51
|
Bahrami M, Valiani A, Amirpour N, Ra Rani MZ, Hashemibeni B. Cartilage Tissue Engineering Via Icariin and Adipose-derived Stem Cells in Fibrin Scaffold. Adv Biomed Res 2018. [PMID: 29531934 PMCID: PMC5840972 DOI: 10.4103/2277-9175.225925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: Nowadays, cartilage tissue engineering is the best candidate for regeneration of cartilage defects. This study evaluates the function of herbal extracts icariin (ICA), the major pharmacological constituent of herba Epimedium, compared with transforming growth factor β3 (TGFβ3) to prove its potential effect for cartilage tissue engineering. Materials and Methods: ICA, TGFβ3, and TGFβ3 + ICA were added fibrin-cell constructions derived from adipose tissue stem cells. After 14 days, cell viability analyzed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H- tetrazolium bromide assay and the expression of cartilage genes was evaluated with real-time polymerase chain reaction (RT-PCR). Results: The results showed ICA, TGFβ3, and TGFβ3 + ICA increased the rate of proliferation and viability of cells; but there were no significant differences between them (P > 0.05). Furthermore, quantitative RT-PCR analysis demonstrated that cooperation of ICA with TGFβ3 showed a better effect in expression of cartilaginous specific genes and increased Sox9, type II collagen, and aggrecan expression significantly. Furthermore, the results of the expression of type I and X collagens revealed that TGFβ3 increased the expression of them (P < 0.01); However, treatment with ICA + TGFβ3 down regulated the expression of these genes significantly. Conclusion: The results indicated ICA could be a potential factor for chondrogenesis and in cooperation with TGFβ3 could reduce its hypertrophic effects and it is a promising factor for cartilage tissue engineering.
Collapse
Affiliation(s)
- Maryam Bahrami
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Valiani
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Noushin Amirpour
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Zamani Ra Rani
- Department of Anatomical Sciences, Faculty of Medicine, Hormozgan University of Medical Sciences, Hormozgan, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Torabinejad Dental Research Center, Dental School, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
52
|
Wang S, Bao Y, Guan Y, Zhang C, Liu H, Yang X, Gao L, Guo T, Chen Q. Strain distribution of repaired articular cartilage defects by tissue engineering under compression loading. J Orthop Surg Res 2018; 13:19. [PMID: 29382342 PMCID: PMC5791196 DOI: 10.1186/s13018-018-0726-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/19/2018] [Indexed: 01/07/2023] Open
Abstract
Background It is difficult to repair cartilage damage when cartilage undergoes trauma or degeneration. Cartilage tissue engineering is an ideal treatment method to repair cartilage defects, but at present, there are still some uncertainties to be researched in cartilage tissue engineering including the mechanical properties of the repaired region. Methods In this study, using an agarose gel as artificial cartilage implanted into the cartilage defect and gluing the agarose gel to cartilage by using the medical bio-adhesive, the full-thickness and half-thickness defects models of articular cartilage in vitro repaired by tissue engineering were constructed. Strain behaviors of the repaired region were analyzed by the digital correlation technology under 5, 10, 15, and 20% compressive load. Results The axial normal strain (Ex) perpendicular to the surface of the cartilage and lateral normal strain (Ey) as well as shear strain (Exy) appeared obviously heterogeneous in the repaired region. In the full-defect model, Ex showed depth-dependent strain profiles where maximum Ex occurs at the low middle zone while in the half-defect mode, Ex showed heterogeneous strain profiles where maximum Ex occurs at the near deep zone. Ey and Exy at the interface site of both models present significantly differed from the host cartilage site. Ey and Exy exhibited region-specific change at the host, interface, and artificial cartilage sites in the superficial, middle, and deep zones due to the artificial cartilage implantation. Conclusion Both defect models of cartilage exhibited a heterogeneous strain field due to the engineered cartilage tissue implant. The abnormal strain field can cause the cells within the repaired area to enter complex mechanical states which will affect the restoration of cartilage defects.
Collapse
Affiliation(s)
- Shilei Wang
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin, 300384, China
| | - Yan Bao
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin, 300384, China
| | - Yinjie Guan
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, 1 Hoppin St., Ste. 402, Providence, RI, 02903, USA
| | - Chunqiu Zhang
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin, 300384, China. .,Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, 1 Hoppin St., Ste. 402, Providence, RI, 02903, USA.
| | - Haiying Liu
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin, 300384, China
| | - Xu Yang
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, 1 Hoppin St., Ste. 402, Providence, RI, 02903, USA
| | - Lilan Gao
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin, 300384, China
| | - Tongtong Guo
- Nature Science Department, Harbin Institute of Technology, Shenzhen Campus, Shenzhen, 518055, China.
| | - Qian Chen
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, 1 Hoppin St., Ste. 402, Providence, RI, 02903, USA
| |
Collapse
|
53
|
Shen H, Lin H, Sun AX, Song S, Zhang Z, Dai J, Tuan RS. Chondroinductive factor-free chondrogenic differentiation of human mesenchymal stem cells in graphene oxide-incorporated hydrogels. J Mater Chem B 2018; 6:908-917. [PMID: 32254371 DOI: 10.1039/c7tb02172k] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Graphene-based nanomaterials have been applied as biomaterials to enhance stem cell adhesion, growth and differentiation by serving as nanocarriers for growth factors or other small molecules. However, the direct effect of graphene oxide (GO) itself on stem cells, in the absence of exogenous differentiation inductive factors, has not been tested. In this study, we loaded GO nanosheets and human bone marrow-derived mesenchymal stem cells (hBMSC) into a photopolymerizable poly-d,l-lactic acid/polyethylene glycol (PDLLA) hydrogel, a robust chondrosupportive scaffold recently developed in our laboratory, and assessed hBMSC differentiation along the chondrogenic lineage without supplemental chondroinductive factors. We first examined the effect of GO incorporation on the mechanical properties of constructs, and observed that the GO-containing constructs (GO/PDLLA) exhibited enhanced compressive modulus in a GO concentration dependent manner. hBMSCs cultured in GO/PDLLA maintained high cell viability (>95%), indicating minimal cytotoxicity of GO. Importantly, compared to those encapsulated in PDLLA hydrogel, hBMSCs within GO/PDLLA showed significantly higher level of gene expression of the cartilage matrix genes, aggrecan and collagen type II, and produced more cartilage matrix. In addition, the pro-chondrogenesis effect of GO increased with increasing GO concentration. Immunohistochemical results suggested that GO-enhanced hBMSC chondrogenesis was correlated with enriched sequestration of insulin, a necessary supplement known to have pro-chondrogenesis effects on hBMSC. Taken together, these findings demonstrate the utility of using GO to improve the mechanical properties and chondrogenic differentiation state of MSC-laden, engineered hydrogel constructs, without the use of exogenous growth factors, thus representing a potentially promising, biologics-free approach for cartilage tissue engineering.
Collapse
Affiliation(s)
- He Shen
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| | | | | | | | | | | | | |
Collapse
|
54
|
Duarte ARC, Santo VE, Gomes ME, Reis RL. Supercritical Fluid Technology as a Tool to Prepare Gradient Multifunctional Architectures Towards Regeneration of Osteochondral Injuries. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1058:265-278. [PMID: 29691826 DOI: 10.1007/978-3-319-76711-6_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Platelet lysates (PLs) are a natural source of growth factors (GFs) known for its stimulatory role on stem cells which can be obtained after activation of platelets from blood plasma. The possibility to use PLs as growth factor source for tissue healing and regeneration has been pursued following different strategies. Platelet lysates are an enriched pool of growth factors which can be used as either a GFs source or as a three-dimensional (3D) hydrogel. However, most of current PLs-based hydrogels lack stability, exhibiting significant shrinking behavior. This chapter focuses on the application of supercritical fluid technology to develop three-dimensional architectures of PL constructs, crosslinked with genipin. The proposed technology allows in a single step operation the development of mechanically stable porous structures, through chemical crosslinking of the growth factors present in the PL pool, followed by supercritical drying of the samples. Furthermore gradient structures of PL-based structures with bioactive glass are also presented and are described as an interesting approach to the treatment of osteochondral defects.
Collapse
Affiliation(s)
- Ana Rita C Duarte
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco/Guimarães, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Vitor E Santo
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco/Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Manuela E Gomes
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco/Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco/Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
55
|
Pot MW, de Kroon LMG, van der Kraan PM, van Kuppevelt TH, Daamen WF. Unidirectional BMP2-loaded collagen scaffolds induce chondrogenic differentiation. ACTA ACUST UNITED AC 2017; 13:015007. [PMID: 29165318 DOI: 10.1088/1748-605x/aa8960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Microfracture surgery may be improved by the implantation of unidirectional collagen scaffolds that provide a template for mesenchymal stem cells to regenerate cartilage. Incorporation of growth factors in unidirectional scaffolds may further enhance cartilage regeneration. In scaffolds, immobilization of growth factors is required to prolong in vivo activity, to limit diffusion and to reduce the amount of growth factor needed for safe clinical application. We investigated the immobilization of bone morphogenetic protein 2 (BMP2) to unidirectional collagen scaffolds and the effect on in vitro chondrogenesis. C3H10T1/2 cells were seeded on unidirectional collagen scaffolds with and without covalently attached heparin, and with and without incubation with BMP2 (1 and 10 μg), or with BMP2 present in the culture medium (10-200 ng ml-1). Culturing was for 2 weeks and readout parameters included histology, immunohistochemistry, biochemical analysis and molecular biological analysis. The unidirectional pores facilitated the distribution of C3H10T1/2 cells and matrix formation throughout scaffolds. The effective dose of medium supplementation with BMP2 was 100 ng ml-1 (total exposure 1 μg BMP2), and similar production of cartilage-specific molecules chondroitin sulfate (CS) and type II collagen was found for scaffolds pre-incubated with 10 μg BMP2. Pre-incubation with 1 μg BMP2 resulted in less cartilage matrix formation. The conjugation of heparin to the scaffolds resulted in more CS and less type II collagen deposition compared to scaffolds without heparin. In conclusion, unidirectional collagen scaffolds pre-incubated with 10 μg BMP2 supported chondrogenesis in vitro and may be suitable for prolonged cartilage matrix synthesis in vivo.
Collapse
Affiliation(s)
- Michiel W Pot
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
56
|
Coculture of allogenic DBM and BMSCs in the knee joint cavity of rabbits for cartilage tissue engineering. Biosci Rep 2017; 37:BSR20170804. [PMID: 28931727 PMCID: PMC5968190 DOI: 10.1042/bsr20170804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/03/2017] [Accepted: 09/15/2017] [Indexed: 01/02/2023] Open
Abstract
The present study aims to assess coculture of allogenic decalcified bone matrix (DBM) and bone marrow mesenchymal stem cells (BMSCs) in the knee joint cavity of rabbits for cartilage tissue engineering. Rabbits were assigned to an in vitro group, an in vivo group, and a blank control group. At the 4th, 8th, and 12th week, samples from all groups were collected for hematoxylin–eosin (HE) staining and streptavidin–peroxidase (SP) method. The morphological analysis software was used to calculate the average absorbance value (A value). SP and flow cytometry demonstrated that BMSCs were induced into chondrocytes. DBM scaffold showed honeycomb-shaped porous and three-dimensional structure, while the surface pores are interlinked with the deep pores. At the 4th week, in the blank control group, DBM scaffold structure was clear, and cells analogous to chondrocytes were scattered in the interior of DBM scaffolds. At the 8th week, in the in vivo group, there were a large amount of cells, mainly mature chondrocytes, and the DBM scaffolds were partially absorbed. At the 12th week, in the in vitro group, the interior of scaffolds was filled up with chondrocytes with partial fibrosis, but arranged in disorder. In the in vivo group, the chondrocytes completely infiltrated into the interior of scaffolds and were arranged in certain stress direction. The in vivo group showed higher A value than the in vitro and blank control groups at each time point. Allogenic DBM combined BMSCs in the knee joint cavity of rabbits could provide better tissue-engineered cartilage than that cultivated in vitro.
Collapse
|
57
|
do Amaral RJFC, Almeida HV, Kelly DJ, O'Brien FJ, Kearney CJ. Infrapatellar Fat Pad Stem Cells: From Developmental Biology to Cell Therapy. Stem Cells Int 2017; 2017:6843727. [PMID: 29018484 PMCID: PMC5606137 DOI: 10.1155/2017/6843727] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/03/2017] [Indexed: 12/20/2022] Open
Abstract
The ideal cell type to be used for cartilage therapy should possess a proven chondrogenic capacity, not cause donor-site morbidity, and should be readily expandable in culture without losing their phenotype. There are several cell sources being investigated to promote cartilage regeneration: mature articular chondrocytes, chondrocyte progenitors, and various stem cells. Most recently, stem cells isolated from joint tissue, such as chondrogenic stem/progenitors from cartilage itself, synovial fluid, synovial membrane, and infrapatellar fat pad (IFP) have gained great attention due to their increased chondrogenic capacity over the bone marrow and subcutaneous adipose-derived stem cells. In this review, we first describe the IFP anatomy and compare and contrast it with other adipose tissues, with a particular focus on the embryological and developmental aspects of the tissue. We then discuss the recent advances in IFP stem cells for regenerative medicine. We compare their properties with other stem cell types and discuss an ontogeny relationship with other joint cells and their role on in vivo cartilage repair. We conclude with a perspective for future clinical trials using IFP stem cells.
Collapse
Affiliation(s)
- Ronaldo J. F. C. do Amaral
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Henrique V. Almeida
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Cathal J. Kearney
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin & Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
58
|
Mazor M, Cesaro A, Ali M, Best TM, Lespessaille E, Toumi H. Progenitor Cells from Cartilage: Grade Specific Differences in Stem Cell Marker Expression. Int J Mol Sci 2017; 18:ijms18081759. [PMID: 28805694 PMCID: PMC5578148 DOI: 10.3390/ijms18081759] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 01/09/2023] Open
Abstract
Recent research has confirmed the presence of Mesenchymal stem cell (MSC)-like progenitors (MPC) in both normal and osteoarthritic cartilage. However, there is only limited information concerning how MPC markers are expressed with osteoarthritis (OA) progression. The purpose of this study was to compare the prevalence of various MPC markers in different OA grades. Human osteoarthritic tibial plateaus were obtained from ten patients undergoing total knee replacement. Each sample had been classified into a mild or severe group according to OARSI scoring. Tissue was taken from each specimen and mRNA expression levels of CD105, CD166, Notch 1, Sox9, Acan and Col II A1 were measured at day 0 and day 14 (2 weeks in vitro). Furthermore, MSC markers: Nucleostemin, CD90, CD73, CD166, CD105 and Notch 1 were studied by immunofluorescence. mRNA levels of MSC markers did not differ between mild and severe OA at day 0. At day 14, protein analysis showed that proliferated cells from both sources expressed all 6 MSC markers. Only cells from the mild OA subjects resulted in a significant increase of mRNA CD105 and CD166 after in vitro expansion. Moreover, cells from the mild OA subjects showed significantly higher levels of CD105, Sox9 and Acan compared with those from severe OA specimens. Results confirmed the presence of MSC markers in mild and severe OA tissue at both mRNA and protein levels. We found significant differences between cells obtained from mild compared to severe OA specimens suggests that mild OA derived cells may have a greater MSC potential.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antigens, CD/analysis
- Antigens, CD/genetics
- Biomarkers/analysis
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cell Adhesion Molecules, Neuronal/analysis
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Differentiation
- Endoglin/analysis
- Endoglin/genetics
- Fetal Proteins/analysis
- Fetal Proteins/genetics
- Humans
- Knee Joint/metabolism
- Knee Joint/pathology
- Mesenchymal Stem Cells/metabolism
- Mesenchymal Stem Cells/pathology
- Middle Aged
- Osteoarthritis, Knee/genetics
- Osteoarthritis, Knee/pathology
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- SOX9 Transcription Factor/analysis
- SOX9 Transcription Factor/genetics
- Transcriptome
Collapse
Affiliation(s)
- Marija Mazor
- Department of Sciences, University of Orleans, I3MTO, EA 4708, Orleans F-45032, France.
| | - Annabelle Cesaro
- Department of Sciences, University of Orleans, I3MTO, EA 4708, Orleans F-45032, France.
| | - Mazen Ali
- Service chirurgie orthopédique et traumatologique Centre Hospitalier Régional d'Orléans, La Source 45000, France.
| | - Thomas M Best
- UHealth Sports Medicine Institute, Department of Orthopedics, Division of Sports Medicine, U of Miami, Coral Gables, FL 33146, USA.
| | - Eric Lespessaille
- Department of Sciences, University of Orleans, I3MTO, EA 4708, Orleans F-45032, France.
- EA4708/I3MTO, Service de Rhumatologie, Centre Hospitalier Régional d'Orléans, La Source 45000, France.
| | - Hechmi Toumi
- Department of Sciences, University of Orleans, I3MTO, EA 4708, Orleans F-45032, France.
- EA4708/I3MTO, Service de Rhumatologie, Centre Hospitalier Régional d'Orléans, La Source 45000, France.
| |
Collapse
|
59
|
Nam Y, Rim YA, Ju JH. Chondrogenic Pellet Formation from Cord Blood-derived Induced Pluripotent Stem Cells. J Vis Exp 2017. [PMID: 28654049 DOI: 10.3791/55988] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Human articular cartilage lacks the ability to repair itself. Cartilage degeneration is thus treated not by curative but by conservative treatments. Currently, efforts are being made to regenerate damaged cartilage with ex vivo expanded chondrocytes or bone marrow-derived mesenchymal stem cells (BMSCs). However, the restricted viability and instability of these cells limit their application in cartilage reconstruction. Human induced pluripotent stem cells (hiPSCs) have received scientific attention as a new alternative for regenerative applications. With unlimited self-renewal ability and multipotency, hiPSCs have been highlighted as a new replacement cell source for cartilage repair. However, obtaining a high quantity of high-quality chondrogenic pellets is a major challenge to their clinical application. In this study, we used embryoid body (EB)-derived outgrowth cells for chondrogenic differentiation. Successful chondrogenesis was confirmed by PCR and staining with alcian blue, toluidine blue, and antibodies against collagen types I and II (COL1A1 and COL2A1, respectively). We provide a detailed method for the differentiation of cord blood mononuclear cell-derived iPSCs (CBMC-hiPSCs) into chondrogenic pellets.
Collapse
Affiliation(s)
- Yoojun Nam
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Division of Rheumatology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea
| | - Yeri Alice Rim
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Division of Rheumatology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea
| | - Ji Hyeon Ju
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea;
| |
Collapse
|
60
|
Biostable scaffolds of polyacrylate polymers implanted in the articular cartilage induce hyaline-like cartilage regeneration in rabbits. Int J Artif Organs 2017; 40:350-357. [PMID: 28574106 PMCID: PMC6379805 DOI: 10.5301/ijao.5000598] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2017] [Indexed: 11/24/2022]
Abstract
Purpose To study the influence of scaffold properties on the organization of in vivo cartilage regeneration. Our hypothesis was that stress transmission to the cells seeded inside the pores of the scaffold or surrounding it, which is highly dependent on the scaffold properties, determines the differentiation of both mesenchymal cells and dedifferentiated autologous chondrocytes. Methods 4 series of porous scaffolds made of different polyacrylate polymers, previously seeded with cultured rabbit chondrocytes or without cells, were implanted in cartilage defects in rabbits. Subchondral bone was injured during the surgery to allow blood to reach the implantation site and fill the scaffold pores. Results At 3 months after implantation, excellent tissue regeneration was obtained, with a well-organized layer of hyaline-like cartilage at the condylar surface in most cases of the hydrophobic or slightly hydrophilic series. The most hydrophilic material induced the poorest regeneration. However, no statistically significant difference was observed between preseeded and non-preseeded scaffolds. All of the materials used were biocompatible, biostable polymers, so, in contrast to some other studies, our results were not perturbed by possible effects attributable to material degradation products or to the loss of scaffold mechanical properties over time due to degradation. Conclusions Cartilage regeneration depends mainly on the properties of the scaffold, such as stiffness and hydrophilicity, whereas little difference was observed between preseeded and non-preseeded scaffolds.
Collapse
|
61
|
Lin H, Zhou J, Cao L, Wang HR, Dong J, Chen ZR. Tissue-engineered cartilage constructed by a biotin-conjugated anti-CD44 avidin binding technique for the repairing of cartilage defects in the weight-bearing area of knee joints in pigs. Bone Joint Res 2017; 6:284-295. [PMID: 28515058 PMCID: PMC5457648 DOI: 10.1302/2046-3758.65.bjr-2016-0277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/20/2017] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES The lack of effective treatment for cartilage defects has prompted investigations using tissue engineering techniques for their regeneration and repair. The success of tissue-engineered repair of cartilage may depend on the rapid and efficient adhesion of transplanted cells to a scaffold. Our aim in this study was to repair full-thickness defects in articular cartilage in the weight-bearing area of a porcine model, and to investigate whether the CD44 monoclonal antibody biotin-avidin (CBA) binding technique could provide satisfactory tissue-engineered cartilage. METHODS Cartilage defects were created in the load-bearing region of the lateral femoral condyle of mini-type pigs. The defects were repaired with traditional tissue-engineered cartilage, tissue-engineered cartilage constructed with the biotin-avidin (BA) technique, tissue-engineered cartilage constructed with the CBA technique and with autologous cartilage. The biomechanical properties, Western blot assay, histological findings and immunohistochemical staining were explored. RESULTS The CBA group showed similar results to the autologous group in biomechanical properties, Moran's criteria, histological tests and Wakitani histological scoring. CONCLUSIONS These results suggest that tissue-engineered cartilage constructed using the CBA technique could be used effectively to repair cartilage defects in the weight-bearing area of joints.Cite this article: H. Lin, J. Zhou, L. Cao, H. R. Wang, J. Dong, Z. R. Chen. Tissue-engineered cartilage constructed by a biotin-conjugated anti-CD44 avidin binding technique for the repairing of cartilage defects in the weight-bearing area of knee joints in pigs. Bone Joint Res 2017;6:-295. DOI: 10.1302/2046-3758.65.BJR-2016-0277.
Collapse
Affiliation(s)
- H Lin
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - J Zhou
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - L Cao
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - H R Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - J Dong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Z R Chen
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
62
|
Merlin Rajesh Lal LP, Suraishkumar GK, Nair PD. Chitosan-agarose scaffolds supports chondrogenesis of Human Wharton's Jelly mesenchymal stem cells. J Biomed Mater Res A 2017; 105:1845-1855. [DOI: 10.1002/jbm.a.36054] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 11/11/2022]
Affiliation(s)
- L. P. Merlin Rajesh Lal
- Department of Biotechnology; IIT Madras; Chennai Tamil Nadu 600036 India
- Division of Tissue Engineering and Regeneration Technologies; Sree Chitra Tirunal Institute for Medical Sciences and Technology; BMT Wing Trivandrum Kerala 695012 India
| | - G. K. Suraishkumar
- Division of Tissue Engineering and Regeneration Technologies; Sree Chitra Tirunal Institute for Medical Sciences and Technology; BMT Wing Trivandrum Kerala 695012 India
| | - Prabha D. Nair
- Division of Tissue Engineering and Regeneration Technologies; Sree Chitra Tirunal Institute for Medical Sciences and Technology; BMT Wing Trivandrum Kerala 695012 India
| |
Collapse
|
63
|
Evaluating the cartilage adjacent to the site of repair surgery with glycosaminoglycan-specific magnetic resonance imaging. INTERNATIONAL ORTHOPAEDICS 2017; 41:969-974. [DOI: 10.1007/s00264-017-3434-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 02/28/2017] [Indexed: 01/17/2023]
|
64
|
McKee C, Hong Y, Yao D, Chaudhry GR. Compression Induced Chondrogenic Differentiation of Embryonic Stem Cells in Three-Dimensional Polydimethylsiloxane Scaffolds. Tissue Eng Part A 2017; 23:426-435. [PMID: 28103756 DOI: 10.1089/ten.tea.2016.0376] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Embryonic stem cells (ESCs) are an ideal source for chondrogenic progenitors for the repair of damaged cartilage tissue. It is currently difficult to induce uniform and scalable ESC differentiation in vitro, a process required for stem cell therapy. This is partly because stem cell fate is determined by complex interactions with the native microenvironment and mechanical properties of the extracellular matrix. Mechanical signaling is considered to be one of the major factors regulating the proliferation and differentiation of chondrogenic cells both in vitro and in vivo. We used biocompatible and elastic polydimethylsiloxane (PDMS) scaffolds, capable of transducing mechanical signals, including compressive stress in vitro. ESCs seeded into the PDMS scaffolds and subjected to mechanical loading resulted in induction of differentiation. Differentiated ESC derivatives in three-dimensional (3-D) PDMS scaffolds exhibited elongated single cell rather than round clonal ESC morphology. They expressed chondrogenic marker, Col2, with concomitant reduction in the expression of pluripotent marker, Oct4. Immunocytochemical analysis also showed that the expression of COL2 protein was significantly higher in ESCs in 3-D scaffolds subjected to compressive stress. Further analysis showed that compressive stress also resulted in expression of early chondrogenic makers, Sox9 and Acan, but not hypertrophic chondrogenic markers, Runx2, Col10, and Mmp13. Compressive stress induced differentiation caused a reduction in the expression of β-Catenin and an increase in the expression of genes, Rhoa, Yap, and Taz, which are known to be affected by mechanosignaling. The chondroinductive role of RhoA was confirmed by its downregulation with simultaneous decrease in the transcriptional and translational expression of early chondrogenic markers, SOX9, COL2, and ACAN, when ESCs in PDMS scaffolds were subjected to compressive stress and treated with RhoA inhibitor, CCG-1432. Based on these observations, a model for compression induced chondrogenic differentiation of ESCs in 3-D scaffolds was proposed.
Collapse
Affiliation(s)
- Christina McKee
- 1 Department of Biological Sciences, Oakland University , Rochester, Michigan.,2 OU-WB Institute for Stem Cell and Regenerative Medicine , Rochester, Michigan
| | - Yifeng Hong
- 3 School of Materials Science and Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - Donggang Yao
- 3 School of Materials Science and Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - G Rasul Chaudhry
- 1 Department of Biological Sciences, Oakland University , Rochester, Michigan.,2 OU-WB Institute for Stem Cell and Regenerative Medicine , Rochester, Michigan
| |
Collapse
|
65
|
Abstract
Articular cartilage is a load-bearing tissue that lines the surface of bones in diarthrodial joints. Unfortunately, this avascular tissue has a limited capacity for intrinsic repair. Treatment options for articular cartilage defects include microfracture and arthroplasty; however, these strategies fail to generate tissue that adequately restores damaged cartilage. Limitations of current treatments for cartilage defects have prompted the field of cartilage tissue engineering, which seeks to integrate engineering and biological principles to promote the growth of new cartilage to replace damaged tissue. To date, a wide range of scaffolds and cell sources have emerged with a focus on recapitulating the microenvironments present during development or in adult tissue, in order to induce the formation of cartilaginous constructs with biochemical and mechanical properties of native tissue. Hydrogels have emerged as a promising scaffold due to the wide range of possible properties and the ability to entrap cells within the material. Towards improving cartilage repair, hydrogel design has advanced in recent years to improve their utility. Some of these advances include the development of improved network crosslinking (e.g. double-networks), new techniques to process hydrogels (e.g. 3D printing) and better incorporation of biological signals (e.g. controlled release). This review summarises these innovative approaches to engineer hydrogels towards cartilage repair, with an eye towards eventual clinical translation.
Collapse
Affiliation(s)
| | | | - J A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104,
| |
Collapse
|
66
|
Nam Y, Rim YA, Jung SM, Ju JH. Cord blood cell-derived iPSCs as a new candidate for chondrogenic differentiation and cartilage regeneration. Stem Cell Res Ther 2017; 8:16. [PMID: 28129782 PMCID: PMC5273802 DOI: 10.1186/s13287-017-0477-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 01/03/2017] [Accepted: 01/07/2017] [Indexed: 12/12/2022] Open
Abstract
Background The native articular cartilage lacks the ability to heal. Currently, ex vivo expanded chondrocytes or bone marrow-derived mesenchymal stem cells are used to regenerate the damaged cartilage. With unlimited self-renewal ability and multipotency, human induced pluripotent stem cells (hiPSCs) have been highlighted as a new replacement cell source for cartilage repair. Still, further research is needed on cartilage regeneration using cord blood mononuclear cell-derived hiPSCs (CBMC-hiPSCs). Methods Human iPSCs were generated from CBMCs using the Sendai virus. The characterization of CBMC-hiPSCs was performed by various assays. Embryonic bodies (EBs) were obtained using CBMC-hiPSCs, and outgrowth cells were induced by plating the EBs onto a gelatin-coated plate. Expanded outgrowth cells were detached and dissociated for chondrogenic differentiation. Outgrowth cells were differentiated into chondrogenic lineage with pellet culture. Chondrogenic pellets were maintained for 30 days. The quality of chondrogenic pellets was evaluated using various staining and genetic analysis of cartilage-specific markers. Results Reprogramming was successfully done using CBMCs. CBMC-hiPSCs (n = 3) showed high pluripotency and normal karyotype. Chondrogenic pellets were generated from the outgrowth cells derived from CBMC-hiPSC EBs. The generated chondrogenic pellets showed high expression of chondrogenic genetic markers such as ACAN, COMP, COL2A1, and SOX9. The production of extracellular matrix (ECM) proteins was confirmed by safranin O, alcian blue and toluidine blue staining. Expression of collagen type II and aggrecan was detected in the accumulated ECM by immunohistological staining. Chondrogenic pellets showed low expression of fibrotic and hypertrophic cartilage marker, collagen type I and X. Conclusions This study reveals the potential of CBMC-hiPSCs as a promising candidate for cartilage regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0477-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yoojun Nam
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, #505, Banpo-Dong, Seocho-Gu, Seoul, 137-701, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, #505, Banpo-Dong, Seocho-Gu, Seoul, 137-701, Republic of Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Yonsei University, Seoul, 120-749, Republic of Korea
| | - Ji Hyeon Ju
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea. .,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, #505, Banpo-Dong, Seocho-Gu, Seoul, 137-701, Republic of Korea.
| |
Collapse
|
67
|
Vira S, Ramme AJ, Chapman C, Xia D, Regatte RR, Chang G. Juvenile Particulate Osteochondral Allograft for Treatment of Osteochondral Lesions of the Talus: Detection of Altered Repair Tissue Biochemical Composition Using 7 Tesla MRI and T2 Mapping. J Foot Ankle Surg 2017; 56:26-29. [PMID: 27989341 DOI: 10.1053/j.jfas.2016.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Indexed: 02/03/2023]
Abstract
During the previous 2 decades, numerous surgical procedures have become available to treat osteochondral lesions of the talus. The objective of the present study was to use 7 Tesla (7T) magnetic resonance imaging (MRI) to quantify and compare T2 values (a marker of collagen architecture) of native tibiotalar cartilage and cartilage repair tissue in patients treated with a juvenile particulate allograft for osteochondral lesions of the talus. The institutional review board approved the present study, and all subjects provided written informed consent. We scanned the ankles of 7 cartilage repair patients using a 7T MRI scanner with a multi-echo spin-echo sequence to measure the cartilage T2 values. We assessed the cartilage T2 values in the talar repair tissue, adjacent native talar cartilage, and overlying tibial cartilage. We compared the differences between groups using the paired t test. The talar cartilage repair tissue demonstrated greater mean T2 relaxation times compared with the native adjacent talar cartilage (64.88 ± 12.23 ms versus 49.56 ± 7.82 ms; p = .043). The tibial cartilage regions overlying these talar cartilage regions demonstrated a trend toward greater T2 relaxation times (77.00 ± 31.29 ms versus 59.52 ± 7.89 ms; p = .067). 7T MRI can detect differences in T2 values in cartilage repair tissue compared with native cartilage and could be useful for monitoring the status of cartilage health after surgical intervention.
Collapse
Affiliation(s)
- Shaleen Vira
- Resident Physician, Department of Orthopaedic Surgery, NYU Hospital for Joint Diseases, New York, NY
| | - Austin J Ramme
- Resident Physician, Department of Orthopaedic Surgery, NYU Hospital for Joint Diseases, New York, NY
| | - Cary Chapman
- Assistant Professor, Department of Orthopaedic Surgery, NYU Hospital for Joint Diseases, New York, NY
| | - Ding Xia
- Research Scientist, Center for Biomedical Imaging, Department of Radiology, NYU Langone Medical Center, New York, NY
| | - Ravinder R Regatte
- Professor, Center for Biomedical Imaging, Department of Radiology, NYU Langone Medical Center, New York, NY
| | - Gregory Chang
- Associate Professor, Center for Biomedical Imaging, Department of Radiology, NYU Langone Medical Center, New York, NY.
| |
Collapse
|
68
|
Challenges for Cartilage Regeneration. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2017. [DOI: 10.1007/978-3-662-53574-5_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
69
|
Koh JL, Logli AL. Retropatellar Accessory Portals for Improved Access to the Patella: Anatomic Description and Evaluation of Safety. Orthop J Sports Med 2016; 4:2325967116672445. [PMID: 27900340 PMCID: PMC5122835 DOI: 10.1177/2325967116672445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background: Standard knee arthroscopy portals are frequently used to address retropatellar chondral pathology. Alternative portals may offer a safe and simple substitute, particularly when reaching the base of deep lesions is required. Purpose: To describe and assess the safety of accessory retropatellar portals. Study Design: Descriptive laboratory study. Methods: An anatomic study was performed on 10 fresh-frozen cadaveric knees. Medial and lateral retropatellar portals were created, and the distance to adjacent neurovascular structures (common peroneal nerve [CPN] and infrapatellar branch of the saphenous nerve [IPBSN]) and bony landmarks (medial and lateral epicondyles and margins of the midpatella) was recorded. A clinical account of portal use is provided through a retrospective chart review. Results: The medial and lateral retropatellar portals were a mean 15.8 ± 15.5 mm and 53.8 ± 7.2 mm from the IPBSN and CPN, respectively. No nerves were contacted in any specimens. Clinically, the portals have been used in 109 cases over a 7-year period without complication. Conclusion: The high level of IPBSN variability poses some risk to medial portal use; however, there is little to no risk laterally. Clinical experience with portals suggests that they can be used safely. We recommend careful blunt dissection to minimize chance of iatrogenic nerve injury. Clinical Relevance: Retropatellar portals may offer improved access to chondral lesions of the patella, thereby safely allowing one to maintain an arthroscopic approach.
Collapse
Affiliation(s)
- Jason L Koh
- Department of Orthopaedic Surgery, NorthShore University HealthSystem, Evanston, Illinois, USA.; University of Chicago Medical Center, Chicago, Illinois, USA
| | - Anthony L Logli
- University of Illinois College of Medicine at Rockford, Rockford, Illinois, USA
| |
Collapse
|
70
|
Broguiere N, Cavalli E, Salzmann GM, Applegate LA, Zenobi-Wong M. Factor XIII Cross-Linked Hyaluronan Hydrogels for Cartilage Tissue Engineering. ACS Biomater Sci Eng 2016; 2:2176-2184. [DOI: 10.1021/acsbiomaterials.6b00378] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nicolas Broguiere
- ETH Zürich, Cartilage Engineering and
Regeneration Laboratory, HPL J20, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Emma Cavalli
- ETH Zürich, Cartilage Engineering and
Regeneration Laboratory, HPL J20, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | | | - Lee Ann Applegate
- Department
of Musculoskeletal Medicine, Regenerative Therapy Unit, University Hospital of Lausanne, 1011 Lausanne, Switzerland
| | - Marcy Zenobi-Wong
- ETH Zürich, Cartilage Engineering and
Regeneration Laboratory, HPL J20, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| |
Collapse
|
71
|
|
72
|
Miranda-Nieves D, Chaikof EL. Collagen and Elastin Biomaterials for the Fabrication of Engineered Living Tissues. ACS Biomater Sci Eng 2016; 3:694-711. [PMID: 33440491 DOI: 10.1021/acsbiomaterials.6b00250] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Collagen and elastin represent the two most predominant proteins in the body and are responsible for modulating important biological and mechanical properties. Thus, the focus of this review is the use of collagen and elastin as biomaterials for the fabrication of living tissues. Considering the importance of both biomaterials, we first propose the notion that many tissues in the human body represent a reinforced composite of collagen and elastin. In the rest of the review, collagen and elastin biosynthesis and biophysics, as well as molecular sources and biomaterial fabrication methodologies, including casting, fiber spinning, and bioprinting, are discussed. Finally, we summarize the current attempts to fabricate a subset of living tissues and, based on biochemical and biomechanical considerations, suggest that future tissue-engineering efforts consider direct incorporation of collagen and elastin biomaterials.
Collapse
Affiliation(s)
- David Miranda-Nieves
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, United States
| | - Elliot L Chaikof
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| |
Collapse
|
73
|
Abstract
BACKGROUND Mandibular condyle cartilage (MCC) has a unique structure among articular cartilages; however, little is known about its nanoscale collagen network architecture, hampering design of regeneration therapies and rigorous evaluation of regeneration experiment outcomes in preclinical research. Helium ion microscopy is a novel technology with a long depth of field that is uniquely suited to imaging open 3D collagen networks at multiple scales without obscuring conductive coatings. OBJECTIVE The objective of this research was to image, at the micro- and nanoscales, the depth-dependent MCC collagen network architecture. DESIGN MCC was collected from New Zealand white rabbits. Images of MCC zones were acquired using helium ion, transmission electron, and light microscopy. Network fibril and canal diameters were measured. RESULTS For the first time, the MCC was visualized as a 3D collagen fibril structure at the nanoscale, the length scale of network assembly. Fibril diameters ranged from 7 to 110 nm and varied by zone. The articular surface was composed of a fine mesh that was woven through thin layers of larger fibrils. The fibrous zone was composed of approximately orthogonal lamellae of aligned fibrils. Fibrocyte processes surrounded collagen bundles forming extracellular compartments. The proliferative, mature, and hypertrophic zones were composed of a branched network that was progressively remodeled to accommodate chondrocyte hypertrophy. Osteoid fibrils were woven around osteoblast cytoplasmic processes to create numerous canals similar in size to canaliculi of mature bone. CONCLUSION This multiscale investigation advances our foundational understanding of the complex, layered 3D architecture of the MCC collagen network.
Collapse
Affiliation(s)
- Wendy S. Vanden Berg-Foels
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
74
|
Bauer C, Berger M, Baumgartner RR, Höller S, Zwickl H, Niculescu-Morzsa E, Halbwirth F, Nehrer S. A Novel Cross-Linked Hyaluronic Acid Porous Scaffold for Cartilage Repair: An In Vitro Study With Osteoarthritic Chondrocytes. Cartilage 2016; 7:265-73. [PMID: 27375842 PMCID: PMC4918062 DOI: 10.1177/1947603515611949] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
PURPOSE An important feature of biomaterials used in cartilage regeneration is their influence on the establishment and stabilization of a chondrocytic phenotype of embedded cells. The purpose of this study was to examine the effects of a porous 3-dimensional scaffold made of cross-linked hyaluronic acid on the expression and synthesis performance of human articular chondrocytes. MATERIALS AND METHODS Osteoarthritic chondrocytes from 5 patients with a mean age of 74 years were passaged twice and cultured within the cross-linked hyaluronic acid scaffolds for 2 weeks. Analyses were performed at 3 different time points. For estimation of cell content within the scaffold, DNA-content (CyQuant cell proliferation assay) was determined. The expression of chondrocyte-specific genes by embedded cells as well as the total amount of sulfated glycosaminoglycans produced during the culture period was analyzed in order to characterize the synthesis performance and differentiation status of the cells. RESULTS Cells showed a homogenous distribution within the scaffold. DNA quantification revealed a reduction of the cell number. This might be attributed to loss of cells from the scaffold during media exchange connected with a stop in cell proliferation. Indeed, the expression of cartilage-specific genes and the production of sulfated glycosaminoglycans were increased and the differentiation index was clearly improved. CONCLUSIONS These results suggest that the attachment of osteoarthritic P2 chondrocytes to the investigated material enhanced the chondrogenic phenotype as well as promoted the retention.
Collapse
Affiliation(s)
- Christoph Bauer
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria,Christoph Bauer, Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr.-Karl-Dorrek-Strasse 30, Krems, 3500, Austria.
| | - Manuela Berger
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| | | | | | - Hannes Zwickl
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| | - Eugenia Niculescu-Morzsa
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| | - Florian Halbwirth
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| | - Stefan Nehrer
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| |
Collapse
|
75
|
Zhang C, Qiu L, Gao L, Guan Y, Xu Q, Zhang X, Chen Q. A novel dual-frequency loading system for studying mechanobiology of load-bearing tissue. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:262-7. [PMID: 27612712 DOI: 10.1016/j.msec.2016.06.080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 05/30/2016] [Accepted: 06/25/2016] [Indexed: 01/04/2023]
Abstract
In mechanobiological research, an appropriate loading system is an essential tool to mimic mechanical signals in a native environment. To achieve this goal, we have developed a novel loading system capable of applying dual-frequency loading including both a low-frequency high-amplitude loading and a high-frequency low-amplitude loading, according to the mechanical conditions experienced by bone and articular cartilage tissues. The low-frequency high-amplitude loading embodies the main force from muscular contractions and/or reaction forces while the high-frequency low-amplitude loading represents an assistant force from small muscles, ligaments and/or other tissue in order to maintain body posture during human activities. Therefore, such dual frequency loading system may reflect the natural characteristics of complex mechanical load on bone or articular cartilage than the single frequency loading often applied during current mechanobiological experiments. The dual-frequency loading system is validated by experimental tests using precision miniature plane-mirror interferometers. The dual-frequency loading results in significantly more solute transport in articular cartilage than that of the low-frequency high-amplitude loading regiment alone, as determined by quantitative fluorescence microscopy of tracer distribution in articular cartilage. Thus, the loading system can provide a new method to mimic mechanical environment in bone and cartilage, thereby revealing the in vivo mechanisms of mechanosensation, mechanotransduction and mass-transport, and improving mechanical conditioning of cartilage and/or bone constructs for tissue engineering.
Collapse
Affiliation(s)
- Chunqiu Zhang
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin 300384, China; Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA.
| | - Lulu Qiu
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin 300384, China
| | - Lilan Gao
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin 300384, China
| | - Yinjie Guan
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Qiang Xu
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin 300384, China
| | - Xizheng Zhang
- Tianjin Key Laboratory of the Design and Intelligent Control of the Advanced Mechatronical System, Tianjin University of Technology, Tianjin 300384, China
| | - Qian Chen
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA.
| |
Collapse
|
76
|
Popa EG, Reis RL, Gomes ME. Seaweed polysaccharide-based hydrogels used for the regeneration of articular cartilage. Crit Rev Biotechnol 2016; 35:410-24. [PMID: 24646368 DOI: 10.3109/07388551.2014.889079] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This manuscript provides an overview of the in vitro and in vivo studies reported in the literature focusing on seaweed polysaccharides based hydrogels that have been proposed for applications in regenerative medicine, particularly, in the field of cartilage tissue engineering. For a better understanding of the main requisites for these specific applications, the main aspects of the native cartilage structure, as well as recognized diseases that affect this tissue are briefly described. Current available treatments are also presented to emphasize the need for alternative techniques. The following part of this review is centered on the description of the general characteristics of algae polysaccharides, as well as relevant properties required for designing hydrogels for cartilage tissue engineering purposes. An in-depth overview of the most well known seaweed polysaccharide, namely agarose, alginate, carrageenan and ulvan biopolymeric gels, that have been proposed for engineering cartilage is also provided. Finally, this review describes and summarizes the translational aspect for the clinical application of alternative systems emphasizing the importance of cryopreservation and the commercial products currently available for cartilage treatment.
Collapse
Affiliation(s)
- Elena Geta Popa
- a 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark , Guimarães , Portugal and
| | | | | |
Collapse
|
77
|
Shang J, Wang H, Fan X, Shangguan L, Liu H. A genome wide analysis of alternative splicing events during the osteogenic differentiation of human cartilage endplate-derived stem cells. Mol Med Rep 2016; 14:1389-96. [PMID: 27278552 DOI: 10.3892/mmr.2016.5359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 05/12/2016] [Indexed: 11/06/2022] Open
Abstract
Low back pain is a prevalent disease, which leads to suffering and disabilities in a vast number of individuals. Degenerative disc diseases are usually the underlying causes of low back pain. However, the pathogenesis of degenerative disc diseases is highly complex and difficult to determine. Current therapies for degenerative disc diseases are various. In particular, cell-based therapies have proven to be effective and promising. Our research group has previously isolated and identified the cartilage endplate‑derived stem cells. In addition, alternative splicing is a sophisticated regulatory mechanism, which greatly increases cellular complexity and phenotypic diversity of eukaryotic organisms. The present study continued to investigate alternative splicing events in osteogenic differentiation of cartilage endplate‑derived stem cells. An Affymetrix Human Transcriptome Array 2.0 was used to detect splicing changes between the control and differentiated samples. Additionally, molecular function and pathway analysis were also performed. Following rigorous bioinformatics analysis of the data, 3,802 alternatively spliced genes were identified, and 10 of these were selected for validation by reverse transcription‑polymerase chain reaction. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway analysis also revealed numerous enriched GO terms and signaling pathways. To the best of our knowledge, the present study is the first to investigate alternative splicing mechanisms in osteogenic differentiation of stem cells on a genome‑wide scale. The illumination of molecular mechanisms of stem cell osteogenic differentiation may assist the development novel bioengineered methods to treat degenerative disc diseases.
Collapse
Affiliation(s)
- Jin Shang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Honggang Wang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Xin Fan
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Lei Shangguan
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Huan Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
78
|
Raghothaman D, Leong MF, Lim TC, Wan ACA, Ser Z, Lee EH, Yang Z. Cell type dependent morphological adaptation in polyelectrolyte hydrogels governs chondrogenic fate. Biomed Mater 2016; 11:025013. [DOI: 10.1088/1748-6041/11/2/025013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
79
|
Maturavongsadit P, Luckanagul JA, Metavarayuth K, Zhao X, Chen L, Lin Y, Wang Q. Promotion of In Vitro Chondrogenesis of Mesenchymal Stem Cells Using In Situ Hyaluronic Hydrogel Functionalized with Rod-Like Viral Nanoparticles. Biomacromolecules 2016; 17:1930-8. [DOI: 10.1021/acs.biomac.5b01577] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Panita Maturavongsadit
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| | - Jittima Amie Luckanagul
- Department
of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Wangmai, Pathumwan, Bangkok, 10330, Thailand
| | - Kamolrat Metavarayuth
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| | - Xia Zhao
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People’s Republic of China
| | - Limin Chen
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People’s Republic of China
| | - Yuan Lin
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People’s Republic of China
| | - Qian Wang
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| |
Collapse
|
80
|
Corradetti B, Taraballi F, Minardi S, Van Eps J, Cabrera F, Francis LW, Gazze SA, Ferrari M, Weiner BK, Tasciotti E. Chondroitin Sulfate Immobilized on a Biomimetic Scaffold Modulates Inflammation While Driving Chondrogenesis. Stem Cells Transl Med 2016; 5:670-82. [PMID: 27013739 DOI: 10.5966/sctm.2015-0233] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 01/04/2016] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Costs associated with degenerative inflammatory conditions of articular cartilage are exponentially increasing in the aging population, and evidence shows a strong clinical need for innovative therapies. Stem cell-based therapies represent a promising strategy for the treatment of innumerable diseases. Their regenerative potential is undeniable, and it has been widely exploited in many tissue-engineering approaches, especially for bone and cartilage repair. Their immune-modulatory capacities in particular make stem cell-based therapeutics an attractive option for treating inflammatory diseases. However, because of their great plasticity, mesenchymal stem cells (MSCs) are susceptible to different external factors. Biomaterials capable of concurrently providing physical support to cells while acting as synthetic extracellular matrix have been established as a valuable strategy in cartilage repair. Here we propose a chondroitin sulfate-based biomimetic scaffold that recapitulates the physicochemical features of the chondrogenic niche and retains MSC immunosuppressive potential in vitro, either in response to a proinflammatory cytokine or in the presence of stimulated peripheral blood mononuclear cells. In both cases, a significant increase in the production of molecules associated with immunosuppression (nitric oxide and prostaglandins), as well as in the expression of their inducible enzymes (iNos, Pges, Cox-2, and Tgf-β). When implanted subcutaneously in rats, our scaffold revealed a reduced infiltration of leukocytes at 24 hours, which correlated with a greater upregulation of genes involved in inflammatory cell apoptotic processes. In support of its effective use in tissue-engineering applications of cartilage repair, the potential of the proposed platform to drive chondrogenic and osteogenic differentiation of MSC was also proven. SIGNIFICANCE Recently, increasing clinical evidence has highlighted the important role of proinflammatory mediators and infiltrating inflammatory cell populations inducing chronic inflammation and diseases in damaged cartilage. This work should be of broad interest because it proposes an implantable biomimetic material, which holds the promise for a variety of medical conditions that necessitate the functional restoration of damaged cartilage tissue (such as trauma, diseases, deformities, or cancer).
Collapse
Affiliation(s)
- Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Francesca Taraballi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Silvia Minardi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA Institute of Science and Technology for Ceramics, National Research Council of Italy, Faenza, Italy
| | - Jeffrey Van Eps
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA Department of Surgery, Houston Methodist Hospital, Houston, Texas, USA
| | - Fernando Cabrera
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Lewis W Francis
- Centre for NanoHealth, Swansea University Medical School, Swansea University Bay, Singleton Park, Wales, United Kingdom
| | - Salvatore A Gazze
- Centre for NanoHealth, Swansea University Medical School, Swansea University Bay, Singleton Park, Wales, United Kingdom
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Bradley K Weiner
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA Department of Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
81
|
D'Este M, Sprecher CM, Milz S, Nehrbass D, Dresing I, Zeiter S, Alini M, Eglin D. Evaluation of an injectable thermoresponsive hyaluronan hydrogel in a rabbit osteochondral defect model. J Biomed Mater Res A 2016; 104:1469-78. [PMID: 26833870 DOI: 10.1002/jbm.a.35673] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/14/2016] [Accepted: 01/29/2016] [Indexed: 01/06/2023]
Abstract
Articular cartilage displays very little self-healing capabilities, generating a major clinical need. Here, we introduce a thermoresponsive hyaluronan hydrogel for cartilage repair obtained by covalently grafting poly(N-isopropylacrylamide) to hyaluronan, to give a brush co-polymer HpN. The gel is fluid at room temperature and becomes gel at body temperature. In this pilot study HpN safety and repair response were evaluated in an osteochondral defect model in rabbit. Follow-up was of 1 week and 12 weeks and the empty defect served as a control, for a total of four experimental groups. At 12 weeks the defect sites were evaluated macroscopically and histologically. Local lymph nodes, spleen, liver, and kidneys were analyzed for histopathological evaluation. HpN could be easily injected and remained into the defect throughout the study. The macroscopic score was statistically superior for HpN versus empty. Histological score gave opposite trend but not statistically significant. A slight tissue reaction was observed around HpN, however, vascularization and subchondral bone formation were not impeded. An upper proteoglycans rich fibro-cartilaginous tissue with fairly good continuity and lateral integration into the existing articular cartilage was observed in all cases. No signs of local or systemic acute or subacute toxicity were observed. In conclusion, HpN is easily injectable, remains into an osteochondral defect within a moving synovial joint, is biocompatible and does not interfere with the intrinsic healing response of osteochondral defects in a rabbit model. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1469-1478, 2016.
Collapse
Affiliation(s)
- Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | | | - Stefan Milz
- Department of Anatomy II-Neuroanatomy, Ludwig-Maximilian-University of Munich, Bavaria, Germany
| | - Dirk Nehrbass
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | - Iska Dresing
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | - Stephan Zeiter
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| |
Collapse
|
82
|
Goodrich LR, Chen AC, Werpy NM, Williams AA, Kisiday JD, Su AW, Cory E, Morley PS, McIlwraith CW, Sah RL, Chu CR. Addition of Mesenchymal Stem Cells to Autologous Platelet-Enhanced Fibrin Scaffolds in Chondral Defects: Does It Enhance Repair? J Bone Joint Surg Am 2016; 98:23-34. [PMID: 26738900 PMCID: PMC4697360 DOI: 10.2106/jbjs.o.00407] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The chondrogenic potential of culture-expanded bone-marrow-derived mesenchymal stem cells (BMDMSCs) is well described. Numerous studies have also shown enhanced repair when BMDMSCs, scaffolds, and growth factors are placed into chondral defects. Platelets provide a rich milieu of growth factors and, along with fibrin, are readily available for clinical use. The objective of this study was to determine if the addition of BMDMSCs to an autologous platelet-enriched fibrin (APEF) scaffold enhances chondral repair compared with APEF alone. METHODS A 15-mm-diameter full-thickness chondral defect was created on the lateral trochlear ridge of both stifle joints of twelve adult horses. In each animal, one defect was randomly assigned to receive APEF+BMDMSCs and the contralateral defect received APEF alone. Repair tissues were evaluated one year later with arthroscopy, histological examination, magnetic resonance imaging (MRI), micro-computed tomography (micro-CT), and biomechanical testing. RESULTS The arthroscopic findings, MRI T2 map, histological scores, structural stiffness, and material stiffness were similar (p > 0.05) between the APEF and APEF+BMDMSC-treated repairs at one year. Ectopic bone was observed within the repair tissue in four of twelve APEF+BMDMSC-treated defects. Defects repaired with APEF alone had less trabecular bone edema (as seen on MRI) compared with defects repaired with APEF+BMDMSCs. Micro-CT analysis showed thinner repair tissue in defects repaired with APEF+BMDMSCs than in those treated with APEF alone (p < 0.05). CONCLUSIONS APEF alone resulted in thicker repair tissue than was seen with APEF+BMDMSCs. The addition of BMDMSCs to APEF did not enhance cartilage repair and stimulated bone formation in some cartilage defects. CLINICAL RELEVANCE APEF supported repair of critical-size full-thickness chondral defects in horses, which was not improved by the addition of BMDMSCs. This work supports further investigation to determine whether APEF enhances cartilage repair in humans.
Collapse
Affiliation(s)
- Laurie R. Goodrich
- Gail Holmes Equine Orthopedic Research Center, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523
| | - Albert C. Chen
- Department of Bioengineering, Mail Code 0412, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0412
| | - Natasha M. Werpy
- Large Animal Clinical Sciences, 2015 S.W. 16th Avenue, Gainesville, FL 32608
| | - Ashley A. Williams
- Department of Orthopedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063
| | - John D. Kisiday
- Gail Holmes Equine Orthopedic Research Center, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523
| | - Alvin W. Su
- Department of Bioengineering, Mail Code 0412, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0412
| | - Esther Cory
- Department of Bioengineering, Mail Code 0412, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0412
| | - Paul S. Morley
- Gail Holmes Equine Orthopedic Research Center, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523
| | - C. Wayne McIlwraith
- Gail Holmes Equine Orthopedic Research Center, Colorado State University, 300 West Drake Road, Fort Collins, CO 80523
| | - Robert L. Sah
- Department of Bioengineering, Mail Code 0412, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0412
| | - Constance R. Chu
- Department of Orthopedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA 94063.,E-mail address for C.R. Chu:
| |
Collapse
|
83
|
Temporomandibular joint disorders treated with articular injection: the effectiveness of plasma rich in growth factors-Endoret. J Craniofac Surg 2016; 26:709-13. [PMID: 25974777 DOI: 10.1097/scs.0000000000001534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The objective of this study was to evaluate the effectiveness of the temporomandibular joint (TMJ) osteoarthritis treatment through articular injections of plasma rich in growth factors (PGRF)-Endoret. Thirteen patients (median age, 47.64 y; SD, 7.51; range, 40-64 y; male-female ratio, 2:11) with osteoarthritis of TMJ associated to chronic pain have been selected. They were treated with articular injections of PRGF-Endoret, measuring the maximum mouth opening and pain level before the first injection (t0), 30 days after just before the second (t1), and after 6 months (t2). Data were analyzed using the paired Student's t-test data. The visual analogue scale score at t0 is 7.69 (range, 4-10; SD, 1.9), whereas that at t1 is 1.54 (range, 0-5; SD, 1.74) and that at t2 is 0.23 (range, 0-2; SD, 0.65). These differences in the results are statistically highly significant (P < 0.0001 comparison t0-t1 and t0-t2 and P < 0.01 comparison t1-t2). In terms of maximum mouth opening, it reduced from 30.15 mm at t0 (range, 26-40 mm; SD, 4.44) to 37.54 mm at t1 (range, 31-51 mm; SD, 5.10), with an increase of 7.38 mm (range, 4-11 mm; SD, 2.02) and a highly significant difference (P < 0.0001). At t2, it was 39.54 mm (range, 34-51; SD, 4.55) with an increase of 9.38 mm (range, 5-12 mm; SD, 2.21) compared with t0 and that of 2.00 mm compared with t1. Both differences in the results are statistically significant (P < 0.0001 and P < 0.01, respectively). The articular injections of PRGF-Endoret represent a very efficient method to control pain and to improve the TMJ mobility.
Collapse
|
84
|
Singhal R, Gupta K. A Review: Tailor-made Hydrogel Structures (Classifications and Synthesis Parameters). POLYMER-PLASTICS TECHNOLOGY AND ENGINEERING 2016; 55:54-70. [DOI: 10.1080/03602559.2015.1050520] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
85
|
Kim IL, Pfeifer CG, Fisher MB, Saxena V, Meloni GR, Kwon MY, Kim M, Steinberg DR, Mauck RL, Burdick JA. Fibrous Scaffolds with Varied Fiber Chemistry and Growth Factor Delivery Promote Repair in a Porcine Cartilage Defect Model. Tissue Eng Part A 2015; 21:2680-90. [PMID: 26401910 DOI: 10.1089/ten.tea.2015.0150] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Current clinically approved methods for cartilage repair are generally based on either endogenous cell recruitment (e.g., microfracture) or chondrocyte delivery (e.g., autologous chondrocyte implantation). However, both methods culminate in repair tissue with inferior mechanical properties and the addition of biomaterials to these clinical interventions may improve their efficacy. To this end, the objective of this study was to investigate the ability of multipolymer acellular fibrous scaffolds to improve cartilage repair when combined with microfracture in a large animal (i.e., minipig) model. Composite scaffolds were formulated from a combination of hyaluronic acid (HA) fibers and poly(ɛ-caprolactone) (PCL) fibers, either with or without transforming growth factor-β3 (TGFβ3). After 12 weeks in vivo, material choice and TGFβ3 delivery had a significant impact on outcomes; specifically, PCL scaffolds without TGFβ3 had inferior gross appearance and reduced mechanical properties, whereas HA scaffolds that released TGFβ3 resulted in improved histological scores and increased type 2 collagen content. Importantly, analysis of the overall dataset revealed that histology, but not gross appearance, was a better predictor of mechanical properties. This study highlights the importance of scaffold properties on in vivo cartilage repair as well as the need for numerous quantitative outcome measures to fully evaluate treatment methods.
Collapse
Affiliation(s)
- Iris L Kim
- 1 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania
| | - Christian G Pfeifer
- 2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania.,3 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Matthew B Fisher
- 2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania.,3 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Vishal Saxena
- 2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania.,3 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Gregory R Meloni
- 2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania.,3 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Mi Y Kwon
- 1 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Minwook Kim
- 2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania.,3 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - David R Steinberg
- 2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania.,3 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Robert L Mauck
- 1 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania.,3 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Jason A Burdick
- 1 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Translational Musculoskeletal Research Center, Philadelphia VA Medical Center , Philadelphia, Pennsylvania
| |
Collapse
|
86
|
Dang PN, Solorio LD, Alsberg E. Driving cartilage formation in high-density human adipose-derived stem cell aggregate and sheet constructs without exogenous growth factor delivery. Tissue Eng Part A 2015; 20:3163-75. [PMID: 24873753 DOI: 10.1089/ten.tea.2012.0551] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
An attractive cell source for cartilage tissue engineering, human adipose-derived stem cells (hASCs) can be easily expanded and signaled to differentiate into chondrocytes. This study explores the influence of growth factor distribution and release kinetics on cartilage formation within 3D hASC constructs incorporated with transforming growth factor-β1 (TGF-β1)-loaded gelatin microspheres. The amounts of microspheres, TGF-β1 concentration, and polymer degradation rate were varied within hASC aggregates. Microsphere and TGF-β1 loading concentrations were identified that resulted in glycosaminoglycan (GAG) production comparable to those of control aggregates cultured in TGF-β1-containing medium. Self-assembling hASC sheets were then engineered for the production of larger, more clinically relevant constructs. Chondrogenesis was observed in hASC-only sheets cultured with exogenous TGF-β1 at 3 weeks. Importantly, sheets with incorporated TGF-β1-loaded microspheres achieved GAG production similar to sheets treated with exogenous TGF-β1. Cartilage formation was confirmed histologically via observation of cartilage-like morphology and GAG staining. This is the first demonstration of the self-assembly of hASCs into high-density cell sheets capable of forming cartilage in the presence of exogenous TGF-β1 or with TGF-β1-releasing microspheres. Microsphere incorporation may bypass the need for extended in vitro culture, potentially enabling hASC sheets to be implanted more rapidly into defects to regenerate cartilage in vivo.
Collapse
Affiliation(s)
- Phuong N Dang
- 1 Department of Biomedical Engineering, Case Western Reserve University , Cleveland, Ohio
| | | | | |
Collapse
|
87
|
Pustlauk W, Paul B, Brueggemeier S, Gelinsky M, Bernhardt A. Modulation of chondrogenic differentiation of human mesenchymal stem cells in jellyfish collagen scaffolds by cell density and culture medium. J Tissue Eng Regen Med 2015; 11:1710-1722. [PMID: 26178016 DOI: 10.1002/term.2065] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 05/05/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022]
Abstract
Studies on tissue-engineering approaches for the regeneration of traumatized cartilage focus increasingly on multipotent human mesenchymal stem cells (hMSCs) as an alternative to autologous chondrocytes. The present study applied porous scaffolds made of collagen from the jellyfish Rhopilema esculentum for the in vitro chondrogenic differentiation of hMSCs. Culture conditions in those scaffolds differ from conditions in high-density pellet cultures, making a re-examination of these data necessary. We systematically investigated the influence of seeding density, basic culture media [Dulbecco's modified Eagle's medium (DMEM), α-minimum essential medium (α-MEM)] with varying glucose content and supplementation with fetal calf serum (FCS) or bovine serum albumin (BSA) on the chondrogenic differentiation of hMSCs. Gene expression analyses of selected markers for chondrogenic differentiation and hypertrophic development were conducted. Furthermore, the production of cartilage extracellular matrix (ECM) was analysed by quantification of sulphated glycosaminoglycan and collagen type II contents. The strongest upregulation of chondrogenic markers, along with the highest ECM deposition was observed in scaffolds seeded with 2.4 × 106 cells/cm3 after cultivation in high-glucose DMEM and 0.125% BSA. Lower seeding densities compared to high-density pellet cultures were sufficient to induce in vitro chondrogenic differentiation of hMSCs in collagen scaffolds, which reduces the amount of cells required for the seeding of scaffolds and thus the monolayer expansion period. Furthermore, examination of the impact of FCS and α-MEM on chondrogenic MSC differentiation is an important prerequisite for the development of an osteochondral medium for simultaneous osteogenic and chondrogenic differentiation in biphasic scaffolds for osteochondral tissue regeneration. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- W Pustlauk
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - B Paul
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - S Brueggemeier
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - M Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | - A Bernhardt
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital, and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| |
Collapse
|
88
|
do Amaral RJ, Matsiko A, Tomazette MR, Rocha WK, Cordeiro-Spinetti E, Levingstone TJ, Farina M, O'Brien FJ, El-Cheikh MC, Balduino A. Platelet-rich plasma releasate differently stimulates cellular commitment toward the chondrogenic lineage according to concentration. J Tissue Eng 2015; 6:2041731415594127. [PMID: 26380066 PMCID: PMC4555349 DOI: 10.1177/2041731415594127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/03/2015] [Indexed: 01/22/2023] Open
Abstract
Platelet-rich plasma has been used to treat articular cartilage defects, with the expectations of anabolic and anti-inflammatory effects. However, its role on cellular chondrogenic or fibrogenic commitment is still a controversy. Herein, the role of platelet-rich plasma releasate, the product obtained following platelet-rich plasma activation, on cellular commitment toward the chondrogenic lineage was evaluated in vitro. Human nasoseptal chondrogenic cells and human bone marrow mesenchymal stromal cells were used as cell types already committed to the chondrogenic lineage and undifferentiated cells, respectively, as different concentrations of platelet-rich plasma releasate were tested in comparison to commonly used fetal bovine serum. Low concentration of platelet-rich plasma releasate (2.5%) presented similar effects on cellular growth compared to 10% fetal bovine serum, for both cell types. In a three-dimensional culture system, platelet-rich plasma releasate alone did not induce full nasoseptal chondrogenic cells cartilage-like pellet formation. Nonetheless, platelet-rich plasma releasate played a significant role on cell commitment as high-passage nasoseptal chondrogenic cells only originated cartilage-like pellets when expanded in the presence of platelet-rich plasma releasate rather than fetal bovine serum. Histological analyses and measurements of pellet area demonstrated that even low concentrations of platelet-rich plasma releasate were enough to prevent nasoseptal chondrogenic cells from losing their chondrogenic potential due to in vitro expansion thereby promoting their recommitment. Low concentration of platelet-rich plasma releasate supplemented in chondrogenic medium also increased the chondrogenic potential of mesenchymal stromal cells seeded on collagen-hyaluronic acid scaffolds, as observed by an increase in chondrogenic-related gene expression, sulfated glycosaminoglycan production, and compressive modulus following in vitro culture. On the contrary, higher concentration of platelet-rich plasma releasate (10%) hampered some of these features. In conclusion, platelet-rich plasma releasate was able to prevent cellular chondrogenic capacity loss, inducing regain of their phenotype, and modulate cell commitment. Our data support the hypothesis of platelet-rich plasma chondrogenic potential, allowing fetal bovine serum substitution for platelet-rich plasma releasate at specific concentrations in culture medium when chondrogenic commitment is desired on specific cell types and moments of culture.
Collapse
Affiliation(s)
- Ronaldo Jfc do Amaral
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brasil ; Excellion Serviços Biomédicos, Amil/UnitedHealth Group, Petrópolis, Brasil
| | - Amos Matsiko
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland ; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland ; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Marcel Rp Tomazette
- Laboratório de Biologia e Tecnologia Celular, Universidade Veiga de Almeida, Rio de Janeiro, Brasil
| | - Wanessa Kr Rocha
- Instituto Estadual de Hematologia Arthur de Siqueira Cavalcanti, Rio de Janeiro, Brasil
| | - Eric Cordeiro-Spinetti
- Laboratório de Biologia e Tecnologia Celular, Universidade Veiga de Almeida, Rio de Janeiro, Brasil
| | - Tanya J Levingstone
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland ; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland ; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Marcos Farina
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brasil
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland ; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland ; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Marcia C El-Cheikh
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brasil
| | - Alex Balduino
- Excellion Serviços Biomédicos, Amil/UnitedHealth Group, Petrópolis, Brasil ; Laboratório de Biologia e Tecnologia Celular, Universidade Veiga de Almeida, Rio de Janeiro, Brasil
| |
Collapse
|
89
|
SHAYAN MAHDIS, CHUN YOUNGJAE, LIM WOOCHUL, LEE MINUK, LEE TAEHEE, MIN BYUNGHYUN, LEE DONGGUN. COMPUTATIONAL ANALYSIS OF THE REGENERATED KNEE STRUCTURE AFTER BONE MARROW STIMULATION TECHNIQUES. J MECH MED BIOL 2015. [DOI: 10.1142/s0219519415500360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Bone marrow stimulation techniques, such as abrasion arthroplasty or microfracture, have been widely used for repairing cartilage; however, the mechanical stress analysis of these surgical techniques has not been fully investigated. In this study, finite element analysis was used to investigate stresses produced in complex structures (e.g., cartilage, subchondral bone and trabecular bone) using 2D knee structural models. Abrasion arthroplasty creates global damages only in subchondral bone, but, microfracture technique creates local damages in both trabecular and subchondral regions. Although stresses do not significantly change in trabecular bones as 50% recovery occurs in both abrasion and microfacture samples, significant changes are observed in both subchondral bone and cartilage layer depending on the procedure. The maximum stress levels in the microfractured bone represent approximately a 10.48% increase in cartilage and a 38.25% increase in subchondral bones compared to normal conditions. After 150% recovery, however, all three layers increase their stress levels in microfractured samples. Therefore, the 2D computational analysis results suggest that the microfracture technique should be cautiously used.
Collapse
Affiliation(s)
- MAHDIS SHAYAN
- Department of Industrial Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - YOUNGJAE CHUN
- Department of Industrial Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - WOOCHUL LIM
- School of Mechanical Engineering & Department of Automotive Engineering, Hanyang University, Seoul 133–791, South Korea
| | - MINUK LEE
- School of Mechanical Engineering & Department of Automotive Engineering, Hanyang University, Seoul 133–791, South Korea
| | - TAE HEE LEE
- School of Mechanical Engineering & Department of Automotive Engineering, Hanyang University, Seoul 133–791, South Korea
| | - BYUNG-HYUN MIN
- Department of Orthopedics, Ajou University Medical Center, Suwon, Gyenggi 442–749, South Korea
| | - DONG-GUN LEE
- Department of Energy & Electrical Engineering, Korea Polytechnic University, Siheung, Gyeonggi 429–793, South Korea
| |
Collapse
|
90
|
Particulated Juvenile Articular Cartilage Allograft Transplantation With Bone Marrow Aspirate Concentrate for Treatment of Talus Osteochondral Defects. TECHNIQUES IN FOOT AND ANKLE SURGERY 2015. [DOI: 10.1097/btf.0000000000000092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
91
|
Time evolution of in vivo articular cartilage repair induced by bone marrow stimulation and scaffold implantation in rabbits. Int J Artif Organs 2015; 38:210-23. [PMID: 25952995 DOI: 10.5301/ijao.5000404] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2015] [Indexed: 11/20/2022]
Abstract
PURPOSE Tissue engineering techniques were used to study cartilage repair over a 12-month period in a rabbit model. METHODS A full-depth chondral defect along with subchondral bone injury were originated in the knee joint, where a biostable porous scaffold was implanted, synthesized of poly(ethyl acrylate-co-hydroxyethyl acrylate) copolymer. Morphological evolution of cartilage repair was studied 1 and 2 weeks, and 1, 3, and 12 months after implantation by histological techniques. The 3-month group was chosen to compare cartilage repair to an additional group where scaffolds were preseeded with allogeneic chondrocytes before implantation, and also to controls, who underwent the same surgery procedure, with no scaffold implantation. RESULTS Neotissue growth was first observed in the deepest scaffold pores 1 week after implantation, which spread thereafter; 3 months later scaffold pores were filled mostly with cartilaginous tissue in superficial and middle zones, and with bone tissue adjacent to subchondral bone. Simultaneously, native chondrocytes at the edges of the defect started to proliferate 1 week after implantation; within a month those edges had grown centripetally and seemed to embed the scaffold, and after 3 months, hyaline-like cartilage was observed on the condylar surface. Preseeded scaffolds slightly improved tissue growth, although the quality of repair tissue was similar to non-preseeded scaffolds. Controls showed that fibrous cartilage was mainly filling the repair area 3 months after surgery. In the 12-month group, articular cartilage resembled the untreated surface. CONCLUSIONS Scaffolds guided cartilaginous tissue growth in vivo, suggesting their importance in stress transmission to the cells for cartilage repair.
Collapse
|
92
|
Tower DE, Wood RW, Vaardahl MD. Talocalcaneal Joint Middle Facet Coalition Resection With Interposition of a Juvenile Hyaline Cartilage Graft. J Foot Ankle Surg 2015; 54:1178-82. [PMID: 25922335 DOI: 10.1053/j.jfas.2015.02.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Indexed: 02/03/2023]
Abstract
Talocalcaneal joint middle facet coalition is the most common tarsal coalition, occurring in ≤2% of the population. Fewer than 50% of involved feet obtain lasting relief of symptoms after nonoperative treatment, and surgical intervention is commonly used to relieve symptoms, increase the range of motion, improve function, reconstruct concomitant pes planovalgus, and prevent future arthrosis from occurring at the surrounding joints. Several approaches to surgical intervention are available for patients with middle facet coalitions, ranging from resection to hindfoot arthrodesis. We present a series of 4 cases, in 3 adolescent patients, of talocalcaneal joint middle facet coalition resection with interposition of a particulate juvenile hyaline cartilaginous allograft (DeNovo(®) NT Natural Tissue Graft, Zimmer, Inc., Warsaw, IN). With a mean follow-up period of 42.8 ± 2.9 (range 41 to 47) months, the 3 adolescent patients in the present series were doing well with improved subtalar joint motion and decreased pain, and 1 foot showed no bony regrowth on a follow-up computed tomography scan. The use of a particulate juvenile hyaline cartilaginous allograft as interposition material after talocalcaneal middle facet coalition resection combined with adjunct procedures to address concomitant pes planovalgus resulted in good short-term outcomes in 4 feet in 3 adolescent patients.
Collapse
Affiliation(s)
- Dyane E Tower
- Podiatrist, Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, Lebanon, NH; Assistant Professor of Orthopaedics, Geisel School of Medicine at Dartmouth, Hanover, NH.
| | - Ryan W Wood
- Podiatrist, Coachella Valley Foot and Ankle, Rancho Mirage, CA
| | - Michael D Vaardahl
- Podiatrist, Foot and Ankle Center of Northern Colorado, Greeley, CO; Residency Director, North Colorado Medical Center, Greeley, CO
| |
Collapse
|
93
|
Kučera L, Weinfurterová R, Dvořákova J, Kučera J, Pravda M, Foglarová M, Švík K, Klein P, Velebný V, Kubala L. Chondrocyte Cultivation in Hyaluronan-Tyramine Cross-Linked Hydrogel. INT J POLYM MATER PO 2015. [DOI: 10.1080/00914037.2014.996715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
94
|
Mačiulaitis J, Deveikytė M, Rekštytė S, Bratchikov M, Darinskas A, Šimbelytė A, Daunoras G, Laurinavičienė A, Laurinavičius A, Gudas R, Malinauskas M, Mačiulaitis R. Preclinical study of SZ2080 material 3D microstructured scaffolds for cartilage tissue engineering made by femtosecond direct laser writing lithography. Biofabrication 2015; 7:015015. [PMID: 25797444 DOI: 10.1088/1758-5090/7/1/015015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Over the last decade DLW employing ultrafast pulsed lasers has become a well-established technique for the creation of custom-made free-form three-dimensional (3D) microscaffolds out of a variety of materials ranging from proteins to biocompatible glasses. Its potential applications for manufacturing a patient's specific scaffold seem unlimited in terms of spatial resolution and geometry complexity. However, despite few exceptions in which live cells or primitive organisms were encapsulated into a polymer matrix, no demonstration of an in vivo study case of scaffolds generated with the use of such a method was performed. Here, we report a preclinical study of 3D artificial microstructured scaffolds out of hybrid organic-inorganic (HOI) material SZ2080 fabricated using the DLW technique. The created 2.1 × 2.1 × 0.21 mm(3) membrane constructs are tested both in vitro by growing isolated allogeneic rabbit chondrocytes (Cho) and in vivo by implanting them into rabbit organisms for one, three and six months. An ex vivo histological examination shows that certain pore geometry and the pre-growing of Cho prior to implantation significantly improves the performance of the created 3D scaffolds. The achieved biocompatibility is comparable to the commercially available collagen membranes. The successful outcome of this study supports the idea that hexagonal-pore-shaped HOI microstructured scaffolds in combination with Cho seeding may be successfully implemented for cartilage tissue engineering.
Collapse
Affiliation(s)
- Justinas Mačiulaitis
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian Health Science University, Mickevičiaus 9, LT 44307 Kaunas, Lithuania. Institute of Sports, Medical Academy, Lithuanian University of Health Science, Kalniečių 231, LT 44307 Kaunas, Lithuania. Orthopaedic and Trauma Department, Lithuanian Health Science University, Mickevičiaus 9, LT 44307 Kaunas, Lithuania
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Wei B, Yao Q, Guo Y, Mao F, Liu S, Xu Y, Wang L. Three-dimensional polycaprolactone-hydroxyapatite scaffolds combined with bone marrow cells for cartilage tissue engineering. J Biomater Appl 2015; 30:160-70. [PMID: 25766036 DOI: 10.1177/0885328215575762] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The goal of this study was to investigate the chondrogenic potential of three-dimensional polycaprolactone-hydroxyapatite (PCL-HA) scaffolds loaded with bone marrow cells in vitro and the effect of PCL-HA scaffolds on osteochondral repair in vivo. Here, bone marrow was added to the prepared PCL-HA scaffolds and cultured in chondrogenic medium for 10 weeks. Osteochondral defects were created in the trochlear groove of 29 knees in 17 New Zealand white rabbits, which were then divided into four groups that underwent: implantation of PCL-HA scaffolds (left knee, n = 17; Group 1), microfracture (right knee, n = 6; Group 2), autologous osteochondral transplantation (right knee, n = 6; Group 3), and no treatment (right knee, n = 5; Control). Extracellular matrix produced by bone marrow cells covered the surface and filled the pores of PCL-HA scaffolds after 10 weeks in culture. Moreover, many cell-laden cartilage lacunae were observed, and cartilage matrix was concentrated in the PCL-HA scaffolds. After a 12-week repair period, Group 1 showed excellent vertical and lateral integration with host bone, but incomplete cartilage regeneration and matrix accumulation. An uneven surface of regenerated cartilage and reduced distribution of cartilage matrix were observed in Group 2. In addition, abnormal bone growth and unstable integration between repaired and host tissues were detected. For Group 3, the integration between transplanted and host cartilage was interrupted. Our findings indicate that the PCL-HA scaffolds loaded with bone marrow cells improved chondrogenesis in vitro and implantation of PCL-HA scaffolds for osteochondral repairenhanced integration with host bone. However, cartilage regeneration remained unsatisfactory. The addition of trophic factors or the use of precultured cell-PCL-HA constructs for accelerated osteochondral repair requires further investigation.
Collapse
Affiliation(s)
- Bo Wei
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yang Guo
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Fengyong Mao
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shuai Liu
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Xu
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liming Wang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
96
|
Lopa S, Piraino F, Kemp RJ, Di Caro C, Lovati AB, Di Giancamillo A, Moroni L, Peretti GM, Rasponi M, Moretti M. Fabrication of multi-well chips for spheroid cultures and implantable constructs through rapid prototyping techniques. Biotechnol Bioeng 2015; 112:1457-71. [PMID: 25678107 DOI: 10.1002/bit.25557] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 01/29/2023]
Abstract
Three-dimensional (3D) culture models are widely used in basic and translational research. In this study, to generate and culture multiple 3D cell spheroids, we exploited laser ablation and replica molding for the fabrication of polydimethylsiloxane (PDMS) multi-well chips, which were validated using articular chondrocytes (ACs). Multi-well ACs spheroids were comparable or superior to standard spheroids, as revealed by glycosaminoglycan and type-II collagen deposition. Moreover, the use of our multi-well chips significantly reduced the operation time for cell seeding and medium refresh. Exploiting a similar approach, we used clinical-grade fibrin to generate implantable multi-well constructs allowing for the precise distribution of multiple cell types. Multi-well fibrin constructs were seeded with ACs generating high cell density regions, as shown by histology and cell fluorescent staining. Multi-well constructs were compared to standard constructs with homogeneously distributed ACs. After 7 days in vitro, expression of SOX9, ACAN, COL2A1, and COMP was increased in both constructs, with multi-well constructs expressing significantly higher levels of chondrogenic genes than standard constructs. After 5 weeks in vivo, we found that despite a dramatic size reduction, the cell distribution pattern was maintained and glycosaminoglycan content per wet weight was significantly increased respect to pre-implantation samples. In conclusion, multi-well chips for the generation and culture of multiple cell spheroids can be fabricated by low-cost rapid prototyping techniques. Furthermore, these techniques can be used to generate implantable constructs with defined architecture and controlled cell distribution, allowing for in vitro and in vivo investigation of cell interactions in a 3D environment.
Collapse
Affiliation(s)
- Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Via R. Galeazzi 4, 20161, Milan, Italy
| | - Francesco Piraino
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, 20133, Italy
| | - Raymond J Kemp
- Tissue Regeneration Department, University of Twente, 7522 NB, Enschede, The Netherlands
| | - Clelia Di Caro
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, 20133, Italy
| | - Arianna B Lovati
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Via R. Galeazzi 4, 20161, Milan, Italy
| | | | - Lorenzo Moroni
- Tissue Regeneration Department, University of Twente, 7522 NB, Enschede, The Netherlands
- Department of Complex Tissue Regeneration, Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Giuseppe M Peretti
- IRCCS Galeazzi Orthopaedic Institute, Milan, 20161, Italy
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, 20161, Italy
| | - Marco Rasponi
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, 20133, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Via R. Galeazzi 4, 20161, Milan, Italy.
| |
Collapse
|
97
|
Rivera AL, Baskaran H. The Effect of Biomolecular Gradients on Mesenchymal Stem Cell Chondrogenesis under Shear Stress. MICROMACHINES 2015; 6:330-346. [PMID: 34026281 PMCID: PMC8138782 DOI: 10.3390/mi6030330] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tissue engineering is viewed as a promising option for long-term repair of cartilage lesions, but current engineered cartilage constructs fail to match the mechanical properties of native tissue. The extracellular matrix of adult human articular cartilage contains highly organized collagen fibrils that enhance the mechanical properties of the tissue. Unlike articular cartilage, mesenchymal stem cell (MSC) based tissue engineered cartilage constructs lack this oriented microstructure and therefore display much lower mechanical strength. The goal of this study was to investigate the effect of biomolecular gradients and shear stress on MSCs undergoing chondrogenesis within a microfluidic device. Via poly(dimethyl siloxane) soft-lithography, microfluidic devices containing a gradient generator were created. Human MSCs were seeded within these chambers and exposed to flow-based transforming growth factor β1 (TGF-β1) gradients. When the MSCs were both confluent and exposed to shear stress, the cells aligned along the flow direction. Exposure to TGF-β1 gradients led to chondrogenesis of MSCs, indicated by positive type II collagen staining. These results, together with a previous study that showed that aligned MSCs produce aligned collagen, suggest that oriented cartilage tissue structures with superior mechanical properties can be obtained by aligning MSCs along the flow direction and exposing MSCs to chondrogenic gradients.
Collapse
Affiliation(s)
- Alexander L. Rivera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-216-368-1029; Fax: +1-216-368-3016
| |
Collapse
|
98
|
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
|
99
|
Hunziker EB, Lippuner K, Keel MJB, Shintani N. An educational review of cartilage repair: precepts & practice--myths & misconceptions--progress & prospects. Osteoarthritis Cartilage 2015; 23:334-50. [PMID: 25534362 DOI: 10.1016/j.joca.2014.12.011] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 10/29/2014] [Accepted: 12/12/2014] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The repair of cartilaginous lesions within synovial joints is still an unresolved and weighty clinical problem. Although research activity in this area has been indefatigably sustained, no significant progress has been made during the past decade. The aim of this educational review is to heighten the awareness amongst students and scientists of the basic issues that must be tackled and resolved before we can hope to escape from the whirlpool of stagnation into which we have fallen: cartilage repair redivivus! DESIGN Articular-cartilage lesions may be induced traumatically (e.g., by sports injuries and occupational accidents) or pathologically during the course of a degenerative disease (e.g., osteoarthritis). This review addresses the biological basis of cartilage repair and surveys current trends in treatment strategies, focussing on those that are most widely adopted by orthopaedic surgeons [viz., abrasive chondroplasty, microfracturing/microdrilling, osteochondral grafting and autologous-chondrocyte implantation (ACI)]. Also described are current research activities in the field of cartilage-tissue engineering, which, as a therapeutic principle, holds more promise for success than any other experimental approach. RESULTS AND CONCLUSIONS Tissue engineering aims to reconstitute a tissue both structurally and functionally. This process can be conducted entirely in vitro, initially in vitro and then in vivo (in situ), or entirely in vivo. Three key constituents usually form the building blocks of such an approach: a matrix scaffold, cells, and signalling molecules. Of the proposed approaches, none have yet advanced beyond the phase of experimental development to the level of clinical induction. The hurdles that need to be surmounted for ultimate success are discussed.
Collapse
Affiliation(s)
- E B Hunziker
- Departments of Osteoporosis, Orthopaedic Surgery and Clinical Research, Inselspital, University of Bern, Bern, Switzerland.
| | - K Lippuner
- Departments of Osteoporosis, Orthopaedic Surgery and Clinical Research, Inselspital, University of Bern, Bern, Switzerland.
| | - M J B Keel
- Departments of Osteoporosis, Orthopaedic Surgery and Clinical Research, Inselspital, University of Bern, Bern, Switzerland.
| | - N Shintani
- Departments of Osteoporosis, Orthopaedic Surgery and Clinical Research, Inselspital, University of Bern, Bern, Switzerland.
| |
Collapse
|
100
|
Ren CD, Gao S, Kurisawa M, Ying JY. Cartilage synthesis in hyaluronic acid-tyramine constructs. J Mater Chem B 2015; 3:1942-1956. [PMID: 32262266 DOI: 10.1039/c4tb01229a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The objective of this study was to determine the potential for cartilage production within a hyaluronic acid-tyramine (HA-Tyr) hydrogel scaffold. Chondrocytes were encapsulated within HA-Tyr hydrogels and subcutaneously implanted in mice. The HA-Tyr hydrogels were formed by the oxidative coupling of Tyr moieties catalyzed by hydrogen peroxide (H2O2) and horseradish peroxidase (HRP). Harvested constructs were shown to achieve a glycosaminoglycan (GAG) content of 1.2 wt%, and they demonstrated 40% of the collagen content of normal articular cartilage, including the presence of Type II collagen, which is the characteristic of articular cartilage. Matrix production was found to be influenced by the initial cell density, scaffold degradation rate and Type II collagen concentration. Injectability was also imparted to the system by delivering HRP through thermoresponsive liposomes. The method of HRP delivery, either by simple addition or through thermoresponsive liposomes, was not shown to have an effect on matrix production.
Collapse
Affiliation(s)
- Cindy D Ren
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669.
| | | | | | | |
Collapse
|