1
|
Torabi Rahvar P, Abdekhodaie MJ, Jooybar E, Gantenbein B. An enzymatically crosslinked collagen type II/hyaluronic acid hybrid hydrogel: A biomimetic cell delivery system for cartilage tissue engineering. Int J Biol Macromol 2024; 279:134614. [PMID: 39127277 DOI: 10.1016/j.ijbiomac.2024.134614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
This study presents new injectable hydrogels based on hyaluronic acid and collagen type II that mimic the polysaccharide-protein structure of natural cartilage. After collagen isolation from chicken sternal cartilage, tyramine-grafted hyaluronic acid and collagen type II (HA-Tyr and COL-II-Tyr) were synthesized. Hybrid hydrogels were prepared with different ratios of HA-Tyr/COL-II-Tyr using horseradish peroxidase and noncytotoxic concentrations of hydrogen peroxide to encapsulate human bone marrow-derived mesenchymal stromal cells (hBM-MSCs). The findings showed that a higher HA-Tyr content resulted in a higher storage modulus and a lower hydrogel shrinkage, resulting in hydrogel swelling. Incorporating COL-II-Tyr into HA-Tyr hydrogels induced a more favorable microenvironment for hBM-MSCs chondrogenic differentiation. Compared to HA-Tyr alone, the hybrid HA-Tyr/COL-II-Tyr hydrogel promoted enhanced chondrocyte adhesion, spreading, proliferation, and upregulation of cartilage-related gene expression. These results highlight the promising potential of injectable HA-Tyr/COL-II-Tyr hybrid hydrogels to deliver cells for cartilage regeneration.
Collapse
Affiliation(s)
- Parisa Torabi Rahvar
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Mohammad J Abdekhodaie
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Environmental and Applied Science Management, Yeates School of Graduate Studies, Toronto Metropolitan University, Toronto, Canada.
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland; Inselspital, Bern University Hospital, Department of Orthopedic Surgery & Traumatology, Bern, Switzerland
| |
Collapse
|
2
|
Zhang Z, Hu X, Jin M, Mu Y, Zhou H, Ma C, Ma L, Liu B, Yao H, Huang Y, Wang DA. Collagen Type II-Based Injectable Materials for In situ Repair and Regeneration of Articular Cartilage Defect. Biomater Res 2024; 28:0072. [PMID: 39220112 PMCID: PMC11362811 DOI: 10.34133/bmr.0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
Repairing and regenerating articular cartilage defects (ACDs) have long been challenging for physicians and scientists. The rise of injectable materials provides a novel strategy for minimally invasive surgery to repair ACDs. In this study, we successfully developed injectable materials based on collagen type II, achieving hyaline cartilage repair and regeneration of ACDs. Analysis was conducted on the regenerated cartilage after materials injection. The histology staining demonstrated complete healing of the ACDs with the attainment of a hyaline cartilage phenotype. The biochemical and biomechanical properties are similar to the adjacent native cartilage without noticeable adverse effects on the subchondral bone. Further transcriptome analysis found that compared with the Native cartilage adjacent to the defect area, the Regenerated cartilage in the defect area repaired with type II collagen-based injection materials showed changes in cartilage-related pathways, as well as down-regulation of T cell receptor signaling pathways and interleukin-17 signaling pathways, which changed the immune microenvironment of the ACD area. Overall, these findings offer a promising injectable approach to treating ACDs, providing a potential solution to the challenges associated with achieving hyaline cartilage in situ repair and regeneration while minimizing damage to the surrounding cartilage.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Xu Hu
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Min Jin
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine,
HKSTP, Sha Tin, Hong Kong SAR
| | - Yulei Mu
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Huiqun Zhou
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine,
HKSTP, Sha Tin, Hong Kong SAR
| | - Cheng Ma
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine,
HKSTP, Sha Tin, Hong Kong SAR
| | - Liang Ma
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Bangheng Liu
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine,
HKSTP, Sha Tin, Hong Kong SAR
| | - Hang Yao
- School of Chemistry and Chemical Engineering,
Yangzhou University, Yangzhou, China
| | - Ye Huang
- Knee Preservation Clinical and Research Center,
Beijing Jishuitan Hospital, Beijing, China
| | - Dong-An Wang
- Department of Biomedical Engineering,
City University of Hong Kong, Kowloon, Hong Kong SAR
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine,
HKSTP, Sha Tin, Hong Kong SAR
- Center for Neuromusculoskeletal Restorative Medicine,
HKSTP, Shatin, Hong Kong SAR
| |
Collapse
|
3
|
Hu X, Jin M, Sun K, Zhang Z, Wu Z, Shi J, Liu P, Yao H, Wang DA. Type II collagen scaffolds repair critical-sized osteochondral defects under induced conditions of osteoarthritis in rat knee joints via inhibiting TGF-β-Smad1/5/8 signaling pathway. Bioact Mater 2024; 35:416-428. [PMID: 38384986 PMCID: PMC10879694 DOI: 10.1016/j.bioactmat.2024.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/13/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
The bidirectional relationship between osteochondral defects (OCD) and osteoarthritis (OA), with each condition exacerbating the other, makes OCD regeneration in the presence of OA challenging. Type II collagen (Col2) is important in OCD regeneration and the management of OA, but its potential applications in cartilage tissue engineering are significantly limited. This study investigated the regeneration capacity of Col2 scaffolds in critical-sized OCDs under surgically induced OA conditions and explored the underlying mechanisms that promoted OCD regeneration. Furthermore, the repair potential of Col2 scaffolds was validated in over critical-sized OCD models. After 90 days or 150 days since scaffold implantation, complete healing was observed histologically in critical-sized OCD, evidenced by the excellent integration with surrounding native tissues. The newly formed tissue biochemically resembled adjacent natural tissue and exhibited comparable biomechanical properties. The regenerated OA tissue demonstrated lower expression of genes associated with cartilage degradation than native OA tissue but comparable expression of genes related to osteochondral anabolism compared with normal tissue. Additionally, transcriptome and proteome analysis revealed the hindrance of TGF-β-Smad1/5/8 in regenerated OA tissue. In conclusion, the engrafting of Col2 scaffolds led to the successful regeneration of critical-sized OCDs under surgically induced OA conditions by inhibiting the TGF-β-Smad1/5/8 signaling pathway.
Collapse
Affiliation(s)
- Xu Hu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Min Jin
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
| | - Kang Sun
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
| | - Zhonglian Wu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, PR China
| | - Junli Shi
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, PR China
| | - Peilai Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, 107 Wenhua Xilu, Jinan, PR China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, PR China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, PR China
| |
Collapse
|
4
|
Zhou H, Mu Y, Ma C, Zhang Z, Tao C, Wang DA. Rejuvenating Hyaline Cartilaginous Phenotype of Dedifferentiated Chondrocytes in Collagen II Scaffolds: A Mechanism Study Using Chondrocyte Membrane Nanoaggregates as Antagonists. ACS NANO 2024; 18:2077-2090. [PMID: 38194361 DOI: 10.1021/acsnano.3c09033] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Joint cartilage lesions affect the global population in the current aging society. Maintenance and rejuvenation of articular cartilage with hyaline phenotype remains a challenge as the underlying mechanism has not been completely understood. Here, we have designed and performed a mechanism study using scaffolds made of type II collagen (Col2) as the 3D cell cultural platforms, on some of which nanoaggregates comprising extracts of chondrocyte membrane (CCM) were coated as the antagonist of Col2. Dedifferentiated chondrocytes were, respectively, seeded into these Col2 based scaffolds with (antCol2S) or without (Col2S) CCM coating. After 6 weeks, in Col2S, the chondrocytes were rejuvenated to regain hyaline phenotype, whereas this redifferentiation effect was attenuated in antCol2S. Transcriptomic and proteomic profiling indicated that the Wnt/β-catenin signaling pathway, which is an opponent to maintenance of the hyaline cartilaginous phenotype, was inhibited in Col2S, but it was contrarily upregulated in antCol2S due to the antagonism and shielding against Col2 by the CCM coating. Specifically, in antCol2S, since the coated CCM nanoaggregates contain the same components as those present on the surface of the seeded chondrocytes, the corresponding ligand sites on Col2 had been preoccupied and saturated by CCM coating before exposure to the seeded cells. The results indicated that the ligation between Col2 ligands and integrin α5 receptors on the surface of the seeded chondrocytes in antCol2S was antagonized by the CCM coating, which facilitates the Wnt/β-catenin signaling toward the loss of hyaline cartilaginous phenotype. This finding reveals the contribution of Col2 for maintenance and rejuvenation of the hyaline cartilaginous phenotype in chondrocytes.
Collapse
Affiliation(s)
- Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
| | - Cheng Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Chao Tao
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 999077, P. R. China
| |
Collapse
|
5
|
Dewey MJ, Collins AJ, Tiffany A, Barnhouse VR, Lu C, Kolliopoulos V, Mutreja I, Hickok NJ, Harley BAC. Evaluation of bacterial attachment on mineralized collagen scaffolds and addition of manuka honey to increase mesenchymal stem cell osteogenesis. Biomaterials 2023; 294:122015. [PMID: 36701999 PMCID: PMC9928779 DOI: 10.1016/j.biomaterials.2023.122015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/06/2023] [Accepted: 01/14/2023] [Indexed: 01/21/2023]
Abstract
The design of biomaterials to regenerate bone is likely to increasingly require modifications that reduce bacterial attachment and biofilm formation as infection during wound regeneration can significantly impede tissue repair and typically requires surgical intervention to restart the healing process. Further, much research on infection prevention in bone biomaterials has focused on modeling of non-resorbable metal alloy materials, whereas an expanding direction of bone regeneration has focused on development of bioresorbable materials. This represents a need for the prevention and understanding of infection in resorbable biomaterials. Here, we investigate the ability of a mineralized collagen biomaterial to natively resist infection and examine how the addition of manuka honey, previously identified as an antimicrobial agent, affects gram positive and negative bacterial colonization and mesenchymal stem cell osteogenesis and vasculature formation. We incorporate manuka honey into these scaffolds via either direct fabrication into the scaffold microarchitecture or via soaking the scaffold in a solution of manuka honey after fabrication. Direct incorporation results in a change in the surface characteristics and porosity of mineralized collagen scaffolds. Soaking scaffolds in honey concentrations higher than 10% had significant negative effects on mesenchymal stem cell metabolic activity. Soaking or incorporating 5% honey had no impact on endothelial cell tube formation. Although solutions of 5% honey reduced metabolic activity of mesenchymal stem cells, MSC-seeded scaffolds displayed increased calcium and phosphorous mineral formation, osteoprotegerin release, and alkaline phosphatase activity. Bacteria cultured on mineralized collagen scaffolds demonstrated surfaces covered in bacteria and no method of preventing infection, and using 10 times the minimal inhibitory concentration of antibiotics did not completely kill bacteria within the mineralized collagen scaffolds, indicating bioresorbable scaffold materials may act to shield bacteria from antibiotics. The addition of 5% manuka honey to scaffolds was not sufficient to prevent P. aeruginosa attachment or consistently reduce the activity of methicillin resistant staphylococcus aureus, and concentrations above 7% manuka honey are likely necessary to impact MRSA. Together, our results suggest bioresorbable scaffolds may create an environment conducive to bacterial growth, and potential trade-offs exist for the incorporation of low levels of honey in scaffolds to increase osteogenic potential of osteoprogenitors while high-levels of honey may be sufficient to reduce gram positive or negative bacteria activity but at the cost of reduced osteogenesis.
Collapse
Affiliation(s)
- Marley J Dewey
- Dept. of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Alan J Collins
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Aleczandria Tiffany
- Dept. of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Victoria R Barnhouse
- Dept. of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Crislyn Lu
- School of Chemical Sciences, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Vasiliki Kolliopoulos
- Dept. of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Isha Mutreja
- Department of Restorative Science, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Noreen J Hickok
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Brendan A C Harley
- Dept. of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA; Dept. of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA; Dept. of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
6
|
Voga M, Majdic G. Articular Cartilage Regeneration in Veterinary Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1401:23-55. [DOI: 10.1007/5584_2022_717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
7
|
Higher Chondrogenic Potential of Extracellular Vesicles Derived from Mesenchymal Stem Cells Compared to Chondrocytes-EVs In Vitro. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9011548. [PMID: 34938811 PMCID: PMC8687842 DOI: 10.1155/2021/9011548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/09/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022]
Abstract
The inability of cartilage to self-repair necessitates an effective therapeutic approach to restore damaged tissues. Extracellular vesicles (EVs) are attractive options because of their roles in cellular communication and tissue repair where they regulate the cellular processes of proliferation, differentiation, and recruitment. However, it is a challenge to determine the relevant cell sources for isolation of EVs with high chondrogenic potential. The current study aims to evaluate the chondrogenic potential of EVs derived from chondrocytes (Cho-EV) and mesenchymal stem cells (MSC-EV). The EVs were separately isolated from conditioned media of both rabbit bone marrow MSCs and chondrocyte cultures. The isolated vesicles were assessed in terms of size, morphology, and surface marker expression. The chondrogenic potential of MSCs in the presence of different concentrations of EVs (50, 100, and 150 μg/ml) was evaluated during 21 days, and chondrogenic surface marker expressions were checked by qRT-PCR and histologic assays. The extracted vesicles had a spherical morphology and a size of 44.25 ± 8.89 nm for Cho-EVs and 112.1 ± 10.10 nm for MSC-EVs. Both groups expressed the EV-specific surface markers CD9 and CD81. Higher expression of chondrogenic specified markers, especially collagen type II (COL II), and secretion of glycosaminoglycans (GAGs) and proteoglycans were observed in MSCs treated with 50 and 100 μg/ml MSC-EVs compared to the Cho-EVs. The results from the use of EVs, particularly MSC-EVs, with high chondrogenic ability will provide a basis for developing therapeutic agents for cartilage repair.
Collapse
|
8
|
Wu Z, Korntner SH, Mullen AM, Zeugolis DI. Collagen type II: From biosynthesis to advanced biomaterials for cartilage engineering. BIOMATERIALS AND BIOSYSTEMS 2021; 4:100030. [PMID: 36824570 PMCID: PMC9934443 DOI: 10.1016/j.bbiosy.2021.100030] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/02/2021] [Accepted: 11/19/2021] [Indexed: 12/11/2022] Open
Abstract
Collagen type II is the major constituent of cartilage tissue. Yet, cartilage engineering approaches are primarily based on collagen type I devices that are associated with suboptimal functional therapeutic outcomes. Herein, we briefly describe cartilage's development and cellular and extracellular composition and organisation. We also provide an overview of collagen type II biosynthesis and purification protocols from tissues of terrestrial and marine species and recombinant systems. We then advocate the use of collagen type II as a building block in cartilage engineering approaches, based on safety, efficiency and efficacy data that have been derived over the years from numerous in vitro and in vivo studies.
Collapse
Affiliation(s)
- Z Wu
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - SH Korntner
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - AM Mullen
- Teagasc Research Centre, Ashtown, Ireland
| | - DI Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway (NUI Galway), Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
- Correspondence author at: REMODEL, NUI Galway & UCD.
| |
Collapse
|
9
|
Dewey MJ, Milner DJ, Weisgerber D, Flanagan CL, Rubessa M, Lotti S, Polkoff KM, Crotts S, Hollister SJ, Wheeler MB, Harley BAC. Repair of critical-size porcine craniofacial bone defects using a collagen-polycaprolactone composite biomaterial. Biofabrication 2021; 14. [PMID: 34663761 DOI: 10.1088/1758-5090/ac30d5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022]
Abstract
Regenerative medicine approaches for massive craniomaxillofacial (CMF) bone defects face challenges associated with the scale of missing bone, the need for rapid graft-defect integration, and challenges related to inflammation and infection. Mineralized collagen scaffolds have been shown to promote mesenchymal stem cell osteogenesis due to their porous nature and material properties, but are mechanically weak, limiting surgical practicality. Previously, these scaffolds were combined with 3D-printed polycaprolactone (PCL) mesh to form a scaffold-mesh composite to increase strength and promote bone formation in sub-critical sized porcine ramus defects. Here, we compare the performance of mineralized collagen-PCL composites to the PCL mesh in a critical-sized porcine ramus defect model. While there were no differences in overall healing response between groups, our data demonstrated broadly variable metrics of healing regarding new bone infiltration and fibrous tissue formation. Abscesses were present surrounding some implants and PCL polymer was still present after 9-10 months of implantation. Overall, while there was limited successful healing, with 2 of 22 implants showed substantial levels of bone regeneration, and others demonstrating some form of new bone formation, the results suggest targeted improvements to improve repair of large animal models to more accurately represent CMF bone healing. Notably, strategies to increase osteogenesis throughout the implant, modulate the immune system to support repair, and employ shape-fitting tactics to avoid implant micromotion and resultant fibrosis. Improvements to the mineralized collagen scaffolds involve changes in pore size and shape to increase cell migration and osteogenesis and inclusion or delivery of factors to aid vascular ingrowth and bone regeneration.
Collapse
Affiliation(s)
- Marley J Dewey
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Derek J Milner
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Daniel Weisgerber
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Colleen L Flanagan
- Department of Bioengineering, University of Michigan, Ann Arbor, MI, 30332, United States of America
| | - Marcello Rubessa
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Sammi Lotti
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Kathryn M Polkoff
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Sarah Crotts
- Center for 3D Medical Fabrication, Wallace A. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, United States of America
| | - Scott J Hollister
- Center for 3D Medical Fabrication, Wallace A. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, United States of America
| | - Matthew B Wheeler
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Brendan A C Harley
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.,Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| |
Collapse
|
10
|
Liao S, Meng H, Li J, Zhao J, Xu Y, Wang A, Xu W, Peng J, Lu S. Potential and recent advances of microcarriers in repairing cartilage defects. J Orthop Translat 2021; 27:101-109. [PMID: 33520655 PMCID: PMC7810913 DOI: 10.1016/j.jot.2020.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 11/11/2022] Open
Abstract
Articular cartilage regeneration is one of the challenges faced by orthopedic surgeons. Microcarrier applications have made great advances in cartilage tissue engineering in recent years and enable cost-effective cell expansion, thus providing permissive microenvironments for cells. In addition, microcarriers can be loaded with proteins, factors, and drugs for cartilage regeneration. Some microcarriers also have the advantages of injectability and targeted delivery. The application of microcarriers with these characteristics can overcome the limitations of traditional methods and provide additional advantages. In terms of the transformation potential, microcarriers have not only many advantages, such as providing sufficient and beneficial cells, factors, drugs, and microenvironments for cartilage regeneration, but also many application characteristics; for example, they can be injected to reduce invasiveness, transplanted after microtissue formation to increase efficiency, or combined with other stents to improve mechanical properties. Therefore, this technology has enormous potential for clinical transformation. In this review, we focus on recent advances in microcarriers for cartilage regeneration. We compare the characteristics of microcarriers with other methods for repairing cartilage defects, provide an overview of the advantages of microcarriers, discuss the potential of microcarrier systems, and present an outlook for future development. Translational potential of this article We reviewed the advantages and recent advances of microcarriers for cartilage regeneration. This review could give many scholars a better understanding of microcarriers, which can provide doctors with potential methods for treating patients with cartilage injure.
Collapse
Affiliation(s)
- Sida Liao
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Haoye Meng
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Junkang Li
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jun Zhao
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yichi Xu
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Aiyuan Wang
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wenjing Xu
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiang Peng
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| | - Shibi Lu
- Institute of Orthopedics/ Beijing Key Laboratory of Regenerative Medicine in Orthopedics/ Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
11
|
Chen YT, Lee HS, Hsieh DJ, Periasamy S, Yeh YC, Lai YP, Tarng YW. 3D composite engineered using supercritical CO 2 decellularized porcine cartilage scaffold, chondrocytes, and PRP: Role in articular cartilage regeneration. J Tissue Eng Regen Med 2020; 15:163-175. [PMID: 33258246 DOI: 10.1002/term.3162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/18/2020] [Accepted: 11/22/2020] [Indexed: 02/01/2023]
Abstract
At present, no definitive treatment for articular cartilage defects has been perfected. Most of the previous treatments involved multiple drilling and microfracture over defect sites with repair-related substances, which poses a limited therapeutic effect. End-stage therapy includes artificial knee joint replacement. In this study, we prepared a novel decellularized natural cartilage scaffold from porcine articular cartilage by supercritical CO2 extraction technology and three-dimensional (3D) composites made using decellularized porcine cartilage graft (dPCG) as scaffolds, platelet-rich plasma (PRP), thrombin as signals and chondrocytes as cells for the treatment of articular cartilage defects. In this study, in vitro and in vivo cartilage regeneration and the expression of chondrogenic markers were examined. Decellularized cartilage graft (dPCG) was evaluated for the extent of cell and DNA removal. Residual cartilage ECM structure was confirmed to be type II collagen by SDS PAGE and immunostaining. The new 3D composite with dPCG (100 mg and 2 × 106 chondrocytes) scaffold promotes chondrogenic marker expression in vitro. We found that the in vivo 3D composite implanted cartilage defect showed significant regeneration relative to the blank and control implant. Immunohistochemical staining showed increase of expression including Collagen type II and aggrecan in 3D composite both in vitro and in vivo studies. In this study, the bioengineered 3D composite by combining dPCG scaffold, chondrocytes, and PRP facilitated the chondrogenic marker expression in both in vitro and in vivo models with accelerated cartilage regeneration. This might serve the purpose of clinical treatment of large focal articular cartilage defects in humans in the near future.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Department of Orthopedic, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China.,Graduate Institute of Medical Science, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Herng-Sheng Lee
- Department of Pathology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China
| | - Dar-Jen Hsieh
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung City, Taiwan, Republic of China
| | - Srinivasan Periasamy
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung City, Taiwan, Republic of China
| | - Yi-Chun Yeh
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung City, Taiwan, Republic of China
| | - Yi-Ping Lai
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung City, Taiwan, Republic of China
| | - Yih-Wen Tarng
- Department of Orthopedic, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan, Republic of China.,Graduate Institute of Medical Science, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| |
Collapse
|
12
|
Dewey MJ, Johnson EM, Weisgerber DW, Wheeler MB, Harley BAC. Shape-fitting collagen-PLA composite promotes osteogenic differentiation of porcine adipose stem cells. J Mech Behav Biomed Mater 2019; 95:21-33. [PMID: 30953806 DOI: 10.1016/j.jmbbm.2019.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/17/2018] [Accepted: 03/17/2019] [Indexed: 10/27/2022]
Abstract
Craniomaxillofacial bone defects can occur as a result of congenital, post-oncologic, and high-energy impact conditions. The scale and irregularity of such defects motivate new biomaterials to promote regeneration of the damaged bone. We have recently described a mineralized collagen scaffold capable of instructing stem cell osteogenic differentiation and new bone infill in the absence of traditional osteogenic supplements. Herein, we report the integration of a millimeter-scale reinforcing poly (lactic acid) frame fabricated via 3D-printing into the mineralized collagen scaffold with micron-scale porosity to form a multi-scale mineralized collagen-PLA composite. We describe modifications to the PLA frame design to increase the compressive strength (Young's Modulus, ultimate stress and strain) of the composite. A critical challenge beyond increasing the compressive strength of the collagen scaffold is addressing challenges inherent with the irregularity of clinical defects. As a result, we examined the potential for modifying the frame architecture to render the composite with increased compressive strength in one axis or radial compressibility and shape-fitting capacity in an orthogonal axis. A library of mineralized collagen-PLA composites was mechanically characterized via compression testing and push-out test to describe mechanical performance and shape-fitting capacity. We also report in vitro comparison of the bioactivity of porcine adipose derived stem cells in the mineralized collagen-PLA composite versus the mineralized collagen scaffold via metabolic activity, gene expression, and functional matrix synthesis. The results suggest that incorporation of the PLA reinforcing frame does not negatively influence the osteoinductive nature of the mineralized collagen scaffold. Together, these findings suggest a strategy to address often competing bioactivity, mechanical strength, and shape-fitting design requirements for biomaterials for craniomaxillofacial bone regeneration.
Collapse
Affiliation(s)
- Marley J Dewey
- Dept. of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Eileen M Johnson
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Daniel W Weisgerber
- Dept. of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Matthew B Wheeler
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan A C Harley
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Dept. of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
13
|
Sun Y, Yan L, Chen S, Pei M. Functionality of decellularized matrix in cartilage regeneration: A comparison of tissue versus cell sources. Acta Biomater 2018; 74:56-73. [PMID: 29702288 PMCID: PMC7307012 DOI: 10.1016/j.actbio.2018.04.048] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 01/12/2023]
Abstract
Increasing evidence indicates that decellularized extracellular matrices (dECMs) derived from cartilage tissues (T-dECMs) or chondrocytes/stem cells (C-dECMs) can support proliferation and chondrogenic differentiation of cartilage-forming cells. However, few review papers compare the differences between these dECMs when they serve as substrates for cartilage regeneration. In this review, after an introduction of cartilage immunogenicity and decellularization methods to prepare T-dECMs and C-dECMs, a comprehensive comparison focuses on the effects of T-dECMs and C-dECMs on proliferation and chondrogenic differentiation of chondrocytes/stem cells in vitro and in vivo. Key factors within dECMs, consisting of microarchitecture characteristics and micromechanical properties as well as retained insoluble and soluble matrix components, are discussed in-depth for potential mechanisms underlying the functionality of these dECMs in regulating chondrogenesis. With this information, we hope to benefit dECM based cartilage engineering and tissue regeneration for future clinical application. STATEMENT OF SIGNIFICANCE The use of decellularized extracellular matrix (dECM) is becoming a promising approach for tissue engineering and regeneration. Compared to dECM derived from cartilage tissue, recently reported dECM from cell sources exhibits a distinct role in cell based cartilage regeneration. In this review paper, for the first time, tissue and cell based dECMs are comprehensively compared for their functionality in cartilage regeneration. This information is expected to provide an update for dECM based cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Lianqi Yan
- Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan 610083, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
14
|
Pulkkinen H, Tiitu V, Valonen P, Hämäläinen ER, Lammi M, Kiviranta I. Recombinant human type II collagen as a material for cartilage tissue engineering. Int J Artif Organs 2018; 31:960-9. [DOI: 10.1177/039139880803101106] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose Collagen type II is the major component of cartilage and would be an optimal scaffold material for reconstruction of injured cartilage tissue. In this study, the feasibility of recombinant human type II collagen gel as a 3-dimensional culture system for bovine chondrocytes was evaluated in vitro. Methods Bovine chondrocytes (4x106 cells) were seeded within collagen gels and cultivated for up to 4 weeks. The gels were investigated with confocal microscopy, histology, and biochemical assays. Results Confocal microscopy revealed that the cells maintained their viability during the entire cultivation period. The chondrocytes were evenly distributed inside the gels, and the number of cells and the amount of the extracellular matrix increased during cultivation. The chondrocytes maintained their round phenotype during the 4-week cultivation period. The glycosaminoglycan levels of the tissue increased during the experiment. The relative levels of aggrecan and type II collagen mRNA measured with realtime polymerase chain reaction (PCR) showed an increase at 1 week. Conclusion Our results imply that recombinant human type II collagen is a promising biomaterial for cartilage tissue engineering, allowing homogeneous distribution in the gel and biosynthesis of extracellular matrix components.
Collapse
Affiliation(s)
- H.J. Pulkkinen
- Institute of Biomedicine, Department of Anatomy, University of Kuopio, Kuopio - Finland
- Department of Orthopaedics and Traumatology, Jyväskylä Central Hospital, Jyväskylä - Finland
| | - V. Tiitu
- Institute of Biomedicine, Department of Anatomy, University of Kuopio, Kuopio - Finland
- Department of Orthopaedics and Traumatology, Jyväskylä Central Hospital, Jyväskylä - Finland
| | - P. Valonen
- Institute of Biomedicine, Department of Anatomy, University of Kuopio, Kuopio - Finland
- Department of Orthopaedics and Traumatology, Jyväskylä Central Hospital, Jyväskylä - Finland
| | - E.-R. Hämäläinen
- Bioprocess Engineering Laboratory, University of Oulu, Oulu - Finland
| | - M.J. Lammi
- Institute of Biomedicine, Department of Anatomy, University of Kuopio, Kuopio - Finland
- Department of Biosciences, Applied Biotechnology, University of Kuopio, Kuopio - Finland
| | - I. Kiviranta
- Department of Orthopaedics and Traumatology, Jyväskylä Central Hospital, Jyväskylä - Finland
- Department of Orthopaedics and Traumatology, Helsinki University Hospital, Helsinki - Finland
| |
Collapse
|
15
|
Cigan AD, Durney KM, Nims RJ, Vunjak-Novakovic G, Hung CT, Ateshian GA. Nutrient Channels Aid the Growth of Articular Surface-Sized Engineered Cartilage Constructs. Tissue Eng Part A 2016; 22:1063-74. [PMID: 27481330 DOI: 10.1089/ten.tea.2016.0179] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Symptomatic osteoarthritic lesions span large regions of joint surfaces and the ability to engineer cartilage constructs at clinically relevant sizes would be highly desirable. We previously demonstrated that nutrient transport limitations can be mitigated by the introduction of channels in 10 mm diameter cartilage constructs. In this study, we scaled up our previous system to cast and cultivate 40 mm diameter constructs (2.3 mm overall thickness); 4 mm diameter and channeled 10 mm diameter constructs were studied for comparison. Furthermore, to assess whether prior results using primary bovine cells are applicable for passaged cells-a more clinically realistic scenario-we cast constructs of each size with primary or twice-passaged cells. Constructs were assessed mechanically for equilibrium compressive Young's modulus (EY), dynamic modulus at 0.01 Hz (G*), and friction coefficient (μ); they were also assessed biochemically, histologically, and immunohistochemically for glycosaminoglycan (GAG) and collagen contents. By maintaining open channels, we successfully cultured robust constructs the size of entire human articular cartilage layers (growing to ∼52 mm in diameter, 4 mm thick, mass of 8 g by day 56), representing a 100-fold increase in scale over our 4 mm diameter constructs, without compromising their functional properties. Large constructs reached EY of up to 623 kPa and GAG contents up to 8.9%/ww (% of wet weight), both within native cartilage ranges, had G* >2 MPa, and up to 3.5%/ww collagen. Constructs also exhibited some of the lowest μ reported for engineered cartilage (0.06-0.11). Passaged cells produced tissue of lower quality, but still exhibited native EY and GAG content, similar to their smaller controls. The constructs produced in this study are, to our knowledge, the largest engineered cartilage constructs to date which possess native EY and GAG, and are a testament to the effectiveness of nutrient channels in overcoming transport limitations in cartilage tissue engineering.
Collapse
Affiliation(s)
- Alexander D Cigan
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Krista M Durney
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Robert J Nims
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Gordana Vunjak-Novakovic
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
- 2 Department of Medicine, Columbia University , New York, New York
| | - Clark T Hung
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Gerard A Ateshian
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
- 3 Department of Mechanical Engineering, Columbia University , New York, New York
| |
Collapse
|
16
|
Ge Y, Gong YY, Xu Z, Lu Y, Fu W. The Application of Sheet Technology in Cartilage Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:114-24. [DOI: 10.1089/ten.teb.2015.0189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Yang Ge
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yi Yi Gong
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Zhiwei Xu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yanan Lu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Wei Fu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
17
|
Luo L, Chu JYJ, Eswaramoorthy R, Mulhall KJ, Kelly DJ. Engineering Tissues That Mimic the Zonal Nature of Articular Cartilage Using Decellularized Cartilage Explants Seeded with Adult Stem Cells. ACS Biomater Sci Eng 2016; 3:1933-1943. [PMID: 33440551 DOI: 10.1021/acsbiomaterials.6b00020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Articular cartilage (AC) possesses uniquely complex mechanical properties; for example its stiffness increases with depth through the tissue and it softens when compressed. These properties are integral to the function of AC and can be attributed to the tissue's collagen network and how it interacts with negatively charged proteoglycans. In this study, scaffolds containing arrays of channels were produced from decellularized AC explants derived from skeletally immature and mature pigs. These scaffolds were then repopulated with human infrapatellar fat pad derived stem cells (FPSCs). After 4 weeks in culture, FPSCs filled channels within the decellularized explants with a matrix rich in proteoglycans and collagen. Cellular and neo-matrix alignment within these scaffolds appeared to be influenced by the underlying collagen architecture of the decellularized cartilage. Repopulating scaffolds derived from decellularized skeletally mature cartilage with FPSCs led to the development of engineered cartilage with depth-dependent mechanical properties mimicking aspects of native tissue. Furthermore, these constructs displayed the characteristic strain softening behavior of AC. These findings highlight the importance of the collagen network to engineering mechanically functional cartilage grafts.
Collapse
Affiliation(s)
- Lu Luo
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Johnnie Y J Chu
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Rajalakshmanan Eswaramoorthy
- Department of Biomedical Sciences, Sri Ramachandra University, No.1, Ramachandra Nagar, Porur, Chennai, Tamil Nadu 600116, India
| | - Kevin J Mulhall
- Department of Orthopaedic Surgery, Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Daniel J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, College Green, Dublin 2, Ireland.,Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Naughton Institute, Royal College of Surgeons in Ireland and Trinity College Dublin, College Green, Dublin 2, Ireland
| |
Collapse
|
18
|
Spector M, Lim TC. Injectable biomaterials: a perspective on the next wave of injectable therapeutics. ACTA ACUST UNITED AC 2016; 11:014110. [PMID: 26836246 DOI: 10.1088/1748-6041/11/1/014110] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We are experiencing a new wave of injectable therapeutics (namely/injectable biomaterials) to complement injectable drugs and injectable biologics, and to serve as the basis for injectable combinatorial therapeutics. Injectable biomaterials contribute to the treatment of the fluid-filled defects which often result from disease and injury, by providing the missing physical framework (i.e. the stroma). However, while injectable matrices may be necessary for the successful treatment of certain lesions, they will not likely be sufficient. Chemoattractants for select endogenous cells, or cells themselves, may need to be incorporated into the matrix prior to its injection to ensure the necessary cellular repopulation of the cavitary defect. These agents and others (drugs and biologics) delivered by the matrix represent the new category of injectable combinatorial therapeutics.
Collapse
Affiliation(s)
- Myron Spector
- Tissue Engineering, VA Boston Healthcare System, Boston, MA 02130, USA. Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
19
|
Tiruvannamalai Annamalai R, Mertz DR, Daley ELH, Stegemann JP. Collagen Type II enhances chondrogenic differentiation in agarose-based modular microtissues. Cytotherapy 2016; 18:263-77. [PMID: 26794716 PMCID: PMC4724061 DOI: 10.1016/j.jcyt.2015.10.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/04/2015] [Accepted: 10/13/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND AIMS Cell-based therapies have made an impact on the treatment of osteoarthritis; however, the repair and regeneration of thick cartilage defects is an important and growing clinical problem. Next-generation therapies that combine cells with biomaterials may provide improved outcomes. We have developed modular microenvironments that mimic the composition of articular cartilage as a delivery system for consistently differentiated cells. METHODS Human bone marrow-derived mesenchymal stem cells (MSC) were embedded in modular microbeads consisting of agarose (AG) supplemented with 0%, 10% and 20% collagen Type II (COL-II) using a water-in-oil emulsion technique. AG and AG/COL-II microbeads were characterized in terms of their structural integrity, size distribution and protein content. The viability of embedded MSC and their ability to differentiate into osteogenic, adipogenic and chondrogenic lineages over 3 weeks in culture were also assessed. RESULTS Microbeads made with <20% COL-II were robust, generally spheroidal in shape and 80 ± 10 µm in diameter. MSC viability in microbeads was consistently high over a week in culture, whereas viability in corresponding bulk hydrogels decreased with increasing COL-II content. Osteogenic differentiation of MSC was modestly supported in both AG and AG/COL-II microbeads, whereas adipogenic differentiation was strongly inhibited in COL-II containing microbeads. Chondrogenic differentiation of MSC was clearly promoted in microbeads containing COL-II, compared with pure AG matrices. CONCLUSIONS Inclusion of collagen Type II in agarose matrices in microbead format can potentiate chondrogenic differentiation of human MSC. Such compositionally tailored microtissues may find utility for cell delivery in next-generation cartilage repair therapies.
Collapse
Affiliation(s)
| | - David R Mertz
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Ethan L H Daley
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
20
|
Kontturi LS, Järvinen E, Muhonen V, Collin EC, Pandit AS, Kiviranta I, Yliperttula M, Urtti A. An injectable, in situ forming type II collagen/hyaluronic acid hydrogel vehicle for chondrocyte delivery in cartilage tissue engineering. Drug Deliv Transl Res 2015; 4:149-58. [PMID: 25786729 DOI: 10.1007/s13346-013-0188-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this study, chondrocytes were encapsulated into an injectable, in situ forming type II collagen/hyaluronic acid (HA) hydrogel cross-linked with poly(ethylene glycol) ether tetrasuccinimidyl glutarate (4SPEG) and supplemented with the transforming growth factor β1 (TGFβ1). The chondrocyte-hydrogel constructs were cultured in vitro for 7 days and studied for cell viability and proliferation, morphology, glycosaminoglycan production, and gene expression. Type II collagen/HA/4SPEG formed a strong and stable hydrogel, and the chondrocytes remained viable during the encapsulation process and for the 7-day culture period. In addition, the encapsulated cells showed spherical morphology characteristic for chondrocytic phenotype. The cells were able to produce glycosaminoglycans into their extracellular matrix, and the gene expression of type II collagen and aggrecan, genes specific for differentiated chondrocytes, increased over time. The results indicate that the studied composite hydrogel with incorporated chondrogenic growth factor TGFβ1 is able to maintain chondrocyte viability and characteristics, and thus, it can be regarded as potential injectable cell delivery vehicle for cartilage tissue engineering.
Collapse
Affiliation(s)
- Leena-Stiina Kontturi
- Centre for Drug Research, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, 00790, Helsinki, Finland,
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Ren CD, Gao S, Kurisawa M, Ying JY. Cartilage synthesis in hyaluronic acid-tyramine constructs. J Mater Chem B 2015; 3:1942-1956. [PMID: 32262266 DOI: 10.1039/c4tb01229a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The objective of this study was to determine the potential for cartilage production within a hyaluronic acid-tyramine (HA-Tyr) hydrogel scaffold. Chondrocytes were encapsulated within HA-Tyr hydrogels and subcutaneously implanted in mice. The HA-Tyr hydrogels were formed by the oxidative coupling of Tyr moieties catalyzed by hydrogen peroxide (H2O2) and horseradish peroxidase (HRP). Harvested constructs were shown to achieve a glycosaminoglycan (GAG) content of 1.2 wt%, and they demonstrated 40% of the collagen content of normal articular cartilage, including the presence of Type II collagen, which is the characteristic of articular cartilage. Matrix production was found to be influenced by the initial cell density, scaffold degradation rate and Type II collagen concentration. Injectability was also imparted to the system by delivering HRP through thermoresponsive liposomes. The method of HRP delivery, either by simple addition or through thermoresponsive liposomes, was not shown to have an effect on matrix production.
Collapse
Affiliation(s)
- Cindy D Ren
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669.
| | | | | | | |
Collapse
|
22
|
3D dynamic culture of rabbit articular chondrocytes encapsulated in alginate gel beads using spinner flasks for cartilage tissue regeneration. BIOMED RESEARCH INTERNATIONAL 2014; 2014:539789. [PMID: 25506593 PMCID: PMC4260432 DOI: 10.1155/2014/539789] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 09/11/2014] [Accepted: 09/14/2014] [Indexed: 11/17/2022]
Abstract
Cell-based therapy using chondrocytes for cartilage repair suffers from chondrocyte dedifferentiation. In the present study, the effects of an integrated three-dimensional and dynamic culture on rabbit articular chondrocytes were investigated. Cells (passages 1 and 4) were encapsulated in alginate gel beads and cultured in spinner flasks in chondrogenic and chondrocyte growth media. Subcutaneous implantation of the cell-laden beads was performed to evaluate the ectopic chondrogenesis. It was found that cells remained viable after 35 days in the three-dimensional dynamic culture. Passage 1 cells demonstrated a proliferative growth in both media. Passage 4 cells showed a gradual reduction in DNA content in growth medium, which was attenuated in chondrogenic medium. Deposition of glycosaminoglycans (GAG) was found in all cultures. While passage 1 cells generally produced higher amounts of GAG than passage 4 cells, GAG/DNA became similar on day 35 for both cells in growth media. Interestingly, GAG/DNA in growth medium was greater than that in chondrogenic medium for both cells. Based on GAG quantification and gene expression analysis, encapsulated passage 1 cells cultured in growth medium displayed the best ectopic chondrogenesis. Taken together, the three-dimensional and dynamic culture for chondrocytes holds great potential in cartilage regeneration.
Collapse
|
23
|
Xu HG, Zhang W, Zheng Q, Yu YF, Deng LF, Wang H, Liu P, Zhang M. Investigating conversion of endplate chondrocytes induced by intermittent cyclic mechanical unconfined compression in three-dimensional cultures. Eur J Histochem 2014; 58:2415. [PMID: 25308847 PMCID: PMC4194395 DOI: 10.4081/ejh.2014.2415] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/10/2014] [Accepted: 06/12/2014] [Indexed: 12/25/2022] Open
Abstract
Mechanical stimulation is known to regulate the calcification of endplate chondrocytes. The Ank protein has a strong influence on anti-calcification by transports intracellular inorganic pyrophosphate (PPi) to the extracellular matrix. It is known that TGF-β1 is able to induce Ank gene expression and protect chondrocyte calcification. Intermittent cyclic mechanical tension (ICMT) could induce calcification of endplate chondrocytes by decrease the expression of Ank gene. In this study, we investigated the relation of intermittent cyclic mechanical unconfined compression (ICMC) and Ank gene expression. We found that ICMC decreased the Ank gene expression in the endplate chondrocytes, and there was an decreased in the TGF-β1 expression after ICMC stimulation. The Ank gene expression significantly increased when treated by transforming growth factor alpha 1 (TGF-β1) in a dose-dependent manner and decreased when treated by SB431542 (ALK inhibitor) in a dose-dependent manner. Our results implicate that ICMC-induced downregulation of Ank gene expression may be regulated by TGF-β1 in end-plate chondrocytes.
Collapse
|
24
|
Yeh HY, Lin TY, Lin CH, Yen BL, Tsai CL, Hsu SH. Neocartilage formation from mesenchymal stem cells grown in type II collagen-hyaluronan composite scaffolds. Differentiation 2014; 86:171-83. [PMID: 24462469 DOI: 10.1016/j.diff.2013.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 11/06/2013] [Accepted: 11/07/2013] [Indexed: 12/11/2022]
Abstract
Three-dimensional (3D) collagen type II-hyaluronan (HA) composite scaffolds (CII-HA) which mimics the extracellular environment of natural cartilage were fabricated in this study. Rheological measurements demonstrated that the incorporation of HA increased the compression modulus of the scaffolds. An initial in vitro evaluation showed that scaffolds seeded with porcine chondrocytes formed cartilaginous-like tissue after 8 weeks, and HA functioned to promote the growth of chondrocytes into scaffolds. Placenta-derived multipotent cells (PDMC) and gingival fibroblasts (GF) were seeded on tissue culture polystyrene (TCPS), CII-HA films, and small intestinal submucosa (SIS) sheets for comparing their chondrogenesis differentiation potentials with those of adipose-derived adult stem cells (ADAS) and bone marrow-derived mesenchymal stem cells (BMSC). Among different cells, PDMC showed the greatest chondrogenic differentiation potential on both CII-HA films and SIS sheets upon TGF-β3 induction, followed by GF. This was evidenced by the up-regulation of chondrogenic genes (Sox9, aggrecan, and collagen type II), which was not observed for cells grown on TCPS. This finding suggested the essential role of substrate materials in the chondrogenic differentiation of PDMC and GF. Neocartilage formation was more obvious in both PDMC and GF cells plated on CII-HA composite scaffolds vs. 8-layer SIS at 28 days in vitro. Finally, implantation of PDMC/CII-HA constructs into NOD-SCID mice confirmed the formation of tissue-engineered cartilage in vivo.
Collapse
Affiliation(s)
- Hsi-Yi Yeh
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Ting-Yu Lin
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Chen-Huan Lin
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | - B Linju Yen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ching-Lin Tsai
- Department of Orthopaedics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Engineering, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
25
|
Rich H, Odlyha M, Cheema U, Mudera V, Bozec L. Effects of photochemical riboflavin-mediated crosslinks on the physical properties of collagen constructs and fibrils. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:11-21. [PMID: 24006048 PMCID: PMC3890585 DOI: 10.1007/s10856-013-5038-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 08/27/2013] [Indexed: 05/27/2023]
Abstract
The use of collagen scaffold in tissue engineering is on the rise, as modifications to mechanical properties are becoming more effective in strengthening constructs whilst preserving the natural biocompatibility. The combined technique of plastic compression and cross-linking is known to increase the mechanical strength of the collagen construct. Here, a modified protocol for engineering these collagen constructs is used to bring together a plastic compression method, combined with controlled photochemical crosslinking using riboflavin as a photoinitiator. In order to ascertain the effects of the photochemical crosslinking approach and the impact of the crosslinks created upon the properties of the engineered collagen constructs, the constructs were characterized both at the macroscale and at the fibrillar level. The resulting constructs were found to have a 2.5 fold increase in their Young's modulus, reaching a value of 650 ± 73 kPa when compared to non-crosslinked control collagen constructs. This value is not yet comparable to that of native tendon, but it proves that combining a crosslinking methodology to collagen tissue engineering may offer a new approach to create stronger, biomimetic constructs. A notable outcome of crosslinking collagen with riboflavin is the collagen's greater affinity for water; it was demonstrated that riboflavin crosslinked collagen retains water for a longer period of time compared to non-cross-linked control samples. The affinity of the cross-linked collagen to water also resulted in an increase of individual collagen fibrils' cross-sectional area as function of the crosslinking. These changes in water affinity and fibril morphology induced by the process of crosslinking could indicate that the crosslinked chains created during the photochemical crosslinking process may act as intermolecular hydrophilic nanosprings. These intermolecular nanosprings would be responsible for a change in the fibril morphology to accommodate variable volume of water within the fibril.
Collapse
Affiliation(s)
- Harvey Rich
- Division of Surgery and Interventional Science, UCL Tissue Repair and Engineering Centre, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, UK
| | - Marianne Odlyha
- Department of Biological Sciences Birkbeck, Institute of Structural and Molecular Biology, University of London, London, UK
| | - Umber Cheema
- Division of Surgery and Interventional Science, UCL Tissue Repair and Engineering Centre, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, UK
| | - Vivek Mudera
- Division of Surgery and Interventional Science, UCL Tissue Repair and Engineering Centre, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, UK
| | - Laurent Bozec
- Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, London, UK
| |
Collapse
|
26
|
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
|
27
|
Xu H, Zhang X, Wang H, Zhang Y, Shi Y, Zhang X. Continuous cyclic mechanical tension increases ank expression in endplate chondrocytes through the TGF-β1 and p38 pathway. Eur J Histochem 2013; 57:e28. [PMID: 24085277 PMCID: PMC3794359 DOI: 10.4081/ejh.2013.e28] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 08/22/2013] [Accepted: 08/23/2013] [Indexed: 01/06/2023] Open
Abstract
The normal ANK protein has a strong influence on anti-calcification. It is known that TGF-β1 is also able to induce extracellular inorganic pyrophosphate (ePPi) elaboration via the TGF-β1-induced ank gene expression and the mitogen-activated protein kinase (MAPK) signaling acts as a downstream effector of TGF-β1. We hypothesized that the expression of the ank gene is regulated by mechanics through TGF-β1-p38 pathway. In this study, we investigated the mechanism of short-time mechanical tension-induced ank gene expression. We found that the continuous cyclic mechanical tension (CCMT) increased the ank gene expression in the endplate chondrocytes, and there was an increase in the TGF-β1 expression after CCMT stimulation. The ank gene expression significantly increased when treated by TGF-β1 in a dose-dependent manner and decreased when treated by SB431542 (ALK inhibitor) in a dose-dependent manner. Our study results indicate that CCMT-induced ank gene expressions may be regulated by TGF-β1 and p38 MAPK pathway.
Collapse
|
28
|
Grogan SP, Chen X, Sovani S, Taniguchi N, Colwell CW, Lotz MK, D'Lima DD. Influence of cartilage extracellular matrix molecules on cell phenotype and neocartilage formation. Tissue Eng Part A 2013; 20:264-74. [PMID: 23962090 DOI: 10.1089/ten.tea.2012.0618] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Interaction between chondrocytes and the cartilage extracellular matrix (ECM) is essential for maintaining the cartilage's role as a low-friction and load-bearing tissue. In this study, we examined the influence of cartilage zone-specific ECM on human articular chondrocytes (HAC) in two-dimensional and three-dimensional (3D) environments. Two culture systems were used. SYSTEM 1: HAC were cultured on cell-culture plates that had been precoated with the following ECM molecules for 7 days: decorin, biglycan, tenascin C (superficial zone), collagen type II, hyaluronan (HA) (middle and deep zones), and osteopontin (deep zone). Uncoated standard culture plates were used as controls. Expanded cells were examined for phenotypic changes using real-time polymerase chain reaction. In addition, expanded cells were placed into high-density pellet cultures for 14 days. Neocartilage formation was assessed via gene expression and histology evaluations. SYSTEM 2: HAC that were cultured on untreated plates and encapsulated in a 3D alginate scaffold were mixed with one of the zone-specific ECM molecules. Cell viability, gene expression, and histology assessments were conducted on 14-day-old tissues. In HAC monolayer culture, exposure to decorin, HA, and osteopontin increased COL2A1 and aggrecan messenger RNA (mRNA) levels compared with controls. Biglycan up-regulated aggrecan without a significant impact on COL2A1 expression; Tenascin C reduced COL2A1 expression. Neocartilage formed after preculture on tenascin C and collagen type II expressed higher COL2A1 mRNA compared with control pellets. Preculture of HAC on HA decreased both COL2A1 and aggrecan expression levels compared with controls, which was consistent with histology. Reduced proteoglycan 4 (PRG4) mRNA levels were observed in HAC pellets that had been precultured with biglycan and collagen type II. Exposing HAC to HA directly in 3D-alginate culture most effectively induced neocartilage formation, showing increased COL2A1 and aggrecan, and reduced COL1A1 compared with controls. Decorin treatments increased HAC COL2A1 mRNA levels. These data indicate that an appropriate exposure to cartilage-specific ECM proteins could be used to enhance cartilage formation and to even induce the formation of zone-specific phenotypes to improve cartilage regeneration.
Collapse
Affiliation(s)
- Shawn P Grogan
- 1 Shiley Center for Orthopaedic Research and Education , Scripps Clinic, La Jolla, California
| | | | | | | | | | | | | |
Collapse
|
29
|
Abou Neel EA, Bozec L, Knowles JC, Syed O, Mudera V, Day R, Hyun JK. Collagen--emerging collagen based therapies hit the patient. Adv Drug Deliv Rev 2013; 65:429-56. [PMID: 22960357 DOI: 10.1016/j.addr.2012.08.010] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 08/10/2012] [Accepted: 08/28/2012] [Indexed: 12/11/2022]
Abstract
The choice of biomaterials available for regenerative medicine continues to grow rapidly, with new materials often claiming advantages over the short-comings of those already in existence. Going back to nature, collagen is one of the most abundant proteins in mammals and its role is essential to our way of life. It can therefore be obtained from many sources including porcine, bovine, equine or human and offer a great promise as a biomimetic scaffold for regenerative medicine. Using naturally derived collagen, extracellular matrices (ECMs), as surgical materials have become established practice for a number of years. For clinical use the goal has been to preserve as much of the composition and structure of the ECM as possible without adverse effects to the recipient. This review will therefore cover in-depth both naturally and synthetically produced collagen matrices. Furthermore the production of more sophisticated three dimensional collagen scaffolds that provide cues at nano-, micro- and meso-scale for molecules, cells, proteins and bulk fluids by inducing fibrils alignments, embossing and layered configuration through the application of plastic compression technology will be discussed in details. This review will also shed light on both naturally and synthetically derived collagen products that have been available in the market for several purposes including neural repair, as cosmetic for the treatment of dermatologic defects, haemostatic agents, mucosal wound dressing and guided bone regeneration membrane. There are other several potential applications of collagen still under investigations and they are also covered in this review.
Collapse
|
30
|
Yonenaga K, Nishizawa S, Fujihara Y, Asawa Y, Kanazawa S, Nagata S, Takato T, Hoshi K. Application of floating cells for improved harvest in human chondrocyte culture. Biomed Res 2012; 33:281-9. [PMID: 23124248 DOI: 10.2220/biomedres.33.281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cell culture medium, which must be discarded during medium change, may contain many cells that do not attach to culture plates. In the present study, we focused on these floating cells and attempted to determine their usefulness for cartilage regeneration. We counted the number of floating cells discarded during medium change and compared the proliferation and differentiation between floating cells and their adherent counterparts. Chondrocyte monolayer culture at a density of 5 × 103 cells/cm(2) produced viable floating cells at a rate of 2.7-3.2 × 10(3) cells/cm(2) per primary culture. When only the floating cells from one dish were harvested and replated in another dish, the number of cells was 2.8 × 10(4) cells/cm(2) (approximately half confluency) on culture day 7. The number of cells was half of that obtained by culturing only adherent cells (5 × 10(4) cells/cm(2)). The floating and adherent cells showed similar proliferation and differentiation properties. The recovery of floating cells from the culture medium could provide an approximately 1.5-fold increase in cell number over conventional monolayer culture. Thus, the collection of floating cells may be regarded as a simple, easy, and reliable method to increase the cell harvest for chondrocytes.
Collapse
Affiliation(s)
- Kazumichi Yonenaga
- Department of 1 Cartilage & Bone Regeneration (Fujisoft), The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Bae SE, Bhang SH, Kim BS, Park K. Self-assembled extracellular macromolecular matrices and their different osteogenic potential with preosteoblasts and rat bone marrow mesenchymal stromal cells. Biomacromolecules 2012; 13:2811-20. [PMID: 22813212 DOI: 10.1021/bm300791h] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Extracellular environment is a physical support that is critical to cell adhesion, migration, and differentiation. In this work, cell-derived matrices (CDMs) were obtained by separately culturing fibroblasts, preosteoblasts, and chondrocytes. The cells were grown on a coverslip and subjected to decellularization using detergents and enzymes. The resulting matrices were named fibroblast-derived matrix (FDM), preosteoblast-derived matrix (PDM), and chondrocyte-derived matrix (CHDM). We hypothesize that the unique compositional and structural feature of each CDM provides cells with a distinct microenvironment capable of functioning as a different signaling cue in the regulation of preosteoblast and rat bone marrow mesenchymal stromal cell (BMSC) osteogenic differentiation. SEM images show that each cell type creates its unique surface texture in a fibrillar structure. Three major macromolecules, fibronectin, type I collagen, and laminin, were clearly identified using both immunofluorescence and Western blot, in which FDM exhibited a much stronger signal of each ECM component than that of PDM or CHDM. For early cell morphology, BMSCs on the CDMs were highly elongated in a spindle-like shape. Both preosteoblasts and BMSCs proliferated well on CDMs comparable to the control. Once preosteoblasts were cultured for 2 weeks, their osteogenic activity was significantly different depending on the type of CDM. Using Alizarin red and von Kossa staining, we found that the cells on the FDM were much more osteogenic than the other groups. Furthermore, FDM was the most effective in upregulating the osteogenic markers, such as alkaline phosphatase (ALP), osteopontin, osteocalcin, and type I collagen. In particular, we observed a 2.5-fold increase in ALP activity with FDM compared to that of control and CHDM. In stark contrast, CHDM was very poor in stimulating osteogenic differentiation of preosteoblasts. Interestingly, these results were reproducible with the use of BMSCs, which are much more heterogeneous in cell populations than preosteoblasts. CHDM was still very weak in triggering the osteogenesis of BMSCs, whereas both FDM and PDM were equally competitive. This study demonstrates that a combination of factors (surface texture and composition) shape a unique cellular microenvironment, which serves as a physical cue toward the osteogenic differentiation of preosteoblasts and BMSCs.
Collapse
Affiliation(s)
- Soon Eon Bae
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 136-791, Republic of Korea
| | | | | | | |
Collapse
|
32
|
Park GR, Lee JG, Chun HJ, Han DK, Park K. Characterization of naturally derived macromolecular matrix and its osteogenic activity with preosteoblasts. Macromol Res 2012. [DOI: 10.1007/s13233-012-0119-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
33
|
Han E, Ge C, Chen AC, Schumacher BL, Sah RL. Compaction enhances extracellular matrix content and mechanical properties of tissue-engineered cartilaginous constructs. Tissue Eng Part A 2012; 18:1151-60. [PMID: 22372815 PMCID: PMC3360506 DOI: 10.1089/ten.tea.2011.0300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 01/27/2012] [Indexed: 11/13/2022] Open
Abstract
Many cell-based tissue-engineered cartilaginous constructs are mechanically softer than native tissue and have low content and abnormal proportions of extracellular matrix (ECM) constituents. We hypothesized that the load-bearing mechanical properties of cartilaginous constructs improve with the inclusion of collagen (COL) and proteoglycan (PG) during assembly. The objectives of this work were to determine (1) the effect of addition of PG, COL, or COL+PG on compressive properties of 2% agarose constructs and (2) the ability of mechanical compaction to concentrate matrix content and improve the compressive properties of such constructs. The inclusion of COL+PG improved the compressive properties of hydrogel constructs compared with PG or COL alone. Mechanical compaction increased the PG and COL concentrations in and compressive stiffness of the constructs. Chondrocytes included in the constructs maintained high viability after compaction. These results support the concepts that the assembly of cartilaginous constructs with COL+PG and application of mechanical compaction enhance the ECM content and compressive properties of engineered cartilaginous constructs.
Collapse
Affiliation(s)
- EunHee Han
- Department of Bioengineering, University of California-San Diego, La Jolla, California
| | - Chenghao Ge
- Department of Bioengineering, University of California-San Diego, La Jolla, California
| | - Albert C. Chen
- Department of Bioengineering, University of California-San Diego, La Jolla, California
| | - Barbara L. Schumacher
- Department of Bioengineering, University of California-San Diego, La Jolla, California
| | - Robert L. Sah
- Department of Bioengineering, University of California-San Diego, La Jolla, California
- Center for Musculoskeletal Research, Institute of Engineering in Medicine, University of California-San Diego, La Jolla, California
| |
Collapse
|
34
|
Berendsen AD, Vonk LA, Zandieh-Doulabi B, Everts V, Bank RA. Contraction-induced Mmp13 and -14 expression by goat articular chondrocytes in collagen type I but not type II gels. J Tissue Eng Regen Med 2011; 6:721-30. [PMID: 21948715 DOI: 10.1002/term.477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2010] [Revised: 01/22/2011] [Accepted: 07/05/2011] [Indexed: 11/11/2022]
Abstract
Collagen gels are promising scaffolds to prepare an implant for cartilage repair but several parameters, such as collagen concentration and composition as well as cell density, should be carefully considered, as they are reported to affect phenotypic aspects of chondrocytes. In this study we investigated whether the presence of collagen type I or II in gel lattices affects matrix contraction and relative gene expression levels of matrix proteins, MMPs and the subsequent degradation of collagen by goat articular chondrocytes. Only floating collagen I gels, and not those attached or composed of type II collagen, contracted during a culture period of 12 days. This coincided with an upregulation of both Mmp13 and -14 gene expression, whereas Mmp1 expression was not affected. The release of hydroxyproline in the culture medium, indicating matrix degradation, was increased five-fold in contracted collagen I gels compared to collagen II gels without contraction. Furthermore, blocking contraction of collagen I gels by cytochalasin B inhibited Mmp13 and -14 expression and the release of hydroxyproline. The expression of cartilage-specific ECM genes was decreased in contracted collagen I gels, with increased numbers of cells with an elongated morphology, suggesting that matrix contraction induces dedifferentiation of chondrocytes into fibroblast-like cells. We conclude that the collagen composition of the gels affects matrix contraction by articular chondrocytes and that matrix contraction induces an increased Mmp13 and -14 expression as well as matrix degradation.
Collapse
Affiliation(s)
- Agnes D Berendsen
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Research Institute MOVE, Amsterdam, The Netherlands
| | - Lucienne A Vonk
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Research Institute MOVE, Amsterdam, The Netherlands
| | | | - Vincent Everts
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Research Institute MOVE, Amsterdam, The Netherlands
| | - Ruud A Bank
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Research Institute MOVE, Amsterdam, The Netherlands.,Stem Cell and Tissue Engineering Research Group, Medical Biology Section, University Medical Centre Groningen, The Netherlands
| |
Collapse
|
35
|
Salzmann GM, Sauerschnig M, Berninger MT, Kaltenhauser T, Schönfelder M, Vogt S, Wexel G, Tischer T, Sudkamp N, Niemeyer P, Imhoff AB, Schöttle PB. The dependence of autologous chondrocyte transplantation on varying cellular passage, yield and culture duration. Biomaterials 2011; 32:5810-8. [PMID: 21592563 DOI: 10.1016/j.biomaterials.2011.04.073] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 04/24/2011] [Indexed: 10/18/2022]
Abstract
Matrix-assisted chondrocyte transplantation (m-ACI) still lacks any standardization in its execution in terms of cell passage (P), cell yield (C) and in vitro membrane-holding time (T). It was the goal of this study to analyze the effect of shifting cell culture parameters (P, C, T) on the in vitro as well as in vivo effort of a regulated animal m-ACI. Autologous rabbit knee articular chondrocytes were seeded within bilayer collagen I/III 3-D matrices in variation of P, C and T. Each time, 2 PCT-identical by 2 PCT-identical cell-matrix-constructs (CMC)/animal were created. Simultaneously 2 (PCT-distinct) were re-implanted (CMC-e) autologous into artificial trochlear pristine chondral defects in vivo to remain for 12 weeks while the remaining 2 were harvested (CMC-i) for immediate in vitro analysis at the time of transplantation of their identical twins. mRNA of both, CMC-e regenerates and CMC-i membranes, was analyzed for Collagen-1,-2,-10, COMP, Aggrecan, Sox9 expression by use of a mixed linear model, multiple regression analysis. Generally, CMC-i values were higher than CMC-e values for differentiation targets; the opposite was true for dedifferentiation targets. Regarding individual gene expression, in vivo regenerate cell-matrix properties were significantly dependent on initial cell-matrix in vitro values as a sign of linearity. The parameter membrane-holding time (T) had strongest effects on the resulting mRNA expression with slightly less impact of the parameter passage (P), whereas cell yield (C) had clearly less effects. Noting differences between in vitro and in vivo data, in general, optimal expression patterns concerning chondrogenic differentiation were achieved by few passages, medium cellular yield, short membrane-holding time. Clinical m-ACI may benefit from optimal orchestration of the cell culture parameters passage, yield and time.
Collapse
Affiliation(s)
- Gian M Salzmann
- Department of Orthopaedic and Trauma Surgery, University Medical Center, Albert-Ludwigs University Freiburg, Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhang Y, Su J, Yu J, Bu X, Ren T, Liu X, Yao L. An essential role of discoidin domain receptor 2 (DDR2) in osteoblast differentiation and chondrocyte maturation via modulation of Runx2 activation. J Bone Miner Res 2011; 26:604-17. [PMID: 20734453 DOI: 10.1002/jbmr.225] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Discoidin domain receptor 2 (DDR2) belongs to receptor tyrosine kinase (RTK) family and is activated by collagen binding. Although the bone defects in Ddr2 null mice have been reported for a decade, the molecular mechanism remains unclear. This study sought to investigate the function and detailed mechanism of DDR2 in osteogenic and chondrogenic differentiation. Herein we found that in preosteoblastic cells, DDR2 activation was enhanced by osteogenic induction but was not paralleled with the alteration of DDR2 expression. Under differentiated condition, downregulation of endogenous DDR2 through specific shRNA dramatically repressed osteoblastic marker gene expression and osteogenic differentiation. Enforced expression of constitutively activated DDR2 increased the expression of bone markers in both undifferentiated and differentiated osteoblasts. Importantly, molecular evidence showed that DDR2 regulated the transactivity of Runx2, a master transcription factor involved in skeletal development, by modulating its phosphorylation. Analysis of candidate protein kinases indicated that extracellular signal-regulated kinase (ERK) activation is responsive to DDR2 signaling and involved in DDR2 regulation of Runx2 phosphorylation and transcriptional activity. Notably, a gain-of-function mutant of Runx2 with enhanced ERK-independent phosphorylation rescued the impaired osteogenic phenotypes observed in Ddr2-silenced cells, whereas a Runx2 mutant devoid of phosphorylation regulation by ERK inhibited DDR2 induction of osteogenesis. In addition, DDR2 facilitated Runx2 transactivation and type X collagen expression in hypertrophic chondrocytes. Thus this study reveals for the first time that DDR2 plays an essential role in osteoblast and chondrocyte differentiation. The mechanism disclosure may provide therapeutic targets for human genetic disorders caused by DDR2 deficiency.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Skotak M, Noriega S, Larsen G, Subramanian A. Electrospun cross-linked gelatin fibers with controlled diameter: the effect of matrix stiffness on proliferative and biosynthetic activity of chondrocytes cultured in vitro. J Biomed Mater Res A 2011; 95:828-36. [PMID: 20824648 DOI: 10.1002/jbm.a.32850] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Nanofibrous scaffolds were prepared from gelatin solutions and were further cross-linked with glutaraldehyde (GA). The fiber diameter was varied from 100 to 1000 nm by controlling the applied voltage (4-15 kV) and the concentration of the gelatin solution (4-15%). The tensile moduli and the tensile strength of the noncross-linked scaffolds varied from 20 to 120 MPa and 0.5 to 3.5 MPa, respectively. Cross-linking with GA led to an increase in both the tensile modulus and strength and correlated with cross-linker concentration. Gelatin-based matrices were characterized by Fourier transform infrared spectroscopy and differential scanning calorimetry. High cellular viabilities and rounded morphology of chondrocytes was observed at the end of 7 days in culture with added matrix deposition and flattening of cells at 15 days. Matrix stiffness was noted to impact cell densities and the expression of chondrocytic markers, especially aggrecan. The ratios of collagen-II (C-II) to collagen-I (C-I) of 0.62 and 1.33 were noted on gelatin nanofibrous scaffolds cross-linked with 0.1% GA at the end of 7 and 15 days in culture, respectively. C-II/C-I ratios of 1.30 and 2.58 were noted on scaffolds cross-linked with 1.0% GA at the end of 7 and 15 days in culture, respectively.
Collapse
Affiliation(s)
- Maciej Skotak
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Nebraska 68588-0643, USA
| | | | | | | |
Collapse
|
38
|
Chang SH, Hsiao YW, Lin HY. Low-frequency electromagnetic field exposure accelerates chondrocytic phenotype expression on chitosan substrate. Orthopedics 2011; 34:20. [PMID: 21210623 DOI: 10.3928/01477447-20101123-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
To find whether low-frequency pulsed electromagnetic fields help repair larger cartilage defects with the assistance of tissue-engineered scaffolds, we tested their effect on the behavior of chondrocyte cells cultured on chitosan films. Primary porcine chondrocytes growing on chitosan films were exposed to low-frequency pulsed electromagnetic fields (frequency=75 Hz; impulse width=1.3 ms; strength=1.8-3 mT) 2 hours a day for 3 weeks. The cells that were not exposed to low-frequency pulsed electromagnetic fields served as controls. For 3 weeks, cell proliferation, viability, and expressions of type II collagen and glycosaminoglycan were measured weekly. Cell morphology and histological stains of glycosaminoglycan and type II collagen were performed at the end of the test. The cell proliferation and viability of the low-frequency pulsed electromagnetic fields group and the control were similar each week. By the end of the third week, cells in the low-frequency pulsed electromagnetic fields group deposited 28% more glycosaminoglycan than the control cells. The amounts of type II collagen deposited in the low-frequency pulsed electromagnetic fields group were 24% and 27% higher than those of the control group by week 2 and 3, respectively. Histological and immunohistochemical analyses confirmed the releases of glycosaminoglycan and type II collagen. Cells from both groups grew in aggregates and possessed a spherical shape after 3 weeks. These results suggest that low-frequency pulsed electromagnetic fields can enhance extracellular matrix production on chitosan substrate. Combining tissue engineering and low-frequency pulsed electromagnetic fields could further accelerate cartilage repair.
Collapse
|
39
|
Bölgen N, Yang Y, Korkusuz P, Güzel E, El Haj AJ, Pişkin E. 3D ingrowth of bovine articular chondrocytes in biodegradable cryogel scaffolds for cartilage tissue engineering. J Tissue Eng Regen Med 2010; 5:770-9. [PMID: 22002920 DOI: 10.1002/term.375] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 09/02/2010] [Indexed: 11/08/2022]
Abstract
A feasibility study was undertaken to examine the potential of biodegradable HEMA-lactate-dextran (HEMA-LLA-D)-based cryogels as scaffolds for cartilage tissue engineering. This was a preliminary in vitro study giving essential information on the biocompatibility of cryogels with cartilage cells. HEMA-lactate (HEMA-LLA) and HEMA-LLA-D were synthesized and characterized by different techniques. Cryogel scaffolds with supermacroporous structures were produced by cryogenic treatment of these macromers. Chondrocytes obtained from bovine articular cartilage were seeded onto cylindrical cryogels and cultured. The samples were examined by several microcopical techniques for cell viability and morphological analyses were performed at two culture points. Histological study of the constructs revealed the cells' growth on the surface and within the scaffolds. Confocal microscopical images demonstrated that the majority of live vs. dead cells had been attached to and integrated with the pores of the scaffold. SEM analysis showed round to oval-shaped chondrocytic cells interconnected with each other by communicating junctions. The chondrocytes rapidly proliferated in the cryogels, manifesting that they fully covered the scaffold surface after 9 days and almost filled the spaces in the pores of the scaffold after 15 days of culture. Chondrocytes secreted significant amount of extracellular matrix in the scaffolds and exhibited highly interconnective morphology. Light and transmission electron microscopy revealed groups of active cartilage cells closely apposed to the cryogel. We concluded that cryogel scaffolds could be excellent candidates for cartilage tissue regeneration with their extraordinary properties, including soft, elastic nature, highly open interconnected pore structure and very rapid, controllable swellability.
Collapse
Affiliation(s)
- N Bölgen
- Department of Chemical Engineering, Faculty of Engineering, Mersin University, Mersin, Turkey.
| | | | | | | | | | | |
Collapse
|
40
|
Stenhamre H, Nannmark U, Lindahl A, Gatenholm P, Brittberg M. Influence of pore size on the redifferentiation potential of human articular chondrocytes in poly(urethane urea) scaffolds. J Tissue Eng Regen Med 2010; 5:578-88. [DOI: 10.1002/term.350] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 07/08/2010] [Indexed: 01/16/2023]
|
41
|
Lee TJ, Bhang SH, La WG, Yang HS, Seong JY, Lee H, Im GI, Lee SH, Kim BS. Spinner-flask culture induces redifferentiation of de-differentiated chondrocytes. Biotechnol Lett 2010; 33:829-36. [PMID: 21125413 DOI: 10.1007/s10529-010-0488-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 11/17/2010] [Indexed: 01/17/2023]
Abstract
Implantation of chondrocytes isolated from patients and expanded in number in vitro is being used to treat patients with cartilage injuries. However, chondrocytes de-differentiate during culture with several passages, and cartilage regenerated by implantation of de-differentiated chondrocytes may be suboptimal. Here, we show that a spinner-flask culture system induces formation of chondrocyte aggregates and redifferentiate de-differentiated chondrocytes. Spinner-flask cultures induced the aggregate formation of chondrocytes with passage 1 or 4. Importantly, spinner-flask cultures induced redifferentiation of the de-differentiated chondrocytes, as type I collagen expression was significantly lower and type II collagen expression was significantly higher in spinner flask-cultured chondrocytes than in monolayer-cultured chondrocytes. This system is easily scalable and could be feasible for clinical setting.
Collapse
Affiliation(s)
- Tae-Jin Lee
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Vickers SM, Gotterbarm T, Spector M. Cross-linking affects cellular condensation and chondrogenesis in type II collagen-GAG scaffolds seeded with bone marrow-derived mesenchymal stem cells. J Orthop Res 2010; 28:1184-92. [PMID: 20225321 DOI: 10.1002/jor.21113] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The formation of cartilaginous tissue by chondroprogenitor cells, whether in vivo or in vitro, appears to require a critical initial stage of "condensation" in which intercellular space is reduced through an aggregation of cells, leading to development of cell-to-cell junctions followed by chondrocytic differentiation. The objective of this study was to investigate the association of aggregation (condensation) of mesenchymal stem cell (MSCs) and chondrogenesis in vitro. Previous work with chondrocytes indicated that the cross-link density and related cell-mediated contraction of collagen scaffolds significantly affects cartilaginous tissue formation within the cell-seeded construct. Based on this finding, we hypothesized that the cell-aggregating effect of the contraction of MSC-seeded collagen scaffolds of lower cross-link density favors chondrogenesis; scaffolds of higher cross-link density, which resist cell-mediated contraction, would demonstrate a lower cell number density (i.e., subcritical packing density) and less cartilage formation. Type II collagen-GAG scaffolds, chemically cross-linked to achieve a range of cross-link densities, were seeded with caprine MSCs and cultured for 4 weeks. Constructs with low cross-link densities experienced cell-mediated contraction, increased cell number densities, and a greater degree of chondrogenesis (indicated by the chondrocytic morphology of cells, and synthesis of GAG and type II collagen) compared to more highly cross-linked scaffolds that resisted cellular contraction. These results provide a foundation for further investigation of the mechanisms by which condensation of mesenchymal cells induces chondrogenesis in this in vitro model, and may inform cross-linking protocols for collagen scaffolds for use in cartilage tissue engineering.
Collapse
Affiliation(s)
- Scott M Vickers
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
43
|
Babalola OM, Bonassar LJ. Effects of Seeding Density on Proteoglycan Assembly of Passaged Mesenchymal Stem Cells. Cell Mol Bioeng 2010. [DOI: 10.1007/s12195-010-0107-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
44
|
Zhang L, Spector M. Comparison of three types of chondrocytes in collagen scaffolds for cartilage tissue engineering. Biomed Mater 2009; 4:045012. [PMID: 19636108 DOI: 10.1088/1748-6041/4/4/045012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The objective of this study was to compare the chondrogenesis in type I and II collagen scaffolds seeded with chondrocytes from three types of cartilage, after four weeks of culture: auricular (AU), articular (AR) and meniscal (ME). Related aims were to investigate the expression of a contractile muscle actin isoform, alpha-smooth muscle actin (SMA), in the cells in the scaffold and to determine the presence of a lubricating glycoprotein, lubricin, in the constructs. Adult goat AU, AR and ME chondrocytes were seeded into two types of collagen scaffolds: type II collagen and type I/III collagen. After four weeks of culture, the constructs were prepared for histochemical and immunohistochemical analysis of the distribution of glycosaminoglycan (GAG), types I and II collagen, elastin, SM and lubricin. AU constructs contained substantially more tissue than the AR and ME samples. The AU constructs exhibited neocartilage, but no elastin. There were no notable differences between the type I and II collagen scaffolds. Novel findings were the expression of SMA by the AU cells in the scaffolds and the presence of lubricin in the AR and AU constructs. AU cells have the capability to produce cartilage in collagen scaffolds under conditions in which there is little histogenesis by AR and ME cells.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Tissue Engineering Center, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | | |
Collapse
|
45
|
Saddiq ZA, Barbenel JC, Grant MH. The mechanical strength of collagen gels containing glycosaminoglycans and populated with fibroblasts. J Biomed Mater Res A 2009; 89:697-706. [DOI: 10.1002/jbm.a.32007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
46
|
Richter A, Hauschild G, Murua Escobar H, Nolte I, Bullerdiek J. Application of high-mobility-group-A proteins increases the proliferative activity of chondrocytes in vitro. Tissue Eng Part A 2009; 15:473-7. [PMID: 18721076 DOI: 10.1089/ten.tea.2007.0308] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The low capability of self-repair in hyaline cartilage tissue and chondrocytes de-differentiating when grown in vitro (e.g., for tissue engineering approaches) limits articular cartilage repair. It has been shown that the embryonic architectural transcription factors of the high-mobility-group-A (HMGA) protein family affect the regulation of cell differentiation by influencing the state of cell chromatin and are involved in hyaline cartilage development by affecting the expression of chondrocyte-specific marker genes. Thus, the control of cartilage cell proliferation and differentiation by HMGA proteins promises to be an important aspect in cartilage tissue repair. To elucidate the effects on the proliferative activity of hyaline chondrocytes, HMGA proteins were recombinantly expressed, highly purified, and applied to porcine hyaline cartilage cells growing in in vitro monolayer cell culture. Direct application of HMGA1a, HMGA1b, and HMGA2 proteins onto porcine chondrocytes was shown to have a highly significant influence on cell proliferation. Greater proliferation of chondrocytes was achieved than in the untreated control group, indicating a promising approach to enhancing cartilage tissue repair.
Collapse
Affiliation(s)
- Andreas Richter
- Center for Human Genetics, University of Bremen, Bremen, Germany
| | | | | | | | | |
Collapse
|
47
|
Nah SS, Choi IY, Lee CK, Oh JS, Kim YG, Moon HB, Yoo B. Effects of advanced glycation end products on the expression of COX-2, PGE2 and NO in human osteoarthritic chondrocytes. Rheumatology (Oxford) 2008; 47:425-31. [DOI: 10.1093/rheumatology/kem376] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
48
|
Regulatory Effects of Mechanical Strain on the Chondrogenic Differentiation of MSCs in a Collagen-GAG Scaffold: Experimental and Computational Analysis. Ann Biomed Eng 2007; 36:185-94. [DOI: 10.1007/s10439-007-9416-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 11/26/2007] [Indexed: 11/27/2022]
|
49
|
Steinert AF, Palmer GD, Capito R, Hofstaetter JG, Pilapil C, Ghivizzani SC, Spector M, Evans CH. Genetically enhanced engineering of meniscus tissue using ex vivo delivery of transforming growth factor-beta 1 complementary deoxyribonucleic acid. ACTA ACUST UNITED AC 2007; 13:2227-37. [PMID: 17561802 DOI: 10.1089/ten.2006.0270] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
To investigate the use of a scaffold seeded with genetically modified meniscal cells or mesenchymal stem cells (MSCs) for the healing of meniscal lesions, primary meniscus cells and bone marrow-derived MSCs were isolated from bovine calves and transduced with first-generation adenoviral vectors encoding green fluorescent protein, luciferase, or transforming growth factor (TGF)-beta1 complementary deoxyribonucleic acid (cDNA). The genetically modified cells were seeded in type I collagen-glycosaminoglycan (GAG) matrices and transplanted into tears of the avascular zone of bovine menisci. After 3 weeks of in vitro culture, constructs and repair tissues were analyzed histologically, biochemically, and using reverse transcriptase polymerase chain reaction. Recombinant adenovirus readily transduced meniscal cells and MSCs, and transgene expression remained high after the cells were incorporated into collagen-GAG matrices. Transfer of TGF-beta1 cDNA increased cellularitiy and the synthesis of GAG/DNA [microg/microg]. It also led to stronger staining for proteoglycans and type II collagen and enhanced expression of meniscal genes. Transplantation of the TGF-beta1 transduced constructs into meniscal lesions of the avascular zone resulted in filling of the lesions with repair tissue after 3 weeks of in vitro culture. These results indicate that TGF-beta1 cDNA delivery may affect cell-based meniscus repair approaches in vivo.
Collapse
Affiliation(s)
- Andre F Steinert
- Center for Molecular Orthopedics, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wu YN, Yang Z, Hui JHP, Ouyang HW, Lee EH. Cartilaginous ECM component-modification of the micro-bead culture system for chondrogenic differentiation of mesenchymal stem cells. Biomaterials 2007; 28:4056-67. [PMID: 17590431 DOI: 10.1016/j.biomaterials.2007.05.039] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 05/29/2007] [Indexed: 01/22/2023]
Abstract
In this study a 3-D alginate microbead platform was coated with cartilaginous extracellular matrix (ECM) components to emulate chondrogenic microenvironment in vivo for the differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). BMSCs were seeded onto the microbead surface and the effect of the modified microbead on BMSC adhesion, proliferation and chondrogenic differentiation was studied, and compared to chondrogenesis in conventional pellet culture. Our results indicated that microbead system promoted BMSC proliferation and protein deposition resulting in the formation of bigger aggregates compared to conventional pellet culture. Analysis of the aggregates indicated that chondroitin sulfate (CS)- and Col2-coated microbeads enhanced the chondrogenic differentiation of hBMSCs, with increasing formation of glycosaminoglycan (GAG) and collagen II deposition in histology, immunohistochemistry and real time PCR analysis. In addition, Col2-coated microbeads resulted in hypertrophic maturation of the differentiated chondrocytes, similar to conventional pellet culture, while CS-coated microbeads were able to retain the pre-hypertrophy state of the differentiated cells. Our result suggested that provision of suitable cartilaginous microenvironment in a 3-D system can promote the chondrogenic differentiation of BMSC and influence the phenotype of resulting chondrocytes. Our microbead system provides an easy method of processing a 3-D alginate system that allows the possibility of scaling up chondrogenic pellet production for clinical application, while the modifiable microbeads also provide an adjustable 3-D platform for the study of co-interaction of ECM and differentiation factors during the stem cell differentiation.
Collapse
Affiliation(s)
- Ying-Nan Wu
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore NUS Tissue Engineering Program (NUSTEP), National University Hospital, Lower Kent Ridge Road, Singapore 119074, Republic of Singapore
| | | | | | | | | |
Collapse
|