101
|
Kim J, Lin B, Kim S, Choi B, Evseenko D, Lee M. TGF-β1 conjugated chitosan collagen hydrogels induce chondrogenic differentiation of human synovium-derived stem cells. J Biol Eng 2015; 9:1. [PMID: 25745515 PMCID: PMC4350967 DOI: 10.1186/1754-1611-9-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/24/2014] [Indexed: 12/27/2022] Open
Abstract
Background Unlike bone tissue, articular cartilage regeneration has not been very successful and has many challenges ahead. We have previously developed injectable hydrogels using photopolymerizable chitosan (MeGC) that supported growth of chondrocytes. In this study, we demonstrate a biofunctional hydrogel for specific use in cartilage regeneration by conjugating transforming growth factor-β1 (TGF-β1), a well-documented chondrogenic factor, to MeGC hydrogels impregnating type II collagen (Col II), one of the major cartilaginous extracellular matrix (ECM) components. Results TGF-β1 was delivered from MeGC hydrogels in a controlled manner with reduced burst release by chemically conjugating the protein to MeGC. The hydrogel system did not compromise viability of encapsulated human synovium-derived mesenchymal stem cells (hSMSCs). Col II impregnation and TGF-β1 delivery significantly enhanced cellular aggregation and deposition of cartilaginous ECM by the encapsulated cells, compared with pure MeGC hydrogels. Conclusions This study demonstrates successful engineering of a biofunctional hydrogel with a specific microenvironment tailored to promote chondrogenesis. This hydrogel system can provide promising efficacious therapeutics in the treatment of cartilage defects. Electronic supplementary material The online version of this article (doi:10.1186/1754-1611-9-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinku Kim
- Department of Bio and Chemical Engineering, Hongik University, Sejong, 339-701 South Korea
| | - Brian Lin
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA
| | - Soyon Kim
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Bogyu Choi
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095 USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA ; Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
102
|
Drissi H, Gibson JD, Guzzo RM, Xu RH. Derivation and Chondrogenic Commitment of Human Embryonic Stem Cell-Derived Mesenchymal Progenitors. Methods Mol Biol 2015; 1340:65-78. [PMID: 26445831 DOI: 10.1007/978-1-4939-2938-2_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The induction of human embryonic stem cells to a mesenchymal-like progenitor population constitutes a developmentally relevant approach for efficient directed differentiation of human embryonic stem (hES) cells to the chondrogenic lineage. The initial enrichment of a hemangioblast intermediate has been shown to yield a replenishable population of highly purified progenitor cells that exhibit the typical mesenchymal stem cell (MSC) surface markers as well as the capacity for multilineage differentiation to bone, fat, and cartilage. Herein, we provide detailed methodologies for the derivation and characterization of potent mesenchymal-like progenitors from hES cells and describe in vitro assays for bone morphogenetic protein (BMP)-2-mediated differentiation to the chondrogenic lineage.
Collapse
Affiliation(s)
- Hicham Drissi
- Department of Orthopaedic Surgery, UConn Health, 263 Farmington Avenue, Farmington, CT, 06030-4037, USA.
- Department of Genetics and Genome Sciences, UConn Health, 400 Farmington Avenue, Farmington, CT, 06030-6403, USA.
- Stem Cell Institute, UConn Health, 400 Farmington Avenue, Farmington, CT, 06030-6403, USA.
| | - Jason D Gibson
- Department of Orthopaedic Surgery, UConn Health, 263 Farmington Avenue, Farmington, CT, 06030-4037, USA
| | - Rosa M Guzzo
- Department of Orthopaedic Surgery, UConn Health, 263 Farmington Avenue, Farmington, CT, 06030-4037, USA
- Stem Cell Institute, UConn Health, 400 Farmington Avenue, Farmington, CT, 06030-6403, USA
| | - Ren-He Xu
- Department of Genetics and Genome Sciences, UConn Health, 400 Farmington Avenue, Farmington, CT, 06030-6403, USA
- Stem Cell Institute, UConn Health, 400 Farmington Avenue, Farmington, CT, 06030-6403, USA
| |
Collapse
|
103
|
Visible-light-initiated hydrogels preserving cartilage extracellular signaling for inducing chondrogenesis of mesenchymal stem cells. Acta Biomater 2015; 12:30-41. [PMID: 25462526 DOI: 10.1016/j.actbio.2014.10.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/10/2014] [Accepted: 10/13/2014] [Indexed: 11/23/2022]
Abstract
Hydrogels have a unique opportunity to regenerate damaged cartilage tissues by introducing mesenchymal stem cells (MSCs) in a highly swollen environment similar to articular cartilage. During cartilage development, collagen-cell interactions play an important role in mediating early mesenchymal condensation and chondrogenesis with transforming growth factor-β1 (TGF-β1) stimulation. Here, a hydrogel environment that can enhance cell-matrix interactions and chondrogenesis by stabilizing type-II collagen (Col II) and TGF-β1 into photopolymerizable (methacrylated) chitosan (MeGC) with simple entrapment and affinity binding is demonstrated. The MeGC hydrogel was designed to gel upon initiation by exposure to visible blue light in the presence of riboflavin, an aqueous initiator from natural vitamin. The incorporation of Col II into MeGC hydrogels increased cellular condensation and deposition of cartilaginous extracellular matrix by encapsulated chondrocytes. MeGC hydrogels containing Col II supported the release of TGF-β1 in a controlled manner over time in chondrogenic medium and the incorporated TGF-β1 further enhanced chondrogenesis of encapsulated chondrocytes and MSCs, especially synovial MSCs. Subcutaneous implantation of hydrogel cultures showed greatly improved neocartilage formation in constructs loaded with TGF-β1 compared with controls. These findings suggest that cartilage mimetic hydrogels have a high potential for cartilage repair.
Collapse
|
104
|
Articular cartilage repair using marrow stimulation augmented with a viable chondral allograft: 9-month postoperative histological evaluation. Case Rep Orthop 2015; 2015:617365. [PMID: 25628907 PMCID: PMC4299361 DOI: 10.1155/2015/617365] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/19/2014] [Indexed: 01/09/2023] Open
Abstract
Marrow stimulation is frequently employed to treat focal chondral defects of the knee. However, marrow stimulation typically results in fibrocartilage repair tissue rather than healthy hyaline cartilage, which, over time, predisposes the repair to failure. Recently, a cryopreserved viable chondral allograft was developed to augment marrow stimulation. The chondral allograft is comprised of native viable chondrocytes, chondrogenic growth factors, and extracellular matrix proteins within the superficial, transitional, and radial zones of hyaline cartilage. Therefore, host mesenchymal stem cells that infiltrate the graft from the underlying bone marrow following marrow stimulation are provided with the optimal microenvironment to undergo chondrogenesis. The present report describes treatment of a trochlear defect with marrow stimulation augmented with this novel chondral allograft, along with nine month postoperative histological results. At nine months, the patient demonstrated complete resolution of pain and improvement in function, and the repair tissue consisted of 85% hyaline cartilage. For comparison, a biopsy obtained from a patient 8.2 months after treatment with marrow stimulation alone contained only 5% hyaline cartilage. These outcomes suggest that augmenting marrow stimulation with the viable chondral allograft can eliminate pain and improve outcomes, compared with marrow stimulation alone.
Collapse
|
105
|
Wei B, Guo Y, Xu Y, Mao F, Yao Q, Jin C, Gu Q, Wang L. Composite scaffolds composed of bone marrow mesenchymal stem cell-derived extracellular matrix and marrow clots promote marrow cell retention and proliferation. J Biomed Mater Res A 2014; 103:2374-82. [PMID: 25410417 DOI: 10.1002/jbm.a.35373] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/03/2014] [Accepted: 11/17/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Bo Wei
- Department of Orthopaedic Surgery; Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
| | - Yang Guo
- Department of Orthopaedic Surgery; Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
| | - Yan Xu
- Department of Orthopaedic Surgery; Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
| | - Fengyong Mao
- Department of Orthopaedic Surgery; Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery; Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
| | - Chengzhe Jin
- Department of Orthopaedic Surgery; Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
| | - Qiangrong Gu
- Department of Orthopaedic Surgery; Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
| | - Liming Wang
- Department of Orthopaedic Surgery; Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
- China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University; Nanjing 210006 People's Republic of China
| |
Collapse
|
106
|
Colombini A, Ceriani C, Banfi G, Brayda-Bruno M, Moretti M. Fibrin in Intervertebral Disc Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:713-21. [DOI: 10.1089/ten.teb.2014.0158] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Alessandra Colombini
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Cristina Ceriani
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Giuseppe Banfi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Marco Brayda-Bruno
- Department of Orthopedics and Traumatology–Vertebral Surgery III–Scoliosis, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| |
Collapse
|
107
|
Florine EM, Miller RE, Liebesny PH, Mroszczyk KA, Lee RT, Patwari P, Grodzinsky AJ. Delivering heparin-binding insulin-like growth factor 1 with self-assembling peptide hydrogels. Tissue Eng Part A 2014; 21:637-46. [PMID: 25231349 DOI: 10.1089/ten.tea.2013.0679] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Heparin-binding insulin-like growth factor 1 (HB-IGF-1) is a fusion protein of IGF-1 with the HB domain of heparin-binding epidermal growth factor-like growth factor. A single dose of HB-IGF-1 has been shown to bind specifically to cartilage and to promote sustained upregulation of proteoglycan synthesis in cartilage explants. Achieving strong integration between native cartilage and tissue-engineered cartilage remains challenging. We hypothesize that if a growth factor delivered by the tissue engineering scaffold could stimulate enhanced matrix synthesis by both the cells within the scaffold and the adjacent native cartilage, integration could be enhanced. In this work, we investigated methods for adsorbing HB-IGF-1 to self-assembling peptide hydrogels to deliver the growth factor to encapsulated chondrocytes and cartilage explants cultured with growth factor-loaded hydrogels. We tested multiple methods for adsorbing HB-IGF-1 in self-assembling peptide hydrogels, including adsorption prior to peptide assembly, following peptide assembly, and with/without heparan sulfate (HS, a potential linker between peptide molecules and HB-IGF-1). We found that HB-IGF-1 and HS were retained in the peptide for all tested conditions. A subset of these conditions was then studied for their ability to stimulate increased matrix production by gel-encapsulated chondrocytes and by chondrocytes within adjacent native cartilage. Adsorbing HB-IGF-1 or IGF-1 prior to peptide assembly was found to stimulate increased sulfated glycosaminoglycan per DNA and hydroxyproline content of chondrocyte-seeded hydrogels compared with basal controls at day 10. Cartilage explants cultured adjacent to functionalized hydrogels had increased proteoglycan synthesis at day 10 when HB-IGF-1 was adsorbed, but not IGF-1. We conclude that delivery of HB-IGF-1 to focal defects in cartilage using self-assembling peptide hydrogels is a promising technique that could aid cartilage repair via enhanced matrix production and integration with native tissue.
Collapse
Affiliation(s)
- Emily M Florine
- 1 Biological Engineering, Massachusetts Institute of Technology , Cambridge, Massachusetts
| | | | | | | | | | | | | |
Collapse
|
108
|
Giza E, Delman C, Coetzee JC, Schon LC. Arthroscopic treatment of talus osteochondral lesions with particulated juvenile allograft cartilage. Foot Ankle Int 2014; 35:1087-94. [PMID: 25223301 DOI: 10.1177/1071100714548704] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
UNLABELLED Osteochondral lesions of the talus (OLT) are commonly associated with traumatic injury to the ankle joint. Treatment options depend on the grade, location, and size of the lesion. Operative intervention is frequently required with initial management involving marrow stimulation techniques, such as microfracture/curettage. Larger lesions often require a secondary procedure, such as osteochondral transplantation or autologous chondrocyte implantation. The advent of particulated juvenile articular cartilage (PJAC) provides an alternative method for OLTs refractory to traditional treatments. This article describes the technique of PJAC transplantation for the treatment of osteochondral lesions of the talus. LEVEL OF EVIDENCE Level V, expert opinion.
Collapse
Affiliation(s)
- Eric Giza
- Department of Orthopaedics, University of California, Davis, Sacramento, CA, USA
| | - Connor Delman
- Department of Orthopaedics, University of California, Davis, Sacramento, CA, USA
| | - J Chris Coetzee
- Minnesota Orthopedic Sports Medicine Institute, a division of Twin Cities Orthopedics, Eden Prairie, MN, USA
| | | |
Collapse
|
109
|
Li JJ, Kaplan DL, Zreiqat H. Scaffold-based regeneration of skeletal tissues to meet clinical challenges. J Mater Chem B 2014; 2:7272-7306. [PMID: 32261954 DOI: 10.1039/c4tb01073f] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The management and reconstruction of damaged or diseased skeletal tissues have remained a significant global healthcare challenge. The limited efficacy of conventional treatment strategies for large bone, cartilage and osteochondral defects has inspired the development of scaffold-based tissue engineering solutions, with the aim of achieving complete biological and functional restoration of the affected tissue in the presence of a supporting matrix. Nevertheless, significant regulatory hurdles have rendered the clinical translation of novel scaffold designs to be an inefficient process, mainly due to the difficulties of arriving at a simple, reproducible and effective solution that does not rely on the incorporation of cells and/or bioactive molecules. In the context of the current clinical situation and recent research advances, this review will discuss scaffold-based strategies for the regeneration of skeletal tissues, with focus on the contribution of bioactive ceramic scaffolds and silk fibroin, and combinations thereof, towards the development of clinically viable solutions.
Collapse
Affiliation(s)
- Jiao Jiao Li
- Biomaterials and Tissue Engineering Research Unit, School of AMME, University of Sydney, Sydney, NSW 2006, Australia.
| | | | | |
Collapse
|
110
|
Zhu S, Chen P, Wu Y, Xiong S, Sun H, Xia Q, Shi L, Liu H, Ouyang HW. Programmed Application of Transforming Growth Factor β3 and Rac1 Inhibitor NSC23766 Committed Hyaline Cartilage Differentiation of Adipose-Derived Stem Cells for Osteochondral Defect Repair. Stem Cells Transl Med 2014; 3:1242-51. [PMID: 25154784 DOI: 10.5966/sctm.2014-0042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hyaline cartilage differentiation is always the challenge with application of stem cells for joint repair. Transforming growth factors (TGFs) and bone morphogenetic proteins can initiate cartilage differentiation but often lead to hypertrophy and calcification, related to abnormal Rac1 activity. In this study, we developed a strategy of programmed application of TGFβ3 and Rac1 inhibitor NSC23766 to commit the hyaline cartilage differentiation of adipose-derived stem cells (ADSCs) for joint cartilage repair. ADSCs were isolated and cultured in a micromass and pellet culture model to evaluate chondrogenic and hypertrophic differentiation. The function of Rac1 was investigated with constitutively active Rac1 mutant and dominant negative Rac1 mutant. The efficacy of ADSCs with programmed application of TGFβ3 and Rac1 inhibitor for cartilage repair was studied in a rat model of osteochondral defects. The results showed that TGFβ3 promoted ADSCs chondro-lineage differentiation and that NSC23766 prevented ADSC-derived chondrocytes from hypertrophy in vitro. The combination of ADSCs, TGFβ3, and NSC23766 promoted quality osteochondral defect repair in rats with much less chondrocytes hypertrophy and significantly higher International Cartilage Repair Society macroscopic and microscopic scores. The findings have illustrated that programmed application of TGFβ3 and Rac1 inhibitor NSC23766 can commit ADSCs to chondro-lineage differentiation and improve the efficacy of ADSCs for cartilage defect repair. These findings suggest a promising stem cell-based strategy for articular cartilage repair.
Collapse
Affiliation(s)
- Shouan Zhu
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Pengfei Chen
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yan Wu
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Si Xiong
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Heng Sun
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qingqing Xia
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Libing Shi
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Huanhuan Liu
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Hong Wei Ouyang
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
111
|
Designer functionalised self-assembling peptide nanofibre scaffolds for cartilage tissue engineering. Expert Rev Mol Med 2014; 16:e12. [PMID: 25089851 DOI: 10.1017/erm.2014.13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Owing to the limited regenerative capacity of cartilage tissue, cartilage repair remains a challenge in clinical treatment. Tissue engineering has emerged as a promising and important approach to repair cartilage defects. It is well known that material scaffolds are regarded as a fundamental element of tissue engineering. Novel biomaterial scaffolds formed by self-assembling peptides consist of nanofibre networks highly resembling natural extracellular matrices, and their fabrication is based on the principle of molecular self-assembly. Indeed, peptide nanofibre scaffolds have obtained much progress in repairing various damaged tissues (e.g. cartilage, bone, nerve, heart and blood vessel). This review outlines the rational design of peptide nanofibre scaffolds and their potential in cartilage tissue engineering.
Collapse
|
112
|
Man Z, Yin L, Shao Z, Zhang X, Hu X, Zhu J, Dai L, Huang H, Yuan L, Zhou C, Chen H, Ao Y. The effects of co-delivery of BMSC-affinity peptide and rhTGF-β1 from coaxial electrospun scaffolds on chondrogenic differentiation. Biomaterials 2014; 35:5250-60. [DOI: 10.1016/j.biomaterials.2014.03.031] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 03/14/2014] [Indexed: 01/03/2023]
|
113
|
Potier E, Rivron NC, Van Blitterswijk CA, Ito K. Micro-aggregates do not influence bone marrow stromal cell chondrogenesis. J Tissue Eng Regen Med 2014; 10:1021-1032. [PMID: 24700552 DOI: 10.1002/term.1887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 11/04/2013] [Accepted: 02/24/2014] [Indexed: 12/31/2022]
Abstract
Although bone marrow stromal cells (BMSCs) appear promising for cartilage repair, current clinical results are suboptimal and the success of BMSC-based therapies relies on a number of methodological improvements, among which is better understanding and control of their differentiation pathways. We investigated here the role of the cellular environment (paracrine vs juxtacrine signalling) in the chondrogenic differentiation of BMSCs. Bovine BMSCs were encapsulated in alginate beads, as dispersed cells or as small micro-aggregates, to create different paracrine and juxtacrine signalling conditions. BMSCs were then cultured for 21 days with TGFβ3 added for 0, 7 or 21 days. Chondrogenic differentiation was assessed at the gene (type II and X collagens, aggrecan, TGFβ, sp7) and matrix (biochemical assays and histology) levels. The results showed that micro-aggregates had no beneficial effects over dispersed cells: matrix production was similar, whereas chondrogenic marker gene expression was lower for the micro-aggregates, under all TGFβ conditions tested. This weakened chondrogenic differentiation might be explained by a different cytoskeleton organization at day 0 in the micro-aggregates. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- E Potier
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - N C Rivron
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - C A Van Blitterswijk
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - K Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
114
|
Frappier J, Stanish W, Brittberg M, Steinwachs M, Crowe L, Castelo D, Restrepo A. Economic evaluation of BST-CarGel as an adjunct to microfracture vs microfracture alone in knee cartilage surgery. J Med Econ 2014; 17:266-78. [PMID: 24601747 DOI: 10.3111/13696998.2014.897626] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVES Knee cartilage damage is a common cause of referral for orthopedic surgery. Treatment aims to reduce pain and symptoms by repairing cartilage. Microfracture, the current standard of care, yields good short-term clinical outcomes; however, treatment might fail after 2-3 years. A Chitosan-Beta glycerolphosphate-based medical device (BST-CarGel) is used as an adjunct to microfracture and demonstrates improvements in quantity and quality of repaired tissue, potentially reducing the risk of treatment failure. This study aimed to establish the economic value of BST-CarGel vs microfracture alone in knee cartilage repair from the societal perspective, using Germany as the reference market. METHODS A decision tree with a 20-year time-horizon was constructed, in which undesirable clinical events were inferred following initial surgery. These events consisted of pain management, surgery, and total knee replacement. Clinical outcomes were taken from the pivotal clinical trial, supplemented by other literature. Data and assumptions were validated by a Delphi panel. All relevant resource use and costs for procedures and events were considered. RESULTS In a group of patients with all lesion sizes, the model inferred that BST-CarGel yields a positive return on investment at year 4 (with 20-year cumulative cost savings of €6448). Reducing the incremental risk of treatment failure gap between the device and microfracture by 25-50% does not alter this conclusion. Cost savings are greatest for patients with large lesions; results for patients with small lesions are more modest. LIMITATIONS Clinical evidence for microfracture and other interventions varies in quality. Comparative long-term data are lacking. The comparison is limited to microfracture and looks only at costs without considering quality-of-life. CONCLUSION BST-CarGel potentially represents a cost-saving alternative for patients with knee cartilage injury by reducing the risk of clinical events through regeneration of chondral tissue with hyaline characteristics. Since the burden of this condition is high, both to the patient and society, an effective and economically viable alternative is of importance.
Collapse
|
115
|
Haaparanta AM, Järvinen E, Cengiz IF, Ellä V, Kokkonen HT, Kiviranta I, Kellomäki M. Preparation and characterization of collagen/PLA, chitosan/PLA, and collagen/chitosan/PLA hybrid scaffolds for cartilage tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:1129-1136. [PMID: 24375147 DOI: 10.1007/s10856-013-5129-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 12/15/2013] [Indexed: 06/03/2023]
Abstract
In this study, three-dimensional (3D) porous scaffolds were developed for the repair of articular cartilage defects. Novel collagen/polylactide (PLA), chitosan/PLA, and collagen/chitosan/PLA hybrid scaffolds were fabricated by combining freeze-dried natural components and synthetic PLA mesh, where the 3D PLA mesh gives mechanical strength, and the natural polymers, collagen and/or chitosan, mimic the natural cartilage tissue environment of chondrocytes. In total, eight scaffold types were studied: four hybrid structures containing collagen and/or chitosan with PLA, and four parallel plain scaffolds with only collagen and/or chitosan. The potential of these types of scaffolds for cartilage tissue engineering applications were determined by the analysis of the microstructure, water uptake, mechanical strength, and the viability and attachment of adult bovine chondrocytes to the scaffolds. The manufacturing method used was found to be applicable for the manufacturing of hybrid scaffolds with highly porous 3D structures. All the hybrid scaffolds showed a highly porous structure with open pores throughout the scaffold. Collagen was found to bind water inside the structure in all collagen-containing scaffolds better than the chitosan-containing scaffolds, and the plain collagen scaffolds had the highest water absorption. The stiffness of the scaffold was improved by the hybrid structure compared to plain scaffolds. The cell viability and attachment was good in all scaffolds, however, the collagen hybrid scaffolds showed the best penetration of cells into the scaffold. Our results show that from the studied scaffolds the collagen/PLA hybrids are the most promising scaffolds from this group for cartilage tissue engineering.
Collapse
Affiliation(s)
- Anne-Marie Haaparanta
- Department of Electronics and Communications Engineering, Tampere University of Technology, Korkeakoulunkatu 3, 33720, Tampere, Finland,
| | | | | | | | | | | | | |
Collapse
|
116
|
Zhang W, Heng BC, Jiang YZ, Ouyang HW. Clinical translation of autologous cell-based tissue engineering techniques as Class III therapeutics in China: Taking cartilage tissue engineering as an example. J Orthop Translat 2014. [DOI: 10.1016/j.jot.2014.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
117
|
Wang LS, Du C, Toh WS, Wan AC, Gao SJ, Kurisawa M. Modulation of chondrocyte functions and stiffness-dependent cartilage repair using an injectable enzymatically crosslinked hydrogel with tunable mechanical properties. Biomaterials 2014; 35:2207-17. [DOI: 10.1016/j.biomaterials.2013.11.070] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/22/2013] [Indexed: 12/25/2022]
|
118
|
Bianchera A, Salomi E, Pezzanera M, Ruwet E, Bettini R, Elviri L. Chitosan hydrogels for chondroitin sulphate controlled release: an analytical characterization. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2014; 2014:808703. [PMID: 25614850 PMCID: PMC4295592 DOI: 10.1155/2014/808703] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/11/2014] [Accepted: 12/15/2014] [Indexed: 05/04/2023]
Abstract
This paper provides an analytical characterization of chitosan scaffolds obtained by freeze-gelation toward the uptake and the controlled release of chondroitin sulphate (CS), as cartilage repair agent, under different pH conditions. Scanning electron microscopy (SEM), attenuated total reflectance-Fourier transform infrared spectroscopy (ATR-FTIR), and liquid chromatography-UV spectrophotometry (LC-UV) techniques were exploited to obtain qualitative and quantitative descriptions of polymer and drug behaviour in the biomaterial. As for morphology, SEM analysis allowed the evaluation of scaffold porosity in terms of pore size and distribution both at the surface (Feret diameter 58 ± 19 μm) and on the cross section (Feret diameter 106 ± 51 μm). LC and ATR-FTIR evidenced a pH-dependent CS loading and release behaviour, strongly highlighting the role of electrostatic forces on chitosan/chondroitin sulphate interactions.
Collapse
Affiliation(s)
- Annalisa Bianchera
- Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Enrico Salomi
- Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Matteo Pezzanera
- Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Elisabeth Ruwet
- Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Ruggero Bettini
- Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Lisa Elviri
- Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
- *Lisa Elviri:
| |
Collapse
|
119
|
Abstract
Biomaterials are playing a vital role in our day-to-day life. Hyaluronan (hyaluronic acid), a biomaterial, receives special attention among them. Hyaluronic acid (HA) is a polyanionic natural polymer occurring as linear polysaccharide composed of glucuronic acid and N-acetylglucosamine repeats via a β-1,4 linkage. It is the most versatile macromolecule present in the connective tissues of all vertebrates. Hyaluronic acid has a wide range of applications with its excellent physicochemical properties such as biodegradability, biocompatibility, nontoxicity, and nonimmunogenicity and serves as an excellent tool in biomedical applications such as osteoarthritis surgery, ocular surgery, plastic surgery, tissue engineering, and drug delivery. It plays a key role in cushioning and lubricating the body and is abundant in the eyes, joints, and heart valves. A powerful antioxidant, hyaluronic acid is perhaps best known for its ability to bond water to tissue. Hyaluronan production increases in proliferating cells, and the polymer may play a role in mitosis. This chapter gives an overview of hyaluronic acid and its physicochemical properties and applications. This chapter gives a deep understanding on the special benefits of hyaluronic acid in the fields of pharmaceutical, medical, and environmental applications. Hyaluronic acid paves the way for beneficial research and applications to the welfare of life forms.
Collapse
Affiliation(s)
- Prasad N Sudha
- PG and Research Department of Chemistry, DKM College for Women, Thiruvalluvar University, Vellore, Tamil Nadu, India.
| | - Maximas H Rose
- Department of Biology, Sri Sai Vidyasharam, Vellore, Tamil Nadu, India
| |
Collapse
|
120
|
Abstract
![]()
Cells carefully control the transit of compounds through their membranes using “gated” protein channels that respond to chemical stimuli. Connexin gap junctions, which are high conductance cell-to-cell channels, are a remarkable class of “gated” channel with multiple levels of assembly. A gap junction between adhering cells comprises two half-channels in each cell membrane that adhere to each other to form a continuous cell-to-cell channel. Each half-channel is a hexameric assembly of six protein transmembrane subunits. These gap junctions display both intramembrane assembly and intermembrane assembly, making them an attractive target for biomimetic studies. Although many examples of self-assembled channels have been developed, few can also mediate intermembrane adhesion. Developing systems that combine membrane adhesion with controlled transit across the membrane would not only provide a better understanding of self-assembly in and around the membrane, but would also provide a route towards smart biomaterials, targeted drug delivery and an interface with nanotechnology. This Account describes our biomimetic approaches to combining membrane adhesion with membrane transport, using both self-assembled “sticky” pores and “sticky” nanoparticles to trigger transit across membranes. This combination links both fundamental and applied research, acting as a bridge between molecular level assembly and the formation of functional biomaterials. The ultimate goal is to create complex self-assembled systems in biological or biomimetic environments that can both interface with cells and transport compounds across bilayers in response to remote chemical or electromagnetic signals. Our research in this area started with fundamental studies of intramembrane and intermembrane self-assembly, building upon previously known channel-forming compounds to create self-assembled channels that were switchable or able to mediate vesicle–vesicle adhesion. Subsequently, nanoparticles with a “sticky” coating were used to mediate adhesion between vesicles. Combining these adhesive properties with the unique characteristics of nanosized magnetite allowed a noninvasive magnetic signal to trigger transport of compounds out of magnetic nanoparticle-vesicle assemblies. Adding an extravesicular matrix produced new responsive biomaterials for use in tissue engineering. These biomaterials can be magnetically patterned and can deliver drugs upon receipt of a magnetic signal, allowing spatiotemporal control over cellular responses.
Collapse
Affiliation(s)
- Simon J. Webb
- Manchester Institute of Biotechnology and the School of Chemistry, University of Manchester, 131 Princess St, Manchester M1 7DN, United Kingdom
| |
Collapse
|
121
|
Deponti D, Di Giancamillo A, Gervaso F, Domenicucci M, Domeneghini C, Sannino A, Peretti GM. Collagen scaffold for cartilage tissue engineering: the benefit of fibrin glue and the proper culture time in an infant cartilage model. Tissue Eng Part A 2013; 20:1113-26. [PMID: 24152291 DOI: 10.1089/ten.tea.2013.0171] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
This study (i) developed a scaffold made of collagen I designed for hosting the autologous chondrocytes, (ii) focused on the optimization of chondrocytes seeding by the addition of the fibrin glue, and (iii) investigated the culture time for the ideal scaffold maturation in vitro. In the first part of the study, fresh chondrocytes were isolated from infant swine articular cartilage, and immediately seeded onto the collagen sponges either in medium or in fibrinogen in order to show the contribute of fibrin glue in cell seeding and survival into the scaffold. In the second part of the study, chondrocytes were first expanded in vitro and then resuspended in fibrinogen, seeded in collagen sponges, and cultured for 1, 3, and 5 weeks in order to identify the optimal time for the rescue of cell phenotype and for the scaffold maturation into a tissue with chondral properties. The histological and immunohistochemical data from the first part of the study (study with primary chondrocytes) demonstrated that the presence of fibrin glue ameliorated cell distribution and survival into the chondral composites. The second part of this work (study with dedifferentiated chondrocytes) showed that the prolongation of the culture to 3 weeks promoted a significant restoration of the cell phenotype, resulting in a composite with proper morphological features, biochemical composition, and mechanical integrity. In conclusion, this study developed a collagenic-fibrin glue scaffold that was able to support chondrocyte survival and synthetic activity in a static culture; in particular, this model was able to turn the engineered samples into a tissue with chondral-like properties when cultured in vitro for at least 3 weeks.
Collapse
|
122
|
Dvořáková J, Kučera L, Kučera J, Švík K, Foglarová M, Muthný T, Pravda M, Němcová M, Velebný V, Kubala L. Chondrogenic differentiation of mesenchymal stem cells in a hydrogel system based on an enzymatically crosslinked tyramine derivative of hyaluronan. J Biomed Mater Res A 2013; 102:3523-30. [PMID: 24243864 DOI: 10.1002/jbm.a.35033] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 10/24/2013] [Accepted: 11/01/2013] [Indexed: 12/12/2022]
Abstract
Hyaluronan-based tissue substitutes are promising materials in cartilage reconstruction surgery. Herein, the chondrogenesis of human mesenchymal stem cells (MSC) in a hydrogel based on a tyramine derivative of hyaluronan crosslinked by hydrogen peroxidase (HA-TA) was evaluated. Human MSC seeded in the scaffold were incubated in standard chondrogenic medium and medium enriched with bone morphogenetic protein-6 (BMP6). Cell viability, the gene expression of selected markers (collagen type II, aggrecan, SOX9, collagen type X, and osteopontin), and the histological characteristics were examined during three weeks of in vitro cultivation. The tissue reaction of both unseeded and MSC seeded HA-TA scaffolds were tested in vivo after subcutaneous application in rats for 12 weeks. The data showed that cells resisted the process of crosslinking and remained viable for the whole time while exhibiting changes in cell organization. Human MSC cultivated in HA-TA hydrogel expressed genes of both chondrogenic and osteogenic differentiation and the addition of BMP6 revealed a tendency to potentiate both processes. Histological analysis of HA-TA in vivo implants did not reveal a chronic inflammatory reaction. In both cases, in vivo HA-TA implants were continuously degraded and MSC-seeded hydrogels tended to form clusters similar to in vitro samples. In conclusion, MSC chondrogenic differentiation may proceed in a HA-TA scaffold that is biocompatible. © 2013 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 102A: 3523-3530, 2014.
Collapse
Affiliation(s)
- Jana Dvořáková
- Contipro Biotech s.r.o., 401, 561 02, Dolní Dobrouč, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Cissell DD, Hu JC, Griffiths LG, Athanasiou KA. Antigen removal for the production of biomechanically functional, xenogeneic tissue grafts. J Biomech 2013; 47:1987-96. [PMID: 24268315 DOI: 10.1016/j.jbiomech.2013.10.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/16/2013] [Accepted: 10/19/2013] [Indexed: 10/26/2022]
Abstract
Xenogeneic tissues are derived from other animal species and provide a source of material for engineering mechanically functional tissue grafts, such as heart valves, tendons, ligaments, and cartilage. Xenogeneic tissues, however, contain molecules, known as antigens, which invoke an immune reaction following implantation into a patient. Therefore, it is necessary to remove the antigens from a xenogeneic tissue to prevent immune rejection of the graft. Antigen removal can be accomplished by treating a tissue with solutions and/or physical processes that disrupt cells and solubilize, degrade, or mask antigens. However, processes used for cell and antigen removal from tissues often have deleterious effects on the extracellular matrix (ECM) of the tissue, rendering the tissue unsuitable for implantation due to poor mechanical properties. Thus, the goal of an antigen removal process should be to reduce the antigen content of a xenogeneic tissue while preserving its mechanical functionality. To expand the clinical use of antigen-removed xenogeneic tissues as biomechanically functional grafts, it is essential that researchers examine tissue antigen content, ECM composition and architecture, and mechanical properties as new antigen removal processes are developed.
Collapse
Affiliation(s)
- Derek D Cissell
- Department of Orthopaedic Surgery, University of California, Davis, CA, USA; Department of Surgical and Radiological Sciences, University of California, Davis, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Leigh G Griffiths
- Department of Veterinary Medicine: Medicine and Epidemiology, University of California, Davis, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California, Davis, CA, USA; Department of Orthopaedic Surgery, University of California, Davis, CA, USA.
| |
Collapse
|
124
|
Frank RM, Cole BJ. Complex cartilage cases in the athletic patient: advances in malalignment, instability, articular defects, and meniscal insufficiency. PHYSICIAN SPORTSMED 2013; 41:41-52. [PMID: 24231596 DOI: 10.3810/psm.2013.11.2035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
For sports medicine specialists, patients presenting with multiple coexisting knee pathologies can be some of the most difficult to treat patients, especially those with cartilage pathology. In particular, articular cartilage lesions are often incidental findings, and the decision to treat patients must be based on the lesions' confirmed contribution to patient symptomatology. The combination of malalignment, ligamentous instability, and chondral/meniscal damage in patients is challenging to treat because of the difficulty in determining both the relative contribution of each of the pathologies to patient clinical condition and the timing of performing specific corrective procedures. Corrective operations performed in isolation to treat each of the pathologies have historically produced reasonable results in patients; however, combined procedures for treatment of combined pathologies may prove essential for the success of any single procedure. This review describes the clinical evaluation of patients with multiple coexisting knee pathologies and highlights current evidence-based treatment strategies for the management of these combined disorders.
Collapse
Affiliation(s)
- Rachel M Frank
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL.
| | | |
Collapse
|
125
|
Olee T, Grogan SP, Lotz MK, Colwell CW, D'Lima DD, Snyder EY. Repair of cartilage defects in arthritic tissue with differentiated human embryonic stem cells. Tissue Eng Part A 2013; 20:683-92. [PMID: 24028447 DOI: 10.1089/ten.tea.2012.0751] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chondrocytes have been generated in vitro from a range of progenitor cell types and by a number of strategies. However, achieving reconstitution of actual physiologically relevant, appropriately-laminated cartilage in situ that would be applicable to conditions, such as arthritis and cartilage degeneration remains elusive. This lack of success is multifactorial and includes limited cell source, decreased proliferation rate of mature chondrocytes, lack of maintenance of phenotype, reduced matrix synthesis, and poor integration with host tissue. We report an efficient approach for deriving mesenchymal chondroprogenitor cells from human embryonic stem cells. These cells generated tissue containing cartilage-specific matrix proteins that integrated in situ in a partial-thickness defect in ex vivo articular cartilage harvested from human arthritic joints. Given that stem cells provide a virtually inexhaustible supply of starting material and that our technique is easily scalable, cartilaginous tissue primed and grafted in this manner could be suitable for clinical translation.
Collapse
Affiliation(s)
- Tsaiwei Olee
- 1 Shiley Center for Orthopaedic Research and Education at Scripps Health , La Jolla, California
| | | | | | | | | | | |
Collapse
|
126
|
Preliminary results of a novel single-stage cartilage restoration technique: particulated juvenile articular cartilage allograft for chondral defects of the patella. Arthroscopy 2013; 29:1661-70. [PMID: 23876608 DOI: 10.1016/j.arthro.2013.05.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 05/16/2013] [Accepted: 05/22/2013] [Indexed: 02/02/2023]
Abstract
PURPOSE To evaluate outcomes and magnetic resonance imaging (MRI) findings after use of particulated juvenile cartilage for the treatment of focal Outerbridge grade 4 articular cartilage defects of the patella. METHODS From 2007 to 2011, 16 patients (2 bilateral) underwent a novel single-stage articular cartilage restoration procedure using particulated juvenile articular cartilage allograft. We enrolled 15 knees (13 patients) in this study. The mean age at surgery was 26.4 ± 9.1 years, and the mean postoperative follow-up was 28.8 ± 10.2 months. A musculoskeletal radiologist evaluated each knee with postoperative MRI for the International Cartilage Repair Society cartilage repair assessment score, graft hypertrophy, bony changes around the graft, and percent fill of the defect. All patients also completed the Knee Injury and Osteoarthritis Outcome Score, International Knee Documentation Committee Subjective Knee Evaluation, and Kujala clinical outcome survey (scale, 0 to 100), as well as the Tegner activity scale and visual analog pain scale (scale, 0 to 10). RESULTS The mean International Cartilage Repair Society cartilage repair assessment score on MRI was 8.0 ± 2.8, a nearly normal assessment. Of 15 knees, 11 (73%) were found to have normal or nearly normal cartilage repair. Three patients had mild graft hypertrophy whereas 2 had gross graft hypertrophy, 2 of whom required arthroscopic debridement because of symptoms. The mean fill of the defect at follow-up was 89% ± 19.6%, with 12 of 15 knees (80%) showing at least 90% defect coverage. The mean clinical outcome score at follow-up was 73.3 ± 17.6 for the International Knee Documentation Committee evaluation, and the mean scores for each subdomain of the Knee Injury and Osteoarthritis Outcome Score were as follows: 84.2 ± 14.2 for pain, 85.0 ± 12.3 for symptoms and stiffness, 88.9 ± 12.9 for activities of daily living, 62.0 ± 25.1 for sports and recreation, and 60.8 ± 28.6 for quality of life. The median score for the Kujala survey was 79 (range, 55 to 99). The median score on the Tegner activity scale was 5 (range, 3 to 9), and the mean score on the visual analog scale was 1.9 ± 1.4, indicating minimal pain. CONCLUSIONS Preliminary results suggest that cartilage restoration using particulated juvenile articular cartilage allograft offers a viable option for patients with focal grade 4 articular cartilage defects of the patella.
Collapse
|
127
|
Zheng L, Lu HQ, Fan HS, Zhang XD. Reinforcement and chemical cross-linking in collagen-based scaffolds in cartilage tissue engineering: a comparative study. IRANIAN POLYMER JOURNAL 2013. [DOI: 10.1007/s13726-013-0182-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
128
|
Fini M, Pagani S, Giavaresi G, De Mattei M, Ongaro A, Varani K, Vincenzi F, Massari L, Cadossi M. Functional Tissue Engineering in Articular Cartilage Repair: Is There a Role for Electromagnetic Biophysical Stimulation? TISSUE ENGINEERING PART B-REVIEWS 2013; 19:353-67. [DOI: 10.1089/ten.teb.2012.0501] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Milena Fini
- Laboratory of Preclinical and Surgical Studies, Technological Innovations, and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
- Laboratory of Biocompatibility, Technological Innovations, and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Stefania Pagani
- Laboratory of Preclinical and Surgical Studies, Technological Innovations, and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
- Laboratory of Biocompatibility, Technological Innovations, and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Gianluca Giavaresi
- Laboratory of Preclinical and Surgical Studies, Technological Innovations, and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
- Laboratory of Biocompatibility, Technological Innovations, and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Monica De Mattei
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Alessia Ongaro
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Katia Varani
- Department of Clinical and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Fabrizio Vincenzi
- Department of Clinical and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Leo Massari
- Department of Biomedical Sciences and Advanced Therapies, St. Anna Hospital, Ferrara, Italy
| | - Matteo Cadossi
- II Orthopaedics and Trauma Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|
129
|
Santo VE, Gomes ME, Mano JF, Reis RL. Controlled release strategies for bone, cartilage, and osteochondral engineering--Part I: recapitulation of native tissue healing and variables for the design of delivery systems. TISSUE ENGINEERING. PART B, REVIEWS 2013; 19:308-26. [PMID: 23268651 PMCID: PMC3690094 DOI: 10.1089/ten.teb.2012.0138] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 12/11/2012] [Indexed: 12/12/2022]
Abstract
The potential of growth factors to stimulate tissue healing through the enhancement of cell proliferation, migration, and differentiation is undeniable. However, critical parameters on the design of adequate carriers, such as uncontrolled spatiotemporal presence of bioactive factors, inadequate release profiles, and supraphysiological dosages of growth factors, have impaired the translation of these systems onto clinical practice. This review describes the healing cascades for bone, cartilage, and osteochondral interface, highlighting the role of specific growth factors for triggering the reactions leading to tissue regeneration. Critical criteria on the design of carriers for controlled release of bioactive factors are also reported, focusing on the need to provide a spatiotemporal control over the delivery and presentation of these molecules.
Collapse
Affiliation(s)
- Vítor E. Santo
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Manuela E. Gomes
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João F. Mano
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
130
|
Wu L, Gonzalez S, Shah S, Kyupelyan L, Petrigliano FA, McAllister DR, Adams JS, Karperien M, Tuan TL, Benya PD, Evseenko D. Extracellular matrix domain formation as an indicator of chondrocyte dedifferentiation and hypertrophy. Tissue Eng Part C Methods 2013; 20:160-8. [PMID: 23758619 DOI: 10.1089/ten.tec.2013.0056] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cartilage injury represents one of the most significant clinical conditions. Implantation of expanded autologous chondrocytes from noninjured compartments of the joint is a typical strategy for repairing cartilage. However, two-dimensional culture causes dedifferentiation of chondrocytes, making them functionally inferior for cartilage repair. We hypothesized that functional exclusion of dedifferentiated chondrocytes can be achieved by the selective mapping of collagen molecules deposited by chondrogenic cells in a three-dimensional environment. Freshly isolated and in vitro expanded human fetal or adult articular chondrocytes were cultured in a thermoreversible hydrogel at density of 1 × 10(7) cells/mL for 24 h. Chondrocytes were released from the gel, stained with antibodies against collagen type 2 (COL II) or COL I or COL X and sorted by fluorescence activated cell sorting. Imaging flow cytometry, immunohistochemistry, quantitative polymerase chain reaction, and glycosaminoglycan (GAG) assays were performed to evaluate the differences between COL II domain forming and COL II domain-negative cells. Freshly dissected periarticular chondrocytes robustly formed domains that consisted of the extracellular matrix surrounding cells in the hydrogel as a capsule clearly detectable by imaging flow cytometry (ImageStream) and confocal microscopy. These domains were almost exclusively formed by COL II. In contrast to that, a significant percentage of freshly isolated growth plate pre-hypertrophic and hyperdrophic chondrocytes deposited matrix domains positive for COL II, COL I, and COL X. The proportion of the cells producing COL II domains decreased with the increased passage of in vitro expanded periarticular fetal or adult articular chondrocytes. Sorted COL II domain forming cells deposited much higher levels of COL II and GAGs in pellet assays than COL II domain-negative cells. COL II domain forming cells expressed chondrogenic genes at higher levels than negative cells. We report a novel method that allows separation of functionally active chondrogenic cells, which deposit high levels of COL II from functionally inferior dedifferentiated cells or hypertrophic chondrocytes producing COL X. This approach may significantly improve current strategies used for cartilage repair.
Collapse
Affiliation(s)
- Ling Wu
- 1 Department of Orthopaedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles , Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Baptista LS, Silva KR, Pedrosa CSG, Amaral RJFC, Belizário JV, Borojevic R, Granjeiro JM. Bioengineered cartilage in a scaffold-free method by human cartilage-derived progenitor cells: a comparison with human adipose-derived mesenchymal stromal cells. Artif Organs 2013; 37:1068-75. [PMID: 23865470 DOI: 10.1111/aor.12121] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The objective of our study was to investigate chondrogenesis potential of human adipose-derived mesenchymal stromal cells (MSCs), using as a positive control a human source of cartilage-derived progenitor cells (PCs). This source of PCs was recently described by our group and dwells on the surface of nasoseptal cartilage. Histological analysis using Safranin O staining and immunofluorescence for actin filaments and collagen type II was performed on three-dimensional (3D) pellet cultures. Cartilage PCs and adipose MSCs showed similarities in monolayer culture related to cell morphology and proliferation. Our 3D pellet cultures substantially reduced the actin stress and after 21 days under chondrogenic medium, we observed an increase in the pellet diameter for cartilage PCs (7.4%) and adipose MSCs (21.2%). Adipose-derived MSCs responded to chondrogenic stimulus, as seen by positive areas for collagen type II, but they were not able to recreate a mature extracellular matrix. Using semi-quantitative analysis, we observed a majority of Safranin O areas rising from blue (no stain) to orange (moderate staining) and no changes in fibroblastic morphology (P < 0.0001). For cartilage PCs, chondrogenic induction is responsible for morphological changes and a high percentage of matrix area/number of cells (P ≤ 0.0001), evaluated by computerized histomorphometry. Morphological analyses reveal that adipose-derived MSCs were not able to recreate a bioengineered cartilage. The cost of culture was reduced, as the cartilage PCs under growth-factor free medium exhibit a high score for cartilage formation compared with the induced adipose mesenchymal stromal cells (P = 0.0021). Using a pellet 3D culture, our cartilage PCs were able to produce a cartilage tissue in vitro, leading to the future development of bioengineered products.
Collapse
Affiliation(s)
- Leandra S Baptista
- Federal University of Rio de Janeiro/Xerém, Duque de Caxias, Brazil; Division of Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | | | | | | | | | | | | |
Collapse
|
132
|
Kim IL, Khetan S, Baker BM, Chen CS, Burdick JA. Fibrous hyaluronic acid hydrogels that direct MSC chondrogenesis through mechanical and adhesive cues. Biomaterials 2013; 34:5571-80. [PMID: 23623322 PMCID: PMC3652578 DOI: 10.1016/j.biomaterials.2013.04.004] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 04/03/2013] [Indexed: 12/15/2022]
Abstract
Electrospinning has recently gained much interest due to its ability to form scaffolds that mimic the nanofibrous nature of the extracellular matrix, such as the size and depth-dependent alignment of collagen fibers within hyaline cartilage. While much progress has been made in developing bulk, isotropic hydrogels for tissue engineering and understanding how the microenvironment of such scaffolds affects cell response, these effects have not been extensively studied in a nanofibrous system. Here, we show that the mechanics (through intrafiber crosslink density) and adhesivity (through RGD density) of electrospun hyaluronic acid (HA) fibers significantly affect human mesenchymal stem cell (hMSC) interactions and gene expression. Specifically, hMSC spreading, proliferation, and focal adhesion formation were dependent on RGD density, but not on the range of fiber mechanics investigated. Moreover, traction-mediated fiber displacements generally increased with more adhesive fibers. The expression of chondrogenic markers, unlike trends in cell spreading and cytoskeletal organization, was influenced by both fiber mechanics and adhesivity, in which softer fibers and lower RGD densities generally enhanced chondrogenesis. This work not only reveals concurrent effects of mechanics and adhesivity in a fibrous context, but also highlights fibrous HA hydrogels as a promising scaffold for future cartilage repair strategies.
Collapse
Affiliation(s)
- Iris L. Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA [Tel: 215-898-8537; Fax: 215-573-2071]
| | - Sudhir Khetan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA [Tel: 215-898-8537; Fax: 215-573-2071]
| | - Brendon M. Baker
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA [Tel: 215-898-8537; Fax: 215-573-2071]
| | - Christopher S. Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA [Tel: 215-898-8537; Fax: 215-573-2071]
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA [Tel: 215-898-8537; Fax: 215-573-2071]
| |
Collapse
|
133
|
One-stage focal cartilage defect treatment with bone marrow mononuclear cells and chondrocytes leads to better macroscopic cartilage regeneration compared to microfracture in goats. Osteoarthritis Cartilage 2013; 21:950-6. [PMID: 23583464 DOI: 10.1016/j.joca.2013.03.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/24/2013] [Accepted: 03/31/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The combination of chondrocytes and mononuclear fraction (MNF) cells might solve the expansion induced dedifferentiation problem of reimplanted cells in autologous chondrocytes implantation as sufficient cells would be available for direct, one-stage, implantation. Earlier in vitro work already showed a positive stimulation of cartilage specific matrix production when chondrocytes and MNF cells were combined. Therefore, this study aimed to evaluate cartilage regeneration using a one-stage procedure combining MNF cells and primary chondrocytes for the treatment of focal cartilage lesions in goats compared to microfracture treatment. DESIGN Freshly created focal cartilage defects were treated with either a combination of chondrocytes and MNF cells embedded in fibrin glue or microfracture treatment. After 6 months follow-up local regeneration as well as the general joint cartilage health were evaluated using validated scores and biochemical assays. RESULTS Macroscopic (P = 0.015) scores for the cartilage surface at the treated defect were, after 6 months, significantly higher for the chondrocyteMNF treatment compared to microfracture-treated defects, but microscopic scores were not (P = 0.067). The articulating cartilage showed more (P = 0.005) degeneration following microfracture treatment compared to chondrocyteMNF treatment. Biochemical glycosaminoglycans (GAG) evaluation did not reveal differences between the treatments. Both treatments had resulted in a slight to moderate cartilage degeneration at other locations in the joint. CONCLUSION In conclusion, treatment of focal articular cartilage lesions in goats using a combination of MNF cells from bone marrow and unexpanded chondrocytes leads to better macroscopic regeneration compared to microfracture, however needs further fine-tuning to decrease the negative influence on other joint compartments.
Collapse
|
134
|
Gadjanski I, Yodmuang S, Spiller K, Bhumiratana S, Vunjak-Novakovic G. Supplementation of exogenous adenosine 5'-triphosphate enhances mechanical properties of 3D cell-agarose constructs for cartilage tissue engineering. Tissue Eng Part A 2013; 19:2188-200. [PMID: 23651296 DOI: 10.1089/ten.tea.2012.0352] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Formation of tissue-engineered cartilage is greatly enhanced by mechanical stimulation. However, direct mechanical stimulation is not always a suitable method, and the utilization of mechanisms underlying mechanotransduction might allow for a highly effective and less aggressive alternate means of stimulation. In particular, the purinergic, adenosine 5'-triphosphate (ATP)-mediated signaling pathway is strongly implicated in mechanotransduction within the articular cartilage. We investigated the effects of transient and continuous exogenous ATP supplementation on mechanical properties of cartilaginous constructs engineered using bovine chondrocytes and human mesenchymal stem cells (hMSCs) encapsulated in an agarose hydrogel. For both cell types, we have observed significant increases in equilibrium and dynamic compressive moduli after transient ATP treatment applied in the fourth week of cultivation. Continuous ATP treatment over 4 weeks of culture only slightly improved the mechanical properties of the constructs, without major changes in the total glycosaminoglycan (GAG) and collagen content. Structure-function analyses showed that transiently ATP-treated constructs, and in particular those based on hMSCs, had the highest level of correlation between compositional and mechanical properties. Transiently treated groups showed intense staining of the territorial matrix for GAGs and collagen type II. These results indicate that transient ATP treatment can improve functional mechanical properties of cartilaginous constructs based on chondrogenic cells and agarose hydrogels, possibly by improving the structural organization of the bulk phase and territorial extracellular matrix (ECM), that is, by increasing correlation slopes between the content of the ECM components (GAG, collagen) and mechanical properties of the construct.
Collapse
Affiliation(s)
- Ivana Gadjanski
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
135
|
Kotecha M, Ravindran S, Schmid TM, Vaidyanathan A, George A, Magin RL. Application of sodium triple-quantum coherence NMR spectroscopy for the study of growth dynamics in cartilage tissue engineering. NMR IN BIOMEDICINE 2013; 26:709-17. [PMID: 23378198 PMCID: PMC3634872 DOI: 10.1002/nbm.2916] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 10/23/2012] [Accepted: 12/09/2012] [Indexed: 06/01/2023]
Abstract
We studied the tissue growth dynamics of tissue-engineered cartilage at an early growth stage after cell seeding for four weeks using sodium triple-quantum coherence NMR spectroscopy. The following tissue-engineering constructs were studied: 1) bovine chondrocytes cultured in alginate beads; 2) bovine chondrocytes cultured as pellets (scaffold-free chondrocyte pellets); and 3) human marrow stromal cells (HMSCs) seeded in collagen/chitosan based biomimetic scaffolds. We found that the sodium triple-quantum coherence spectroscopy could differentiate between different tissue-engineered constructs and native tissues based on the fast and slow components of relaxation rate as well as on the average quadrupolar coupling. Both fast (Tf ) and slow (Ts ) relaxation times were found to be longer in chondrocyte pellets and biomimetic scaffolds compared to chondrocytes suspended in alginate beads and human articular cartilage tissues. In all cases, it was found that relaxation rates and motion of sodium ions measured from correlation times were dependent on the amount of macromolecules, high cell density and anisotropy of the cartilage tissue-engineered constructs. Average quadrupolar couplings were found to be lower in the engineered tissue compared to native tissue, presumably due to the lack of order in collagen accumulated in the engineered tissue. These results support the use of sodium triple-quantum coherence spectroscopy as a tool to investigate anisotropy and growth dynamics of cartilage tissue-engineered constructs in a simple and reliable way.
Collapse
Affiliation(s)
- Mrignayani Kotecha
- Department of Bioengineering, University of Illinois at Chicago, IL 60607, USA.
| | | | | | | | | | | |
Collapse
|
136
|
Centola M, Abbruzzese F, Scotti C, Barbero A, Vadalà G, Denaro V, Martin I, Trombetta M, Rainer A, Marsano A. Scaffold-based delivery of a clinically relevant anti-angiogenic drug promotes the formation of in vivo stable cartilage. Tissue Eng Part A 2013; 19:1960-71. [PMID: 23611597 DOI: 10.1089/ten.tea.2012.0455] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Standard cartilage tissue engineering approaches, for example, matrix-induced autologous chondrocyte implantation (MACI), consist of the implantation of cell-based constructs whose survival and further development first depend on the degree of graft maturity at the time of surgery (e.g., matrix production) and, subsequently, on initial host reaction. Indeed, blood vessel ingrowth and macrophage migration within the implant may endanger graft stability of immature constructs; so, control of angiogenesis was proposed as an adjuvant of cellular therapy for the treatment of cartilage defects. In this study, we hypothesized that engineered constructs with no in vitro precultivation, but functionalized to block angiogenesis right on implantation, might result in better survival, as well as superior long-term cartilaginous quality. Here, we propose a clinically compatible fibrin/hyaluronan scaffold seeded with nasal chondrocytes (NC) and functionalized with an FDA-approved anti-angiogenic drug (bevacizumab; Avastin(®)), which sequestrates vascular endothelial growth factor from the surrounding environment. Our results show that the sustained bevacizumab release from NC-loaded scaffolds after subcutaneous implantation in nude mice efficiently blocked host vessels ingrowth (five times lower CD31(+) cells infiltration vs. control group, at 3 weeks after implant), and enhanced constructs survival rate (75% vs. 18% for the control, at 6 weeks after implant). In vitro assays, developed to elucidate the role of specific construct components in the in vivo remodeling, allowed to determine that fibrin degradation products enhanced the in vitro endothelial cell proliferation, as well as the macrophage migration; whereas the presence of bevacizumab was capable of counteracting these effects. The proposed pharmacological control of angiogenesis by a therapeutic drug released from a scaffold might enhance cartilage regeneration by MACI approaches, possibly allowing it to bypass the complex and costly phase of graft preculture to gain increased functionality.
Collapse
Affiliation(s)
- Matteo Centola
- Tissue Engineering Laboratory, Center for Integrated Research, Università Campus Bio-Medico di Roma, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Dai W, Yao Z, Dong J, Kawazoe N, Zhang C, Chen G. Cartilage tissue engineering with controllable shape using a poly(lactic-co-glycolic acid)/collagen hybrid scaffold. J BIOACT COMPAT POL 2013. [DOI: 10.1177/0883911513484205] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
It is of critical importance to prepare three-dimensional biodegradable porous scaffolds for cartilage tissue engineering. We developed a poly(lactic- co-glycolic acid)/collagen hybrid scaffold, which combined the advantages of natural type I collagen and synthetic polymer polylactin 910 knitted mesh (90:10 copolymer of glycolic acid and lactic acid), and a method using this poly(lactic- co-glycolic acid)/collagen scaffold to regenerate cartilage with controllable shape. The mechanically strong poly(lactic- co-glycolic acid) mesh served as a mechanical skeleton supporting the scaffold, and the collagen benefited cell seeding, distribution, and tissue formation. Bovine chondrocytes were cultured in the hybrid scaffold and transplanted into the subcutaneous sites of nude mice for 4 weeks. All the samples showed spatially uniform cell distribution, natural chondrocyte morphology, and deposition of abundant cartilaginous extracellular matrices such as type II collagen and aggrecan. Production of glycosaminoglycans per DNA reached 74.63% of the natural articular cartilage. The mechanical strength was 62.76% and 71.43% in Young’s modulus and stiffness, respectively, compared to the native articular cartilage. All the samples successfully maintained the original shapes. Our method provides a new strategy for regeneration of cartilage tissue with designed shapes.
Collapse
Affiliation(s)
- Wenda Dai
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenjun Yao
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Naoki Kawazoe
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Ibaraki, Japan
| | - Chi Zhang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoping Chen
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Ibaraki, Japan
| |
Collapse
|
138
|
Zhang W, Chen J, Tao J, Hu C, Chen L, Zhao H, Xu G, Heng BC, Ouyang HW. The promotion of osteochondral repair by combined intra-articular injection of parathyroid hormone-related protein and implantation of a bi-layer collagen-silk scaffold. Biomaterials 2013; 34:6046-57. [PMID: 23702148 DOI: 10.1016/j.biomaterials.2013.04.055] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 04/27/2013] [Indexed: 01/01/2023]
Abstract
The repair of osteochondral defects can be enhanced with scaffolds but is often accompanied with undesirable terminal differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Parathyroid hormone-related protein (PTHrP) has been shown to inhibit aberrant differentiation, but administration at inappropriate time points would have adverse effects on chondrogenesis. This study aims to develop an effective tissue engineering strategy by combining PTHrP and collagen-silk scaffold for osteochondral defect repair. The underlying mechanisms of the synergistic effect of combining PTHrP administration with collagen-silk scaffold implantation for rabbit knee joint osteochondral defect repair were investigated. In vitro studies showed that PTHrP treatment significantly reduced Alizarin Red staining and expression of terminal differentiation-related markers. This is achieved in part through blocking activation of the canonical Wnt/β-catenin signaling pathway. For the in vivo repair study, intra-articular injection of PTHrP was carried out at three different time windows (4-6, 7-9 and 10-12 weeks) together with implantation of a bi-layer collagen-silk scaffold. Defects treated with PTHrP at the 4-6 weeks time window exhibited better regeneration (reconstitution of cartilage and subchondral bone) with minimal terminal differentiation (hypertrophy, ossification and matrix degradation), as well as enhanced chondrogenesis (cell shape, Col2 and GAG accumulation) compared with treatment at other time windows. Furthermore, the timing of PTHrP administration also influenced PTHrP receptor expression, thus affecting the treatment outcome. Our results demonstrated that intra-articular injection of PTHrP at 4-6 weeks post-injury together with collagen-silk scaffold implantation is an effective strategy for inhibiting terminal differentiation and enhancing chondrogenesis, thus improving cartilage repair and regeneration in a rabbit model.
Collapse
Affiliation(s)
- Wei Zhang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Fernandes AM, Herlofsen SR, Karlsen TA, Küchler AM, Fløisand Y, Brinchmann JE. Similar properties of chondrocytes from osteoarthritis joints and mesenchymal stem cells from healthy donors for tissue engineering of articular cartilage. PLoS One 2013; 8:e62994. [PMID: 23671648 PMCID: PMC3650033 DOI: 10.1371/journal.pone.0062994] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 03/30/2013] [Indexed: 12/16/2022] Open
Abstract
Lesions of hyaline cartilage do not heal spontaneously, and represent a therapeutic challenge. In vitro engineering of articular cartilage using cells and biomaterials may prove to be the best solution. Patients with osteoarthritis (OA) may require tissue engineered cartilage therapy. Chondrocytes obtained from OA joints are thought to be involved in the disease process, and thus to be of insufficient quality to be used for repair strategies. Bone marrow (BM) derived mesenchymal stem cells (MSCs) from healthy donors may represent an alternative cell source. We have isolated chondrocytes from OA joints, performed cell culture expansion and tissue engineering of cartilage using a disc-shaped alginate scaffold and chondrogenic differentiation medium. We performed real-time reverse transcriptase quantitative PCR and fluorescence immunohistochemistry to evaluate mRNA and protein expression for a range of molecules involved in chondrogenesis and OA pathogenesis. Results were compared with those obtained by using BM-MSCs in an identical tissue engineering strategy. Finally the two populations were compared using genome-wide mRNA arrays. At three weeks of chondrogenic differentiation we found high and similar levels of hyaline cartilage-specific type II collagen and fibrocartilage-specific type I collagen mRNA and protein in discs containing OA and BM-MSC derived chondrocytes. Aggrecan, the dominant proteoglycan in hyaline cartilage, was more abundantly distributed in the OA chondrocyte extracellular matrix. OA chondrocytes expressed higher mRNA levels also of other hyaline extracellular matrix components. Surprisingly BM-MSC derived chondrocytes expressed higher mRNA levels of OA markers such as COL10A1, SSP1 (osteopontin), ALPL, BMP2, VEGFA, PTGES, IHH, and WNT genes, but lower levels of MMP3 and S100A4. Based on the results presented here, OA chondrocytes may be suitable for tissue engineering of articular cartilage.
Collapse
Affiliation(s)
- Amilton M. Fernandes
- The Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Sarah R. Herlofsen
- The Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tommy A. Karlsen
- The Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Axel M. Küchler
- The Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Yngvar Fløisand
- Department of Hematology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jan E. Brinchmann
- The Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
140
|
Vahdati A, Wagner DR. Implant size and mechanical properties influence the failure of the adhesive bond between cartilage implants and native tissue in a finite element analysis. J Biomech 2013; 46:1554-60. [DOI: 10.1016/j.jbiomech.2013.03.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/13/2013] [Accepted: 03/16/2013] [Indexed: 10/26/2022]
|
141
|
Reverte-Vinaixa MM, Joshi N, Diaz-Ferreiro EW, Teixidor-Serra J, Dominguez-Oronoz R. Medium-term outcome of mosaicplasty for grade III-IV cartilage defects of the knee. J Orthop Surg (Hong Kong) 2013; 21:4-9. [PMID: 23629978 DOI: 10.1177/230949901302100104] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PURPOSE To evaluate the medium-term outcome of mosaicplasty for full-thickness cartilage defects of the knee joint in 17 patients. METHODS Records of 12 men and 5 women aged 16 to 57 (mean, 35) years who underwent mosaicplasty for grade III/IV osteochondral defects in the lateral (n=14) or medial (n=3) femoral condyle were reviewed. 12 of the patients had undergone knee surgeries. The mean size of the defects was 3.4 (range, 1-4) cm(2). Three patients had defects of >2 cm(2). All operations were performed by a single surgeon using mini-arthrotomy. The lateral edge of the trochlea was the donor site. Graft integration and the presence of any abnormality at the articular surface were assessed using magnetic resonance imaging (MRI). In addition, patients were evaluated using the International Knee Documentation Committee (IKDC) rating scale, the SF-36 health questionnaire, visual analogue scale (VAS) score for pain. RESULTS Two of the 17 patients developed necrosis and cystic degeneration of the grafts and underwent conversion to unicompartmental knee arthroplasty within 2 years. They were older than 45 years and had defects of >2 cm(2). Respectively in years 4 and 7, one and 4 patients were lost to follow-up, the mean IKDC score was 75% and 88%, the SF-36 score was 83% and 90%, and the VAS score was ≤3 in 13 of 14 patients at year 4 and in all 11 patients at year 7. At the 7-year follow-up, patient satisfaction with mosaicplasty was excellent in 8 patients, good in 3, and poor in 2 (who underwent unicompartmental knee arthroplasty). At year 4, MRI showed integration of the cartilage repair tissue and incorporation of the osseous portion of the graft into the bone in 13 of the 14 patients. The remaining patient had osteoarthritis at the graft donor site. At year 7, MRI showed good integration of the implant in all 11 available patients, but fissures were seen on the cartilage surface in 3 patients. CONCLUSION The medium-term outcome of autologous mosaicplasty for symptomatic osteochondral defects in the femoral condyle is good. Longer follow-up is needed to determine the structural and functional integrity of the graft over time.
Collapse
Affiliation(s)
- Maria Mercedes Reverte-Vinaixa
- Department of Trauma and Orthopaedic Surgery, Hospital Universitari Vall d'Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
142
|
Li B, Yang J, Ma L, Li F, Tu Z, Gao C. Fabrication of poly(lactide-co-glycolide) scaffold filled with fibrin gel, mesenchymal stem cells, and poly(ethylene oxide)-b-poly(L-lysine)/TGF-β1 plasmid DNA complexes for cartilage restoration in vivo. J Biomed Mater Res A 2013; 101:3097-108. [PMID: 23529956 DOI: 10.1002/jbm.a.34618] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 12/19/2012] [Accepted: 01/16/2013] [Indexed: 12/15/2022]
Abstract
A poly (lactide-co-glycolide) (PLGA) scaffold filled with fibrin gel, mesenchymal stem cells (MSCs) and poly(ethylene oxide)-b-poly (L-lysine) (PEO-b-PLL)/pDNA-TGF-β1 complexes was fabricated and applied in vivo for synchronized regeneration of cartilage and subchondral bone. The PEO-b-PLL/pDNA-TGF-β1 complexes could transfect MSCs in vitro to produce TGF-β1 in situ and up regulate the expression of chondrogenesis-related genes in the construct. The expression of heterogeneous TGF-β1 in vivo declined along with the prolongation of implantation time, and lasted for 3 and 6 weeks in the mRNA and protein levels, respectively. The constructs (Experimental group) of PLGA/fibrin gel/MSCs/(PEO-b-PLL/pDNA-TGF-β1 complexes) were implanted into the osteochondral defects of rabbits to restore the functional cartilages, with gene-absent constructs as the Control. After 12 weeks, the Experimental group regenerated the neo-cartilage and subchondral bone with abundant deposition of glycosaminoglycans (GAGs) and type II collagen. The regenerated tissues had good integration with the host tissues too. By contrast, the defects were only partially repaired by the Control constructs. qRT-PCR results demonstrated that expression of the chondrogenesis-marker genes in the Experimental group was significantly higher than that of the Control group, and was very close to that of the normal cartilage tissue.
Collapse
Affiliation(s)
- Bo Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering Zhejiang University, Hangzhou, 310027, China
| | | | | | | | | | | |
Collapse
|
143
|
Cerrato R. Particulated juvenile articular cartilage allograft transplantation for osteochondral lesions of the talus. Foot Ankle Clin 2013; 18:79-87. [PMID: 23465950 DOI: 10.1016/j.fcl.2012.12.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Osteochondral lesions of the talus can present a challenge to the orthopedic surgeon. Because of its avascular nature, articular cartilage has a poor capacity for self-repair and regeneration. A wide variety of strategies have been developed to restore the structure and function of injured cartilage. Surgical strategies range from repair of cartilage through the formation of fibrocartilage to a variety of restorative procedures, including tissue-engineering-based strategies. A novel treatment option involves the implantation of particulated articular cartilage obtained from a juvenile allograft donor, the DeNovo NT graft. This article reviews the DeNovo NT graft, its usage, and surgical technique.
Collapse
Affiliation(s)
- Rebecca Cerrato
- Institute for Foot & Ankle Reconstruction, Mercy Medical Center, 301 St. Paul Place, Baltimore, MD 21202, USA.
| |
Collapse
|
144
|
Herlofsen SR, Bryne JC, Høiby T, Wang L, Issner R, Zhang X, Coyne MJ, Boyle P, Gu H, Meza-Zepeda LA, Collas P, Mikkelsen TS, Brinchmann JE. Genome-wide map of quantified epigenetic changes during in vitro chondrogenic differentiation of primary human mesenchymal stem cells. BMC Genomics 2013; 14:105. [PMID: 23414147 PMCID: PMC3620534 DOI: 10.1186/1471-2164-14-105] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 02/12/2013] [Indexed: 01/01/2023] Open
Abstract
Background For safe clinical application of engineered cartilage made from mesenchymal stem cells (MSCs), molecular mechanisms for chondrogenic differentiation must be known in detail. Changes in gene expression and extracellular matrix synthesis have been extensively studied, but the epigenomic modifications underlying these changes have not been described. To this end we performed whole-genome chromatin immunoprecipitation and deep sequencing to quantify six histone modifications, reduced representation bisulphite sequencing to quantify DNA methylation and mRNA microarrays to quantify gene expression before and after 7 days of chondrogenic differentiation of MSCs in an alginate scaffold. To add to the clinical relevance of our observations, the study is based on primary bone marrow-derived MSCs from four donors, allowing us to investigate inter-individual variations. Results We see two levels of relationship between epigenetic marking and gene expression. First, a large number of genes ontogenetically linked to MSC properties and the musculoskeletal system are epigenetically prepatterned by moderate changes in H3K4me3 and H3K9ac near transcription start sites. Most of these genes remain transcriptionally unaltered. Second, transcriptionally upregulated genes, more closely associated with chondrogenesis, are marked by H3K36me3 in gene bodies, highly increased H3K4me3 and H3K9ac on promoters and 5' end of genes, and increased H3K27ac and H3K4me1 marking in at least one enhancer region per upregulated gene. Within the 7-day time frame, changes in promoter DNA methylation do not correlate significantly with changes in gene expression. Inter-donor variability analysis shows high level of similarity between the donors for this data set. Conclusions Histone modifications, rather than DNA methylation, provide the primary epigenetic control of early differentiation of MSCs towards the chondrogenic lineage.
Collapse
Affiliation(s)
- Sarah R Herlofsen
- Institute of Immunology and Norwegian Center for Stem Cell Research, Oslo University Hospital Rikshospitalet, Oslo 0424, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Ganesh N, Hanna C, Nair SV, Nair LS. Enzymatically cross-linked alginic-hyaluronic acid composite hydrogels as cell delivery vehicles. Int J Biol Macromol 2013; 55:289-94. [PMID: 23357799 DOI: 10.1016/j.ijbiomac.2012.12.045] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 12/26/2012] [Accepted: 12/31/2012] [Indexed: 12/16/2022]
Abstract
An injectable composite gel was developed from alginic and hyaluronic acid. The enzymatically cross-linked injectable gels were prepared via the oxidative coupling of tyramine modified sodium algiante and sodium hyaluronate in the presence of horse radish peroxidase (HRP) and hydrogen peroxide (H2O2). The composite gels were prepared by mixing equal parts of the two tyraminated polymer solutions in 10U HRP and treating with 1.0% H2O2. The properties of the alginate gels were significantly affected by the addition of hyaluronic acid. The percentage water absorption and storage modulus of the composite gels were found to be lower than the alginate gels. The alginate and composite gels showed lower protein release compared to hyaluronate gels in the absence of hyaluronidase. Even hyaluronate gels showed only approximately 10% protein release after 14 days incubation in phosphate buffer solution. ATDC-5 cells encapsulated in the injectable gels showed high cell viability. The composite gels showed the presence of enlarged spherical cells with significantly higher metabolic activity compared to cells in hyaluronic and alginic acid gels. The results suggest the potential of the composite approach to develop covalently cross-linked hydrogels with tuneable physical, mechanical, and biological properties.
Collapse
Affiliation(s)
- Nitya Ganesh
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | | | | | | |
Collapse
|
146
|
Sharma B, Fermanian S, Gibson M, Unterman S, Herzka DA, Cascio B, Coburn J, Hui AY, Marcus N, Gold GE, Elisseeff JH. Human cartilage repair with a photoreactive adhesive-hydrogel composite. Sci Transl Med 2013; 5:167ra6. [PMID: 23303605 PMCID: PMC3972413 DOI: 10.1126/scitranslmed.3004838] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Surgical options for cartilage resurfacing may be significantly improved by advances and application of biomaterials that direct tissue repair. A poly(ethylene glycol) diacrylate (PEGDA) hydrogel was designed to support cartilage matrix production, with easy surgical application. A model in vitro system demonstrated deposition of cartilage-specific extracellular matrix in the hydrogel biomaterial and stimulation of adjacent cartilage tissue development by mesenchymal stem cells. For translation to the joint environment, a chondroitin sulfate adhesive was applied to covalently bond and adhere the hydrogel to cartilage and bone tissue in articular defects. After preclinical testing in a caprine model, a pilot clinical study was initiated where the biomaterials system was combined with standard microfracture surgery in 15 patients with focal cartilage defects on the medial femoral condyle. Control patients were treated with microfracture alone. Magnetic resonance imaging showed that treated patients achieved significantly higher levels of tissue fill compared to controls. Magnetic resonance spin-spin relaxation times (T(2)) showed decreasing water content and increased tissue organization over time. Treated patients had less pain compared with controls, whereas knee function [International Knee Documentation Committee (IKDC)] scores increased to similar levels between the groups over the 6 months evaluated. No major adverse events were observed over the study period. With further clinical testing, this practical biomaterials strategy has the potential to improve the treatment of articular cartilage defects.
Collapse
Affiliation(s)
- Blanka Sharma
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Matthew Gibson
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Shimon Unterman
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Daniel A. Herzka
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brett Cascio
- Department of Orthopedics, Lake Charles Memorial Hospital, Lake Charles, LA 70601, USA
| | - Jeannine Coburn
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alexander Y. Hui
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Norman Marcus
- Private Practice Orthopedic Surgery, Springfield, VA 22151, USA
| | - Garry E. Gold
- Department of Radiology, Bioengineering, and Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
147
|
Giavaresi G, Bondioli E, Melandri D, Giardino R, Tschon M, Torricelli P, Cenacchi G, Rotini R, Castagna A, Veronesi F, Pagani S, Fini M. Response of human chondrocytes and mesenchymal stromal cells to a decellularized human dermis. BMC Musculoskelet Disord 2013; 14:12. [PMID: 23294867 PMCID: PMC3547812 DOI: 10.1186/1471-2474-14-12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 12/31/2012] [Indexed: 12/20/2022] Open
Abstract
Background Although progress has been made in the treatment of articular cartilage lesions, they are still a major challenge because current techniques do not provide satisfactory long-term outcomes. Tissue engineering and the use of functional biomaterials might be an alternative regenerative strategy and fulfill clinical needs. Decellularized extracellular matrices have generated interest as functional biologic scaffolds, but there are few studies on cartilage regeneration. The aim of this study was to evaluate in vitro the biological influence of a newly developed decellularized human dermal extracellular matrix on two human primary cultures. Methods Normal human articular chondrocytes (NHAC-kn) and human mesenchymal stromal cells (hMSC) from healthy donors were seeded in polystyrene wells as controls (CTR), and on decellularized human dermis batches (HDM_derm) for 7 and 14 days. Cellular proliferation and differentiation, and anabolic and catabolic synthetic activity were quantified at each experimental time. Histology and scanning electron microscopy were used to evaluate morphology and ultrastructure. Results Both cell cultures had a similar proliferation rate that increased significantly (p < 0.0005) at 14 days. In comparison with CTR, at 14 days NHAC-kn enhanced procollagen type II (CPII, p < 0.05) and aggrecan synthesis (p < 0.0005), whereas hMSC significantly enhanced aggrecan synthesis (p < 0.0005) and transforming growth factor-beta1 release (TGF-β1, p < 0.0005) at both experimental times. Neither inflammatory stimulus nor catabolic activity induction was observed. By comparing data of the two primary cells, NHAC-kn synthesized significantly more CPII than did hMSC at both experimental times (p < 0.005), whereas hMSC synthesized more aggrecan at 7 days (p < 0.005) and TGF-β1 at both experimental times than did NHAC-kn (p < 0.005). Conclusions The results obtained showed that in in vitro conditions HDM_derm behaves as a suitable scaffold for the growth of both well-differentiated chondrocytes and undifferentiated mesenchymal cells, thus ensuring a biocompatible and bioactive substrate. Further studies are mandatory to test the use of HDM_derm with tissue engineering to assess its therapeutic and functional effectiveness in cartilage regeneration.
Collapse
Affiliation(s)
- Gianluca Giavaresi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute IRCCS, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
The use of type 1 collagen scaffold containing stromal cell-derived factor-1 to create a matrix environment conducive to partial-thickness cartilage defects repair. Biomaterials 2013; 34:713-23. [DOI: 10.1016/j.biomaterials.2012.10.027] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 10/08/2012] [Indexed: 11/22/2022]
|
149
|
Ng VY, Jump SS, Santangelo KS, Russell DS, Bertone AL. Genetic engineering of juvenile human chondrocytes improves scaffold-free mosaic neocartilage grafts. Clin Orthop Relat Res 2013; 471:26-38. [PMID: 23008026 PMCID: PMC3528904 DOI: 10.1007/s11999-012-2615-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 09/12/2012] [Indexed: 01/31/2023]
Abstract
BACKGROUND Current cartilage transplantation techniques achieve suboptimal restoration and rely on patient donor cells or living grafts of chondrocytes. PURPOSE We sought to enhance allogeneic grafts by testing mosaics of genetically engineered and naïve juvenile human chondrocytes (jCh). METHODS We obtained specimens from three humans and performed three experiments (two in vitro, one in vivo). We compared neocartilage with and without (1) supplemented serum-free medium (chondrocyte differentiation medium [CDM]), (2) adenoviral BMP-2 (AdBMP-2) transduction, and (3) varying ratios (0.1-1) of transduced and naïve jCh. We compared (4) healing with mosaic grafts with naïve neocartilage or marrow stimulation in immunosuppressed rats. For each of 10 in vitro treatment groups, we had six replicates for each human, and for each of three in vivo treatment groups, we had four replicates for one human. We scored the histology with the semiquantitative Bern score. RESULTS AdBMP-2 and naïve neocartilage growth in CDM were histologically superior (Bern score, 5.2 versus 3.7; 8.0 versus 1.8) and size (8.0 versus 6.1; 7.9 versus 2.2 mg) to standard medium. In CDM, AdBMP-2 decreased viability (76% versus 90%), but increased BMP-2 production (619 ng/mL versus 43 pg/mL). Ten percent and 25% AdBMP-2 transduction had Bern scores of 6.8 and 6.5 and viability of 84% and 83%, respectively. Twenty-five percent mosaic grafts provided better healing histologically than marrow stimulation or naive neocartilage. CONCLUSIONS Low-level AdBMP-2 and CDM augment neocartilage parameters in vitro and vivo. CLINICAL RELEVANCE Genetic augmentation of jCh and creation of mosaic neocartilage may improve graft viability and articular healing compared with naïve neocartilage.
Collapse
Affiliation(s)
- Vincent Y. Ng
- />Department of Orthopaedics, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Seth S. Jump
- />Sports Medicine Center, The Ohio State University Wexner Medical Center, Columbus, OH USA
- />Department of Veterinary Clinical Sciences, The Ohio State University Veterinary Medical Center, Columbus, OH USA
| | - Kelly S. Santangelo
- />Department of Veterinary Biosciences, The Ohio State University, Columbus, OH USA
| | - Duncan S. Russell
- />Department of Veterinary Biosciences, The Ohio State University, Columbus, OH USA
| | - Alicia L. Bertone
- />Department of Orthopaedics, The Ohio State University Wexner Medical Center, Columbus, OH USA
- />Sports Medicine Center, The Ohio State University Wexner Medical Center, Columbus, OH USA
- />Department of Veterinary Clinical Sciences, The Ohio State University Veterinary Medical Center, Columbus, OH USA
- />College of Veterinary Medicine, The Ohio State University, 601 Tharp Street, Columbus, OH 43210 USA
| |
Collapse
|
150
|
Mahmoudifar N, Doran PM. Osteogenic differentiation and osteochondral tissue engineering using human adipose-derived stem cells. Biotechnol Prog 2012; 29:176-85. [PMID: 23125060 DOI: 10.1002/btpr.1663] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 10/18/2012] [Indexed: 12/31/2022]
Abstract
Osteogenesis and the production of composite osteochondral tissues were investigated using human adult adipose-derived stem cells and polyglycolic acid (PGA) mesh scaffolds under dynamic culture conditions. For osteogenesis, cells were expanded with or without osteoinduction factors and cultured in control or osteogenic medium for 2 weeks. Osteogenic medium enhanced osteopontin and osteocalcin gene expression when applied after but not during cell expansion. Osteogenesis was induced and mineralized deposits were present in tissues produced using PGA culture in osteogenic medium. For development of osteochondral constructs, scaffolds seeded with stem cells were precultured in either chondrogenic or osteogenic medium, sutured together, and cultured in dual-chamber stirred bioreactors containing chondrogenic and osteogenic media in separate compartments. After 2 weeks, total collagen synthesis was 2.1-fold greater in the chondroinduced sections of the composite tissues compared with the osteoinduced sections; differentiation markers for cartilage and bone were produced in both sections of the constructs. The results from the dual-chamber bioreactor highlight the challenges associated with achieving simultaneous chondrogenic and osteogenic differentiation in tissue engineering applications using a single stem-cell source.
Collapse
Affiliation(s)
- Nastaran Mahmoudifar
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney NSW 2052, Australia
| | | |
Collapse
|