1
|
Shayan‐Moghadam R, Sherafatvaziri A, Vosoughi F, Mirzamohamadi A, Saffar H, Shafieian M, Zanjani LO, Nematian H, Nabian MH. Optimum storage conditions for osteochondral allograft plugs: An ex vivo comparative study of 12 storage protocols. J Exp Orthop 2025; 12:e70206. [PMID: 40170704 PMCID: PMC11956793 DOI: 10.1002/jeo2.70206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 04/03/2025] Open
Abstract
Objective Physiological storage temperature and chondrogenic supplements can enhance tissue viability, potentially overcoming the limitations associated with osteochondral allograft transplantation. This study aimed to evaluate the quality of macroscopically healthy cartilage across 12 different storage protocols to find optimum storage conditions for osteochondral allograft plugs. Methods Osteochondral plugs were obtained from arthroplasty candidates and divided into 12 groups based on two culture media (Media 1 [supplemented Dulbecco's modified Eagle's medium {DMEM}/Ham's Nutrient Mixture F12] or Media 2 [enriched Media 1 with 10 ng/mL of transforming growth factor-beta {TGF-β}]), two culture conditions (static or dynamic), and three temperatures (-70°C, 4°C and 37°C). Subsequently, samples were evaluated on Days 1, 14, 28 and 60 for biochemical, biomechanical and histopathological characteristics alongside bacterial surveillance. Results A total of 4338 plugs from 843 donors were assessed. Chondrocyte viability and proteoglycan synthesis were highest in the DMEM enriched with TGF-β at 37°C and 4°C. Although biomechanical properties decreased over time in all groups, dynamic culture conditions resulted in smaller decreases compared to other storage protocols. Viscoelasticity was observed in all samples, with dynamic media groups being maintained the most. Histological evaluation showed signs of degeneration, and temperature variations affected the preservation of the tissue differently. Bacterial surveillance identified contamination in specific storage conditions. Conclusion Storing osteochondral allografts at 37°C in TGF-β supplemented media under dynamic conditions may extend the grafting window from 14 to 60 days. This extension could improve tissue availability, reduce costs and minimize graft wastage, thereby advancing joint resurfacing techniques. Further research is needed to confirm the safety and efficacy of this storage protocol. Level of Evidence N/A.
Collapse
Affiliation(s)
- Ramin Shayan‐Moghadam
- Center for Orthopedic Trans‐Disciplinary Applied ResearchTehran University of Medical SciencesTehranIran
| | - Arash Sherafatvaziri
- Department of Orthopedic Surgery, Shariati Hospital and School of MedicineTehran University of Medical SciencesTehranIran
| | - Fardis Vosoughi
- Department of Orthopedic Surgery, Shariati Hospital and School of MedicineTehran University of Medical SciencesTehranIran
| | - Alireza Mirzamohamadi
- Center for Orthopedic Trans‐Disciplinary Applied ResearchTehran University of Medical SciencesTehranIran
| | - Hiva Saffar
- Department of Pathology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Mahdi Shafieian
- Department of Biomedical EngineeringAmirkabir University of Technology (Tehran Polytechnic)TehranIran
| | - Leila Oryadi Zanjani
- Center for Orthopedic Trans‐Disciplinary Applied ResearchTehran University of Medical SciencesTehranIran
| | - Hossein Nematian
- Center for Orthopedic Trans‐Disciplinary Applied ResearchTehran University of Medical SciencesTehranIran
| | - Mohammad Hossein Nabian
- Center for Orthopedic Trans‐Disciplinary Applied ResearchTehran University of Medical SciencesTehranIran
| |
Collapse
|
2
|
Li J, Sun H, Guan J, Li B, Jin C, Xie S, Liu Y. Immunogenicity of chondrocyte sheets: a review. Front Immunol 2025; 16:1529384. [PMID: 40124370 PMCID: PMC11926542 DOI: 10.3389/fimmu.2025.1529384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
The chondrocyte sheet is a sheet-like cell structure obtained by separating in vitro expanded and fused autologous chondrocytes from the bottom of the culture dish by physical means. The cell sheet contains autologous chondrocytes, extracellular matrix secreted by chondrocytes, and connective structures established between cells and matrix, and between cells and cells. In cartilage tissue engineering, chondrocyte sheets technology has great potential for the treatment of cartilage defects. Chondrocyte sheets have a low immunogenicity because they avoid the immune reaction caused by scaffolding materials. However, chondrocyte sheets can still cause severe local tissue swelling in the short term after implantation, resulting in a poor patient experience. In individual cases, an inflammatory reaction may even occur, leading to resorption of the chondrocyte sheet. This may be immunogenetically related to chondrocyte membrane surface-associated antigens, components of the extracellular matrix secreted by chondrocytes, and various bioactive components in the culture medium used during in vitro chondrocyte culture. Therefore, in order to investigate the causes of local tissue swelling and immune-inflammatory reactions induced by the implantation of chondrocyte sheets, this article reviews the immunogenicity of chondrocyte-associated antigens, components of the extracellular matrix of cartilage, and the active components of the cell culture medium.
Collapse
Affiliation(s)
- Juncen Li
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Huilin Sun
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaqi Guan
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Bohui Li
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Jin
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Shanhong Xie
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Liu
- College of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
- National Tissue Engineering Center of China, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Resthetic Biotechnology. Co., Ltd, Shanghai, China
| |
Collapse
|
3
|
Zhang L, Arenas Hoyos I, Helmer A, Banz Y, Haenni B, Lese I, Constantinescu M, Rieben R, Olariu R. Immune Rejection of Cartilage in a Swine Vascularized Composite Allotransplantation Model. Transplant Proc 2024; 56:1896-1903. [PMID: 39242314 DOI: 10.1016/j.transproceed.2024.08.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/24/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Cartilage is a crucial tissue in vascularized composite allotransplantation (VCA) and plays a pivotal role in restoring motor function, especially in joint allotransplantation. Nevertheless, our understanding of immune rejection in cartilage remains limited and contentious. This study seeks to investigate the immune rejection of cartilage in a large animal model of VCA. METHODS Cartilage, including articular cartilage and meniscus, as well as skin, muscle and lymph node, was retrieved from a swine heterotopic VCA graft when the skin of the graft suffered from grade III-IV rejection. Histologic examination, transmission electron microscopy and immunofluorescent staining were used to investigate immune rejection. RESULTS Histologic examination revealed the infiltration of inflammatory cells and tissue destruction in cartilage. Transmission electron microscopy confirmed tissue damage and necrosis in cartilage. However, cartilage exhibited milder tissue damage when compared to rejected skin and muscle. Immunofluorescent staining revealed the activation of both the innate and adaptive immune systems, accompanied by an up-regulation of cell death biomarkers, including apoptosis and pyroptosis, in the rejected cartilage. CONCLUSION Our study demonstrates that cartilage is not immunologically privileged and undergoes immune rejection concurrently with skin and muscle in the VCA graft, though with less severe inflammation and rejection.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Isabel Arenas Hoyos
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Anja Helmer
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Yara Banz
- Institute of Pathology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Beat Haenni
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Ioana Lese
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Mihai Constantinescu
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Radu Olariu
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
4
|
Uto S, Hikita A, Mori D, Sakamoto T, Yano F, Ohba S, Saito T, Takato T, Hoshi K. Subcutaneously Transplanted Fresh Cartilage in Allogeneic and Xenogeneic Immunocompetent Mouse. Tissue Eng Part A 2023; 29:541-556. [PMID: 37548556 DOI: 10.1089/ten.tea.2023.0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
Cartilage is considered to be immune privileged in general. Clinically, live cells are removed from subcutaneously transplanted allogeneic cartilage mainly for preservation and for infection control. However, because maintaining cartilage feature requires live chondrocyte, it would be beneficial to subcutaneously transplant cartilage with live chondrocyte even if it was allogeneic. We harvested femoral head from 3-week-old male C57BL/6 mice, subcutaneously transplanted to 6-week-old male mice, BALB/c, BALB/c nu/nu, or C57BL/6-Tg (enhanced green fluorescent protein [EGFP] under the control of the CMV-IE enhancer, chicken beta-actin promoter, rabbit beta-globin genomic DNA [CAG promoter]), as allogeneic, allogeneic immunodeficient control, or syngeneic transplantation. We also transplanted cartilaginous particles from human induced pluripotent stem cells derived from human leukocyte antigen homozygous donor to 6-week-old male mice either BALB/c and BALB/c nu/nu as xenogeneic or xenogeneic immunodeficient control. The transplantation periods were 1, 2, 3, 4, 8, 12, and 24 weeks. As the result, we did not observe exposure of the transplant or apparent macroscopic inflammatory in all samples. Histological analysis suggested that the femoral head showed focal ossification and thinning in syngeneic transplantation. In allogeneic transplantation, slight invasion of CD3 (+) T cell and the denaturation of the cartilage were observed, suggesting immune reaction against allogeneic cartilage. In xenogeneic transplantation, slight invasion of CD3 (+) cell and CD4 (+) cell and the structure of the perichondrium-like tissue got unclear, suggesting slight immune reaction against xenogeneic cartilage. Our findings suggest that we should carefully investigate for appropriate procedure to control immune reaction against allogeneic cartilage with live chondrocyte and to maintain its cartilage feature for long time.
Collapse
Affiliation(s)
- Sakura Uto
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Division of Tissue Engineering, Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Atsuhiko Hikita
- Division of Tissue Engineering, Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Daisuke Mori
- Department of Bone and Cartilage Regenerative Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomoaki Sakamoto
- Division of Tissue Engineering, Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Fumiko Yano
- Department of Bone and Cartilage Regenerative Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shinsuke Ohba
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Taku Saito
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tsuyoshi Takato
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- JR Tokyo General Hospital, Shibuya-ku, Tokyo, Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Division of Tissue Engineering, Department of Oral-Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
5
|
Vo NX, Che UTT, Ngo TTT, Bui TT. Economic Evaluation of Glucosamine in Knee Osteoarthritis Treatments in Vietnam. Healthcare (Basel) 2023; 11:2502. [PMID: 37761699 PMCID: PMC10531128 DOI: 10.3390/healthcare11182502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Osteoarthritis (OA) is the degeneration of cartilage in joints that results in bones rubbing against each other; it causes uncomfortable symptoms such as pain, swelling, and stiffness and can lead to disability. It usually occurs in the elderly and causes a large medical burden. The aim of this study is to evaluate the cost-effectiveness between the standard treatment for osteoarthritis and standard treatment with added crystalline glucosamine sulfate at various stages. Markov analysis modeling was applied to evaluate the effect of both adding glucosamine compared to standard treatment from a societal perspective during whole patients' lifetimes. Data input was collected from reviews in previous studies. The outcome was measured in quality-adjusted life years (QALYs), and the Incremental Cost-Effectiveness Ratio (ICER) from a societal perspective was applied with 3% and discounted for all costs and outcomes. One-way analysis via the Tornado diagram was performed to investigate the change in factors in the model. In general, adding glucosamine into the standard treatment proved to be more cost-effective compared to the standard treatment. Particularly, the early-stage addition of glucosamine in the treatment was cost-effective compared to the post-stage addition of glucosamine. The addition of supplementing crystalline glucosamine sulfate to the whole regimen at any stage was cost-effective at the willingness-to-pay (WTP) threshold.
Collapse
Affiliation(s)
- Nam Xuan Vo
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam; (U.T.T.C.); (T.T.T.N.)
| | - Uyen Thi Thuc Che
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam; (U.T.T.C.); (T.T.T.N.)
| | - Thanh Thi Thanh Ngo
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam; (U.T.T.C.); (T.T.T.N.)
| | - Tien Thuy Bui
- Faculty of Pharmacy, Le Van Thinh Hospital, Ho Chi Minh City 700000, Vietnam;
| |
Collapse
|
6
|
Zhang L, Hoyos IA, Zubler C, Rieben R, Constantinescu M, Olariu R. Challenges and opportunities in vascularized composite allotransplantation of joints: a systematic literature review. Front Immunol 2023; 14:1179195. [PMID: 37275912 PMCID: PMC10235447 DOI: 10.3389/fimmu.2023.1179195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Background Joint allotransplantation (JA) within the field of vascularized composite allotransplantation (VCA) holds great potential for functional and non-prosthetic reconstruction of severely damaged joints. However, clinical use of JA remains limited due to the immune rejection associated with all forms of allotransplantation. In this study, we aim to provide a comprehensive overview of the current state of JA through a systematic review of clinical, animal, and immunological studies on this topic. Methods We conducted a systematic literature review in accordance with the PRISMA guidelines to identify relevant articles in PubMed, Cochrane Library, and Web of Science databases. The results were analyzed, and potential future prospects were discussed in detail. Results Our review included 14 articles describing relevant developments in JA. Currently, most JA-related research is being performed in small animal models, demonstrating graft survival and functional restoration with short-term immunosuppression. In human patients, only six knee allotransplantations have been performed to date, with all grafts ultimately failing and a maximum graft survival of 56 months. Conclusion Research on joint allotransplantation has been limited over the last 20 years due to the rarity of clinical applications, the complex nature of surgical procedures, and uncertain outcomes stemming from immune rejection. However, the key to overcoming these challenges lies in extending graft survival and minimizing immunosuppressive side effects. With the emergence of new immunosuppressive strategies, the feasibility and clinical potential of vascularized joint allotransplantation warrants further investigation.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Plastic and Reconstructive Surgery, Plastic and Reconstructive Surgery Center, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Isabel Arenas Hoyos
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Cédric Zubler
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Mihai Constantinescu
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Radu Olariu
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
7
|
Yu TH, Yeh TT, Su CY, Yu NY, Chen IC, Fang HW. Preparation and Characterization of Extracellular Matrix Hydrogels Derived from Acellular Cartilage Tissue. J Funct Biomater 2022; 13:jfb13040279. [PMID: 36547539 PMCID: PMC9788521 DOI: 10.3390/jfb13040279] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Decellularized matrices can effectively reduce severe immune rejection with their cells and eliminated nucleic acid material and provide specific environments for tissue repair or tissue regeneration. In this study, we prepared acellular cartilage matrix (ACM) powder through the decellularization method and developed ACM hydrogels by physical, chemical, and enzymatic digestion methods. The results demonstrated that the small size group of ACM hydrogels exhibited better gel conditions when the concentration of ACM hydrogels was 30 and 20 mg/mL in 1N HCl through parameter adjustment. The data also confirmed that the ACM hydrogels retained the main components of cartilage: 61.18% of glycosaminoglycan (GAG) and 78.29% of collagen, with 99.61% of its DNA removed compared to samples without the decellularization procedure (set as 100%). Through turbidimetric gelation kinetics, hydrogel rheological property analysis, and hydrogel tissue physical property testing, this study also revealed that increasing hydrogel concentration is helpful for gelation. Besides, the ex vivo test confirmed that a higher concentration of ACM hydrogels had good adhesive properties and could fill in cartilage defects adequately. This study offers useful information for developing and manufacturing ACM hydrogels to serve as potential alternative scaffolds for future cartilage defect treatment.
Collapse
Affiliation(s)
- Tsong-Hann Yu
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Chenggong Rd., Taipei 114202, Taiwan
| | - Tsu-Te Yeh
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Chenggong Rd., Taipei 114202, Taiwan
| | - Chen-Ying Su
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan
| | - Ni-Yin Yu
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan
| | - I-Cheng Chen
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan
- Accelerator for Happiness and Health Industry, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan
- Correspondence: (I.-C.C.); (H.-W.F.); Tel.: +886-2-2771-2171 (ext. 2521) (H.-W.F.)
| | - Hsu-Wei Fang
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan
- Accelerator for Happiness and Health Industry, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan 35053, Taiwan
- Correspondence: (I.-C.C.); (H.-W.F.); Tel.: +886-2-2771-2171 (ext. 2521) (H.-W.F.)
| |
Collapse
|
8
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
9
|
ECM-Mimicking Hydrogels Loaded with Bone Mesenchymal Stem Cell-Derived Exosomes for the Treatment of Cartilage Defects. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3450672. [PMID: 36387356 PMCID: PMC9649317 DOI: 10.1155/2022/3450672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/20/2022] [Indexed: 11/06/2022]
Abstract
It is well-established that treating articular cartilage injuries is clinically challenging since they lack blood arteries, nerves, and lymphoid tissue. Recent studies have revealed that bone marrow stem cell-derived exosomes (BMSCs-Exos) exert significant chondroprotective effects through paracrine secretions, and hydrogel-based materials can synergize the exosomes through sustained release. Therefore, this research aims to synthesize an ECM (extracellular matrix)-mimicking gelatin methacryloyl (GelMA) hydrogel modified by gelatin combined with BMSCs-derived exosomes to repair cartilage damage. We first isolated and characterized exosomes from BMSCs supernatant and then loaded the exosomes into GelMA hydrogel to investigate cartilage repair effects in in vitro and in vivo experiments. The outcomes showed that the GelMA hydrogel has good biocompatibility with a 3D (three-dimensional) porous structure, exhibiting good carrier characteristics for exosomes. Furthermore, BMSCs-Exos had a significant effect on promoting chondrocyte ECM production and chondrocyte proliferation, and the GelMA hydrogel could enhance this effect through a sustained-release effect. Similarly, in vivo experiments showed that GelMA-Exos promoted cartilage regeneration in rat joint defects and the synthesis of related cartilage matrix proteins.
Collapse
|
10
|
Recent strategies of collagen-based biomaterials for cartilage repair: from structure cognition to function endowment. JOURNAL OF LEATHER SCIENCE AND ENGINEERING 2022. [DOI: 10.1186/s42825-022-00085-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractCollagen, characteristic in biomimetic composition and hierarchical structure, boasts a huge potential in repairing cartilage defect due to its extraordinary bioactivities and regulated physicochemical properties, such as low immunogenicity, biocompatibility and controllable degradation, which promotes the cell adhesion, migration and proliferation. Therefore, collagen-based biomaterial has been explored as porous scaffolds or functional coatings in cell-free scaffold and tissue engineering strategy for cartilage repairing. Among those forming technologies, freeze-dry is frequently used with special modifications while 3D-printing and electrospinning serve as the structure-controller in a more precise way. Besides, appropriate cross-linking treatment and incorporation with bioactive substance generally help the collagen-based biomaterials to meet the physicochemical requirement in the defect site and strengthen the repairing performance. Furthermore, comprehensive evaluations on the repair effects of biomaterials are sorted out in terms of in vitro, in vivo and clinical assessments, focusing on the morphology observation, characteristic production and critical gene expression. Finally, the challenge of biomaterial-based therapy for cartilage defect repairing was summarized, which is, the adaption to the highly complex structure and functional difference of cartilage.
Graphical abstract
Collapse
|
11
|
Ge D, O'Brien MJ, Savoie FH, Gimble JM, Wu X, Gilbert MH, Clark-Patterson GL, Schuster JD, Miller KS, Wang A, Myers L, You Z. Human adipose-derived stromal/stem cells expressing doublecortin improve cartilage repair in rabbits and monkeys. NPJ Regen Med 2021; 6:82. [PMID: 34848747 PMCID: PMC8633050 DOI: 10.1038/s41536-021-00192-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 10/29/2021] [Indexed: 11/25/2022] Open
Abstract
Localized cartilage lesions in early osteoarthritis and acute joint injuries are usually treated surgically to restore function and relieve pain. However, a persistent clinical challenge remains in how to repair the cartilage lesions. We expressed doublecortin (DCX) in human adipose-derived stromal/stem cells (hASCs) and engineered hASCs into cartilage tissues using an in vitro 96-well pellet culture system. The cartilage tissue constructs with and without DCX expression were implanted in the knee cartilage defects of rabbits (n = 42) and monkeys (n = 12). Cohorts of animals were euthanized at 6, 12, and 24 months after surgery to evaluate the cartilage repair outcomes. We found that DCX expression in hASCs increased expression of growth differentiation factor 5 (GDF5) and matrilin 2 in the engineered cartilage tissues. The cartilage tissues with DCX expression significantly enhanced cartilage repair as assessed macroscopically and histologically at 6, 12, and 24 months after implantation in the rabbits and 24 months after implantation in the monkeys, compared to the cartilage tissues without DCX expression. These findings suggest that hASCs expressing DCX may be engineered into cartilage tissues that can be used to treat localized cartilage lesions.
Collapse
Affiliation(s)
- Dongxia Ge
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Michael J O'Brien
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Felix H Savoie
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jeffrey M Gimble
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- LaCell LLC and Obatala Sciences Inc., New Orleans, LA, USA
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Xiying Wu
- LaCell LLC and Obatala Sciences Inc., New Orleans, LA, USA
| | - Margaret H Gilbert
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | | | - Jason D Schuster
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Kristin S Miller
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Alun Wang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Leann Myers
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropic Medicine, New Orleans, LA, USA
| | - Zongbing You
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA.
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Cancer Center and Louisiana Cancer Research Consortium, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, USA.
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA.
| |
Collapse
|
12
|
Jaibaji M, Jaibaji R, Volpin A. Mesenchymal Stem Cells in the Treatment of Cartilage Defects of the Knee: A Systematic Review of the Clinical Outcomes. Am J Sports Med 2021; 49:3716-3727. [PMID: 33555942 DOI: 10.1177/0363546520986812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Osteochondral lesions are a common clinical problem and their management has been historically challenging. Mesenchymal stem cells have the potential to differentiate into chondrocytes and thus restore hyaline cartilage to the defect, theoretically improving clincal outcomes in these patients. They can also be harvested with minimal donor site morbidity. PURPOSE To assess the clinical and functional outcomes of mesenchymal stem cell implantation to treat isolated osteochondral defects of the knee. A secondary purpose is to assess the quality of the current available evidence as well as the radiological and histological outcomes. We also reviewed the cellular preparation and operative techniques for implantation. STUDY DESIGN Systematic review. METHODS A comprehensive literature search of 4 databases was carried out: CINAHL, Embase, MEDLINE, and PubMed. We searched for clinical studies reporting the outcomes on a minimum of 5 patients with at least 12 months of follow-up. Clinical, radiological, and histological outcomes were recorded. We also recorded demographics, stem cell source, culture technique, and operative technique. Methodological quality of each study was assessed using the modified Coleman methodology score, and risk of bias for the randomized controlled studies was assessed using the Cochrane Collaboration tool. RESULTS Seventeen studies were found, encompassing 367 patients. The mean patient age was 35.1 years. Bone marrow was the most common source of stem cells utilized. Mesenchymal stem cell therapy consistently demonstrated good short- to medium-term outcomes in the studies reviewed with no serious adverse events being recorded. There was significant heterogeneity in cell harvesting and preparation as well as in the reporting of outcomes. CONCLUSION Mesenchymal stem cells demonstrated a clinically relevant improvement in outcomes in patients with osteochondral defects of the knee. More research is needed to establish an optimal treatment protocol, long-term outcomes, and superiority over other therapies. REGISTRATION CRD42020179391 (PROSPERO).
Collapse
Affiliation(s)
- Monketh Jaibaji
- Division of Interventional Sciences, University College London, London, UK
| | - Rawan Jaibaji
- Division of Interventional Sciences, University College London, London, UK
| | | |
Collapse
|
13
|
Wei F, Liu S, Chen M, Tian G, Zha K, Yang Z, Jiang S, Li M, Sui X, Chen Z, Guo Q. Host Response to Biomaterials for Cartilage Tissue Engineering: Key to Remodeling. Front Bioeng Biotechnol 2021; 9:664592. [PMID: 34017827 PMCID: PMC8129172 DOI: 10.3389/fbioe.2021.664592] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/14/2021] [Indexed: 12/18/2022] Open
Abstract
Biomaterials play a core role in cartilage repair and regeneration. The success or failure of an implanted biomaterial is largely dependent on host response following implantation. Host response has been considered to be influenced by numerous factors, such as immune components of materials, cytokines and inflammatory agents induced by implants. Both synthetic and native materials involve immune components, which are also termed as immunogenicity. Generally, the innate and adaptive immune system will be activated and various cytokines and inflammatory agents will be consequently released after biomaterials implantation, and further triggers host response to biomaterials. This will guide the constructive remolding process of damaged tissue. Therefore, biomaterial immunogenicity should be given more attention. Further understanding the specific biological mechanisms of host response to biomaterials and the effects of the host-biomaterial interaction may be beneficial to promote cartilage repair and regeneration. In this review, we summarized the characteristics of the host response to implants and the immunomodulatory properties of varied biomaterial. We hope this review will provide scientists with inspiration in cartilage regeneration by controlling immune components of biomaterials and modulating the immune system.
Collapse
Affiliation(s)
- Fu Wei
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China.,Department of Orthopedics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Shuyun Liu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Mingxue Chen
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Guangzhao Tian
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Kangkang Zha
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Zhen Yang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | | | - Muzhe Li
- Department of Orthopedics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xiang Sui
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Zhiwei Chen
- Department of Orthopedics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Quanyi Guo
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries, PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
14
|
Watanabe T, Asawa Y, Watanabe M, Okubo R, Nio M, Takato T, Hoshi K, Hikita A. The usefulness of the decellularized matrix from three-dimensional regenerative cartilage as a scaffold material. Regen Ther 2020; 15:312-322. [PMID: 33426234 PMCID: PMC7770429 DOI: 10.1016/j.reth.2020.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/30/2020] [Accepted: 11/18/2020] [Indexed: 11/30/2022] Open
Abstract
In cartilage tissue engineering, research on materials for three-dimensional (3D) scaffold has attracted attention. Decellularized matrix can be one of the candidates for the scaffold material. In this study, decellularization of regenerated cartilage was carried out and its effectiveness as a scaffold material was examined. Three-dimensionally-cultured cartilage constructs in the differentiation medium containing IGF-1 produced more cartilage matrix than those in the proliferation medium. Detergent-enzymatic method (DEM) could decellularize 3D-cultured cartilage constructs only by 1 cycle without breaking down the structure of the constructs. In vitro, newly-seeded chondrocytes were infiltrated and engrafted into decellularized constructs in the proliferation medium, and newly formed fibers were observed around the surface where newly-seeded cells were attached. Recellularized constructs could mature similarly as those without decellularization in vivo. The decellularized 3D-cultured matrix from regenerative cartilage is expected to be used as a scaffold material in the future.
Collapse
Affiliation(s)
- Tomohiko Watanabe
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Yukiyo Asawa
- Department of Cell & Tissue Engineering (Fujisoft), Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan.,Department of Tissue Engineering, The University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Makoto Watanabe
- Department of Tissue Engineering, The University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ryuji Okubo
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Masaki Nio
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | - Tsuyoshi Takato
- JR Tokyo General Hospital, Yoyogi 2-1-3, Shibuya-ku, Tokyo 151-8528, Japan
| | - Kazuto Hoshi
- Department of Tissue Engineering, The University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan.,Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Atsuhiko Hikita
- Department of Cell & Tissue Engineering (Fujisoft), Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan.,Department of Tissue Engineering, The University of Tokyo Hospital, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
15
|
Serel S, Çerkez C, Işılgan Alpat SE, Yiğit P, Can B, Göktürk H. Non-significant Effects of The Geometric Shape of Autologous Cartilage Grafts on Tissue Healing: An Animal Study. Aesthetic Plast Surg 2020; 44:1845-1853. [PMID: 32766922 DOI: 10.1007/s00266-020-01904-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/26/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND The reconstruction of cartilage defects for cosmetic and/or functional reasons has become routine in plastic and reconstructive surgery. However, it remains challenging due to the slow turnover and low viability of cartilage grafts. Although autologous grafts can be used to determine the shape of the defect in cartilage-reconstruction surgeries, the effect of defect shape on cartilage healing has not been reported. Here, we present the first study aiming to investigate the influence of cartilage graft geometry on healing. METHODS Twelve New Zealand white rabbits were used in the study. Square-, rectangle-, sphere-, and fusiform-shaped cartilage defects were applied to both ears with 1-cm2 geometric templates that completely elevated the cartilage tissue without damaging the opposite perichondrium. As a control, the removed cartilage was sutured back to the right ear, whereas the left ear was sutured back without any graft. Histological examinations were made on samples taken during surgery and those taken four months post-surgery. Chondrocyte production and organisation, chondrocyte vacuolisation, collagen synthesis, proteoglycan levels, vascularisation, focal bleeding, and peripheral proliferation were scored independently by two histologists. RESULTS There was no statistically significant difference in the growth rates of either the control or experimental cartilage tissues when compared with that of the initial cartilage tissue (p = 0.083). Histologic comparisons revealed better outcomes in the grafted cartilage groups compared to those receiving the donor cartilage, but this was not statistically significant. CONCLUSIONS This study demonstrates that the geometric shape of the defect has no significant effect on cartilage healing. LEVEL OF EVIDENCE This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
16
|
Desai S, Jayasuriya CT. Implementation of Endogenous and Exogenous Mesenchymal Progenitor Cells for Skeletal Tissue Regeneration and Repair. Bioengineering (Basel) 2020; 7:E86. [PMID: 32759659 PMCID: PMC7552784 DOI: 10.3390/bioengineering7030086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Harnessing adult mesenchymal stem/progenitor cells to stimulate skeletal tissue repair is a strategy that is being actively investigated. While scientists continue to develop creative and thoughtful ways to utilize these cells for tissue repair, the vast majority of these methodologies can ultimately be categorized into two main approaches: (1) Facilitating the recruitment of endogenous host cells to the injury site; and (2) physically administering into the injury site cells themselves, exogenously, either by autologous or allogeneic implantation. The aim of this paper is to comprehensively review recent key literature on the use of these two approaches in stimulating healing and repair of different skeletal tissues. As expected, each of the two strategies have their own advantages and limitations (which we describe), especially when considering the diverse microenvironments of different skeletal tissues like bone, tendon/ligament, and cartilage/fibrocartilage. This paper also discusses stem/progenitor cells commonly used for repairing different skeletal tissues, and it lists ongoing clinical trials that have risen from the implementation of these cells and strategies. Lastly, we discuss our own thoughts on where the field is headed in the near future.
Collapse
Affiliation(s)
| | - Chathuraka T. Jayasuriya
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and the Rhode Island Hospital, Providence, RI 02903, USA;
| |
Collapse
|
17
|
Calvo R, Espinosa M, Figueroa D, Pozo LM, Conget P. Assessment of Cell Viability of Fresh Osteochondral Allografts in N-Acetylcysteine-Enriched Medium. Cartilage 2020; 11:117-121. [PMID: 29985056 PMCID: PMC6921953 DOI: 10.1177/1947603518786547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE The purpose of this study was to evaluate the effect of N-acetylcysteine (NAC)-enriched storage medium on fresh osteochondral viability at 4°C. Our hypothesis was that the cell viability of chondrocytes obtained from human osteochondral tissue and stored at 4°C significantly improves in the presence of NAC. DESIGN Controlled laboratory study. For this study, 8 samples of femoral condyle osteochondral tissue were obtained from patients undergoing total knee replacement. The samples were stored at either 4°C in phosphate-buffered saline (PBS) or at 3 different concentrations of NAC (NAC 1, 2, and 5 mM). Cell viability was analyzed at time 0 and 4 weeks by flow cytometry. The results of cell viability (median) were analyzed statistically using analysis of variance and Tukey's post hoc test. P values <0.05 were considered statistically significant. RESULTS The viability at time 0 was 95.5% ± 3.7%. At 4 weeks, the cell viability was 56.8% ± 20.1% in the control group (PBS), 83.8% ± 11.9% in the group stored with NAC 1 mM, 73.4% ± 13.6% in the group stored with NAC 2 mM, and 66.4% ± 27.7% in the group stored with NAC 5 mM. A statistically significant difference from the baseline viability (time 0) was observed in the PBS control group (P = 0.0018) but not in the other groups. A statistically significant difference was observed in the NAC 1 mM group compared with the PBS group (P = 0.0255). CONCLUSION The use of NAC at 1 mM concentration improves cell viability after 4 weeks of storage in chondrocytes obtained from human osteochondral tissue.
Collapse
Affiliation(s)
- Rafael Calvo
- Departamento de Traumatología, Facultad
de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Maximiliano Espinosa
- Departamento de Traumatología, Facultad
de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile,Maximiliano Espinosa, Faculty of Medicine,
Clínica Alemana–Universidad del Desarrollo, Vitacura 5951, Santiago, Región
Metropolitana, Chile.
| | - David Figueroa
- Departamento de Traumatología, Facultad
de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Luz María Pozo
- Instituto de Ciencias e Innovación en
Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo,
Santiago, Chile
| | - Paulette Conget
- Instituto de Ciencias e Innovación en
Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo,
Santiago, Chile
| |
Collapse
|
18
|
Elezagic D, Mörgelin M, Hermes G, Hamprecht A, Sengle G, Lau D, Höllriegl S, Wagener R, Paulsson M, Streichert T, Klatt AR. Antimicrobial peptides derived from the cartilage.-specific C-type Lectin Domain Family 3 Member A (CLEC3A) - potential in the prevention and treatment of septic arthritis. Osteoarthritis Cartilage 2019; 27:1564-1573. [PMID: 31279936 DOI: 10.1016/j.joca.2019.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/20/2019] [Accepted: 06/11/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the antimicrobial activity of peptides derived from C-type Lectin Domain Family 3 Member A (CLEC3A), shed light on the mechanism of antimicrobial activity and assess their potential application in prevention and treatment of septic arthritis. DESIGN We performed immunoblot to detect CLEC3A peptides in human cartilage extracts. To investigate their antimicrobial activity, we designed peptides and recombinantly expressed CLEC3A domains and used them to perform viable count assays using E.coli, P.aeruginosa and S.aureus. We investigated the mechanism of their antimicrobial activity by fluorescence and scanning electron microscopy, performed ELISA-style immunoassays and transmission electron microscopy to test for lipopolysaccharide binding and surface plasmon resonance to test for lipoteichoic acid (LTA) binding. We coated CLEC3A peptides on titanium, a commonly used prosthetic material, and performed fluorescence microscopy to quantify bacterial adhesion. Moreover, we assessed the peptides' cytotoxicity against primary human chondrocytes using MTT cell viability assays. RESULTS CLEC3A fragments were detected in human cartilage extracts. Moreover, bacterial supernatants lead to fragmentation of recombinant and cartilage-derived CLEC3A. CLEC3A-derived peptides killed E.coli, P.aeruginosa and S.aureus, permeabilized bacterial membranes and bound lipopolysaccharide and LTA. Coating CLEC3A antimicrobial peptides (AMPs) on titanium lead to significantly reduced bacterial adhesion to the material. In addition, microbicidal concentrations of CLEC3A peptides in vitro displayed no direct cytotoxicity against primary human chondrocytes. CONCLUSIONS We identify cartilage-specific AMPs originating from CLEC3A, resolve the mechanism of their antimicrobial activity and point to a novel approach in the prevention and treatment of septic arthritis using potent, non-toxic, AMPs.
Collapse
Affiliation(s)
- D Elezagic
- Institute for Clinical Chemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - M Mörgelin
- Department of Clinical Sciences, Division of Infection Medicine, Biomedical Center, Lund University, 22184 Lund, Sweden
| | - G Hermes
- Institute for Clinical Chemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - A Hamprecht
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University of Cologne, 50935 Cologne, Germany
| | - G Sengle
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - D Lau
- Institute for Clinical Chemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - S Höllriegl
- Cologne Braunsfeld Trinity Hospital, 50933 Cologne, Germany
| | - R Wagener
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - M Paulsson
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - T Streichert
- Institute for Clinical Chemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - A R Klatt
- Institute for Clinical Chemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
19
|
Meta-Analysis and Evidence Base for the Efficacy of Autologous Bone Marrow Mesenchymal Stem Cells in Knee Cartilage Repair: Methodological Guidelines and Quality Assessment. Stem Cells Int 2019; 2019:3826054. [PMID: 31089328 PMCID: PMC6476108 DOI: 10.1155/2019/3826054] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/13/2019] [Indexed: 02/07/2023] Open
Abstract
The aim of this study is to review all the published clinical trials on autologous bone marrow mesenchymal stem cells (BM-MSCs) in the repair of cartilage lesions of the knee. We performed a comprehensive search in three electronic databases: PubMed, Medline via Ovid, and Web of Science. A systematic review was conducted according to the guidelines of PRISMA protocol and the Cochrane Handbook for Systematic Reviews of Interventions. The modified Coleman methodology score was used to assess the quality of the included studies. Meta-analysis was conducted to estimate the effect size for Pain and function change after receiving BM-MSCs. Thirty-three studies—including 724 patients of mean age 44.2 years—were eligible. 50.7% of the included patients received cultured BM-MSCs for knee cartilage repair. There was improvement in the MINORS quality score over time with a positive correlation with the publication year. Meta-analysis indicated better improvement and statistical significance in the Visual Analog Scale for Pain, IKDC Function, Tegner Activity Scale, and Lysholm Knee Score after administration of noncultured BM-MSCs when compared to evaluation before the treatment. Meanwhile, there was a clear methodological defect in most studies with an average modified Coleman methodology score (MCMS) of 55. BM-MSCs revealed a clinically relevant improvement in pain, function, and histological regeneration.
Collapse
|
20
|
Application of induced pluripotent stem cell transplants: Autologous or allogeneic? Life Sci 2018; 212:145-149. [DOI: 10.1016/j.lfs.2018.09.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 09/30/2018] [Indexed: 12/14/2022]
|
21
|
Akatsu Y, Enomoto T, Yamaguchi S, Tahara M, Fukawa T, Endo J, Hoshi H, Yamamoto Y, Sasaki T, Takahashi K, Akagi R, Sasho T. Age-dependent differences in response to partial-thickness cartilage defects in a rat model as a measure to evaluate the efficacy of interventions for cartilage repair. Cell Tissue Res 2018; 375:425-435. [PMID: 30259137 DOI: 10.1007/s00441-018-2914-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 08/17/2018] [Indexed: 12/18/2022]
Abstract
The objectives of this study are (1) to examine age-dependent longitudinal differences in histological responses after creation of partial-thickness articular cartilage defects (PTCDs) in rats and to use this model (2) to objectively evaluate the effectiveness of interventions for cartilage repair. Linear PTCDs were created at a depth of 100 μm in the weight-bearing region of the medial femoral condyle in rats of different ages (3 weeks, 6 weeks, 10 weeks and 14 weeks). One day, one week, two weeks, four weeks and twelve weeks after PTCD generation, spontaneous healing was evaluated histologically and immunohistochemically. Effects of interventions comprising mesenchymal stem cells (MSCs) or platelet-rich plasma (PRP) or both on 14-week-old PTCD rats were evaluated and compared with natural courses in rats of other ages. Younger rats exhibited better cartilage repair. Cartilage in 3-week-old and 6-week-old rats exhibited nearly normal restoration after 4-12 weeks. Cartilage in 14-week-old rats deteriorated over time and early signs of cartilage degeneration were observed. With injection of MCSs alone or MSCs + PRP, 14-week-old PTCD rats showed almost the same reparative cartilage as 6-week-old rats. With injection of PRP, 14-week-old PTCD rats showed almost the same reparative cartilage as 10-week-old rats. This model will be of great use to objectively compare the effects of interventions for small cartilage lesions and may help to advance the development of disease-modifying osteoarthritis drugs.
Collapse
Affiliation(s)
- Yorikazu Akatsu
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Takahiro Enomoto
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Satoshi Yamaguchi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Masamichi Tahara
- Department of Orthopaedic Surgery, Chiba-East-Hospital, Chiba, Japan
| | - Taisuke Fukawa
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Jun Endo
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Hiroko Hoshi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Yohei Yamamoto
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Toshihide Sasaki
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Kazuhisa Takahashi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Ryuichiro Akagi
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Takahisa Sasho
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan. .,Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan.
| |
Collapse
|
22
|
Melick G, Hayman N, Landsman AS. Mesenchymal Stem Cell Applications for Joints in the Foot and Ankle. Clin Podiatr Med Surg 2018; 35:323-330. [PMID: 29861015 DOI: 10.1016/j.cpm.2018.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The use of mesenchymal stem cell injections is a new approach to the treatment of painful joints, particularly in the foot and ankle. Previous studies performed in the knee have considered allogeneic and xenogeneic injections, and autologous cells expanded in culture. This article considers these applications and considers the possibility of performing these types of injections in the foot and ankle joints.
Collapse
Affiliation(s)
- Garrett Melick
- Cambridge Health Alliance, 1493 Cambridge Street, Cambridge, MA 02139, USA
| | - Najwah Hayman
- Cambridge Health Alliance, 1493 Cambridge Street, Cambridge, MA 02139, USA
| | - Adam S Landsman
- Division of Podiatric Surgery, Department of Surgery, Cambridge Health Alliance, Harvard Medical School, 1493 Cambridge Street, Floor 2, Cambridge, MA 02139, USA.
| |
Collapse
|
23
|
Clinical efficacy and safety of mesenchymal stem cell transplantation for osteoarthritis treatment: A meta-analysis. PLoS One 2017; 12:e0175449. [PMID: 28448518 PMCID: PMC5407776 DOI: 10.1371/journal.pone.0175449] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 03/27/2017] [Indexed: 02/06/2023] Open
Abstract
Purpose The aim of this study was to evaluate the therapeutic efficacy and safety of mesenchymal stem cells (MSCs) for the treatment of patients with knee osteoarthritis (OA). Materials We performed a meta-analysis of relevant published clinical studies. An electronic search was conducted for randomized controlled trials (RCTs) of MSC-based therapy in knee OA. The visual analogue scale (VAS), International Knee Documentation Committee (IKDC) form, Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), Lequesne algofunctional indices (Lequesne), Lysholm knee scale (Lysholm), Tegner activity scale (Tegner) and adverse events (AEs) were evaluated. Results Eleven eligible trials with 582 knee OA patients were included in the present meta-analysis. We demonstrated that MSC treatment could significantly decrease VAS and increase IKDC scoresafter a 24-month follow-up compared with controls (P<0.05). MSC therapy also showed significant decreases in WOMAC and Lequesne scores after the 12-month follow-up (P<0.01). Analysis of Lysholm (24-month) and Tegner (12- and 24-month) scores also demonstrated favorable results for MSC treatment (P<0.05). Conclusion Overall, MSC transplantation treatment was shown to be safe and has great potential as an efficacious clinical therapy for patients with knee OA.
Collapse
|
24
|
Abe S, Nochi H, Ito H. Alloreactivity and immunosuppressive properties of articular chondrocytes from osteoarthritic cartilage. J Orthop Surg (Hong Kong) 2016; 24:232-9. [PMID: 27574269 DOI: 10.1177/1602400222] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PURPOSE To determine whether articular chondrocytes derived from osteoarthritic knee joints could evoke alloreactive proliferation of peripheral blood mononuclear cells (PBMC) and inhibit mitogenic activity of polyclonally activated CD4+ major histocompatibility complex (MHC) class II- restricted T cells in vitro. METHODS Osteoarthritic cartilages of 17 patients aged 61 to 85 years were harvested during total knee arthroplasty. Chondrocytes were cultured for experiments. PBMCs, CD4+ T cells, CD8+ T cells, and CD14+ monocytes from healthy subjects were also used. To investigate the allogeneic response and immunosuppressive properties of chondrocytes, assays for one-way mixed lymphocyte reaction (MLR), apoptosis, activated CD4+ T-cell proliferation, and cytotoxic CD8+ T-cells were performed. Chondrocyte cell-surface antigens were examined using flow cytometry. RESULTS Chondrocytes failed to trigger an allogeneic PBMC reaction and did not induce apoptosis of allogeneic PBMCs in the MLR assay. Chondrocytes inhibited the proliferation of polyclonally activated CD4+ T cells via cell-cell contact and escaped the allogeneic cytotoxic reactivity of CD8+ T cells. Chondrocytes expressed MHC class I but not MHC class II molecules or B7-1/-2-positive co-stimulatory molecules. CONCLUSION Chondrocytes from osteoarthritic knees in older patients exhibited similar immunomodulatory properties in vitro to those in juveniles or adults.
Collapse
Affiliation(s)
- S Abe
- Department of Orthopaedic Surgery, Asahikawa Medical University, Japan
| | | | | |
Collapse
|
25
|
Cokelaere S, Malda J, van Weeren R. Cartilage defect repair in horses: Current strategies and recent developments in regenerative medicine of the equine joint with emphasis on the surgical approach. Vet J 2016; 214:61-71. [PMID: 27387728 DOI: 10.1016/j.tvjl.2016.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 01/26/2016] [Accepted: 02/07/2016] [Indexed: 12/27/2022]
Abstract
Chondral and osteochondral lesions due to injury or other pathology are highly prevalent conditions in horses (and humans) and commonly result in the development of osteoarthritis and progression of joint deterioration. Regenerative medicine of articular cartilage is an emerging clinical treatment option for patients with articular cartilage injury or disease. Functional articular cartilage restoration, however, remains a major challenge, but the field is progressing rapidly and there is an increasing body of supportive clinical and scientific evidence. This review gives an overview of the established and emerging surgical techniques employed for cartilage repair in horses. Through a growing insight in surgical cartilage repair possibilities, surgeons might be more stimulated to explore novel techniques in a clinical setting.
Collapse
Affiliation(s)
- Stefan Cokelaere
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 112, 3584 CM Utrecht, NL, Netherlands.
| | - Jos Malda
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 112, 3584 CM Utrecht, NL, Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, NL, Netherlands
| | - René van Weeren
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 112, 3584 CM Utrecht, NL, Netherlands
| |
Collapse
|