cartilage, the critical properties of scaffolds required for cartilage regeneration, present strategies for scaffold design, and future directions for cartilage regeneration with functional scaffold materials.
Collapse
Affiliation(s)
- Zigang Ge
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, People's Republic of China.
| | | | | | | | | |
Collapse
|
42
|
Yang Z, Zou Y, Guo XM, Tan HS, Denslin V, Yeow CH, Ren XF, Liu TM, Hui JH, Lee EH. Temporal activation of β-catenin signaling in the chondrogenic process of mesenchymal stem cells affects the phenotype of the cartilage generated. Stem Cells Dev 2012; 21:1966-76. [PMID: 22133004 DOI: 10.1089/scd.2011.0376] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Adult mesenchymal stem cells (MSCs) are an attractive cell source for cartilage tissue engineering. In vitro predifferentiation of MSCs has been explored as a means to enhance MSC-based articular cartilage repair. However, there remain challenges to control and prevent the premature progression of MSC-derived chondrocytes to the hypertrophy. This study investigated the temporal effect of transforming growth factor (TGF)-β and β-catenin signaling co-activation during MSC chondrogenic differentiation and evaluated the influence of these predifferentiation conditions to subsequent phenotypic development of the cartilage. MSCs were differentiated in chondrogenic medium that contained either TGFβ alone, TGFβ with transient β-catenin coactivation, or TGFβ with continuous β-catenin coactivation. After in vitro differentiation, the pellets were transplanted into SCID mice. Both coactivation protocols resulted in the enhancement of chondrogenic differentiation of MSCs. Compared with TGFβ activation, transient coactivation of TGFβ-induction with β-catenin activation resulted in heightened hypertrophy and formed highly ossified tissues with marrow-like hematopoietic tissue in vivo. The continuous coactivation of the 2 signaling pathways, however, resulted in inhibition of progression to hypertrophy, marked by the suppression of type X collagen, Runx2, and alkaline phosphatase expression, and did not result in ossified tissue in vivo. Chondrocytes of the continuous co-activation samples secreted significantly more parathyroid hormone-related protein (PTHrP) and expressed cyclin D1. Our results suggest that temporal co-activation of the TGFβ signaling pathway with β-catenin can yield cartilage of different phenotype, represents a potential MSC predifferentiation protocol before clinical implantation, and has potential applications for the engineering of cartilage tissue.
Collapse
Affiliation(s)
- Zheng Yang
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yang Z, Wu Y, Li C, Zhang T, Zou Y, Hui JHP, Ge Z, Lee EH. Improved mesenchymal stem cells attachment and in vitro cartilage tissue formation on chitosan-modified poly(L-lactide-co-epsilon-caprolactone) scaffold. Tissue Eng Part A 2011; 18:242-51. [PMID: 21902611 DOI: 10.1089/ten.tea.2011.0315] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Considering the load-bearing physiological requirement of articular cartilage, scaffold for cartilage tissue engineering should exhibit appropriate mechanical responses as natural cartilage undergoing temporary deformation on loading with little structural collapse, and recovering to the original geometry on unloading. A porous elastomeric poly l-lactide-co-ɛ-caprolactone (PLCL) was generated and crosslinked at the surface to chitosan to improve its wettability. Human bone marrow derived mesenchymal stem cells (MSC) attachment, morphological change, proliferation and in vitro cartilage tissue formation on the chitosan-modified PLCL scaffold were compared with the unmodified PLCL scaffold. Chitosan surface promoted more consistent and even distribution of the seeded MSC within the scaffold. MSC rapidly adopted a distinct spread-up morphology on attachment on the chitosan-modified PLCL scaffold with the formation of F-actin stress fiber which proceeded to cell aggregation; an event much delayed in the unmodified PLCL. Enhanced cartilage formation on the chitosan-modified PLCL was shown by real-time PCR analysis, histological and immunochemistry staining and biochemical assays of the cartilage extracellular matrix components. The Young's modulus of the derived cartilage tissues on the chitosan-modified PLCL scaffold was significantly increased and doubled that of the unmodified PLCL. Our results show that chitosan modification of the PLCL scaffold improved the cell compatibility of the PLCL scaffold without significant alteration of the physical elastomeric properties of PLCL and resulted in the formation of cartilage tissue of better quality.
Collapse
Affiliation(s)
- Zheng Yang
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Hellingman CA, Koevoet W, van Osch GJVM. Can one generate stable hyaline cartilage from adult mesenchymal stem cells? A developmental approach. J Tissue Eng Regen Med 2011; 6:e1-e11. [DOI: 10.1002/term.502] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 05/27/2011] [Accepted: 07/13/2011] [Indexed: 01/30/2023]
Affiliation(s)
- Catharine A. Hellingman
- Department of Otorhinolaryngology, Head and Neck Surgery; Erasmus MC, University Medical Centre Rotterdam; PO Box 2040; 3000; CA; Rotterdam; The Netherlands
| | - Wendy Koevoet
- Department of Otorhinolaryngology, Head and Neck Surgery; Erasmus MC, University Medical Centre Rotterdam; PO Box 2040; 3000; CA; Rotterdam; The Netherlands
| | | |
Collapse
|
45
|
Gauthaman K, Fong CY, Suganya CA, Subramanian A, Biswas A, Choolani M, Bongso A. Extra-embryonic human Wharton's jelly stem cells do not induce tumorigenesis, unlike human embryonic stem cells. Reprod Biomed Online 2011; 24:235-46. [PMID: 22196893 DOI: 10.1016/j.rbmo.2011.10.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Revised: 10/12/2011] [Accepted: 10/12/2011] [Indexed: 12/21/2022]
Abstract
Tumorigenesis is the major obstacle of tissues derived from human embryonic stem cells (ESC) and human induced pluripotent stem cell (IPSC) for transplantation therapy. This prompted a search for other sources of ESC. This study isolated and characterized stem cells from the extra-embryonic human umbilical cord Wharton's jelly (WJSC). These cells are non-controversial, available in abundance, proliferative, multipotent and hypoimmunogenic. However, their tumorigenic potential has not been properly addressed. Their tumour-producing capabilities were compared with human ESC using the immunodeficient mouse model. Unlabelled human ESC+matrigel (2×10(6)cells/site), labelled human WJSC (red fluorescent protein; 5×10(6)cells/site) and unlabelled human WJSC+matrigel (5×10(6)cells/site) were injected via three routes (s.c., i.m. and i.p.). Animals that received human ESC+matrigel developed teratomas in 6 weeks (s.c. 85%; i.m. 75%; i.p. 100%) that contained tissues of ectoderm, mesoderm and endoderm. No animal that received human WJSC developed tumours or inflammatory reactions at the injection sites when maintained for a prolonged period (20 weeks). Human WJSC produced increases in anti-inflammatory cytokines in contrast to human ESC, which increased pro-inflammatory cytokines. Human WJSC, being hypoimmunogenic and non-tumorigenic, have the potential for safe cell-based therapies.
Collapse
Affiliation(s)
- Kalamegam Gauthaman
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
46
|
Choi JS, Kim BS, Kim JD, Choi YC, Lee EK, Park K, Lee HY, Cho YW. In vitro expansion of human adipose-derived stem cells in a spinner culture system using human extracellular matrix powders. Cell Tissue Res 2011; 345:415-23. [PMID: 21866312 DOI: 10.1007/s00441-011-1223-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 07/19/2011] [Indexed: 12/13/2022]
Abstract
Stem cell therapy requires large numbers of stem cells to replace damaged tissues, but only limited numbers of stem cells can be harvested from a single patient. To obtain large quantities of stem cells with differentiation potential, we explored a spinner culture system using human extracellular matrix (hECM) powders. The hECM was extracted from adipose tissue and fabricated into powders. Human adipose-derived stem cells (hASCs) were isolated, seeded on hECM powders, and cultivated in a spinner flask. The 3-D culture system, using hECM powders, was highly effective for promoting cell proliferation. The number of hASCs in the 3-D culture system significantly increased for 10 days, resulting in an approximately 10-fold expansion, whereas a traditional 2-D culture system showed just a 2.8-fold expansion. Surface markers, transcriptional factors, and differentiation potential of hASCs were assayed to identify the characteristics of proliferated cells in 3-D culture system. The hASCs expressed the pluripotency markers, Oct-4 and Sox-2 during 3-D culture and retained their capacity to differentiate into adipogenic, osteogenic, and chondrogenic lineages. These findings demonstrate that the 3-D culture systems using hECM powders provide an efficient in vitro environment for stem cell proliferation, and could act as stem cell delivery carriers for autologous tissue engineering and cell therapy.
Collapse
Affiliation(s)
- Ji Suk Choi
- Departments of Chemical Engineering and Bionanotechnology, Hanyang University, Sangnok-gu, Ansan, Gyeonggi-do, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Erisken C, Kalyon DM, Wang H, Örnek-Ballanco C, Xu J. Osteochondral Tissue Formation Through Adipose-Derived Stromal Cell Differentiation on Biomimetic Polycaprolactone Nanofibrous Scaffolds with Graded Insulin and Beta-Glycerophosphate Concentrations. Tissue Eng Part A 2011; 17:1239-52. [DOI: 10.1089/ten.tea.2009.0693] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Cevat Erisken
- Department of Chemical Engineering and Material Science, Stevens Institute of Technology, Hoboken, New Jersey
| | - Dilhan M. Kalyon
- Department of Chemical Engineering and Material Science, Stevens Institute of Technology, Hoboken, New Jersey
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | - Hongjun Wang
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | - Ceren Örnek-Ballanco
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | - Jiahua Xu
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| |
Collapse
|
48
|
Lim JJ, Hammoudi TM, Bratt-Leal AM, Hamilton SK, Kepple KL, Bloodworth NC, McDevitt TC, Temenoff JS. Development of nano- and microscale chondroitin sulfate particles for controlled growth factor delivery. Acta Biomater 2011; 7:986-95. [PMID: 20965281 DOI: 10.1016/j.actbio.2010.10.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 10/13/2010] [Accepted: 10/14/2010] [Indexed: 01/08/2023]
Abstract
Size scale plays an important role in the release properties and cellular presentation of drug delivery vehicles. Because negatively charged chondroitin sulfate (CS) is capable of electrostatically sequestering positively charged growth factors, CS-derived nanoscale micelles and microscale spheroids were synthesized as potential growth factor carriers to enhance differentiation of stem cells. Particles were characterized for morphology, size distribution, surface charge and cytocompatibility, as well as release of transforming growth factor-β1 (TGF-β1) and tumor necrosis factor-α (TNF-α). CS micelles were spherical and negatively charged with a bimodal distribution of 324.1±8.5 and 73.2±4.4 nm diameters, and CS microspheres possessed a rounded morphology and a diameter of 4.3±0.93 μm. Positively charged TGF-β1 demonstrated minimal release after loading in CS microspheres, while negatively charged TNF-α exhibited substantial release over the first 15 h, suggesting that TGF-β1 electrostatically complexed with CS. The micelles and microparticles were found to be cytocompatible at moderate concentrations with marrow stromal cell monolayers and within embryonic stem cell embryoid bodies. These synthesis techniques, which allow the formation of CS-based carriers over a variety of nano- and microscale sizes, offer versatility for tailored release of positively charged growth factors and controlled CS presentation for a variety of stem cell-based applications in tissue engineering and regenerative medicine.
Collapse
|
49
|
Ke Y, Wang YJ, Ren L, Wu G, Xue W. Bioactive surface modification on amide-photografted poly(3-hydroxybutyrate-co-3-hydroxyvalerate). Biomed Mater 2011; 6:025007. [PMID: 21358029 DOI: 10.1088/1748-6041/6/2/025007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Collagen was chemically immobilized on poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) films via hydrophilic polyacrylamide spacers, aiming to establish the bioactive surface and the inner surface models. The inner surface modified films presented higher wettability than the surface modified films. Wide-angle x-ray diffraction results showed that the d-spacing values of the inner surface model increased compared with those of the surface model, but there was no significant difference between the amide- and collagen-modified PHBV films. The peak melting temperatures of PHBV and the special endotherm around 70 °C were following the order: PHBV > amide-modified PHBV > collagen-modified PHBV. The weight loss of the collagen-modified PHBV (inner surface model) might involve hydrolyzation and mineralization during 360 days of incubation, with a maximum value of 18.24%, while PHBV films did not show significant weight loss. The pH value of the degradation fluids fluctuated in the range of 6.86-7.22, as the initial pH was recorded at 7.20. Based on the surface model, collagen-modified PHBV scaffolds were prepared, which enhanced chondrocyte adhesion and spread on the biomimetic surface. Two surface modification models might develop a protocol with a view to generating a biocompatible and biomechanical scaffold for use in meniscus regeneration.
Collapse
Affiliation(s)
- Yu Ke
- Institute of Life and Health Engineering, Jinan University, Guangzhou, People's Republic of China.
| | | | | | | | | |
Collapse
|
50
|
Li C, Wang L, Yang Z, Kim G, Chen H, Ge Z. A viscoelastic chitosan-modified three-dimensional porous poly(L-lactide-co-ε-caprolactone) scaffold for cartilage tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2011; 23:405-24. [PMID: 21310105 DOI: 10.1163/092050610x551970] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Biomaterials have been playing important roles in cartilage regeneration. Although many scaffolds have been reported to enhance cartilage regeneration, none of the scaffolds available are optimal regarding mechanical properties, integration with host cartilage and providing proper micro-environment for chondrocyte attachment, proliferation and differentiation. In the current study, chitosan-modified poly(L-lactide-co-ε-caprolactone) (PLCL) scaffolds were fabricated to simulate the main biochemical components of cartilage, as well as their interaction with the aim to endow them with viscoelasticity similar to native cartilage. Porous PLCL scaffolds were fabricated with porogen-leaching, freeze-extraction and freeze-gelation before chitosan was cross-linked. The acquired porous scaffolds had pore sizes ranging from 200 to 500 μm and about 85% porosity with good interconnection between individual pores. Chitosan was successfully cross-linked to PLCL scaffolds, as validated by ninhydrin staining and X-ray photoelectron spectroscopy (XPS). The viscoelasticity of the scaffolds was similar to that of bovine cartilage and they had a relatively good recovery ratio from compression deformation, while the Young's modulus was one order of magnitude less than cartilage. Not only could the chitosan-modified PLCL scaffolds promote cell adhesion and proliferation, but also they could significantly enhance excretion of aggrecan and type-II collagen, as testified by both histology and quantitative PCR, compared with PLCL scaffolds. With the fabrication of biomimetic scaffolds, it is possible to make scaffolds for cartilage tissue engineering, which are not only biocompatible, but also have mechanical properties similar to native cartilage.
Collapse
Affiliation(s)
- Chao Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, P. R. China
| | | | | | | | | | | |
Collapse
|