51
|
Martin-Pena A, Porter R, Plumton G, McCarrel T, Morton A, Guijarro M, Ghivizzani S, Sharma B, Palmer G. Lentiviral-based reporter constructs for profiling chondrogenic activity in primary equine cell populations. Eur Cell Mater 2018; 36:156-170. [PMID: 30311630 PMCID: PMC6788286 DOI: 10.22203/ecm.v036a12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Successful clinical translation of mesenchymal stem cell (MSC)-based therapies for cartilage repair will likely require the implementation of standardised protocols and broadly applicable tools to facilitate the comparisons among cell types and chondroinduction methods. The present study investigated the utility of recombinant lentiviral reporter vectors as reliable tools for comparing chondrogenic potential among primary cell populations and distinguishing cellular-level variations of chondrogenic activity in widely used three-dimensional (3D) culture systems. Primary equine MSCs and chondrocytes were transduced with vectors containing combinations of fluorescent and luciferase reporter genes under constitutive cytomeglavirus (CMV) or chondrocyte-lineage (Col2) promoters. Reporter activity was measured by fluorescence imaging and luciferase assay. In 3D cultures of MSC aggregates and polyethylene glycol-hyaluronic acid (PEG-HA) hydrogels, transforming growth factor beta 3 (TGF-β3)-mediated chondroinduction increased Col2 reporter activity, demonstrating close correlation with histology and mRNA expression levels of COL2A1 and SOX9. Comparison of chondrogenic activities among MSC populations using a secretable luciferase reporter revealed enhanced chondrogenesis in bone-marrow-derived MSCs relative to MSC populations from synovium and adipose tissues. A dual fluorescence reporter - enabling discrimination of highly chondrogenic (Col2-GFP) cells within an MSC population (CMV-tdTomato) - revealed marked heterogeneity in differentiating aggregate cultures and identified chondrogenic cells in chondrocyte-seeded PEG-HA hydrogels after 6 weeks in a subcutaneous implant model - indicating stable, long-term reporter expression in vivo. These results suggested that lentiviral reporter vectors may be used to address fundamental questions regarding chondrogenic activity in chondroprogenitor cell populations and accelerate clinical translation of cell-based cartilage repair strategies.
Collapse
Affiliation(s)
- A. Martin-Pena
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA
| | - R.M. Porter
- Department of Orthopaedics, University of Arkansas, Little
Rock, Arkansas, USA
| | - G Plumton
- Department of Biomedical Engineering, University of
Florida, Gainesville, Florida USA
| | - T.M. McCarrel
- College of Veterinary Sciences, University of Florida,
Gainesville, Florida USA
| | - A.J. Morton
- College of Veterinary Sciences, University of Florida,
Gainesville, Florida USA
| | - M.V. Guijarro
- Department of Anatomy and Cell Biology, University of
Florida, Gainesville, Florida USA
| | - S.C. Ghivizzani
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA
| | - B. Sharma
- Department of Orthopaedics, University of Arkansas, Little
Rock, Arkansas, USA
| | - G.D. Palmer
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA,Address for correspondence: Glyn Palmer, Ph.D,
Dept of Orthopaedics and Rehabilitation, University of Florida, 1600 SW Archer
Rd, MSB, M2-235, Gainesville, FL 32610, Telephone: +1 352 273 7087, Fax: +1 352
273 7427,
| |
Collapse
|
52
|
Newell K, Chitty J, Henson FM. "Patient reported outcomes" following experimental surgery-using telemetry to assess movement in experimental ovine models. J Orthop Res 2018; 36:1498-1507. [PMID: 29087600 PMCID: PMC6032879 DOI: 10.1002/jor.23790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/21/2017] [Indexed: 02/04/2023]
Abstract
Many potential treatments for orthopedic disease fail at the animal to human translational hurdle. One reason for this failure is that the majority of pre-clinical outcome measurements emphasize structural changes, such as gross morphology and histology, and do not address pain or its alleviation, which is a key component of treatment success in man. With increasing emphasis on "patient reported outcome measurements (PROM)" in clinical practice, in this study we have used two different telemetric methods (geolocation and Fitbark activity trackers, Kansas City, MO) to measure movement behavior, i.e., an indirect PROM, in an ovine osteoarthritis induction and an osteochondral defect model performed in adult female Welsh Mountain sheep. This study demonstrates that both systems can be used to track movement and activity of experimental sheep before and after surgery and that the Geolocator system recorded a decrease in distance moved and activity at the end of the experimental period in both models. The Fitbark activity tracker also recorded significant alterations in movement behavior at the end of these studies and this method of recording showed a correlation between Fitbark data and radiography, macroscopic and histological scoring (well recognized outcome measurements), particularly in animals with large (10 mm) defects, i.e., more severe pathology. These results suggest that telemetry is able to track movement behavior in experimental sheep and that the methodology should be considered for inclusion in outcome measures in preclinical orthopedic research. © 2017 The Authors. Journal of Orthopaedic Research® Published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 36:1498-1507, 2018.
Collapse
Affiliation(s)
- Karin Newell
- Department of SurgeryUniversity of CambridgeCambridgeUnited Kingdom
| | | | - Frances M. Henson
- Department of SurgeryUniversity of CambridgeCambridgeUnited Kingdom,Department of Veterinary MedicineUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
53
|
Vilela CA, Correia C, da Silva Morais A, Santos TC, Gertrudes AC, Moreira ES, Frias AM, Learmonth DA, Oliveira P, Oliveira JM, Sousa RA, Espregueira-Mendes JD, Reis RL. In vitro
and in vivo
performance of methacrylated gellan gum hydrogel formulations for cartilage repair*. J Biomed Mater Res A 2018; 106:1987-1996. [DOI: 10.1002/jbm.a.36406] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/26/2018] [Accepted: 03/15/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Carlos A. Vilela
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho; Braga Portugal
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics; University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; Guimarães Portugal
- ICVS/3Bs-PT Government Associate Laboratory; Braga/Guimarães Portugal
- Orthopaedic Department; Hospital da Senhora da Oliveira Guimarães EPE; Guimarães Portugal
| | - Cristina Correia
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimarães Portugal
| | - Alain da Silva Morais
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics; University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; Guimarães Portugal
- ICVS/3Bs-PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Tírcia C. Santos
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics; University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; Guimarães Portugal
- ICVS/3Bs-PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Ana C. Gertrudes
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimarães Portugal
| | - Elsa S. Moreira
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimarães Portugal
| | - Ana M. Frias
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimarães Portugal
| | - David A. Learmonth
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimarães Portugal
| | - Pedro Oliveira
- ISUP-EPI Unit, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto; Porto Portugal
| | - Joaquim M. Oliveira
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics; University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; Guimarães Portugal
- ICVS/3Bs-PT Government Associate Laboratory; Braga/Guimarães Portugal
| | - Rui A. Sousa
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimarães Portugal
| | - João D. Espregueira-Mendes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho; Braga Portugal
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics; University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; Guimarães Portugal
- ICVS/3Bs-PT Government Associate Laboratory; Braga/Guimarães Portugal
- Clínica do Dragão, Espregueira-Mendes Sports Centre, FIFA Medical Centre of Excellence and D. Henrique Research Centre; Porto Portugal
| | - Rui L. Reis
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics; University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine; Guimarães Portugal
- ICVS/3Bs-PT Government Associate Laboratory; Braga/Guimarães Portugal
| |
Collapse
|
54
|
Stem Cells for Cartilage Repair: Preclinical Studies and Insights in Translational Animal Models and Outcome Measures. Stem Cells Int 2018. [PMID: 29535784 PMCID: PMC5832141 DOI: 10.1155/2018/9079538] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Due to the restricted intrinsic capacity of resident chondrocytes to regenerate the lost cartilage postinjury, stem cell-based therapies have been proposed as a novel therapeutic approach for cartilage repair. Moreover, stem cell-based therapies using mesenchymal stem cells (MSCs) or induced pluripotent stem cells (iPSCs) have been used successfully in preclinical and clinical settings. Despite these promising reports, the exact mechanisms underlying stem cell-mediated cartilage repair remain uncertain. Stem cells can contribute to cartilage repair via chondrogenic differentiation, via immunomodulation, or by the production of paracrine factors and extracellular vesicles. But before novel cell-based therapies for cartilage repair can be introduced into the clinic, rigorous testing in preclinical animal models is required. Preclinical models used in regenerative cartilage studies include murine, lapine, caprine, ovine, porcine, canine, and equine models, each associated with its specific advantages and limitations. This review presents a summary of recent in vitro data and from in vivo preclinical studies justifying the use of MSCs and iPSCs in cartilage tissue engineering. Moreover, the advantages and disadvantages of utilizing small and large animals will be discussed, while also describing suitable outcome measures for evaluating cartilage repair.
Collapse
|
55
|
Huwe LW, Brown WE, Hu JC, Athanasiou KA. Characterization of costal cartilage and its suitability as a cell source for articular cartilage tissue engineering. J Tissue Eng Regen Med 2018; 12:1163-1176. [PMID: 29286211 DOI: 10.1002/term.2630] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 11/23/2017] [Accepted: 12/11/2017] [Indexed: 12/16/2022]
Abstract
Costal cartilage is a promising donor source of chondrocytes to alleviate cell scarcity in articular cartilage tissue engineering. Limited knowledge exists, however, on costal cartilage characteristics. This study describes the characterization of costal cartilage and articular cartilage properties and compares neocartilage engineered with costal chondrocytes to native articular cartilage, all within a sheep model. Specifically, we (a) quantitatively characterized the properties of costal cartilage in comparison to patellofemoral articular cartilage, and (b) evaluated the quality of neocartilage derived from costal chondrocytes for potential use in articular cartilage regeneration. Ovine costal and articular cartilages from various topographical locations were characterized mechanically, biochemically, and histologically. Costal cartilage was stiffer in compression but softer and weaker in tension than articular cartilage. These differences were attributed to high amounts of glycosaminoglycans and mineralization and a low amount of collagen in costal cartilage. Compared to articular cartilage, costal cartilage was more densely populated with chondrocytes, rendering it an excellent chondrocyte source. In terms of tissue engineering, using the self-assembling process, costal chondrocytes formed articular cartilage-like neocartilage. Quantitatively compared via a functionality index, neocartilage achieved 55% of the medial condyle cartilage mechanical and biochemical properties. This characterization study highlighted the differences between costal and articular cartilages in native forms and demonstrated that costal cartilage is a valuable source of chondrocytes suitable for articular cartilage regeneration strategies.
Collapse
Affiliation(s)
| | - Wendy E Brown
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
56
|
Dias IR, Viegas CA, Carvalho PP. Large Animal Models for Osteochondral Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:441-501. [PMID: 29736586 DOI: 10.1007/978-3-319-76735-2_20] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Namely, in the last two decades, large animal models - small ruminants (sheep and goats), pigs, dogs and horses - have been used to study the physiopathology and to develop new therapeutic procedures to treat human clinical osteoarthritis. For that purpose, cartilage and/or osteochondral defects are generally performed in the stifle joint of selected large animal models at the condylar and trochlear femoral areas where spontaneous regeneration should be excluded. Experimental animal care and protection legislation and guideline documents of the US Food and Drug Administration, the American Society for Testing and Materials and the International Cartilage Repair Society should be followed, and also the specificities of the animal species used for these studies must be taken into account, such as the cartilage thickness of the selected defect localization, the defined cartilage critical size defect and the joint anatomy in view of the post-operative techniques to be performed to evaluate the chondral/osteochondral repair. In particular, in the articular cartilage regeneration and repair studies with animal models, the subchondral bone plate should always be taken into consideration. Pilot studies for chondral and osteochondral bone tissue engineering could apply short observational periods for evaluation of the cartilage regeneration up to 12 weeks post-operatively, but generally a 6- to 12-month follow-up period is used for these types of studies.
Collapse
Affiliation(s)
- Isabel R Dias
- Department of Veterinary Sciences, Agricultural and Veterinary Sciences School, University of Trás-os-Montes e Alto Douro (UTAD), Vila Real, Portugal. .,3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque da Ciência e Tecnologia, Zona Industrial da Gandra, Barco - Guimarães, 4805-017, Portugal. .,Department of Veterinary Medicine, ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Carlos A Viegas
- Department of Veterinary Sciences, Agricultural and Veterinary Sciences School, University of Trás-os-Montes e Alto Douro (UTAD), Vila Real, Portugal.,3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque da Ciência e Tecnologia, Zona Industrial da Gandra, Barco - Guimarães, 4805-017, Portugal.,Department of Veterinary Medicine, ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Pedro P Carvalho
- Department of Veterinary Medicine, University School Vasco da Gama, Av. José R. Sousa Fernandes 197, Lordemão, Coimbra, 3020-210, Portugal.,CIVG - Vasco da Gama Research Center, University School Vasco da Gama, Coimbra, Portugal
| |
Collapse
|
57
|
Su AW, Chen Y, Wailes DH, Wong VW, Cai S, Chen AC, Bugbee WD, Sah RL. Impact insertion of osteochondral grafts: Interference fit and central graft reduction affect biomechanics and cartilage damage. J Orthop Res 2018; 36:377-386. [PMID: 28682003 PMCID: PMC5756525 DOI: 10.1002/jor.23645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/13/2017] [Indexed: 02/04/2023]
Abstract
An osteochondral graft (OCG) is an effective treatment for articular cartilage and osteochondral defects. Impact of an OCG during insertion into the osteochondral recipient site (OCR) can cause chondrocyte death and matrix damage. The aim of the present study was to analyze the effects of graft-host interference fit and a modified OCG geometry on OCG insertion biomechanics and cartilage damage. The effects of interference fit (radius of OCG - radius of OCR), loose (0.00 mm), moderate (0.05 mm), tight (0.10 mm), and of a tight fit with OCG geometry modification (central region of decreased radius), were analyzed for OCG cylinders and OCR blocks from adult bovine knee joints with an instrumented drop tower apparatus. An increasingly tight (OCG - OCR) interference fit led to increased taps for insertion, peak axial force, graft cartilage axial compression, cumulative and total energy delivery to cartilage, lower time of peak axial force, lesser graft advancement during each tap, higher total crack length in the cartilage surface, and lower chondrocyte viability. The modified OCG, with reduction of diameter in the central area, altered the biomechanical insertion variables and biological consequences to be similar to those of the moderate interference fit scenario. Micro-computed tomography confirmed structural interference between the OCR bone and both the proximal and distal bone segments of the OCGs, with the central regions being slightly separated for the modified OCGs. These results clarify OCG insertion biomechanics and mechanobiology, and introduce a simple modification of OCGs that facilitates insertion with reduced energy while maintaining a structural interference fit. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:377-386, 2018.
Collapse
Affiliation(s)
- Alvin W. Su
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA,Program in Materials Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Yunchan Chen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Dustin H. Wailes
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Van W. Wong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Shengqiang Cai
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, USA,Program in Materials Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Albert C. Chen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - William D. Bugbee
- Center for Musculoskeletal Research, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, USA,Department of Orthopaedic Surgery, Scripps Clinic, La Jolla, CA, USA
| | - Robert L. Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA,Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, CA, USA,Program in Materials Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
58
|
Huwe LW, Brown WE, Hu JC, Athanasiou KA. Characterization of costal cartilage and its suitability as a cell source for articular cartilage tissue engineering. J Tissue Eng Regen Med 2017. [PMID: 29286211 DOI: 10.1002/term.2630.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Costal cartilage is a promising donor source of chondrocytes to alleviate cell scarcity in articular cartilage tissue engineering. Limited knowledge exists, however, on costal cartilage characteristics. This study describes the characterization of costal cartilage and articular cartilage properties and compares neocartilage engineered with costal chondrocytes to native articular cartilage, all within a sheep model. Specifically, we (a) quantitatively characterized the properties of costal cartilage in comparison to patellofemoral articular cartilage, and (b) evaluated the quality of neocartilage derived from costal chondrocytes for potential use in articular cartilage regeneration. Ovine costal and articular cartilages from various topographical locations were characterized mechanically, biochemically, and histologically. Costal cartilage was stiffer in compression but softer and weaker in tension than articular cartilage. These differences were attributed to high amounts of glycosaminoglycans and mineralization and a low amount of collagen in costal cartilage. Compared to articular cartilage, costal cartilage was more densely populated with chondrocytes, rendering it an excellent chondrocyte source. In terms of tissue engineering, using the self-assembling process, costal chondrocytes formed articular cartilage-like neocartilage. Quantitatively compared via a functionality index, neocartilage achieved 55% of the medial condyle cartilage mechanical and biochemical properties. This characterization study highlighted the differences between costal and articular cartilages in native forms and demonstrated that costal cartilage is a valuable source of chondrocytes suitable for articular cartilage regeneration strategies.
Collapse
Affiliation(s)
| | - Wendy E Brown
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
59
|
Sheu SY, Wang CH, Pao YH, Fu YT, Liu CH, Yao CH, Kuo TF. The effect of platelet-rich fibrin on autologous osteochondral transplantation: An in vivo porcine model. Knee 2017; 24:1392-1401. [PMID: 29037743 DOI: 10.1016/j.knee.2017.08.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/02/2017] [Accepted: 08/10/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND This work aimed to evaluate the efficacy of cartilage transplantation to the medial femoral condyle±platelet-rich fibrin (PRF) augmentation in a porcine model. The hypothesis of the study was that PRF may act as a bioactive cell scaffold to fill defects and enhance cartilage regeneration. METHODS Thirty-two knees of 16 miniature pigs were randomly assigned to four groups. The critical-size osteochondral defects (8x5mm) in femoral condyle of both knees were treated with one of the following: group 1-untreated controls; group 2-cartilage fragments alone; group 3-PRF alone; group 4-PRFT+cartilage fragments. After completion of the surgical implantation, the periosteal patch harvested from the proximal tibia was sutured onto the cartilage of the medial condyle to cover the implanted defects. Animals were sacrificed at six months after treatment. The regenerated cartilages were assessed by gross inspection and histological examination. RESULTS The best results were obtained with the repair tissue being hyaline-like cartilage (group 4). The grading score of histological evaluation demonstrated that group 4 had better matrix, cell distribution and cartilage mineralization than group 2 and group 3. PRF showed a positive effect on the cartilage repair; the procedure was more effective when PRF was combined with autologous chondrocytes. CONCLUSIONS This approach may provide a successfully employed technique to target cartilage defects in vivo. Larger groups and longer periods of study may provide more definitive and meaningful support for using this therapeutic approach as a new way of cartilage regeneration.
Collapse
Affiliation(s)
- S Y Sheu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Integrated Chinese and Western Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - C H Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Y H Pao
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Y T Fu
- Department of Chinese Medicine, Buddhist Tzu Chi General Hospital, Taichung Branch, Taichung, Taiwan; School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| | - C H Liu
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - C H Yao
- School of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan; Department of Biomedical Informatics, Asia University, Taichung, Taiwan
| | - T F Kuo
- Department of Post-Baccalaureate Veterinary Medicine, Asia University, Taichung, Taiwan.
| |
Collapse
|
60
|
Diederichs S, Renz Y, Hagmann S, Lotz B, Seebach E, Richter W. Stimulation of a calcified cartilage connecting zone by GDF-5-augmented fibrin hydrogel in a novel layered ectopic in vivo model. J Biomed Mater Res B Appl Biomater 2017; 106:2214-2224. [PMID: 29068568 DOI: 10.1002/jbm.b.34027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/26/2017] [Accepted: 10/02/2017] [Indexed: 01/31/2023]
Abstract
Tissue engineering approaches for reconstructing full-depth cartilage defects need to comprise a zone of calcified cartilage to tightly anchor cartilage constructs into the subchondral bone. Here, we investigated whether growth and differentiation factor-5-(GDF-5)-augmented fibrin hydrogel can induce a calcified cartilage-layer in vitro that seamlessly connects cartilage-relevant biomaterials with bone tissue. Human bone marrow stromal cells (BMSCs) were embedded in fibrin hydrogel and subjected to chondrogenesis with TGF-β with or without GDF-5 before constructs were implanted subcutaneously into SCID mice. A novel layered ectopic in vivo model was developed and GDF-5-augmented fibrin with BMSCs was used to glue hydrogel and collagen constructs onto bone disks to investigate formation of a calcified cartilage connecting zone. GDF-5 significantly enhanced ALP activity during in vitro chondrogenesis while ACAN and COL2A1 mRNA, proteoglycan-, collagen-type-II- and collagen-type-X-deposition remained similar to controls. Pellets pretreated with GDF-5 mineralized faster in vivo and formed more ectopic bone. In the novel layered ectopic model, GDF-5 strongly supported calcified cartilage formation that seamlessly connected with the bone. Pro-chondrogenic and pro-hypertrophic activity makes GDF-5-augmented fibrin an attractive bioactive hydrogel with high potential to stimulate a calcified cartilage connecting zone in situ that might promote integration of cartilage scaffolds with bone. Thus, GDF-5-augmented fibrin hydrogel promises to overcome poor fixation of biomaterials in cartilage defects facilitating their long-term regeneration. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 2214-2224, 2018.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Yvonne Renz
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Sébastien Hagmann
- Clinic for Orthopaedics and Trauma Surgery, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Benedict Lotz
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Elisabeth Seebach
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, 69118, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, 69118, Heidelberg, Germany
| |
Collapse
|
61
|
Levato R, Webb WR, Otto IA, Mensinga A, Zhang Y, van Rijen M, van Weeren R, Khan IM, Malda J. The bio in the ink: cartilage regeneration with bioprintable hydrogels and articular cartilage-derived progenitor cells. Acta Biomater 2017; 61:41-53. [PMID: 28782725 PMCID: PMC7116023 DOI: 10.1016/j.actbio.2017.08.005] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/24/2017] [Accepted: 08/03/2017] [Indexed: 01/28/2023]
Abstract
Cell-laden hydrogels are the primary building blocks for bioprinting, and, also termed bioinks, are the foundations for creating structures that can potentially recapitulate the architecture of articular cartilage. To be functional, hydrogel constructs need to unlock the regenerative capacity of encapsulated cells. The recent identification of multipotent articular cartilage-resident chondroprogenitor cells (ACPCs), which share important traits with adult stem cells, represents a new opportunity for cartilage regeneration. However, little is known about the suitability of ACPCs for tissue engineering, especially in combination with biomaterials. This study aimed to investigate the potential of ACPCs in hydrogels for cartilage regeneration and biofabrication, and to evaluate their ability for zone-specific matrix production. Gelatin methacryloyl (gelMA)-based hydrogels were used to culture ACPCs, bone marrow mesenchymal stromal cells (MSCs) and chondrocytes, and as bioinks for printing. Our data shows ACPCs outperformed chondrocytes in terms of neo-cartilage production and unlike MSCs, ACPCs had the lowest gene expression levels of hypertrophy marker collagen type X, and the highest expression of PRG4, a key factor in joint lubrication. Co-cultures of the cell types in multi-compartment hydrogels allowed generating constructs with a layered distribution of collagens and glycosaminoglycans. By combining ACPC- and MSC-laden bioinks, a bioprinted model of articular cartilage was generated, consisting of defined superficial and deep regions, each with distinct cellular and extracellular matrix composition. Taken together, these results provide important information for the use of ACPC-laden hydrogels in regenerative medicine, and pave the way to the biofabrication of 3D constructs with multiple cell types for cartilage regeneration or in vitro tissue models. STATEMENT OF SIGNIFICANCE Despite its limited ability to repair, articular cartilage harbors an endogenous population of progenitor cells (ACPCs), that to date, received limited attention in biomaterials and tissue engineering applications. Harnessing the potential of these cells in 3D hydrogels can open new avenues for biomaterial-based regenerative therapies, especially with advanced biofabrication technologies (e.g. bioprinting). This study highlights the potential of ACPCs to generate neo-cartilage in a gelatin-based hydrogel and bioink. The ACPC-laden hydrogel is a suitable substrate for chondrogenesis and data shows it has a bias in directing cells towards a superficial zone phenotype. For the first time, ACPC-hydrogels are evaluated both as alternative for and in combination with chondrocytes and MSCs, using co-cultures and bioprinting for cartilage regeneration in vitro. This study provides important cues on ACPCs, indicating they represent a promising cell source for the next generation of cartilage constructs with increased biomimicry.
Collapse
Affiliation(s)
- Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | - William R Webb
- Center for Nanohealth, Swansea University Medical School, Wales, United Kingdom
| | - Iris A Otto
- Department of Orthopaedics, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; Department of Plastic and Reconstructive Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anneloes Mensinga
- Department of Orthopaedics, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | - Yadan Zhang
- Center for Nanohealth, Swansea University Medical School, Wales, United Kingdom
| | - Mattie van Rijen
- Department of Orthopaedics, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | - René van Weeren
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Ilyas M Khan
- Center for Nanohealth, Swansea University Medical School, Wales, United Kingdom
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
62
|
McCarrel TM, Pownder SL, Gilbert S, Koff MF, Castiglione E, Saska RA, Bradica G, Fortier LA. Two-Year Evaluation of Osteochondral Repair with a Novel Biphasic Graft Saturated in Bone Marrow in an Equine Model. Cartilage 2017; 8:406-416. [PMID: 28934879 PMCID: PMC5613894 DOI: 10.1177/1947603516675913] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective To evaluate a biphasic cartilage repair device (CRD) for feasibility of arthroscopic implantation, safety, biocompatibility, and efficacy for long-term repair of large osteochondral defects. Methods The CRD was press-fit into defects (10 mm diameter, 10 mm deep) created in the femoral trochlea of 12 horses. In the contralateral limb, 10 mm diameter full-thickness chondral defects were treated with microfracture (MFX). Radiographs were obtained pre- and postoperatively, and at 4, 12, and 24 months. Repeat arthroscopy was performed at 4 and 12 months. Gross assessment, histology, mechanical testing, and magnetic resonance imaging (MRI) were performed at 24 months. Results The CRD was easily placed arthroscopically. There was no evidence of joint infection, inflammation, or degeneration. CRD-treated defects had significantly more sclerosis compared to MFX early ( P = 0.0006), but was not different at 24 months. CRD had better arthroscopic scores at 4 months compared to MFX ( P = 0.0069). At 24 months, there was no difference in repair tissue on histology or mechanical testing. Based on MRI, CRD repair tissue had less proteoglycan (deep P = 0.027, superficial P = 0.015) and less organized collagen (deep P = 0.028) compared to MFX. Cartilage surrounding MFX defects had more fissures compared to CRD. Conclusion The repair tissue formed after CRD treatment of a large osteochondral lesion is fibrocartilage similar to that formed in simple chondral defects treated with MFX. The CRD can be easily placed arthroscopically, is safe, and biocompatible for 24 months. The CRD results in improved early arthroscopic repair scores and may limit fissure formation in adjacent cartilage.
Collapse
Affiliation(s)
- Taralyn M. McCarrel
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, USA,Department of Large Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | | | | | | | | | - Ryan A. Saska
- DSM Biomedical d/b/a Kensey Nash Corporation, Exton, PA, USA
| | - Gino Bradica
- DSM Biomedical d/b/a Kensey Nash Corporation, Exton, PA, USA
| | - Lisa A. Fortier
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, USA,Lisa A. Fortier, VMC C3-181 Cornell University, College of Veterinary Medicine, Ithaca, NY 14853, USA.
| |
Collapse
|
63
|
Bauer C, Jeyakumar V, Niculescu-Morzsa E, Kern D, Nehrer S. Hyaluronan thiomer gel/matrix mediated healing of articular cartilage defects in New Zealand White rabbits-a pilot study. J Exp Orthop 2017; 4:14. [PMID: 28470629 PMCID: PMC5415448 DOI: 10.1186/s40634-017-0089-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/27/2017] [Indexed: 11/13/2022] Open
Abstract
Background Articular cartilage defects are limited to their regenerative potential in human adults. Our current study evaluates tissue regeneration in a surgically induced empty defect site with hyaluronan thiomer as a provisional scaffold in a gel/matrix combination without cells on rabbit models to restore tissue formation. Methods An osteochondral defect of 4 mm in diameter and 5 mm in depth was induced by mechanical drilling in the femoral center of the trochlea in 18 New Zealand White rabbits. Previously evaluated from an in vitro study hyaluronan thiomer matrix, and a hyaluronan thiomer gel was used to treat the defect. As a control, the defect was left untreated. During the whole study, rabbits were clinically examined and after 4 (n = 3) or 12 (n = 3) weeks, the rabbits were sacrificed. Joints were evaluated macroscopically (Brittberg score) and by histology (O’Driscoll score). Synovial cells from the synovial fluid smear were histopathologically evaluated. Results The healing of the defects varied intra-group wise at the first observation period. After 12 weeks the results concerning the cartilage repair score were inhomogeneous within each group, while the macroscopic analysis was more homogenous. In the synovial fluid smear, the mean score of infiltrated synovial and non-synovial cells was slightly increased after 4 weeks and slightly decreased after 12 weeks in both the treatment groups in comparison to the untreated control. Conclusions Taken together with results from the in vivo study indicated that implantation of hyaluronan thiomer as a combination of gel and matrix might enhance articular cartilage regeneration in an empty defect. Despite their benefits, the intrinsic healing capacity of New Zealand rabbits is a limitation for comparative test subject in pre-clinical models of cartilage defects.
Collapse
Affiliation(s)
- Christoph Bauer
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube-University Krems, Dr.-Karl-Dorrek-Strasse 30, Krems, Austria.
| | - Vivek Jeyakumar
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube-University Krems, Dr.-Karl-Dorrek-Strasse 30, Krems, Austria
| | - Eugenia Niculescu-Morzsa
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube-University Krems, Dr.-Karl-Dorrek-Strasse 30, Krems, Austria
| | - Daniela Kern
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube-University Krems, Dr.-Karl-Dorrek-Strasse 30, Krems, Austria
| | - Stefan Nehrer
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube-University Krems, Dr.-Karl-Dorrek-Strasse 30, Krems, Austria
| |
Collapse
|
64
|
Bolaños RV, Cokelaere S, McDermott JE, Benders K, Gbureck U, Plomp S, Weinans H, Groll J, van Weeren P, Malda J. The use of a cartilage decellularized matrix scaffold for the repair of osteochondral defects: the importance of long-term studies in a large animal model. Osteoarthritis Cartilage 2017; 25:413-420. [PMID: 27554995 PMCID: PMC7116104 DOI: 10.1016/j.joca.2016.08.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/15/2016] [Accepted: 08/11/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the effect of decellularized cartilage-derived matrix (CDM) scaffolds, by itself and as a composite scaffold with a calcium phosphate (CaP) base, for the repair of osteochondral defects. It was hypothesized that the chondral defects would heal with fibrocartilaginous tissue and that the composite scaffold would result in better bone formation. METHODS After an 8-week pilot experiment in a single horse, scaffolds were implanted in eight healthy horses in osteochondral defects on the medial trochlear ridge of the femur. In one joint a composite CDM-CaP scaffold was implanted (+P), in the contralateral joint a CDM only (-P) scaffold. After euthanasia at 6 months, tissues were analysed by histology, immunohistochemistry, micro-CT, biochemistry and biomechanical evaluation. RESULTS The 8-week pilot showed encouraging formation of bone and cartilage, but incomplete defect filling. At 6 months, micro-CT and histology showed much more limited filling of the defect, but the CaP component of the +P scaffolds was well integrated with the surrounding bone. The repair tissue was fibrotic with high collagen type I and low type II content and with no differences between the groups. There were also no biochemical differences between the groups and repair tissue was much less stiff than normal tissue (P < 0.0001). CONCLUSIONS The implants failed to produce reasonable repair tissue in this osteochondral defect model, although the CaP base in the -P group integrated well with the recipient bone. The study stresses the importance of long-term in vivo studies to assess the efficacy of cartilage repair techniques.
Collapse
Affiliation(s)
- R.A. Vindas Bolaños
- Cátedra de Cirugìa de Especies Mayores, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - S.M. Cokelaere
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - J.M. Estrada McDermott
- Cátedra de Cirugìa de Especies Mayores, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - K.E.M. Benders
- Department of Orthopaedics, Division of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - U. Gbureck
- Department of Functional Materials in Medicine and Dentistry, University of Wurzburg, Wurzburg, Germany
| | - S.G.M. Plomp
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - H. Weinans
- Department of Orthopaedics, Division of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J. Groll
- Department of Functional Materials in Medicine and Dentistry, University of Wurzburg, Wurzburg, Germany
| | - P.R. van Weeren
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| | - J. Malda
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands,Department of Orthopaedics, Division of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands,Address correspondence and reprint requests to: J. Malda, Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, The Netherlands.
| |
Collapse
|
65
|
Hindle P, Baily J, Khan N, Biant LC, Simpson AHR, Péault B. Perivascular Mesenchymal Stem Cells in Sheep: Characterization and Autologous Transplantation in a Model of Articular Cartilage Repair. Stem Cells Dev 2016; 25:1659-1669. [PMID: 27554322 DOI: 10.1089/scd.2016.0165] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Previous research has indicated that purified perivascular stem cells (PSCs) have increased chondrogenic potential compared to conventional mesenchymal stem cells (MSCs) derived in culture. This study aimed to develop an autologous large animal model for PSC transplantation and to specifically determine if implanted cells are retained in articular cartilage defects. Immunohistochemistry and fluorescence-activated cell sorting were used to ascertain the reactivity of anti-human and anti-ovine antibodies, which were combined and used to identify and isolate pericytes (CD34-CD45-CD146+) and adventitial cells (CD34+CD45-CD146-). The purified cells demonstrated osteogenic, adipogenic, and chondrogenic potential in culture. Autologous ovine PSCs (oPSCs) were isolated, cultured, and efficiently transfected using a green fluorescence protein (GFP) encoding lentivirus. The cells were implanted into articular cartilage defects on the medial femoral condyle using hydrogel and collagen membranes. Four weeks following implantation, the condyle was explanted and confocal laser scanning microscopy demonstrated the presence of oPSCs in the defect repaired with the hydrogel. These data suggest the testability in a large animal of native MSC autologous grafting, thus avoiding possible biases associated with xenotransplantation. Such a setting will be used in priority for indications in orthopedics, at first to model articular cartilage repair.
Collapse
Affiliation(s)
- Paul Hindle
- 1 MRC Centre for Regenerative Medicine, The University of Edinburgh , Edinburgh, United Kingdom
- 2 Department of Trauma and Orthopaedic Surgery, The Royal Infirmary of Edinburgh , Edinburgh, United Kingdom
| | - James Baily
- 1 MRC Centre for Regenerative Medicine, The University of Edinburgh , Edinburgh, United Kingdom
| | - Nusrat Khan
- 1 MRC Centre for Regenerative Medicine, The University of Edinburgh , Edinburgh, United Kingdom
| | - Leela C Biant
- 2 Department of Trauma and Orthopaedic Surgery, The Royal Infirmary of Edinburgh , Edinburgh, United Kingdom
| | - A Hamish R Simpson
- 2 Department of Trauma and Orthopaedic Surgery, The Royal Infirmary of Edinburgh , Edinburgh, United Kingdom
| | - Bruno Péault
- 1 MRC Centre for Regenerative Medicine, The University of Edinburgh , Edinburgh, United Kingdom
- 3 The University of California , Los Angeles, Los Angeles, California
| |
Collapse
|
66
|
Otto I, Breugem C, Malda J, Bredenoord A. Ethical considerations in the translation of regenerative biofabrication technologies into clinic and society. Biofabrication 2016; 8:042001. [PMID: 27716629 PMCID: PMC7116024 DOI: 10.1088/1758-5090/8/4/042001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Biofabrication technologies have the potential to improve healthcare by providing highly advanced and personalized biomedical products for research, treatment and prevention. As the combining of emerging techniques and integrating various biological and synthetic components becomes increasingly complex, it is important that relevant stakeholders anticipate the translation of biofabricated 3D tissue products into patients and society. Ethics is sometimes regarded as a brake on scientific progress, yet from our perspective, ethics in parallel with research anticipates societal impacts of emerging technologies and stimulates responsible innovation. For the ethical assessment, the biofabrication field benefits from similarities to regenerative medicine and an increasing ethical awareness in the development of tissue-engineered products. However, the novelty of the technology itself, the increase in attainable structural complexity, and the potential for automation and personalization are distinguishing facets of biofabrication that call for a specific exploration of the ethics of biofabrication. This review aims to highlight important points of existing ethical discussions, as well as to call attention to emerging issues specific to 3D biofabrication in bench and bedside research and the translation to society.
Collapse
Affiliation(s)
- I.A. Otto
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Plastic and Reconstructive Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - C.C. Breugem
- Department of Plastic and Reconstructive Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J. Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Equine Sciences, Faculty of Veterinary Science, Utrecht University, Utrecht, The Netherlands
| | - A.L. Bredenoord
- Department of Medical Humanities, Julius Center, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
67
|
Malda J, Boere J, van de Lest CHA, van Weeren PR, Wauben MHM. Extracellular vesicles — new tool for joint repair and regeneration. Nat Rev Rheumatol 2016; 12:243-9. [PMID: 26729461 DOI: 10.1038/nrrheum.2015.170] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell-derived extracellular vesicles (EVs), present in synovial fluid and cartilage extracellular matrix (ECM), are involved in joint development and in the regulation of joint homeostasis. Although the exact function of EVs in these processes remains incompletely defined, the knowledge already acquired in this field suggests a role for these EVs as biomarkers of joint disease, and as a new tool to restore joint homeostasis and enhance articular tissue regeneration. In addition to direct injection of therapeutic EVs into the target site, surface coating of scaffolds and embedding of EVs in hydrogels might also lead to novel therapeutic possibilities. Based on the existing literature of EVs in synovial fluid and articular tissues, and investigation of the molecular factors (including microRNAs) active in joint homeostasis (or during its disturbance), we postulate novel perspectives for the implementation of EVs as a regenerative medicine approach in joint repair.
Collapse
|
68
|
Boere J, van de Lest CHA, Libregts SFWM, Arkesteijn GJA, Geerts WJC, Nolte-'t Hoen ENM, Malda J, van Weeren PR, Wauben MHM. Synovial fluid pretreatment with hyaluronidase facilitates isolation of CD44+ extracellular vesicles. J Extracell Vesicles 2016; 5:31751. [PMID: 27511891 PMCID: PMC4980521 DOI: 10.3402/jev.v5.31751] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/07/2016] [Accepted: 07/03/2016] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) in synovial fluid (SF) are gaining increased recognition as important factors in joint homeostasis, joint regeneration, and as biomarkers of joint disease. A limited number of studies have investigated EVs in SF samples of patients with joint disease, but knowledge on the role of EVs in healthy joints is lacking. In addition, no standardized protocol is available for isolation of EVs from SF. Based on the high viscosity of SF caused by high concentrations of hyaluronic acid (HA) - a prominent extracellular matrix component - it was hypothesized that EV recovery could be optimized by pretreatment with hyaluronidase (HYase). Therefore, the efficiency of EV isolation from healthy equine SF samples was tested by performing sequential ultracentrifugation steps (10,000g, 100,000g and 200,000g) in the presence or absence of HYase. Quantitative EV analysis using high-resolution flow cytometry showed an efficient recovery of EVs after 100,000g ultracentrifugation, with an increased yield of CD44+ EVs when SF samples were pretreated with HYase. Morphological analysis of SF-derived EVs with cryo-transmission-electron microscopy did not indicate damage by high-speed ultracentrifugation and revealed that most EVs are spherical with a diameter of 20-200 nm. Further protein characterization by Western blotting revealed that healthy SF-derived EVs contain CD9, Annexin-1, and CD90/Thy1.1. Taken together, these data suggest that EV isolation protocols for body fluids that contain relatively high amounts of HA, such as SF, could benefit from treatment of the fluid with HYase prior to ultracentrifugation. This method facilitates recovery and detection of CD44+ EVs within the HA-rich extracellular matrix. Furthermore, based on the findings presented here, it is recommended to sediment SF-derived EVs with at least 100,000g for optimal EV recovery.
Collapse
Affiliation(s)
- Janneke Boere
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Chris H A van de Lest
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Sten F W M Libregts
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Ger J A Arkesteijn
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Willie J C Geerts
- Department of Cryo-Electron Microscopy, Bijvoet Center for Biomolecular Research, Utrecht, Netherlands
| | - Esther N M Nolte-'t Hoen
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jos Malda
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - P René van Weeren
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Marca H M Wauben
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands;
| |
Collapse
|
69
|
Pfeifer CG, Fisher MB, Carey JL, Mauck RL. Impact of guidance documents on translational large animal studies of cartilage repair. Sci Transl Med 2016; 7:310re9. [PMID: 26491080 DOI: 10.1126/scitranslmed.aac7019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Promising therapies for cartilage repair are translated through large animal models toward human application. To guide this work, regulatory agencies publish recommendations ("guidance documents") to direct pivotal large animal studies. These are meant to aid in study design, outline metrics for judging efficacy, and facilitate comparisons between studies. To determine the penetrance of these documents in the field, we synthesized the recommendations of the American Society for Testing and Materials, U.S. Food and Drug Administration, and European Medicines Agency into a scoring system and performed a systematic review of the past 20 years of preclinical cartilage repair studies. Our hypothesis was that the guidance documents would have a significant impact on how large animal cartilage repair studies were performed. A total of 114 publications meeting our inclusion criteria were reviewed for adherence to 24 categories extracted from the guidance documents, including 11 related to study design and description and 13 related to study outcomes. Overall, a weak positive trend was observed over time (P = 0.004, R(2) = 0.07, slope = 0.63%/year), with overall adherence (the sum of study descriptors and outcomes) ranging from 32 ± 16% to 58 ± 14% in any individual year. There was no impact of the publication of the guidance documents on adherence (P = 0.264 to 0.50). Given that improved adherence would expedite translation, we discuss the reasons for poor adherence and outline approaches to increase and promote their more widespread adoption.
Collapse
Affiliation(s)
- Christian G Pfeifer
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA. Translational Musculoskeletal Research Center, Philadelphia Veterans Affairs Medical Center, Philadelphia, PA 19104, USA. Department of Trauma Surgery, Regensburg University Medical Center, 93053 Regensburg, Germany
| | - Matthew B Fisher
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA. Translational Musculoskeletal Research Center, Philadelphia Veterans Affairs Medical Center, Philadelphia, PA 19104, USA. Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, and North Carolina State University, Raleigh, NC 27695, USA
| | - James L Carey
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA. Translational Musculoskeletal Research Center, Philadelphia Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA. Translational Musculoskeletal Research Center, Philadelphia Veterans Affairs Medical Center, Philadelphia, PA 19104, USA. Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA. Collaborative Research Partner, Acute Cartilage Injury Program of the AO Foundation, 7270 Davos, Switzerland.
| |
Collapse
|
70
|
Caminal M, Peris D, Fonseca C, Barrachina J, Codina D, Rabanal RM, Moll X, Morist A, García F, Cairó JJ, Gòdia F, Pla A, Vives J. Cartilage resurfacing potential of PLGA scaffolds loaded with autologous cells from cartilage, fat, and bone marrow in an ovine model of osteochondral focal defect. Cytotechnology 2016; 68:907-19. [PMID: 25595211 PMCID: PMC4960140 DOI: 10.1007/s10616-015-9842-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 01/08/2015] [Indexed: 12/11/2022] Open
Abstract
Current developments in tissue engineering strategies for articular cartilage regeneration focus on the design of supportive three-dimensional scaffolds and their use in combination with cells from different sources. The challenge of translating initial successes in small laboratory animals into the clinics involves pilot studies in large animal models, where safety and efficacy should be investigated during prolonged follow-up periods. Here we present, in a single study, the long-term (up to 1 year) effect of biocompatible porous scaffolds non-seeded and seeded with fresh ex vivo expanded autologous progenitor cells that were derived from three different cell sources [cartilage, fat and bone marrow (BM)] in order to evaluate their advantages as cartilage resurfacing agents. An ovine model of critical size osteochondral focal defect was used and the test items were implanted arthroscopically into the knees. Evidence of regeneration of hyaline quality tissue was observed at 6 and 12 months post-treatment with variable success depending on the cell source. Cartilage and BM-derived mesenchymal stromal cells (MSC), but not those derived from fat, resulted in the best quality of new cartilage, as judged qualitatively by magnetic resonance imaging and macroscopic assessment, and by histological quantitative scores. Given the limitations in sourcing cartilage tissue and the risk of donor site morbidity, BM emerges as a preferential source of MSC for novel cartilage resurfacing therapies of osteochondral defects using copolymeric poly-D,L-lactide-co-glycolide scaffolds.
Collapse
Affiliation(s)
- M Caminal
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain
| | - D Peris
- Grup d'Enginyeria Cel·lular i Tissular, Departament d'Enginyeria Química, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Edifici Q, Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - C Fonseca
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - J Barrachina
- Hospital ASEPEYO Sant Cugat, Avinguda Alcalde Barnils, 54-60, Sant Cugat del Vallès, 08174, Barcelona, Spain
| | - D Codina
- Hospital ASEPEYO Sant Cugat, Avinguda Alcalde Barnils, 54-60, Sant Cugat del Vallès, 08174, Barcelona, Spain
| | - R M Rabanal
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - X Moll
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - A Morist
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - F García
- Departament de Medicina i Cirurgia Animals, Àrea de Medicina i Cirurgia Animal, Universitat Autònoma de Barcelona, Edifici V, Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - J J Cairó
- Grup d'Enginyeria Cel·lular i Tissular, Departament d'Enginyeria Química, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Edifici Q, Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - F Gòdia
- Grup d'Enginyeria Cel·lular i Tissular, Departament d'Enginyeria Química, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Edifici Q, Campus de la UAB, 08193, Bellaterra, Cerdanyola del Vallès, Spain
| | - A Pla
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain
| | - J Vives
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.
| |
Collapse
|
71
|
Muhonen V, Salonius E, Haaparanta AM, Järvinen E, Paatela T, Meller A, Hannula M, Björkman M, Pyhältö T, Ellä V, Vasara A, Töyräs J, Kellomäki M, Kiviranta I. Articular cartilage repair with recombinant human type II collagen/polylactide scaffold in a preliminary porcine study. J Orthop Res 2016; 34:745-53. [PMID: 26573959 DOI: 10.1002/jor.23099] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/03/2015] [Indexed: 02/04/2023]
Abstract
The purpose of this study was to investigate the potential of a novel recombinant human type II collagen/polylactide scaffold (rhCo-PLA) in the repair of full-thickness cartilage lesions with autologous chondrocyte implantation technique (ACI). The forming repair tissue was compared to spontaneous healing (spontaneous) and repair with a commercial porcine type I/III collagen membrane (pCo). Domestic pigs (4-month-old, n = 20) were randomized into three study groups and a circular full-thickness chondral lesion with a diameter of 8 mm was created in the right medial femoral condyle. After 3 weeks, the chondral lesions were repaired with either rhCo-PLA or pCo together with autologous chondrocytes, or the lesion was only debrided and left untreated for spontaneous repair. The repair tissue was evaluated 4 months after the second operation. Hyaline cartilage formed most frequently in the rhCo-PLA treatment group. Biomechanically, there was a trend that both treatment groups resulted in better repair tissue than spontaneous healing. Adverse subchondral bone reactions developed less frequently in the spontaneous group (40%) and the rhCo-PLA treated group (50%) than in the pCo control group (100%). However, no statistically significant differences were found between the groups. The novel rhCo-PLA biomaterial showed promising results in this proof-of-concept study, but further studies will be needed in order to determine its effectiveness in articular cartilage repair. © 2015 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:745-753, 2016.
Collapse
Affiliation(s)
- Virpi Muhonen
- Department of Orthopaedics and Traumatology, University of Helsinki, Helsinki, Finland
| | - Eve Salonius
- Department of Orthopaedics and Traumatology, University of Helsinki, Helsinki, Finland
| | - Anne-Marie Haaparanta
- Department of Electronics and Communications Engineering, Tampere University of Technology and BioMediTech, Tampere, Finland
| | - Elina Järvinen
- Department of Orthopaedics and Traumatology, University of Helsinki, Helsinki, Finland
| | - Teemu Paatela
- Department of Orthopaedics and Traumatology, University of Helsinki, Helsinki, Finland.,Department of Orthopaedics and Traumatology, Helsinki University Hospital, Helsinki, Finland
| | - Anna Meller
- Laboratory Animal Center, University of Helsinki, Helsinki, Finland
| | - Markus Hannula
- Department of Electronics and Communications Engineering, Tampere University of Technology and BioMediTech, Tampere, Finland
| | - Mimmi Björkman
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Tuomo Pyhältö
- Department of Orthopaedics and Traumatology, Helsinki University Hospital, Helsinki, Finland
| | - Ville Ellä
- Department of Electronics and Communications Engineering, Tampere University of Technology and BioMediTech, Tampere, Finland
| | - Anna Vasara
- Department of Orthopaedics and Traumatology, University of Helsinki, Helsinki, Finland.,Department of Orthopaedics and Traumatology, Helsinki University Hospital, Helsinki, Finland
| | - Juha Töyräs
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland.,Department of Clinical Neurophysiology, Kuopio University Hospital, Kuopio, Finland
| | - Minna Kellomäki
- Department of Electronics and Communications Engineering, Tampere University of Technology and BioMediTech, Tampere, Finland
| | - Ilkka Kiviranta
- Department of Orthopaedics and Traumatology, University of Helsinki, Helsinki, Finland.,Department of Orthopaedics and Traumatology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
72
|
Bornes TD, Jomha NM, Mulet-Sierra A, Adesida AB. Optimal Seeding Densities for In Vitro Chondrogenesis of Two- and Three-Dimensional-Isolated and -Expanded Bone Marrow-Derived Mesenchymal Stromal Stem Cells Within a Porous Collagen Scaffold. Tissue Eng Part C Methods 2016; 22:208-20. [PMID: 26651081 DOI: 10.1089/ten.tec.2015.0365] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal stem cells (BMSCs) are a promising cell source for treating articular cartilage defects. The objective of this study was to assess the impact of cell seeding density within a collagen I scaffold on in vitro BMSC chondrogenesis following isolation and expansion in two-dimensional (2D) and three-dimensional (3D) environments. It was hypothesized that both expansion protocols would produce BMSCs capable of hyaline-like chondrogenesis with an optimal seeding density of 10 × 10(6) cells/cm(3). Ovine BMSCs were isolated in a 2D environment by plastic adherence, expanded to passage two in flasks containing an expansion medium, and seeded within collagen I scaffolds at densities of 50, 10, 5, 1, and 0.5 × 10(6) BMSCs/cm(3). For 3D isolation and expansion, aspirates containing known quantities of mononucleated cells (bone marrow-derived mononucleated cells [BMNCs]) were seeded on scaffolds at 50, 10, 5, 1, and 0.5 × 10(6) BMNCs/cm(3) and cultured in the expansion medium for an equivalent duration to 2D expansion. Constructs were differentiated in vitro in the chondrogenic medium for 21 days and assessed with reverse-transcription quantitative polymerase chain reaction, safranin O staining, histological scoring using the Bern Score, collagen immunofluorescence, and glycosaminoglycan (GAG) quantification. Two-dimensional-expanded BMSCs seeded at all densities were capable of proteoglycan production and displayed increased expressions of aggrecan and collagen II messenger RNA (mRNA) relative to predifferentiation controls. Collagen II deposition was apparent in scaffolds seeded at 0.5-10 × 10(6) BMSCs/cm(3). Chondrogenesis of 2D-expanded BMSCs was most pronounced in scaffolds seeded at 5-10 × 10(6) BMSCs/cm(3) based on aggrecan and collagen II mRNA, safranin O staining, Bern Score, total GAG, and GAG/deoxyribonucleic acid (DNA). For 3D-expanded BMSC-seeded scaffolds, increased aggrecan and collagen II mRNA expressions relative to controls were noted with all densities. Proteoglycan deposition was present in scaffolds seeded at 0.5-50 × 10(6) BMNCs/cm(3), while collagen II deposition occurred in scaffolds seeded at 10-50 × 10(6) BMNCs/cm(3). The highest levels of aggrecan and collagen II mRNA, Bern Score, total GAG, and GAG/DNA occurred with seeding at 50 × 10(6) BMNCs/cm(3). Within a collagen I scaffold, 2D- and 3D-expanded BMSCs are capable of hyaline-like chondrogenesis with optimal cell seeding densities of 5-10 × 10(6) BMSCs/cm(3) and 50 × 10(6) BMNCs/cm(3), respectively.
Collapse
Affiliation(s)
- Troy D Bornes
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Nadr M Jomha
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Aillette Mulet-Sierra
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Adetola B Adesida
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| |
Collapse
|
73
|
Moran CJ, Ramesh A, Brama PAJ, O'Byrne JM, O'Brien FJ, Levingstone TJ. The benefits and limitations of animal models for translational research in cartilage repair. J Exp Orthop 2016; 3:1. [PMID: 26915001 PMCID: PMC4703594 DOI: 10.1186/s40634-015-0037-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/28/2015] [Indexed: 12/31/2022] Open
Abstract
Much research is currently ongoing into new therapies for cartilage defect repair with new biomaterials frequently appearing which purport to have significant regenerative capacity. These biomaterials may be classified as medical devices, and as such must undergo rigorous testing before they are implanted in humans. A large part of this testing involves in vitro trials and biomechanical testing. However, in order to bridge the gap between the lab and the clinic, in vivo preclinical trials are required, and usually demanded by regulatory approval bodies. This review examines the in vivo models in current use for cartilage defect repair testing and the relevance of each in the context of generated results and applicability to bringing the device to clinical practice. Some of the preclinical models currently used include murine, leporine, ovine, caprine, porcine, canine, and equine models. Each of these has advantages and disadvantages in terms of animal husbandry, cartilage thickness, joint biomechanics and ethical and licencing issues. This review will examine the strengths and weaknesses of the various animal models currently in use in preclinical studies of cartilage repair.
Collapse
Affiliation(s)
- Conor J Moran
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Ashwanth Ramesh
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Pieter A J Brama
- Section of Veterinary Clinical Sciences, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - John M O'Byrne
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Cappagh National Orthopaedic Hospital, Finglas, Dublin 11, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Tanya J Levingstone
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland. .,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland. .,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland.
| |
Collapse
|
74
|
Peck Y, He P, Chilla GSVN, Poh CL, Wang DA. A preclinical evaluation of an autologous living hyaline-like cartilaginous graft for articular cartilage repair: a pilot study. Sci Rep 2015; 5:16225. [PMID: 26549401 PMCID: PMC4637897 DOI: 10.1038/srep1622510.1038/srep16225] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 10/12/2015] [Indexed: 11/19/2022] Open
Abstract
In this pilot study, an autologous synthetic scaffold-free construct with hyaline quality, termed living hyaline cartilaginous graft (LhCG), was applied for treating cartilage lesions. Implantation of autologous LhCG was done at load-bearing regions of the knees in skeletally mature mini-pigs for 6 months. Over the course of this study, significant radiographical improvement in LhCG treated sites was observed via magnetic resonance imaging. Furthermore, macroscopic repair was effected by LhCG at endpoint. Microscopic inspection revealed that LhCG engraftment restored cartilage thickness, promoted integration with surrounding native cartilage, produced abundant cartilage-specific matrix molecules, and re-established an intact superficial tangential zone. Importantly, the repair efficacy of LhCG was quantitatively shown to be comparable to native, unaffected cartilage in terms of biochemical composition and biomechanical properties. There were no complications related to the donor site of cartilage biopsy. Collectively, these results imply that LhCG engraftment may be a viable approach for articular cartilage repair.
Collapse
Affiliation(s)
- Yvonne Peck
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 637457, Singapore
| | - Pengfei He
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 637457, Singapore
| | - Geetha Soujanya V N Chilla
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 637457, Singapore
| | - Chueh Loo Poh
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 637457, Singapore
| | - Dong-An Wang
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, 637457, Singapore
| |
Collapse
|
75
|
Peck Y, He P, Chilla GSVN, Poh CL, Wang DA. A preclinical evaluation of an autologous living hyaline-like cartilaginous graft for articular cartilage repair: a pilot study. Sci Rep 2015. [DOI: 10.1038/srep16225] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
76
|
Sridharan B, Sharma B, Detamore MS. A Road Map to Commercialization of Cartilage Therapy in the United States of America. TISSUE ENGINEERING PART B-REVIEWS 2015; 22:15-33. [PMID: 26192161 DOI: 10.1089/ten.teb.2015.0147] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite numerous efforts in cartilage regeneration, few products see the light of clinical translation as the commercialization process is opaque, financially demanding, and requires collaboration with people of varied skill sets. The aim of this review is to introduce, to an academic audience, the different paradigms involved in the commercialization of cartilage regeneration technology, elucidate the different hurdles associated with the use of cells and materials in developing new technologies, discuss potential commercialization strategies, and inform the reader about the current trends observed in both the clinical and laboratory setting for establishing clinical trials. Although there are review articles on articular cartilage tissue engineering, independent reports provided by the Food and Drug Administration, and separate review articles on animal models, this is the first review that encompasses all of these facets and is presented in a format favorable to the academic investigator interested in clinical translation from bench to bedside.
Collapse
Affiliation(s)
| | - Blanka Sharma
- 2 Department of Biomedical Engineering, University of Florida , Gainesville, Florida
| | - Michael S Detamore
- 1 Bioengineering Program, University of Kansas , Lawrence, Kansas.,3 Department of Chemical and Petroleum Engineering, University of Kansas , Lawrence, Kansas
| |
Collapse
|
77
|
Alibardi L. Original and regenerating lizard tail cartilage contain putative resident stem/progenitor cells. Micron 2015; 78:10-18. [DOI: 10.1016/j.micron.2015.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/17/2015] [Accepted: 06/17/2015] [Indexed: 01/10/2023]
|
78
|
Orth P, Peifer C, Goebel L, Cucchiarini M, Madry H. Comprehensive analysis of translational osteochondral repair: Focus on the histological assessment. ACTA ACUST UNITED AC 2015; 50:19-36. [PMID: 26515165 DOI: 10.1016/j.proghi.2015.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/07/2015] [Accepted: 10/07/2015] [Indexed: 12/15/2022]
Abstract
Articular cartilage guarantees for an optimal functioning of diarthrodial joints by providing a gliding surface for smooth articulation, weight distribution, and shock absorbing while the subchondral bone plays a crucial role in its biomechanical and nutritive support. Both tissues together form the osteochondral unit. The structural assessment of the osteochondral unit is now considered the key standard procedure for evaluating articular cartilage repair in translational animal models. The aim of this review is to give a detailed overview of the different methods for a comprehensive evaluation of osteochondral repair. The main focus is on the histological assessment as the gold standard, together with immunohistochemistry, and polarized light microscopy. Additionally, standards of macroscopic, non-destructive imaging such as high resolution MRI and micro-CT, biochemical, and molecular biological evaluations are addressed. Potential pitfalls of analysis are outlined. A second focus is to suggest recommendations for osteochondral evaluation.
Collapse
Affiliation(s)
- Patrick Orth
- Center of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Kirrberger Strasse 100, Building 37, D-66421 Homburg/Saar, Germany; Department of Orthopaedic Surgery, Saarland University Medical Center, Kirrberger Strasse 100, Building 37, D-66421 Homburg/Saar, Germany.
| | - Carolin Peifer
- Center of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Kirrberger Strasse 100, Building 37, D-66421 Homburg/Saar, Germany.
| | - Lars Goebel
- Center of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Kirrberger Strasse 100, Building 37, D-66421 Homburg/Saar, Germany; Department of Orthopaedic Surgery, Saarland University Medical Center, Kirrberger Strasse 100, Building 37, D-66421 Homburg/Saar, Germany.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Kirrberger Strasse 100, Building 37, D-66421 Homburg/Saar, Germany.
| | - Henning Madry
- Center of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Kirrberger Strasse 100, Building 37, D-66421 Homburg/Saar, Germany; Department of Orthopaedic Surgery, Saarland University Medical Center, Kirrberger Strasse 100, Building 37, D-66421 Homburg/Saar, Germany.
| |
Collapse
|
79
|
Barron V, Neary M, Mohamed KMS, Ansboro S, Shaw G, O’Malley G, Rooney N, Barry F, Murphy M. Evaluation of the Early In Vivo Response of a Functionally Graded Macroporous Scaffold in an Osteochondral Defect in a Rabbit Model. Ann Biomed Eng 2015; 44:1832-44. [DOI: 10.1007/s10439-015-1473-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/24/2015] [Indexed: 02/01/2023]
|
80
|
Vilela CA, Correia C, Oliveira JM, Sousa RA, Espregueira-Mendes J, Reis RL. Cartilage Repair Using Hydrogels: A Critical Review of in Vivo Experimental Designs. ACS Biomater Sci Eng 2015; 1:726-739. [DOI: 10.1021/acsbiomaterials.5b00245] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- C. A. Vilela
- 3B’s
Research Group, University of Minho, Guimarães, Portugal
- ICVS/3B’s−PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Life
and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- Orthopaedic
Department, Centro Hospitalar do Alto Ave, Guimarães, Portugal
| | - C. Correia
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Guimarães, Portugal
| | - J. M. Oliveira
- 3B’s
Research Group, University of Minho, Guimarães, Portugal
- ICVS/3B’s−PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - R. A. Sousa
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Guimarães, Portugal
| | - J. Espregueira-Mendes
- 3B’s
Research Group, University of Minho, Guimarães, Portugal
- ICVS/3B’s−PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Life
and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal
- Clínica
do Dragão, Espregueira-Mendes Sports Centre, Porto, Portugal
| | - R. L. Reis
- 3B’s
Research Group, University of Minho, Guimarães, Portugal
- ICVS/3B’s−PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Guimarães, Portugal
| |
Collapse
|
81
|
Vives J, Oliver-Vila I, Pla A. Quality compliance in the shift from cell transplantation to cell therapy in non-pharma environments. Cytotherapy 2015; 17:1009-14. [DOI: 10.1016/j.jcyt.2015.02.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 11/30/2022]
|
82
|
Christensen BB, Foldager CB, Olesen ML, Vingtoft L, Rölfing JHD, Ringgaard S, Lind M. Experimental articular cartilage repair in the Göttingen minipig: the influence of multiple defects per knee. J Exp Orthop 2015; 2:13. [PMID: 26914881 PMCID: PMC4538720 DOI: 10.1186/s40634-015-0031-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 06/12/2015] [Indexed: 11/21/2022] Open
Abstract
Background A gold standard treatment for articular cartilage injuries is yet to be found, and a cost-effective and predictable large animal model is needed to bridge the gap between in vitro studies and clinical studies. Ideally, the animal model should allow for testing of clinically relevant treatments and the biological response should be reproducible and comparable to humans. This allows for a reliable translation of results to clinical studies.This study aimed at verifying the Göttingen minipig as a pre-clinical model for articular cartilage repair by testing existing clinical cartilage repair techniques and evaluating the use of two defects per knee. Methods Sixteen fully mature Göttingen minipigs were used. The minipigs received bilateral trochlear osteochondral drill-hole defects or chondral defects (Ø 6 mm), either one defect per knee or two defects per knee. The defects were treated with one of the following: Matrix-induced autologous chondrocyte implantation (MACI), microfracture (MFx), autologous-dual-tissue transplantation (ADTT), autologous bone graft, autologous cartilage chips. Empty chondral and osteochondral defects were used as controls. MRI and CT were performed 3 and 6 month, histology was performed 6 month postoperative. Results The repair tissue varied in morphology from non-cartilaginous fibrous tissue to fibrocartilaginous tissue as seen on MRI, CT and histology at 6 month. The worst results were seen in the empty controls, while the best results were achieved with the MACI and ADTT treatment. The use of two defects per knee did not have any significant effect on the repair response. Conclusion The outcomes of the applied treatments were consistent with the outcomes in clinical studies and it was possible to apply two defects per knee. The Göttingen minipig model was easy to handle, cost-effective and provided predictable outcome. Based on this study the use of two defects per knee, one in the medial and one in the lateral trochlear facet, in male Göttingen minipigs is recommended.
Collapse
Affiliation(s)
- Bjørn Borsøe Christensen
- Orthopedic Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, building 1A, 1. Floor, Aarhus, Denmark.
| | - Casper Bindzus Foldager
- Orthopedic Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, building 1A, 1. Floor, Aarhus, Denmark.
| | - Morten Lykke Olesen
- Orthopedic Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, building 1A, 1. Floor, Aarhus, Denmark.
| | - Louise Vingtoft
- Orthopedic Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, building 1A, 1. Floor, Aarhus, Denmark.
| | - Jan Hendrik Duedal Rölfing
- Orthopedic Research Laboratory, Aarhus University Hospital, Nørrebrogade 44, building 1A, 1. Floor, Aarhus, Denmark.
| | | | - Martin Lind
- Department of Sports Traumatology, Department of orthopedic surgery, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
83
|
Kon E, Filardo G, Shani J, Altschuler N, Levy A, Zaslav K, Eisman JE, Robinson D. Osteochondral regeneration with a novel aragonite-hyaluronate biphasic scaffold: up to 12-month follow-up study in a goat model. J Orthop Surg Res 2015; 10:81. [PMID: 26018574 PMCID: PMC4486417 DOI: 10.1186/s13018-015-0211-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/04/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The regeneration of articular hyaline cartilage remains an elusive goal despite years of research. Recently, an aragonite-hyaluronate (Ar-HA) biphasic scaffold has been described capable of cartilage regeneration over a 6-month follow-up period. This study was conducted in order to assess the fate of the regenerated osteochondral tissue in a 12-month-long validated caprine model. HYPOTHESIS/PURPOSE The hypothesis was that the implantation of the Ar-HA implant leads to tissue regeneration and maturation. STUDY DESIGN A two-arm caprine model of a critical osteochondral defect compares the fate of acute osteochondral defects (group A) to Ar-HA implanted defects (group B). METHODS Critical 6 mm in diameter and 10-mm in depth osteochondral defects were created in the load-bearing medial femoral condyle of 20 mature goats and randomized into two groups. In group A (n = 6), a blood clot spontaneously filled the defect; in group B (n = 14), a single Ar-HA implant reconstructed the defect. The animals were sacrificed after either 6 or 12 months. Parameters assessed included clinical evaluation, x-rays, micro-CT, ultrasound and histology at both time points, and specimen high-field magnetic resonance imaging with T2 mapping at the 12-month time point. RESULTS In most group A animals, the defects were not reconstructed (1/3 at 6 months, and 0/3 at 12 months). Defects in group B were mostly reconstructed (5/7 at 6 months and 6/7 at 12 months). Group A defects were either empty or contained fibrous repair tissue; while group B filling was compatible with hyaline cartilage and normal bone. CONCLUSION Ar-HA scaffolds implanted in critical osteochondral defects result in hyaline cartilage formation and subchondral bone regeneration. The results improved at the 12-month time point compared to the 6-month time point, indicating a continuous maturation process without deterioration of the repair tissue. CLINICAL RELEVANCE Osteochondral defects are common in humans; the results of the current study suggest that an acellular Ar-HA scaffold might induce cartilage and subchondral bone regeneration.
Collapse
Affiliation(s)
- Elizaveta Kon
- II Orthopedic division and NanoBiotechnology Lab, Rizzoli Orthopedic Institute, Bologna, Italy.
| | - Giuseppe Filardo
- II Orthopedic division and NanoBiotechnology Lab, Rizzoli Orthopedic Institute, Bologna, Italy.
| | | | | | - Andrew Levy
- Center for Advanced Sports Medicine, Knee and Shoulder, Millburn, NJ, USA.
| | - Ken Zaslav
- Cartilage Restoration Center: Advanced Orthopedic Centers and Clinical Prof. Orthopedic Surgery V.C.U. Med. Ctr., Richmond, VA, USA.
| | - John E Eisman
- Osteoporosis and Translational Research, UNSW University, Sydney, NSW, Australia.
| | - Dror Robinson
- Department of Orthopedics, Rabin Medical Center, Petah Tikwa, Israel.
| |
Collapse
|
84
|
Evaluation of Cartilage Repair by Mesenchymal Stem Cells Seeded on a PEOT/PBT Scaffold in an Osteochondral Defect. Ann Biomed Eng 2015; 43:2069-82. [PMID: 25589372 DOI: 10.1007/s10439-015-1246-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/07/2015] [Indexed: 01/03/2023]
Abstract
The main objective of this study was to evaluate the effectiveness of a mesenchymal stem cell (MSC)-seeded polyethylene-oxide-terephthalate/polybutylene-terephthalate (PEOT/PBT) scaffold for cartilage tissue repair in an osteochondral defect using a rabbit model. Material characterisation using scanning electron microscopy indicated that the scaffold had a 3D architecture characteristic of the additive manufacturing fabrication method, with a strut diameter of 296 ± 52 μm and a pore size of 512 ± 22 μm × 476 ± 25 μm × 180 ± 30 μm. In vitro optimisation revealed that the scaffold did not generate an adverse cell response, optimal cell loading conditions were achieved using 50 μg/ml fibronectin and a cell seeding density of 25 × 10(6) cells/ml and glycosaminoglycan (GAG) accumulation after 28 days culture in the presence of TGFβ3 indicated positive chondrogenesis. Cell-seeded scaffolds were implanted in osteochondral defects for 12 weeks, with cell-free scaffolds and empty defects employed as controls. On examination of toluidine blue staining for chondrogenesis and GAG accumulation, both the empty defect and the cell-seeded scaffold appeared to promote repair. However, the empty defect and the cell-free scaffold stained positive for collagen type I or fibrocartilage, while the cell-seeded scaffold stained positive for collagen type II indicative of hyaline cartilage and was statistically better than the cell-free scaffold in the blinded histological evaluation. In summary, MSCs in combination with a 3D PEOT/PBT scaffold created a reparative environment for cartilage repair.
Collapse
|
85
|
Chevrier A, Kouao ASM, Picard G, Hurtig MB, Buschmann MD. Interspecies comparison of subchondral bone properties important for cartilage repair. J Orthop Res 2015; 33:63-70. [PMID: 25242685 DOI: 10.1002/jor.22740] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/25/2014] [Indexed: 02/04/2023]
Abstract
Microfracture repair tissue in young adult humans and in rabbit trochlea is frequently of higher quality than in corresponding ovine or horse models or in the rabbit medial femoral condyle (MFC). This may be related to differences in subchondral properties since repair is initiated from the bone. We tested the hypothesis that subchondral bone from rabbit trochlea and the human MFC are structurally similar. Trochlea and MFC samples from rabbit, sheep, and horse were micro-CT scanned and histoprocessed. Samples were also collected from normal and lesional areas of human MFC. The subchondral bone of the rabbit trochlea was the most similar to human MFC, where both had a relatively thin bone plate and a more porous and less dense character of subchondral bone. MFC from animals all displayed thicker bone plates, denser and less porous bone and thicker trabeculae, which may be more representative of older or osteoarthritic patients, while both sheep trochlear ridges and the horse lateral trochlea shared some structural features with human MFC. Since several cartilage repair procedures rely on subchondral bone for repair, subchondral properties should be accounted for when choosing animal models to study and test procedures that are intended for human cartilage repair.
Collapse
Affiliation(s)
- Anik Chevrier
- Chemical Engineering Department, Ecole Polytechnique de Montreal, PO Box 6079, Succ Centre-Ville, Montreal, Quebec, Canada, H3C 3A7
| | | | | | | | | |
Collapse
|
86
|
Trachtenberg JE, Vo TN, Mikos AG. Pre-clinical characterization of tissue engineering constructs for bone and cartilage regeneration. Ann Biomed Eng 2014; 43:681-96. [PMID: 25319726 DOI: 10.1007/s10439-014-1151-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/06/2014] [Indexed: 12/16/2022]
Abstract
Pre-clinical animal models play a crucial role in the translation of biomedical technologies from the bench top to the bedside. However, there is a need for improved techniques to evaluate implanted biomaterials within the host, including consideration of the care and ethics associated with animal studies, as well as the evaluation of host tissue repair in a clinically relevant manner. This review discusses non-invasive, quantitative, and real-time techniques for evaluating host-materials interactions, quality and rate of neotissue formation, and functional outcomes of implanted biomaterials for bone and cartilage tissue engineering. Specifically, a comparison will be presented for pre-clinical animal models, histological scoring systems, and non-invasive imaging modalities. Additionally, novel technologies to track delivered cells and growth factors will be discussed, including methods to directly correlate their release with tissue growth.
Collapse
Affiliation(s)
- Jordan E Trachtenberg
- Department of Bioengineering, Rice University, MS 142, P.O. Box 1892, Houston, TX, 77251-1892, USA
| | | | | |
Collapse
|
87
|
Benders K, Boot W, Cokelaere S, Van Weeren P, Gawlitta D, Bergman H, Saris D, Dhert W, Malda J. Multipotent Stromal Cells Outperform Chondrocytes on Cartilage-Derived Matrix Scaffolds. Cartilage 2014; 5:221-30. [PMID: 26069701 PMCID: PMC4335771 DOI: 10.1177/1947603514535245] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE Although extracellular matrix (ECM)-derived scaffolds have been extensively studied and applied in a number of clinical applications, the use of ECM as a biomaterial for (osteo)chondral regeneration is less extensively explored. This study aimed at evaluating the chondrogenic potential of cells seeded on cartilage-derived matrix (CDM) scaffolds in vitro. DESIGN Scaffolds were generated from decellularized equine articular cartilage and seeded with either chondrocytes or multipotent stromal cells (MSCs). After 2, 4, and 6 weeks of in vitro culture, CDM constructs were analyzed both histologically (hematoxylin and eosin, Safranin-O, collagen types I and II) and biochemically (glycosaminoglycan [GAG] and DNA content). RESULTS After 4 weeks, both cell types demonstrated chondrogenic differentiation; however, the MSCs significantly outperformed chondrocytes in producing new GAG-containing cartilaginous matrix. CONCLUSION These promising in vitro results underscore the potency of CDM scaffolds in (osteo)chondral defect repair.
Collapse
Affiliation(s)
- K.E.M. Benders
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - W. Boot
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S.M. Cokelaere
- Department of Equine Sciences, Utrecht University, Utrecht, the Netherlands
| | - P.R. Van Weeren
- Department of Equine Sciences, Utrecht University, Utrecht, the Netherlands
| | - D. Gawlitta
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - H.J. Bergman
- Lingehoeve, Veterinary Medicine, Lienden, the Netherlands
| | - D.B.F. Saris
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands,MIRA Institute, Department of Tissue Regeneration, University of Twente, Enschede, the Netherlands
| | - W.J.A. Dhert
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands,Faculty of Veterinary Sciences, Utrecht University, Utrecht, the Netherlands
| | - J. Malda
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands,Department of Equine Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
88
|
Effect of autologous platelet rich fibrin on the healing of experimental articular cartilage defects of the knee in an animal model. BIOMED RESEARCH INTERNATIONAL 2014; 2014:486436. [PMID: 25028656 PMCID: PMC4083211 DOI: 10.1155/2014/486436] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/26/2014] [Indexed: 11/26/2022]
Abstract
The effect of autologous platelet rich fibrin (PRF), a second generation platelet product, on the healing of experimental articular cartilage lesions was evaluated in an animal model. Full thickness cartilage lesions with a diameter of 6 mm and depth of 5 mm were created in the weight bearing area of femoral condyles of both hind limbs in 12 adult mixed breed dogs. Defects in the left hind limb of each dog were repaired by PRF implantation whereas those in the right hind limb were left empty. The animals were euthanized at 4, 16, and 24 weeks following surgery and the resultant repair tissue was investigated macroscopically and microscopically. The results of macroscopic and histological evaluations indicated that there were significant differences between the PRF treated and untreated defects. In conclusion, the present study indicated that the use of platelet rich fibrin as a source of autologous growth factors leads to improvement in articular cartilage repair.
Collapse
|
89
|
Abstract
Cartilage repair in terms of replacement, or
regeneration of damaged or diseased articular cartilage with functional tissue,
is the ‘holy grail’ of joint surgery. A wide spectrum of strategies
for cartilage repair currently exists and several of these techniques
have been reported to be associated with successful clinical outcomes
for appropriately selected indications. However, based on respective
advantages, disadvantages, and limitations, no single strategy, or
even combination of strategies, provides surgeons with viable options
for attaining successful long-term outcomes in the majority of patients.
As such, development of novel techniques and optimisation of current techniques
need to be, and are, the focus of a great deal of research from
the basic science level to clinical trials. Translational research
that bridges scientific discoveries to clinical application involves
the use of animal models in order to assess safety and efficacy
for regulatory approval for human use. This review article provides
an overview of animal models for cartilage repair. Cite this article: Bone Joint Res 2014;4:89–94.
Collapse
Affiliation(s)
- J L Cook
- University of Missouri, ComparativeOrthopaedic Laboratory and Missouri Orthopaedic Institute, Columbia, Missouri, USA
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Chang CH, Hsu YM, Hsiao CN, Kuo TF, Chang MH. CRITICAL-SIZED OSTEOCHONDRAL DEFECTS OF YOUNG MINIATURE PIGS AS A PRECLINICAL MODEL FOR ARTICULAR CARTILAGE REPAIR. BIOMEDICAL ENGINEERING: APPLICATIONS, BASIS AND COMMUNICATIONS 2014. [DOI: 10.4015/s1016237214500033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Purpose: Evaluation by animal models is essential for tissue engineering-based articular cartilage repair techniques. Larger animals are considered to more closely approximate the clinical situations in translational medicine. Therefore, we used miniature pigs and induced full-thickness and osteochondral defects in them. We also studied if there were instances of spontaneous repair for providing baseline data for further cartilage regeneration study. Methods: A total of 12 miniature pigs with average age of 7.4 months were used in this study. Full-thickness and osteochondral defects with 2.7, 4.5 or 8.0 mm diameter were created at medial femoral condyles in the same pig, respectively. The pigs were sacrificed at 8, 16, 24 and 48 weeks. Gross appearances of defects were observed at aforementioned time points, and the histological analyses, including H&E and alcian blue staining, were performed for consequent evaluation as well. Results: The results showed that defects created in the center of the femoral condoyle migrated to periphery, and it implied that the pigs were still growing after 7–8 months of age. However, spontaneous repair was observed in 2.7 mm diameter defects but rarely seen in 4.5 and 8 mm diameter osteochondral defects. On the other hand, osteochondral defects repaired better than defects of full-thickness in the same 2.7 mm diameter. Conclusions: In order to prevent spontaneous repair of osteochondral defect in a young miniature pig animal model, a critical-sized osteochondral defect larger than 40% width of femoral medial condyle (4.5 mm in miniature pig) and observation period for more than 48 weeks are suggested by this study.
Collapse
Affiliation(s)
- Chih-Hung Chang
- Division of Orthopaedics, Department of Surgery, Far Eastern Memorial Hospital, No. 21, Nan-Ya S. Rd., Sec. 2 Pan-Chiao, New Taipei City, Taiwan
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Tao-Yuan, Taiwan
- Department of Orthopedics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yuan-Ming Hsu
- Division of Orthopaedics, Department of Surgery, Far Eastern Memorial Hospital, No. 21, Nan-Ya S. Rd., Sec. 2 Pan-Chiao, New Taipei City, Taiwan
| | - Chun-Ni Hsiao
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Da-an District, Taipei, Taiwan
| | - Tzong-Fu Kuo
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Da-an District, Taipei, Taiwan
| | - Ming-Huang Chang
- Graduate Institute of Veterinary Medicine, College of Agriculture, National Chiayi University, No. 300, Syuefu Rd. East District, Chiayi City, Taiwan
| |
Collapse
|
91
|
Subchondral chitosan/blood implant-guided bone plate resorption and woven bone repair is coupled to hyaline cartilage regeneration from microdrill holes in aged rabbit knees. Osteoarthritis Cartilage 2014; 22:323-33. [PMID: 24361795 DOI: 10.1016/j.joca.2013.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 12/05/2013] [Accepted: 12/10/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Little is known of how to routinely elicit hyaline cartilage repair tissue in middle-aged patients. We tested the hypothesis that in skeletally aged rabbit knees, microdrill holes can be stimulated to remodel the bone plate and induce a more integrated, voluminous and hyaline cartilage repair tissue when treated by subchondral chitosan/blood implants. DESIGN New Zealand White rabbits (13 or 32 months old, N = 7) received two 1.5 mm diameter, 2 mm depth drill holes in each knee, either left to bleed as surgical controls or press-fit with a 10 kDa (distal hole: 10K) or 40 kDa (proximal hole: 40K) chitosan/blood implant with fluorescent chitosan tracer. Post-operative knee effusion was documented. Repair tissues at day 0 (N = 1) and day 70 post-surgery (N = 6) were analyzed by micro-computed tomography, and by histological scoring and histomorphometry (SafO, Col-2, and Col-1) at day 70. RESULTS All chitosan implants were completely cleared after 70 days, without increasing transient post-operative knee effusion compared to controls. Proximal control holes had worse osteochondral repair than distal holes. Both implant formulations induced bone remodeling and improved lateral integration of the bone plate at the hole edge. The 40K implant inhibited further bone repair inside 50% of the proximal holes, while the 10K implant specifically induced a "wound bloom" reaction, characterized by decreased bone plate density in a limited zone beyond the initial hole edge, and increased woven bone (WB) plate repair inside the initial hole (P = 0.016), which was accompanied by a more voluminous and hyaline cartilage repair (P < 0.05 vs control defects). CONCLUSION In a challenging aged rabbit model, bone marrow-derived hyaline cartilage repair can be promoted by treating acute drill holes with a biodegradable subchondral implant that elicits bone plate resorption followed by anabolic WB repair within a 70-day repair period.
Collapse
|
92
|
Schneider-Wald B, von Thaden AK, Schwarz MLR. [Defect models for the regeneration of articular cartilage in large animals]. DER ORTHOPADE 2013; 42:242-53. [PMID: 23575559 DOI: 10.1007/s00132-012-2044-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Several animal models are available for the analysis of regeneration of articular cartilage in large animals, such as sheep, pigs, goats, dogs and horses. The subchondral bone lamella must be considered when ACT and MACT techniques are examined in order to protect the implant against migration of cells from the bone marrow, although recruitment of cells is often desirable in the regeneration of human cartilage. MATERIAL AND METHODS The defects are mainly positioned at the condyles and the trochlea often bilaterally and spontaneous healing should be excluded. The follow-up period for assessment of the effectiveness of cartilage regeneration is 6-12 months. Shorter observation times up to 12 weeks can be used for pilot studies. Scores based on histological, immunohistological and biochemical staining are mostly used for assessing the regenerated tissue. Biomechanical tests with destructive features need isolated specimens from the animal but modern slice imaging techniques can reflect the progression of the healing processes over the time span of the study in vivo. CONCLUSION Approaches to standardize the evaluation of the regeneration of articular cartilage have been sporadically described whereas they are required from the point of view of the approval of new concepts for therapy and the protection of animals.
Collapse
Affiliation(s)
- B Schneider-Wald
- Sektion experimentelle Orthopädie und Unfallchirurgie, Orthopädisch-Unfallchirurgisches Zentrum, Universitätsmedizin Mannheim, Medizinische Fakultät Mannheim, Universität Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Deutschland.
| | | | | |
Collapse
|
93
|
Malda J, McIlwraith CW. Current Trends in Cartilage Science: An Impression from the ICRS World Conference 2012. Cartilage 2013; 4:273-80. [PMID: 26069672 PMCID: PMC4297156 DOI: 10.1177/1947603513479606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, The Netherlands
- Department of Equine Sciences, Utrecht University, The Netherlands
- Scientific Programme Chairs, ICRS Congress 2012, Montreal
| | - C. Wayne McIlwraith
- Gail Holmes Equine Orthopaedic Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Scientific Programme Chairs, ICRS Congress 2012, Montreal
| |
Collapse
|
94
|
Pallante-Kichura AL, Chen AC, Temple-Wong MM, Bugbee WD, Sah RL. In vivo efficacy of fresh versus frozen osteochondral allografts in the goat at 6 months is associated with PRG4 secretion. J Orthop Res 2013; 31:880-6. [PMID: 23362152 PMCID: PMC4076778 DOI: 10.1002/jor.22319] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 01/05/2013] [Indexed: 02/04/2023]
Abstract
The long-term efficacy of osteochondral allografts is due to the presence of viable chondrocytes within graft cartilage. Chondrocytes in osteochondral allografts, especially those at the articular surface that normally produce the lubricant proteoglycan-4 (PRG4), are susceptible to storage-associated death. The hypothesis of this study was that the loss of chondrocytes within osteochondral grafts leads to decreased PRG4 secretion, after graft storage and subsequent implant. The objectives were to determine the effect of osteochondral allograft treatment (FROZEN vs. FRESH) on secretion of functional PRG4 after (i) storage, and (ii) 6 months in vivo in adult goats. FROZEN allograft storage reduced PRG4 secretion from cartilage by ∼85% compared to FRESH allograft storage. After 6 months in vivo, the PRG4-secreting function of osteochondral allografts was diminished with prior FROZEN storage by ∼81% versus FRESH allografts and by ∼84% versus non-operated control cartilage. Concomitantly, cellularity at the articular surface in FROZEN allografts was ∼96% lower than FRESH allografts and non-operated cartilage. Thus, the PRG4-secreting function of allografts appears to be maintained in vivo based on its state after storage. PRG4 secretion may be not only a useful marker of allograft performance, but also a biological process protecting the articular surface of grafts following cartilage repair.
Collapse
Affiliation(s)
| | - Albert C. Chen
- Department of Bioengineering, University of California-San Diego, La Jolla, CA
| | | | - William D. Bugbee
- Department of Orthopaedic Surgery, University of California-San Diego, San Diego, CA,Division of Orthopaedic Surgery, Scripps Clinic, La Jolla, CA
| | - Robert L. Sah
- Department of Bioengineering, University of California-San Diego, La Jolla, CA,Department of Orthopaedic Surgery, University of California-San Diego, San Diego, CA,Institute of Engineering in Medicine, University of California-San Diego, La Jolla, CA,Address correspondence and reprint requests to: R.L. Sah, Department of Bioengineering, University of California-San Diego, 9500 Gilman Drive MC:0412, La Jolla CA 92093-0412, USA. Tel: 858-534-0821; Fax: 858-822-0448;
| |
Collapse
|
95
|
Bell AD, Lascau-Coman V, Sun J, Chen G, Lowerison MW, Hurtig MB, Hoemann CD. Bone-Induced Chondroinduction in Sheep Jamshidi Biopsy Defects with and without Treatment by Subchondral Chitosan-Blood Implant: 1-Day, 3-Week, and 3-Month Repair. Cartilage 2013; 4:131-43. [PMID: 26069656 PMCID: PMC4297102 DOI: 10.1177/1947603512463227] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Delivery of chitosan to subchondral bone is a novel approach for augmented marrow stimulation. We evaluated the effect of 3 presolidified chitosan-blood implant formulations on osteochondral repair progression compared with untreated defects. DESIGN In N = 5 adult sheep, six 2-mm diameter Jamshidi biopsy holes were created bilaterally in the medial femoral condyle and treated with presolidified chitosan-blood implant with fluorescent chitosan tracer (10 kDa, 40 kDa, or 150k Da chitosan, left knee) or left to bleed (untreated, right knee). Implant residency and osteochondral repair were assessed at 1 day (N = 1), 3 weeks (N = 2), or 3 months (N = 2) postoperative using fluorescence microscopy, histomorphometry, stereology, and micro-computed tomography. RESULTS Chitosan implants were retained in 89% of treated Jamshidi holes up to 3 weeks postoperative. At 3 weeks, biopsy sites were variably covered by cartilage flow, and most bone holes contained cartilage flow fragments and heterogeneous granulation tissues with sparse leukocytes, stromal cells, and occasional adipocytes (volume density 1% to 3%). After 3 months of repair, most Jamshidi bone holes were deeper, remodeling at the edges, filled with angiogenic granulation tissue, and lined with variably sized chondrogenic foci fused to bone trabeculae or actively repairing bone plate. The 150-kDa chitosan implant elicited more subchondral cartilage formation compared with 40-kDa chitosan-treated and control defects (P < 0.05, N = 4). Treated defects contained more mineralized repair tissue than control defects at 3 months (P < 0.05, N = 12). CONCLUSION Bone plate-induced chondroinduction is an articular cartilage repair mechanism. Jamshidi biopsy repair takes longer than 3 months and can be influenced by subchondral chitosan-blood implant.
Collapse
Affiliation(s)
- Angela D. Bell
- Department of Clinical Studies, University of Guelph, Guelph, Ontario, Canada
| | - Viorica Lascau-Coman
- Department of Chemical Engineering, École Polytechnique, Montreal, Quebec, Canada
| | - Jun Sun
- BioSyntech/Piramal Healthcare Canada, Montreal, Quebec, Canada
| | - Gaoping Chen
- Department of Chemical Engineering, École Polytechnique, Montreal, Quebec, Canada
| | - Mark W. Lowerison
- Department of Clinical Studies, University of Guelph, Guelph, Ontario, Canada
| | - Mark B. Hurtig
- Department of Clinical Studies, University of Guelph, Guelph, Ontario, Canada
| | - Caroline D. Hoemann
- Department of Chemical Engineering, École Polytechnique, Montreal, Quebec, Canada,Institute of Biomedical Engineering, École Polytechnique, Montreal, Quebec, Canada
| |
Collapse
|
96
|
Aulin C, Jensen-Waern M, Ekman S, Hägglund M, Engstrand T, Hilborn J, Hedenqvist P. Cartilage repair of experimentally 11 induced osteochondral defects in New Zealand White rabbits. Lab Anim 2013; 47:58-65. [DOI: 10.1177/0023677212473716] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Articular cartilage has a limited capacity for self-repair in adult humans, and methods used to stimulate regeneration often result in re-growth of fibrous cartilage, which has lower durability. No current treatment option can provide complete repair. The possibility of growth factor delivery into the joint for cartilage regeneration after injury would be an attractive treatment option. A full thickness osteochondral defect of 4 mm in diameter and 2 mm deep was created by mechanical drilling in the medial femoral condyle in 20 female adult New Zealand White rabbits. In an attempt to improve regeneration a hyaluronic hydrogel system, with or without bone morphogenetic protein-2 (BMP-2) was delivered intraarticularly. The contralateral joint defect was treated with saline as control. Throughout the study, rabbits were clinically examined and after 12 ( n = 6) or 24 ( n = 9) weeks, the rabbits were euthanized and the joints evaluated by histology. The defects healed with fibrocartilage like tissue, and the filling of the defects ranged from less than 25% to complete. The healing of the defects varied both inter- and intra-group wise. Treatment with hyaluronan gel with or without BMP-2 had no effect on cartilage regeneration compared with controls. Instead, severe ectopic bone formation was found in seven joints treated with BMP-2. In conclusion, the present study shows that neither treatment with hyaluronic gel alone, nor in combination with BMP-2, improves the healing of an induced cartilage defect in rabbits. It further shows that BMP-2 can induce ectopic bone formation, which severely affects the functionality of the joint.
Collapse
Affiliation(s)
- C Aulin
- Department of Chemistry, Ångström Laboratory, Polymer Chemistry, Uppsala University, SE-751 21 Uppsala, Sweden
- Department of Medicine, Rheumatology Unit, Karolinska Institute, SE-171 76 Stockholm, Sweden
| | - M Jensen-Waern
- Department of Clinical Sciences, Comparative Physiology and Medicine, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - S Ekman
- Department of Biomedicine and Veterinary Public Health, Division of Pathology, Pharmacology and Toxicology, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - M Hägglund
- Department of Clinical Sciences, Comparative Physiology and Medicine, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| | - T Engstrand
- Department of Chemistry, Ångström Laboratory, Polymer Chemistry, Uppsala University, SE-751 21 Uppsala, Sweden
- Stockholm Craniofacial Centre, Department of Reconstructive Plastic Surgery, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - J Hilborn
- Department of Chemistry, Ångström Laboratory, Polymer Chemistry, Uppsala University, SE-751 21 Uppsala, Sweden
| | - P Hedenqvist
- Department of Clinical Sciences, Comparative Physiology and Medicine, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| |
Collapse
|
97
|
Martin F, Lehmann M, Anderer U. Generation of Scaffold Free 3-D Cartilage-Like Microtissues from Human Chondrocytes. ACTA ACUST UNITED AC 2013. [DOI: 10.4018/978-1-4666-2506-8.ch008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Since our society is characterized by an increasing age of its people on the one hand and a high number of persons dealing with sports on the other hand, the number of patients suffering from traumatic defects or osteoarthritis is growing. In combination with the articular cartilage specific limited capacity to regenerate, a need for suitable therapies is obvious. Thereby, cell-based therapies are of major interest. This type of clinical intervention was introduced to patients at the beginning of the 1990s. During the last years, a technological shift from simple cell suspensions to more complex 3D structures was performed. In order to optimize the scaffold free generation of cartilage, such as microtissues from human chondrocytes, the authors examine the influence of a static or spinner flask culture with respect to differentiation and architecture of the engineered microtissues. Additionally, the impact of the soluble factors TGF-ß2 and ascorbic acid on this process are investigated. The results demonstrate a positive impact of TGF-ß2 and ascorbic acid supplementation with respect to general Type II Collagen and proteoglycan expression for both the static and spinner flask culture.
Collapse
Affiliation(s)
- Frank Martin
- Lausitz University of Applied Sciences (LUAS), Germany
| | - Mario Lehmann
- Lausitz University of Applied Sciences (LUAS), Germany
| | | |
Collapse
|
98
|
Abstract
In the United States, few Food and Drug Administration (FDA)-approved options exist for the treatment of focal cartilage and osteochondral lesions. Developers of products for cartilage repair face many challenges to obtain marketing approval from the FDA. The objective of this review is to discuss the necessary steps for FDA application and approval for a new cartilage repair product. FDA Guidance Documents, FDA Panel Meetings, scientific organization recommendations, and clinicaltrials.gov were reviewed to demonstrate the current thinking of FDA and the scientific community on the regulatory process for cartilage repair therapies. Cartilage repair therapies can receive market approval from FDA as medical devices, drugs, or biologics, and the specific classification of product can affect the nonclinical, clinical, and regulatory strategy to bring the product to market. Recent FDA guidance gives an outline of the required elements to bring a cartilage repair product to market, although these standards are often very general. As a result, companies have to carefully craft their study patient population, comparator group, and clinical endpoint to best showcase their product's attributes. In addition, regulatory strategy and manufacturing process validation need to be considered early in the clinical study process to allow for timely product approval following the completion of clinical study. Although the path to regulatory approval for a cartilage repair therapy is challenging and time-consuming, proper clinical trial planning and attention to the details can eventually save companies time and money by bringing a product to the market in the most expeditious process possible.
Collapse
Affiliation(s)
- Kevin B. McGowan
- Musculoskeletal Clinical Regulatory Advisers, Washington, DC, USA
| | - Glenn Stiegman
- Musculoskeletal Clinical Regulatory Advisers, Washington, DC, USA
| |
Collapse
|
99
|
Chan EF, Liu IL, Semler EJ, Aberman HM, Simon TM, Chen AC, Truncale KG, Sah RL. Association of 3-Dimensional Cartilage and Bone Structure with Articular Cartilage Properties in and Adjacent to Autologous Osteochondral Grafts after 6 and 12 months in a Goat Model. Cartilage 2012; 3. [PMID: 24224069 PMCID: PMC3818730 DOI: 10.1177/1947603511435272] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE The articular cartilage of autologous osteochondral grafts is typically different in structure and function from local host cartilage and thereby presents a remodeling challenge. The hypothesis of this study was that properties of the articular cartilage of trochlear autografts and adjacent femoral condyle are associated with the 3-D geometrical match between grafted and contralateral joints at 6 and 12 months after surgery. DESIGN Autografts were transferred unilaterally from the lateral trochlea (LT) to the medial femoral condyle (MFC) in adult Spanish goats. Operated and contralateral Non-Operated joints were harvested at 6 and 12 months, and analyzed by indentation testing, micro-computed tomography, and histology to compare (1) histological indices of repair, (2) 3-D structure (articular surface deviation, bone-cartilage interface deviation, cartilage thickness), (3) indentation stiffness, and (4) correlations between stiffness and 3-D structure. RESULTS Cartilage deterioration was present in grafts at 6 months and more severe at 12 months. Cartilage thickness and normalized stiffness of Operated MFC were lower than Non-Operated MFC within the graft and proximal adjacent host regions. Operated MFC articular surfaces were recessed relative to Non-Operated MFC and exhibited lower cartilage stiffness with increasing recession. Sites with large bone-cartilage interface deviations, both proud and recessed, were associated with recessed articular surfaces and low cartilage stiffness. CONCLUSION The effectiveness of cartilage repair by osteochondral grafting is associated with the match of 3-D cartilage and bone geometry to the native osteochondral structure.
Collapse
Affiliation(s)
- Elaine F. Chan
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - I-Ling Liu
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | - Albert C. Chen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | - Robert L. Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA,Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
100
|
de Vries RBM, Buma P, Leenaars M, Ritskes-Hoitinga M, Gordijn B. Reducing the number of laboratory animals used in tissue engineering research by restricting the variety of animal models. Articular cartilage tissue engineering as a case study. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:427-35. [PMID: 22571623 DOI: 10.1089/ten.teb.2012.0059] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The use of laboratory animals in tissue engineering research is an important underexposed ethical issue. Several ethical questions may be raised about this use of animals. This article focuses on the possibilities of reducing the number of animals used. Given that there is considerable debate about the adequacy of the current animal models in tissue engineering research, we investigate whether it is possible to reduce the number of laboratory animals by selecting and using only those models that have greatest predictive value for future clinical application of the tissue engineered product. The field of articular cartilage tissue engineering is used as a case study. Based on a study of the scientific literature and interviews with leading experts in the field, an overview is provided of the animal models used and the advantages and disadvantages of each model, particularly in terms of extrapolation to the human situation. Starting from this overview, it is shown that, by skipping the small models and using only one large preclinical model, it is indeed possible to restrict the number of animal models, thereby reducing the number of laboratory animals used. Moreover, it is argued that the selection of animal models should become more evidence based and that researchers should seize more opportunities to choose or create characteristics in the animal models that increase their predictive value.
Collapse
Affiliation(s)
- Rob B M de Vries
- SYRCLE/3R Research Centre, Central Animal Laboratory (Post 231), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|