1
|
Liang W, Zhou C, Deng Y, Fu L, Zhao J, Long H, Ming W, Shang J, Zeng B. The current status of various preclinical therapeutic approaches for tendon repair. Ann Med 2024; 56:2337871. [PMID: 38738394 PMCID: PMC11095292 DOI: 10.1080/07853890.2024.2337871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/27/2024] [Indexed: 05/14/2024] Open
Abstract
Tendons are fibroblastic structures that link muscle and bone. There are two kinds of tendon injuries, including acute and chronic. Each form of injury or deterioration can result in significant pain and loss of tendon function. The recovery of tendon damage is a complex and time-consuming recovery process. Depending on the anatomical location of the tendon tissue, the clinical outcomes are not the same. The healing of the wound process is divided into three stages that overlap: inflammation, proliferation, and tissue remodeling. Furthermore, the curing tendon has a high re-tear rate. Faced with the challenges, tendon injury management is still a clinical issue that must be resolved as soon as possible. Several newer directions and breakthroughs in tendon recovery have emerged in recent years. This article describes tendon injury and summarizes recent advances in tendon recovery, along with stem cell therapy, gene therapy, Platelet-rich plasma remedy, growth factors, drug treatment, and tissue engineering. Despite the recent fast-growing research in tendon recovery treatment, still, none of them translated to the clinical setting. This review provides a detailed overview of tendon injuries and potential preclinical approaches for treating tendon injuries.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, China
| | - Yongjun Deng
- Department of Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Lifeng Fu
- Department of Orthopedics, Shaoxing City Keqiao District Hospital of Traditional Chinese Medicine, Shaoxing, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengguo Long
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenyi Ming
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jinxiang Shang
- Department of Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Bin Zeng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
2
|
Rathod V, Shrivastav S, Gharpinde MR. Knee Arthroscopy in the Era of Precision Medicine: A Comprehensive Review of Tailored Approaches and Emerging Technologies. Cureus 2024; 16:e70932. [PMID: 39502973 PMCID: PMC11537776 DOI: 10.7759/cureus.70932] [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: 09/29/2024] [Accepted: 10/06/2024] [Indexed: 11/08/2024] Open
Abstract
Knee arthroscopy, a minimally invasive procedure, has transformed the treatment of knee pathologies by enabling direct visualization and management with minimal tissue disruption. Recent advances in precision medicine have introduced a new dimension to this field, allowing for highly individualized surgical approaches considering each patient's unique genetic, environmental, and biomechanical characteristics. This review explores the integration of precision medicine into knee arthroscopy, focusing on tailored approaches and emerging technologies. Key innovations such as robotic-assisted surgery, advanced imaging, and patient-specific instrumentation have enhanced surgical accuracy and patient outcomes, reduced recovery times, and minimized postoperative complications. The review also examines the role of biomarkers in guiding personalized treatment strategies, including ligament reconstructions, meniscal repairs, and cartilage restoration, which are now being refined to cater to the specific needs of individual patients. While the benefits of these innovations are clear, there are challenges to widespread adoption, including cost, resource allocation, and the need for further research to validate the efficacy of precision-driven approaches in knee arthroscopy. Moreover, the ethical considerations surrounding personalized medicine, such as patient privacy and genetic data usage, must also be addressed. Despite these barriers, the future of knee arthroscopy in the era of precision medicine holds great promise, with ongoing developments in artificial intelligence, genomics, and biomarker discovery poised to further refine patient-centered care. This comprehensive review provides valuable insights into how precision medicine reshapes knee arthroscopy, offering a glimpse into the future of more targeted and effective orthopedic interventions. By embracing these advancements, surgeons and healthcare providers can ensure optimal outcomes for patients undergoing knee arthroscopy.
Collapse
Affiliation(s)
- Vinit Rathod
- Department of Orthopedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sandeep Shrivastav
- Department of Orthopedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Milind R Gharpinde
- Department of Orthopedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
3
|
Enayati M, Liu W, Madry H, Neisiany RE, Cucchiarini M. Functionalized hydrogels as smart gene delivery systems to treat musculoskeletal disorders. Adv Colloid Interface Sci 2024; 331:103232. [PMID: 38889626 DOI: 10.1016/j.cis.2024.103232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Despite critical advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy based on the delivery of therapeutic genetic sequences has strong value to offer effective, durable options to decisively manage such disorders. Furthermore, scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy, allowing for the spatiotemporal delivery of candidate genes to sites of injury. Among the many scaffolds for musculoskeletal research, hydrogels raised increasing attention in addition to other potent systems (solid, hybrid scaffolds) due to their versatility and competence as drug and cell carriers in tissue engineering and wound dressing. Attractive functionalities of hydrogels for musculoskeletal therapy include their injectability, stimuli-responsiveness, self-healing, and nanocomposition that may further allow to upgrade of them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. Such functionalized hydrogels may also be tuned to successfully transfer therapeutic genes in a minimally invasive manner in order to protect their cargos and allow for their long-term effects. In light of such features, this review focuses on functionalized hydrogels and demonstrates their competence for the treatment of musculoskeletal disorders using gene therapy procedures, from gene therapy principles to hydrogel functionalization methods and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are being discussed in the perspective of translation in patients. STATEMENT OF SIGNIFICANCE: Despite advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy has strong value in offering effective, durable options to decisively manage such disorders. Scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy. Among many scaffolds for musculoskeletal research, hydrogels raised increasing attention. Functionalities including injectability, stimuli-responsiveness, and self-healing, tune them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. This review introduces functionalized hydrogels for musculoskeletal disorder treatment using gene therapy procedures, from gene therapy principles to functionalized hydrogels and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are discussed from the perspective of translation in patients.
Collapse
Affiliation(s)
- Mohammadsaeid Enayati
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Rasoul Esmaeely Neisiany
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland; Department of Polymer Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany.
| |
Collapse
|
4
|
Primorac D, Molnar V, Tsoukas D, Uzieliene I, Tremolada C, Brlek P, Klarić E, Vidović D, Zekušić M, Pachaleva J, Bernotiene E, Wilson A, Mobasheri A. Tissue engineering and future directions in regenerative medicine for knee cartilage repair: a comprehensive review. Croat Med J 2024; 65:268-287. [PMID: 38868973 PMCID: PMC11157252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/26/2024] [Indexed: 06/14/2024] Open
Abstract
This review evaluates the current landscape and future directions of regenerative medicine for knee cartilage repair, with a particular focus on tissue engineering strategies. In this context, scaffold-based approaches have emerged as promising solutions for cartilage regeneration. Synthetic scaffolds, while offering superior mechanical properties, often lack the biological cues necessary for effective tissue integration. Natural scaffolds, though biocompatible and biodegradable, frequently suffer from inadequate mechanical strength. Hybrid scaffolds, combining elements of both synthetic and natural materials, present a balanced approach, enhancing both mechanical support and biological functionality. Advances in decellularized extracellular matrix scaffolds have shown potential in promoting cell infiltration and integration with native tissues. Additionally, bioprinting technologies have enabled the creation of complex, bioactive scaffolds that closely mimic the zonal organization of native cartilage, providing an optimal environment for cell growth and differentiation. The review also explores the potential of gene therapy and gene editing techniques, including CRISPR-Cas9, to enhance cartilage repair by targeting specific genetic pathways involved in tissue regeneration. The integration of these advanced therapies with tissue engineering approaches holds promise for developing personalized and durable treatments for knee cartilage injuries and osteoarthritis. In conclusion, this review underscores the importance of continued multidisciplinary collaboration to advance these innovative therapies from bench to bedside and improve outcomes for patients with knee cartilage damage.
Collapse
Affiliation(s)
- Dragan Primorac
- Dragan Primorac, Poliklinika Sv. Katarina, Branimirova 71E, 10000 Zagreb, Croatia,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Braun CJ, Adames AC, Saur D, Rad R. Tutorial: design and execution of CRISPR in vivo screens. Nat Protoc 2022; 17:1903-1925. [PMID: 35840661 DOI: 10.1038/s41596-022-00700-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 03/22/2022] [Indexed: 11/09/2022]
Abstract
Here we provide a detailed tutorial on CRISPR in vivo screening. Using the mouse as the model organism, we introduce a range of CRISPR tools and applications, delineate general considerations for 'transplantation-based' or 'direct in vivo' screening design, and provide details on technical execution, sequencing readouts, computational analyses and data interpretation. In vivo screens face unique pitfalls and limitations, such as delivery issues or library bottlenecking, which must be counteracted to avoid screening failure or flawed conclusions. A broad variety of in vivo phenotypes can be interrogated such as organ development, hematopoietic lineage decision and evolutionary licensing in oncogenesis. We describe experimental strategies to address various biological questions and provide an outlook on emerging CRISPR applications, such as genetic interaction screening. These technological advances create potent new opportunities to dissect the molecular underpinnings of complex organismal phenotypes.
Collapse
Affiliation(s)
- Christian J Braun
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany. .,Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany. .,Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Andrés Carbonell Adames
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Dieter Saur
- Institute of Experimental Cancer Therapy, Technical University of Munich, Munich, Germany.,Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany.,Department of Medicine II, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany. .,Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany. .,Department of Medicine II, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
6
|
Zaki S, Blaker CL, Little CB. OA foundations - experimental models of osteoarthritis. Osteoarthritis Cartilage 2022; 30:357-380. [PMID: 34536528 DOI: 10.1016/j.joca.2021.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is increasingly recognised as a disease of diverse phenotypes with variable clinical presentation, progression, and response to therapeutic intervention. This same diversity is readily apparent in the many animal models of OA. However, model selection, study design, and interpretation of resultant findings, are not routinely done in the context of the target human (or veterinary) patient OA sub-population or phenotype. This review discusses the selection and use of animal models of OA in discovery and therapeutic-development research. Beyond evaluation of the different animal models on offer, this review suggests focussing the approach to OA-animal model selection on study objective(s), alignment of available models with OA-patient sub-types, and the resources available to achieve valid and translatable results. How this approach impacts model selection is discussed and an experimental design checklist for selecting the optimal model(s) is proposed. This approach should act as a guide to new researchers and a reminder to those already in the field, as to issues that need to be considered before embarking on in vivo pre-clinical research. The ultimate purpose of using an OA animal model is to provide the best possible evidence if, how, when and where a molecule, pathway, cell or process is important in clinical disease. By definition this requires both model and study outcomes to align with and be predictive of outcomes in patients. Keeping this at the forefront of research using pre-clinical OA models, will go a long way to improving the quality of evidence and its translational value.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Australia.
| | - C L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Australia; Murray Maxwell Biomechanics Laboratory, The Kolling Institute, University of Sydney Faculty of Medicine and Health, At Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Australia.
| |
Collapse
|
7
|
Shestovskaya MV, Bozhkova SA, Sopova JV, Khotin MG, Bozhokin MS. Methods of Modification of Mesenchymal Stem Cells and Conditions of Their Culturing for Hyaline Cartilage Tissue Engineering. Biomedicines 2021; 9:biomedicines9111666. [PMID: 34829895 PMCID: PMC8615732 DOI: 10.3390/biomedicines9111666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) for tissue engineering of hyaline cartilage is a topical area of regenerative medicine that has already entered clinical practice. The key stage of this procedure is to create conditions for chondrogenic differentiation of MSCs, increase the synthesis of hyaline cartilage extracellular matrix proteins by these cells and activate their proliferation. The first such works consisted in the indirect modification of cells, namely, in changing the conditions in which they are located, including microfracturing of the subchondral bone and the use of 3D biodegradable scaffolds. The most effective methods for modifying the cell culture of MSCs are protein and physical, which have already been partially introduced into clinical practice. Genetic methods for modifying MSCs, despite their effectiveness, have significant limitations. Techniques have not yet been developed that allow studying the effectiveness of their application even in limited groups of patients. The use of MSC modification methods allows precise regulation of cell culture proliferation, and in combination with the use of a 3D biodegradable scaffold, it allows obtaining a hyaline-like regenerate in the damaged area. This review is devoted to the consideration and comparison of various methods used to modify the cell culture of MSCs for their use in regenerative medicine of cartilage tissue.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Svetlana A. Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
| | - Julia V. Sopova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Mikhail G. Khotin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Mikhail S. Bozhokin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
8
|
Li ZJ, Yang QQ, Zhou YL. Basic Research on Tendon Repair: Strategies, Evaluation, and Development. Front Med (Lausanne) 2021; 8:664909. [PMID: 34395467 PMCID: PMC8359775 DOI: 10.3389/fmed.2021.664909] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/30/2021] [Indexed: 01/07/2023] Open
Abstract
Tendon is a fibro-elastic structure that links muscle and bone. Tendon injury can be divided into two types, chronic and acute. Each type of injury or degeneration can cause substantial pain and the loss of tendon function. The natural healing process of tendon injury is complex. According to the anatomical position of tendon tissue, the clinical results are different. The wound healing process includes three overlapping stages: wound healing, proliferation and tissue remodeling. Besides, the healing tendon also faces a high re-tear rate. Faced with the above difficulties, management of tendon injuries remains a clinical problem and needs to be solved urgently. In recent years, there are many new directions and advances in tendon healing. This review introduces tendon injury and sums up the development of tendon healing in recent years, including gene therapy, stem cell therapy, Platelet-rich plasma (PRP) therapy, growth factor and drug therapy and tissue engineering. Although most of these therapies have not yet developed to mature clinical application stage, with the repeated verification by researchers and continuous optimization of curative effect, that day will not be too far away.
Collapse
Affiliation(s)
- Zhi Jie Li
- Research for Frontier Medicine and Hand Surgery Research Center, The Nanomedicine Research Laboratory, Research Center of Clinical Medicine, Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Medical School of Nantong University, Nantong, China
| | - Qian Qian Yang
- Research for Frontier Medicine and Hand Surgery Research Center, The Nanomedicine Research Laboratory, Research Center of Clinical Medicine, Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Medical School of Nantong University, Nantong, China
| | - You Lang Zhou
- Research for Frontier Medicine and Hand Surgery Research Center, The Nanomedicine Research Laboratory, Research Center of Clinical Medicine, Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Medical School of Nantong University, Nantong, China
| |
Collapse
|
9
|
Morscheid YP, Venkatesan JK, Schmitt G, Orth P, Zurakowski D, Speicher-Mentges S, Menger MD, Laschke MW, Cucchiarini M, Madry H. rAAV-Mediated Human FGF-2 Gene Therapy Enhances Osteochondral Repair in a Clinically Relevant Large Animal Model Over Time In Vivo. Am J Sports Med 2021; 49:958-969. [PMID: 33606561 DOI: 10.1177/0363546521988941] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Osteochondral defects, if left untreated, do not heal and can potentially progress toward osteoarthritis. Direct gene transfer of basic fibroblast growth factor 2 (FGF-2) with the clinically adapted recombinant adeno-associated viral (rAAV) vectors is a powerful tool to durably activate osteochondral repair processes. PURPOSE To examine the ability of an rAAV-FGF-2 construct to target the healing processes of focal osteochondral injury over time in a large translational model in vivo versus a control gene transfer condition. STUDY DESIGN Controlled laboratory study. METHODS Standardized osteochondral defects created in the knee joints of adult sheep were treated with an rAAV human FGF-2 (hFGF-2) vector by direct administration into the defect relative to control (reporter) rAAV-lacZ gene transfer. Osteochondral repair was monitored using macroscopic, histological, immunohistological, and biochemical methods and by micro-computed tomography after 6 months. RESULTS Effective, localized prolonged FGF-2 overexpression was achieved for 6 months in vivo relative to the control condition without undesirable leakage of the vectors outside the defects. Such rAAV-mediated hFGF-2 overexpression significantly increased the individual histological parameter "percentage of new subchondral bone" versus lacZ treatment, reflected in a volume of mineralized bone per unit volume of the subchondral bone plate that was equal to a normal osteochondral unit. Also, rAAV-FGF-2 significantly improved the individual histological parameters "defect filling,""matrix staining," and "cellular morphology" and the overall cartilage repair score versus the lacZ treatment and led to significantly higher cell densities and significantly higher type II collagen deposition versus lacZ treatment. Likewise, rAAV-FGF-2 significantly decreased type I collagen expression within the cartilaginous repair tissue. CONCLUSION The current work shows the potential of direct rAAV-mediated FGF-2 gene therapy to enhance osteochondral repair in a large, clinically relevant animal model over time in vivo. CLINICAL RELEVANCE Delivery of therapeutic (hFGF-2) rAAV vectors in sites of focal injury may offer novel, convenient tools to enhance osteochondral repair in the near future.
Collapse
Affiliation(s)
- Yannik P Morscheid
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| | - Patrick Orth
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| | - David Zurakowski
- Department of Anesthesiology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Susanne Speicher-Mentges
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Homburg/Saar, Germany
| |
Collapse
|
10
|
Uzieliene I, Kalvaityte U, Bernotiene E, Mobasheri A. Non-viral Gene Therapy for Osteoarthritis. Front Bioeng Biotechnol 2021; 8:618399. [PMID: 33520968 PMCID: PMC7838585 DOI: 10.3389/fbioe.2020.618399] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/11/2020] [Indexed: 12/16/2022] Open
Abstract
Strategies for delivering nucleic acids into damaged and diseased tissues have been divided into two major areas: viral and non-viral gene therapy. In this mini-review article we discuss the application of gene therapy for the treatment of osteoarthritis (OA), one of the most common forms of arthritis. We focus primarily on non-viral gene therapy and cell therapy. We briefly discuss the advantages and disadvantages of viral and non-viral gene therapy and review the nucleic acid transfer systems that have been used for gene delivery into articular chondrocytes in cartilage from the synovial joint. Although viral gene delivery has been more popular due to its reported efficiency, significant effort has gone into enhancing the transfection efficiency of non-viral delivery, making non-viral approaches promising tools for further application in basic, translational and clinical studies on OA. Non-viral gene delivery technologies have the potential to transform the future development of disease-modifying therapeutics for OA and related osteoarticular disorders. However, further research is needed to optimize transfection efficiency, longevity and duration of gene expression.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Ursule Kalvaityte
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Centre for Sport, Exercise and Osteoarthritis Versus Arthritis, Queen's Medical Centre, Nottingham, United Kingdom
| |
Collapse
|
11
|
Schreiner AJ, Stoker AM, Bozynski CC, Kuroki K, Stannard JP, Cook JL. Clinical Application of the Basic Science of Articular Cartilage Pathology and Treatment. J Knee Surg 2020; 33:1056-1068. [PMID: 32583400 DOI: 10.1055/s-0040-1712944] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The joint is an organ with each tissue playing critical roles in health and disease. Intact articular cartilage is an exquisite tissue that withstands incredible biologic and biomechanical demands in allowing movement and function, which is why hyaline cartilage must be maintained within a very narrow range of biochemical composition and morphologic architecture to meet demands while maintaining health and integrity. Unfortunately, insult, injury, and/or aging can initiate a cascade of events that result in erosion, degradation, and loss of articular cartilage such that joint pain and dysfunction ensue. Importantly, articular cartilage pathology affects the health of the entire joint and therefore should not be considered or addressed in isolation. Treating articular cartilage lesions is challenging because left alone, the tissue is incapable of regeneration or highly functional and durable repair. Nonoperative treatments can alleviate symptoms associated with cartilage pathology but are not curative or lasting. Current surgical treatments range from stimulation of intrinsic repair to whole-surface and whole-joint restoration. Unfortunately, there is a relative paucity of prospective, randomized controlled, or well-designed cohort-based clinical trials with respect to cartilage repair and restoration surgeries, such that there is a gap in knowledge that must be addressed to determine optimal treatment strategies for this ubiquitous problem in orthopedic health care. This review article discusses the basic science rationale and principles that influence pathology, symptoms, treatment algorithms, and outcomes associated with articular cartilage defects in the knee.
Collapse
Affiliation(s)
- Anna J Schreiner
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri.,BG Center for Trauma and Reconstructive Surgery, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Aaron M Stoker
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| | - Chantelle C Bozynski
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| | - Keiichi Kuroki
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri
| | - James P Stannard
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| | - James L Cook
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri.,Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri
| |
Collapse
|
12
|
Mobasheri A, Choi H, Martín-Vasallo P. Over-Production of Therapeutic Growth Factors for Articular Cartilage Regeneration by Protein Production Platforms and Protein Packaging Cell Lines. BIOLOGY 2020; 9:biology9100330. [PMID: 33050357 PMCID: PMC7599991 DOI: 10.3390/biology9100330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/28/2022]
Abstract
Simple Summary Osteoarthritis (OA) is the most common form of arthritis across the world. Most of the existing drugs for OA treat the symptoms of pain and inflammation. There are no drugs that can dure the disease. There are a number of new treatments for OA including cell therapy and gene therapy. This articles outlines the concept behind TissueGene-C, a new biological drug for OA. This new treatment includes cartilage cells mixed with a genetically modified cell line called GP2-293, which is effectively a “drug factory”, over-producing the growth factors that are important for cartilage regeneration and changing the environment inside joints. The mixture is injected into the affected knee joint. These cells are designed to be short-lived and cannot reproduce. Therefore, after they have done their job, they die and are cleared by immune cells. This is a new and modern approach to treating OA and TissueGene-C is the prototype cell therapy for OA. In the future, it is entirely possible to combine different clones of genetically engineered cells like GP2-293 that have been designed to over-produce a growth factor or biological drug with cells from the cartilage endplate of the intervertebral disc to treat degeneration in the spine. Abstract This review article focuses on the current state-of-the-art cellular and molecular biotechnology for the over-production of clinically relevant therapeutic and anabolic growth factors. We discuss how the currently available tools and emerging technologies can be used for the regenerative treatment of osteoarthritis (OA). Transfected protein packaging cell lines such as GP-293 cells may be used as “cellular factories” for large-scale production of therapeutic proteins and pro-anabolic growth factors, particularly in the context of cartilage regeneration. However, when irradiated with gamma or x-rays, these cells lose their capacity for replication, which makes them safe for use as a live cell component of intra-articular injections. This innovation is already here, in the form of TissueGene-C, a new biological drug that consists of normal allogeneic primary chondrocytes combined with transduced GP2-293 cells that overexpress the growth factor transforming growth factor β1 (TGF-β1). TissueGene-C has revolutionized the concept of cell therapy, allowing drug companies to develop live cells as biological drug delivery systems for direct intra-articular injection of growth factors whose half-lives are in the order of minutes. Therefore, in this paper, we discuss the potential for new innovations in regenerative medicine for degenerative diseases of synovial joints using mammalian protein production platforms, specifically protein packaging cell lines, for over-producing growth factors for cartilage tissue regeneration and give recent examples. Mammalian protein production platforms that incorporate protein packaging eukaryotic cell lines are superior to prokaryotic bacterial expression systems and are likely to have a significant impact on the development of new humanized biological growth factor therapies for treating focal cartilage defects and more generally for the treatment of degenerative joint diseases such as OA, especially when injected directly into the joint.
Collapse
Affiliation(s)
- Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI-90014 Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Versus Arthritis Centre for Sport, Exercise and Osteoarthritis Research, Queen’s Medical Centre, Nottingham NG7 2UH, UK
- Correspondence: or
| | - Heonsik Choi
- Kolon TissueGene, Inc., Rockville, MD 20850, USA;
- Healthcare Research Institute, Kolon Advanced Research Center, Kolon Industries, Inc., Magok-dong, Gangseo-gu, Seoul 07793, Korea
| | - Pablo Martín-Vasallo
- UD of Biochemistry and Molecular Biology, Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, San Cristóbal de La Laguna, 38071 Tenerife, Spain;
| |
Collapse
|
13
|
Weissenberger M, Weissenberger MH, Gilbert F, Groll J, Evans CH, Steinert AF. Reduced hypertrophy in vitro after chondrogenic differentiation of adult human mesenchymal stem cells following adenoviral SOX9 gene delivery. BMC Musculoskelet Disord 2020; 21:109. [PMID: 32066427 PMCID: PMC7026978 DOI: 10.1186/s12891-020-3137-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/12/2020] [Indexed: 01/03/2023] Open
Abstract
Background Mesenchymal stem cell (MSC) based-treatments of cartilage injury are promising but impaired by high levels of hypertrophy after chondrogenic induction with several bone morphogenetic protein superfamily members (BMPs). As an alternative, this study investigates the chondrogenic induction of MSCs via adenoviral gene-delivery of the transcription factor SOX9 alone or in combination with other inducers, and comparatively explores the levels of hypertrophy and end stage differentiation in a pellet culture system in vitro. Methods First generation adenoviral vectors encoding SOX9, TGFB1 or IGF1 were used alone or in combination to transduce human bone marrow-derived MSCs at 5 × 102 infectious particles/cell. Thereafter cells were placed in aggregates and maintained for three weeks in chondrogenic medium. Transgene expression was determined at the protein level (ELISA/Western blot), and aggregates were analysed histologically, immunohistochemically, biochemically and by RT-PCR for chondrogenesis and hypertrophy. Results SOX9 cDNA was superior to that encoding TGFB1, the typical gold standard, as an inducer of chondrogenesis in primary MSCs as evidenced by improved lacuna formation, proteoglycan and collagen type II staining, increased levels of GAG synthesis, and expression of mRNAs associated with chondrogenesis. Moreover, SOX9 modified aggregates showed a markedly lower tendency to progress towards hypertrophy, as judged by expression of the hypertrophy markers alkaline phosphatase, and collagen type X at the mRNA and protein levels. Conclusion Adenoviral SOX9 gene transfer induces chondrogenic differentiation of human primary MSCs in pellet culture more effectively than TGFB1 gene transfer with lower levels of chondrocyte hypertrophy after 3 weeks of in vitro culture. Such technology might enable the formation of more stable hyaline cartilage repair tissues in vivo.
Collapse
Affiliation(s)
- M Weissenberger
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Center for Musculoskeletal Research, Julius-Maximilians-University, Brettreichstrasse 11, D-97074, Würzburg, Germany.
| | - M H Weissenberger
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Center for Musculoskeletal Research, Julius-Maximilians-University, Brettreichstrasse 11, D-97074, Würzburg, Germany.,Department of Pathology, Caritas-Hospital, Bad Mergentheim, Germany
| | - F Gilbert
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Center for Musculoskeletal Research, Julius-Maximilians-University, Brettreichstrasse 11, D-97074, Würzburg, Germany.,Department of Orthopaedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, Würzburg, Germany
| | - J Groll
- Department of Functional Materials in Medicine and Dentistry, Julius-Maximilians-University, Würzburg, Germany
| | - C H Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN, USA
| | - A F Steinert
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Center for Musculoskeletal Research, Julius-Maximilians-University, Brettreichstrasse 11, D-97074, Würzburg, Germany.,Present address: Department of Orthopaedic, Trauma, Shoulder and Arthroplasty Surgery, Rhön-Klinikum Campus Bad Neustadt, Bad Neustadt a.d. Saale, Germany
| |
Collapse
|
14
|
Pferdehirt L, Ross AK, Brunger JM, Guilak F. A Synthetic Gene Circuit for Self-Regulating Delivery of Biologic Drugs in Engineered Tissues. Tissue Eng Part A 2019; 25:809-820. [PMID: 30968743 DOI: 10.1089/ten.tea.2019.0027] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
IMPACT STATEMENT We engineered a synthetic transcription system based on nuclear factor kappa-light-chain-enhancer of activated B cells signaling that can attenuate the effects of the inflammatory cytokine interleukin (IL)-1α in a self-regulating manner. This system responds in a time- and dose-dependent manner to rapidly produce therapeutic levels of IL-1 receptor antagonist (IL-1Ra). The use of lentiviral gene therapy allows this system to be utilized through different transduction methods and in different cell types for a variety of applications. Broadly, this approach may be applicable in developing autoregulated biologic systems for tissue engineering and drug delivery in a range of disease applications.
Collapse
Affiliation(s)
- Lara Pferdehirt
- 1 Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis, St. Louis, Missouri.,3 Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri.,4 Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Alison K Ross
- 1 Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis, St. Louis, Missouri.,3 Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri.,4 Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Jonathan M Brunger
- 5 Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | - Farshid Guilak
- 1 Department of Orthopedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis, St. Louis, Missouri.,3 Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri.,4 Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| |
Collapse
|
15
|
Kim SE, Kim JG, Park K. Biomaterials for the Treatment of Tendon Injury. Tissue Eng Regen Med 2019; 16:467-477. [PMID: 31624702 DOI: 10.1007/s13770-019-00217-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/26/2022] Open
Abstract
Background Most tendon injuries are occurring from a gradual wearing and tearing of the tendon tissues from overuse. Such injuries are usually seen in sports, exercising, or daily activities that involve a high mechanical load and weight bearing. However, owing to the lack of both cellularity and blood vessels in tendons, the process of tendon repair is slow and inefficient. Although various conservative (non-surgical) and surgical management options are conducted by the clinicians, a gold standard of these approaches does not exist. In this regard, the treatment of tendon injuries is challenging. Method Here, we describe the recent advances of biomaterial-based approaches for the treatment of injured tendons. Results Regenerative medicine is an emerging multidisciplinary research that specializes in the repair of damaged tendon tissues through the delivery of regenerative factors by biomaterials. Conclusion Although current biomaterial-based treatment strategies have shown their potential for tendon healing, future research and clinical applications should focused on finding the optimum combinations of regenerative factors with ideal biomaterials for the repair of tendons.
Collapse
Affiliation(s)
- Sung Eun Kim
- 1Department of Orthopedic Surgery and Rare Diseases Institute, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308 Republic of Korea
| | - Jae Gyoon Kim
- Department of Orthopedic Surgery, College of Medicine, Korea University Ansan Hospital, Korea University, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do 15355 Republic of Korea
| | - Kyeongsoon Park
- 3Department of Systems Biotechnology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726 Seodong-daero, Daedeok-myeon, Anseong-si, Gyeonggi-do 17546 Republic of Korea
| |
Collapse
|
16
|
Mesure B, Menu P, Venkatesan JK, Cucchiarini M, Velot É. Biomaterials and Gene Therapy: A Smart Combination for MSC Musculoskeletal Engineering. Curr Stem Cell Res Ther 2019; 14:337-343. [DOI: 10.2174/1574888x14666181205121658] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 11/22/2022]
Abstract
Musculoskeletal pathologies, especially those affecting bones and joints, remain a challenge
for regenerative medicine. The main difficulties affecting bone tissue engineering are the size of the
defects, the need for blood vessels and the synthesis of appropriate matrix elements in the engineered
tissue. Indeed, the cartilage is an avascular tissue and consequently has limited regenerative abilities.
Thanks to their self-renewal, plasticity and immunomodulatory properties, mesenchymal stem cells
(MSCs) became a central player in tissue engineering, and have already been shown to be able to differentiate
towards chondrogenic or osteogenic phenotypes. Whether synthetic (e.g. tricalcium phosphate)
or from natural sources (e.g. hyaluronic acid), biomaterials can be shaped to fit into bone and
cartilage defects to ensure mechanical resistance and may also be designed to control cell spatial distribution
or differentiation. Soluble factors are classically used to promote cell differentiation and to
stimulate extracellular matrix synthesis to achieve the desired tissue production. But as they have a
limited lifetime, transfection using plasmid DNA or transduction via a viral vector of therapeutic genes
to induce the cell secretion of these factors allows to have more lasting effects. Also, the chondrocyte
phenotype may be difficult to control over time, with for example the production of hypertrophic or
osteogenic markers that is undesirable in hyaline cartilage. Thus, tissue regeneration strategies became
more elaborate, with an attempt at associating the benefits of MSCs, biomaterials, and gene therapy to
achieve a proper tissue repair. This minireview focuses on in vitro and in vivo studies combining biomaterials
and gene therapy associated with MSCs for bone and cartilage engineering.
Collapse
Affiliation(s)
- Benjamin Mesure
- UMR 7365 CNRS-UL IMoPA, Universite de Lorraine, Nancy, France
| | - Patrick Menu
- UMR 7365 CNRS-UL IMoPA, Universite de Lorraine, Nancy, France
| | | | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Émilie Velot
- UMR 7365 CNRS-UL IMoPA, Universite de Lorraine, Nancy, France
| |
Collapse
|
17
|
Żylińska B, Silmanowicz P, Sobczyńska-Rak A, Jarosz Ł, Szponder T. Treatment of Articular Cartilage Defects: Focus on Tissue Engineering. In Vivo 2019; 32:1289-1300. [PMID: 30348681 DOI: 10.21873/invivo.11379] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022]
Abstract
The treatment of articular cartilage defects seems to be one of the greatest challenges in modern orthopaedics. From a medical point of view there are 3 main goals to achieve for cartilage trauma treatment: restoration of the joints motion, pain relief and elimination/delay of the onset of osteoarthritis. Treatment can be divided into conservative (including pharmacotherapy, arthrocentesis and physiotherapy) and surgical. The last comprises reparative techniques, regenerative methods and symptomatic treatment. While both are focused on reconstruction of the damaged cartilage, the difference lies in the type of filling tissue. Reparative techniques include: drilling of the subchondral bone, spongiolisation, abrasion, mictrofracture, and autologous matrix induced chondrogenesis (AMIC). Regenerative methods contain: periosteal and perichondral grafts, mosaicplasty, autologous chondrocyte implantation and matrix-induced autologous chondrocyte implantation (MACI). Nowadays tissue engineering, including gene therapy, is emerging as one of the key approaches to cartilage treatment and holds promises for new achievements and better outcomes of many cartilage diseases and traumas.
Collapse
Affiliation(s)
- Beata Żylińska
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Piotr Silmanowicz
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Aleksandra Sobczyńska-Rak
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Łukasz Jarosz
- Department of Epizootiology and Clinic of Infectious Diseases, University of Life Sciences in Lublin, Lublin, Poland
| | - Tomasz Szponder
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| |
Collapse
|
18
|
Current trends in tendinopathy: consensus of the ESSKA basic science committee. Part II: treatment options. J Exp Orthop 2018; 5:38. [PMID: 30251203 PMCID: PMC6153202 DOI: 10.1186/s40634-018-0145-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/26/2018] [Indexed: 01/01/2023] Open
Abstract
The treatment of painful chronic tendinopathy is challenging. Multiple non-invasive and tendon-invasive methods are used. When traditional non-invasive treatments fail, the injections of platelet-rich plasma autologous blood or cortisone have become increasingly favored. However, there is little scientific evidence from human studies supporting injection treatment. As the last resort, intra- or peritendinous open or endoscopic surgery are employed even though these also show varying results. This ESSKA basic science committee current concepts review follows the first part on the biology, biomechanics and anatomy of tendinopathies, to provide a comprehensive overview of the latest treatment options for tendinopathy as reported in the literature.
Collapse
|
19
|
Emerging Concepts in Treating Cartilage, Osteochondral Defects, and Osteoarthritis of the Knee and Ankle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:25-62. [PMID: 29736568 DOI: 10.1007/978-3-319-76735-2_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The management and treatment of cartilage lesions, osteochondral defects, and osteoarthritis remain a challenge in orthopedics. Moreover, these entities have different behaviors in different joints, such as the knee and the ankle, which have inherent differences in function, biology, and biomechanics. There has been a huge development on the conservative treatment (new technologies including orthobiologics) as well as on the surgical approach. Some surgical development upraises from technical improvements including advanced arthroscopic techniques but also from increased knowledge arriving from basic science research and tissue engineering and regenerative medicine approaches. This work addresses the state of the art concerning basic science comparing the knee and ankle as well as current options for treatment. Furthermore, the most promising research developments promising new options for the future are discussed.
Collapse
|
20
|
Lin S, Lee WYW, Xu L, Wang Y, Chen Y, Ho KKW, Qin L, Jiang X, Cui L, Li G. Stepwise preconditioning enhances mesenchymal stem cell-based cartilage regeneration through epigenetic modification. Osteoarthritis Cartilage 2017; 25:1541-1550. [PMID: 28545880 DOI: 10.1016/j.joca.2017.05.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 05/07/2017] [Accepted: 05/09/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study is to investigate the functions and underlying mechanisms of mesenchymal stem cells (MSCs) underwent stepwise preconditioning in chondrogenic medium before expansion, then further explore their therapeutic effects in a surgically induced osteoarthritis (OA) model. METHODS MSCs isolated from the adult rats expressing Green Fluorescence Protein (GFP) were incubated in basal medium or primed in chondrogenic medium before expansion. The multipotency including cell proliferation, differentiation, and survivability was compared between chondrogenic manipulated MSCs (M-MSCs) and untreated MSCs. Methylation modification of Nanog and Oct4 were detected by bisulfite genomic sequencing. Loss-of-function phenotype in M-MSCs induced by shNanog was also observed. Then the therapeutic effect of the cells was evaluated in a surgically induced OA rat model by single intraarticular injection. The injected GFP-labeled cells in the joints were monitored in vivo. These rats were sacrificed and subjected to histological examinations and microstructural analysis after 4 weeks. RESULTS We found that cell clonogenicity, proliferation, survivability, and chondrogenic property were enhanced after stepwise preconditioning. We then further found that the expression level of Nanog and Oct4 was temporarily increased in the M-MSCs. Results of epigenetic analysis revealed that demethylation happened in Nanog and Oct4 after the stepwise preconditioning. Results of in vivo imaging showed more GFP-labeled cells in the M-MSCs-injected group. And results of histology and micro-CT analysis also indicated a superior therapeutic effect of M-MSCs on the surgically induced-OA. CONCLUSION These findings indicated a feasible method to obtain a cell population with high survivability and chondrogenic commitment for the treatment of OA.
Collapse
Affiliation(s)
- S Lin
- Department of Orthopaedics & Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China; Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, China; The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - W Y W Lee
- Department of Orthopaedics & Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China; The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - L Xu
- Department of Orthopaedics & Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China; The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Y Wang
- Department of Orthopaedics & Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Y Chen
- Department of Orthopaedics & Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - K K W Ho
- Department of Orthopaedics & Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - L Qin
- Department of Orthopaedics & Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China; The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - X Jiang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - L Cui
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - G Li
- Department of Orthopaedics & Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China; The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China; Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
21
|
Armbruster N, Krieg J, Weißenberger M, Scheller C, Steinert AF. Rescued Chondrogenesis of Mesenchymal Stem Cells under Interleukin 1 Challenge by Foamyviral Interleukin 1 Receptor Antagonist Gene Transfer. Front Pharmacol 2017; 8:255. [PMID: 28536528 PMCID: PMC5422547 DOI: 10.3389/fphar.2017.00255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/24/2017] [Indexed: 12/15/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) and their chondrogenic differentiation have been extensively investigated in vitro as MSCs provide an attractive source besides chondrocytes for cartilage repair therapies. Here we established prototype foamyviral vectors (FVV) that are derived from apathogenic parent viruses and are characterized by a broad host range and a favorable integration pattern into the cellular genome. As the inflammatory cytokine interleukin 1 beta (IL1β) is frequently present in diseased joints, the protective effects of FVV expressing the human interleukin 1 receptor antagonist protein (IL1RA) were studied in an established in vitro model (aggregate culture system) of chondrogenesis in the presence of IL1β. Materials and Methods: We generated different recombinant FVVs encoding enhanced green fluorescent protein (EGFP) or IL1RA and examined their transduction efficiencies and transgene expression profiles using different cell lines and human primary MSCs derived from bone marrow-aspirates. Transgene expression was evaluated by fluorescence microscopy (EGFP), flow cytometry (EGFP), and ELISA (IL1RA). For evaluation of the functionality of the IL1RA transgene to block the inhibitory effects of IL1β on chondrogenesis of primary MSCs and an immortalized MSC cell line (TERT4 cells), the cells were maintained following transduction as aggregate cultures in standard chondrogenic media in the presence or absence of IL1β. After 3 weeks of culture, pellets were harvested and analyzed by histology and immunohistochemistry for chondrogenic phenotypes. Results: The different FVV efficiently transduced cell lines as well as primary MSCs, thereby reaching high transgene expression levels in 6-well plates with levels of around 100 ng/ml IL1RA. MSC aggregate cultures which were maintained in chondrogenic media without IL1β supplementation revealed a chondrogenic phenotype by means of strong positive staining for collagen type II and matrix proteoglycan (Alcian blue). Addition of IL1β was inhibitory to chondrogenesis in untreated control pellets. In contrast, foamyviral mediated IL1RA expression rescued the chondrogenesis in pellets cultured in the presence of IL1β. Transduced MSC pellets reached thereby very high IL1RA transgene expression levels with a peak of 1087 ng/ml after day 7, followed by a decrease to 194 ng/ml after day 21, while IL1RA concentrations of controls were permanently below 200 pg/ml. Conclusion: Our results indicate that FVV are capable of efficient gene transfer to MSCs, while reaching IL1RA transgene expression levels, that were able to efficiently block the impacts of IL1β in vitro. FVV merit further investigation as a means to study the potential as a gene transfer tool for MSC based therapies for cartilage repair.
Collapse
Affiliation(s)
- Nicole Armbruster
- Institute for Virology and Immunobiology, University of WuerzburgWuerzburg, Germany.,Department of Orthopaedic Surgery, Klinik König-Ludwig-Haus Würzburg - Center for Musculoskeletal Research, University of WuerzburgWuerzburg, Germany
| | - Jennifer Krieg
- Institute for Virology and Immunobiology, University of WuerzburgWuerzburg, Germany.,Department of Orthopaedic Surgery, Klinik König-Ludwig-Haus Würzburg - Center for Musculoskeletal Research, University of WuerzburgWuerzburg, Germany
| | - Manuel Weißenberger
- Department of Orthopaedic Surgery, Klinik König-Ludwig-Haus Würzburg - Center for Musculoskeletal Research, University of WuerzburgWuerzburg, Germany
| | - Carsten Scheller
- Institute for Virology and Immunobiology, University of WuerzburgWuerzburg, Germany
| | - Andre F Steinert
- Department of Orthopaedic Surgery, Klinik König-Ludwig-Haus Würzburg - Center for Musculoskeletal Research, University of WuerzburgWuerzburg, Germany
| |
Collapse
|
22
|
Wang Y, Yang T, Liu Y, Zhao W, Zhang Z, Lu M, Zhang W. Decrease of miR-195 Promotes Chondrocytes Proliferation and Maintenance of Chondrogenic Phenotype via Targeting FGF-18 Pathway. Int J Mol Sci 2017; 18:ijms18050975. [PMID: 28471382 PMCID: PMC5454888 DOI: 10.3390/ijms18050975] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023] Open
Abstract
Slow growth and rapid loss of chondrogenic phenotypes are the major problems affecting chronic cartilage lesions. The role of microRNA-195 (miR-195) and its detailed working mechanism in the fore-mentioned process remains unknown. Fibroblastic growth factor 18 (FGF-18) plays a key role in cartilage homeostasis; whether miR-195 could regulate FGF-18 and its downstream signal pathway in chondrocyte proliferation and maintenance of chondrogenic phenotypes still remains unclear. The present research shows elevated miR-195 but depressed FGF-18 expressed in joint fluid specimens of 20 patients with chronic cartilage lesions and in CH1M and CH3M chondrocytes when compared with that in joint fluid specimens without cartilage lesions and in CH1W and CH2W chondrocytes, respectively. The following loss of function test revealed that downregulation of miR-195 by transfection of miR-195 inhibitors promoted chondrocyte proliferation and expression of a type II collagen α I chain (Col2a1)/aggrecan. Through the online informatics analysis we theoretically predicted that miR-195 could bind to a FGF-18 3' untranslated region (3'UTR), also, we verified that a miR-195 could regulate the FGF-18 and its downstream pathway. The constructed dual luciferase assay further confirmed that FGF-18 was a direct target of miR-195. The executed anti-sense experiment displayed that miR-195 could regulate chondrocyte proliferation and Col2a1/aggrecan expression via the FGF-18 pathway. Finally, through an in vivo anterior cruciate ligament transection (ACLT) model, downregulation of miR-195 presented a significantly protective effect on chronic cartilage lesions. Evaluating all of the outcomes of the current research revealed that a decrease of miR-195 protected chronic cartilage lesions by promoting chondrocyte proliferation and maintenance of chondrogenic phenotypes via the targeting of the FGF-18 pathway and that the miR-195/FGF-18 axis could be a potential target in the treatment of cartilage lesions.
Collapse
Affiliation(s)
- Yong Wang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
- The 4th Department of Orthopedic Surgery, The Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China.
| | - Tao Yang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Yadong Liu
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Wei Zhao
- The 4th Department of Orthopedic Surgery, The Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China.
| | - Zhen Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Ming Lu
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Weiguo Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
23
|
Hydrogel-Based Controlled Delivery Systems for Articular Cartilage Repair. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1215263. [PMID: 27642587 PMCID: PMC5011507 DOI: 10.1155/2016/1215263] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/12/2016] [Indexed: 12/19/2022]
Abstract
Delivery of bioactive factors is a very valuable strategy for articular cartilage repair. Nevertheless, the direct supply of such biomolecules is limited by several factors including rapid degradation, the need for supraphysiological doses, the occurrence of immune and inflammatory responses, and the possibility of dissemination to nontarget sites that may impair their therapeutic action and raise undesired effects. The use of controlled delivery systems has the potential of overcoming these hurdles by promoting the temporal and spatial presentation of such factors in a defined target. Hydrogels are promising materials to develop delivery systems for cartilage repair as they can be easily loaded with bioactive molecules controlling their release only where required. This review exposes the most recent technologies on the design of hydrogels as controlled delivery platforms of bioactive molecules for cartilage repair.
Collapse
|
24
|
Knorpeldefekte. ARTHROSKOPIE 2016. [DOI: 10.1007/s00142-016-0066-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
25
|
Reissis D, Tang QO, Cooper NC, Carasco CF, Gamie Z, Mantalaris A, Tsiridis E. Current clinical evidence for the use of mesenchymal stem cells in articular cartilage repair. Expert Opin Biol Ther 2016; 16:535-57. [PMID: 26798997 DOI: 10.1517/14712598.2016.1145651] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Articular cartilage is renowned for its poor intrinsic capacity for repair. Current treatments for osteoarthritis are limited in their ability to reliably restore the native articular cartilage structure and function. Mesenchymal stem cells (MSCs) present an attractive treatment option for articular cartilage repair, with a recent expansion of clinical trials investigating their use in patients. AREAS COVERED This paper provides a current overview of the clinical evidence on the use of MSCs in articular cartilage repair. EXPERT OPINION The article demonstrates robust clinical evidence that MSCs have significant potential for the regeneration of hyaline articular cartilage in patients. The majority of clinical trials to date have yielded significantly positive results with minimal adverse effects. However the clinical research is still in its infancy. The optimum MSC source, cell concentrations, implantation technique, scaffold, growth factors and rehabilitation protocol for clinical use are yet to be identified. A larger number of randomised control trials are required to objectively compare the clinical efficacy and long-term safety of the various techniques. As the clinical research continues to evolve and address these challenges, it is likely that MSCs may become integrated into routine clinical practice in the near future.
Collapse
Affiliation(s)
- Dimitris Reissis
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Quen Oak Tang
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Nina Catherine Cooper
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Clare Francesca Carasco
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Zakareya Gamie
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Athanasios Mantalaris
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Eleftherios Tsiridis
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK.,b Academic Orthopaedic Unit , Aristotle University Medical School , Thessaloniki , Greece
| |
Collapse
|
26
|
Madry H, Cucchiarini M. Gene therapy for human osteoarthritis: principles and clinical translation. Expert Opin Biol Ther 2015; 16:331-46. [PMID: 26593049 DOI: 10.1517/14712598.2016.1124084] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Osteoarthritis (OA) is the most prevalent chronic joint disease. Its key feature is a progressive articular cartilage loss. Gene therapy for OA aims at delivering gene-based therapeutic agents to the osteoarthritic cartilage, resulting in a controlled, site-specific, long-term presence to rebuild the damaged cartilage. AREAS COVERED An overview is provided of the principles of gene therapy for OA based on a PubMed literature search. Gene transfer to normal and osteoarthritic cartilage in vitro and in animal models in vivo is reviewed. Results from recent clinical gene therapy trials for OA are discussed and placed into perspective. EXPERT OPINION Recombinant adeno-associated viral (rAAV) vectors enable to directly transfer candidate sequences in human articular chondrocytes in situ, providing a potent tool to modulate the structure of osteoarthritic cartilage. However, few preclinical animal studies in OA models have been performed thus far. Noteworthy, several gene therapy clinical trials have been carried out in patients with end-stage knee OA based on the intraarticular injection of human juvenile allogeneic chondrocytes overexpressing a cDNA encoding transforming growth factor-beta-1 via retroviral vectors. In a recent placebo-controlled randomized trial, clinical scores were improved compared with placebo. These translational results provide sufficient reason to proceed with further clinical testing of gene transfer protocols for the treatment of OA.
Collapse
Affiliation(s)
- Henning Madry
- a Center of Experimental Orthopaedics , Saarland University , Homburg/Saar , Germany
| | - Magali Cucchiarini
- a Center of Experimental Orthopaedics , Saarland University , Homburg/Saar , Germany
| |
Collapse
|
27
|
Aguilar IN, Trippel SB, Shi S, Bonassar LJ. Comparison of Efficacy of Endogenous and Exogenous IGF-I in Stimulating Matrix Production in Neonatal and Mature Chondrocytes. Cartilage 2015; 6:264-72. [PMID: 26425264 PMCID: PMC4568729 DOI: 10.1177/1947603515578691] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [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 goal of this study was to compare the efficacy of endogenous upregulation of IGF-I by gene therapy and exogenous addition of insulin-like growth factor I (IGF-I) in enhancing proteoglycan synthesis by skeletally mature and neonatal chondrocytes. Chondrocyte transplantation therapy is a common treatment for focal cartilage lesions, with both mature and neonatal chondrocytes used as a cell source. Additionally, gene therapy strategies to upregulate growth factors such as IGF-I have been proposed to augment chondrocyte transplantation therapies. METHODS Both skeletally mature and neonatal chondrocytes were exposed to either an adeno-associated virus-based plasmid containing the IGF-I gene or exogenous IGF-I. RESULTS Analysis of IGF-I and glycosaminoglycan production using a 4-parameter dose-response model established a clear connection between the amount of IGF-I produced by cells and their biosynthetic response. Both neonatal and mature chondrocytes showed this relationship, but the sensitivities were quite different, with EC50 of 0.57 ng/mL for neonatal chondrocytes and EC50 of 8.70 ng/mL IGF-I for skeletally mature chondrocytes. CONCLUSIONS These data suggest that IGF-I gene therapy may be more effective with younger cell sources. Both cell types were less sensitive to exogenous IGF-I than endogenous IGF-I.
Collapse
Affiliation(s)
- Izath N. Aguilar
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Stephen B. Trippel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shuiliang Shi
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lawrence J. Bonassar
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
28
|
Madry H, Cucchiarini M. Tissue-engineering strategies to repair joint tissue in osteoarthritis: nonviral gene-transfer approaches. Curr Rheumatol Rep 2015; 16:450. [PMID: 25182678 DOI: 10.1007/s11926-014-0450-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Loss of articular cartilage is a common clinical consequence of osteoarthritis (OA). In the past decade, substantial progress in tissue engineering, nonviral gene transfer, and cell transplantation have provided the scientific foundation for generating cartilaginous constructs from genetically modified cells. Combining tissue engineering with overexpression of therapeutic genes enables immediate filling of a cartilage defect with an engineered construct that actively supports chondrogenesis. Several pioneering studies have proved that spatially defined nonviral overexpression of growth-factor genes in constructs of solid biomaterials or hydrogels is advantageous compared with gene transfer or scaffold alone, both in vitro and in vivo. Notably, these investigations were performed in models of focal cartilage defects, because advanced cartilage-repair strategies based on the principles of tissue engineering have not advanced sufficiently to enable resurfacing of extensively degraded cartilage as therapy for OA. These studies serve as prototypes for future technological developments, because they raise the possibility that cartilage constructs engineered from genetically modified chondrocytes providing autocrine and paracrine stimuli could similarly compensate for the loss of articular cartilage in OA. Because cartilage-tissue-engineering strategies are already used in the clinic, combining tissue engineering and nonviral gene transfer could prove a powerful approach to treat OA.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics and Department of Orthopaedic Surgery, Saarland University, 66421, Homburg, Germany,
| | | |
Collapse
|
29
|
Therapeutic applications of nanomedicine in autoimmune diseases: From immunosuppression to tolerance induction. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1003-18. [DOI: 10.1016/j.nano.2014.12.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/17/2014] [Accepted: 12/03/2014] [Indexed: 01/13/2023]
|
30
|
Cucchiarini M, Henrionnet C, Mainard D, Pinzano A, Madry H. New trends in articular cartilage repair. J Exp Orthop 2015; 2:8. [PMID: 26914876 PMCID: PMC4544617 DOI: 10.1186/s40634-015-0026-0] [Citation(s) in RCA: 11] [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] [Received: 12/19/2014] [Accepted: 03/25/2015] [Indexed: 12/31/2022] Open
Abstract
Damage to the articular cartilage is an important, prevalent, and unsolved clinical issue for the orthopaedic surgeon. This review summarizes innovative basic research approaches that may improve the current understanding of cartilage repair processes and lead to novel therapeutic options. In this regard, new aspects of cartilage tissue engineering with a focus on the choice of the best-suited cell source are presented. The importance of non-destructive cartilage imaging is highlighted with the recent availability of adapted experimental tools such as Second Harmonic Generation (SHG) imaging. Novel insights into cartilage pathophysiology based on the involvement of the infrapatellar fat pad in osteoarthritis are also described. Also, recombinant adeno-associated viral vectors are discussed as clinically adapted, efficient tools for potential gene-based medicines in a variety of articular cartilage disorders. Taken as a whole, such advances in basic research in diverse fields of articular cartilage repair may lead to the development of improved therapies in the clinics for an improved, effective treatment of cartilage lesions in a close future.
Collapse
Affiliation(s)
- Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Bldg 37, Kirrbergerstr. 1, D-66421, Homburg, Germany. .,Cartilage Net of the Greater Region, Homburg, Germany.
| | - Christel Henrionnet
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Didier Mainard
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Astrid Pinzano
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Bldg 37, Kirrbergerstr. 1, D-66421, Homburg, Germany. .,Cartilage Net of the Greater Region, Homburg, Germany. .,Department of Orthopaedic Surgery, Saarland University Medical Center and Saarland University, D-66421, Homburg/Saar, Germany.
| |
Collapse
|
31
|
Nonviral gene transfer to human meniscal cells. Part I: transfection analyses and cell transplantation to meniscus explants. INTERNATIONAL ORTHOPAEDICS 2014; 38:1923-30. [PMID: 24962292 DOI: 10.1007/s00264-014-2410-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 12/30/2022]
Abstract
PURPOSE Our aim was to evaluate whether nonviral vectors can genetically modify primary human juvenile and adult meniscal fibrochondrocytes at low toxicity in vitro and to test the hypothesis that transfected human meniscal fibrochondrocytes transplanted into longitudinal defects and onto human medial meniscus explant cultures are capable of expressing transgene products in vitro. METHODS Eighteen nonviral gene transfer systems were examined to identify the best suited method for an efficient transfection of primary cultures of juvenile and adult human meniscal fibrochondrocytes using luciferase and lacZ reporter gene constructs and then transplanted to meniscus explant cultures. RESULTS Gene transfer systems FuGENE 6, GeneJammer, TurboFectin 8, calcium phosphate co-precipitates and GeneJuice led to minimal toxicity in both cell types. Nanofectin 2 and JetPEI resulted in maximal luciferase activity in both cell types. Maximal transfection efficiency based on X-gal staining following lacZ gene transfer was achieved using Lipofectamine 2000, revealing a mean transfection efficiency of 8.6 % in human juvenile and of 8.4 % in adult meniscal fibrochondrocytes. Transfected, transplanted meniscal fibrochondrocytes adhered to the meniscal tissue and continued to express the transgene for at least five days following transfection. CONCLUSIONS Nonviral gene transfer systems are safe and capable of transfecting both juvenile and adult human meniscal fibrochondrocytes, which, when transplanted to meniscal tissue in vitro, permit the expression of selected transgenes to be maintained. These results are of value for combining gene therapy and cell transplantation approaches as a means to enhance meniscal repair.
Collapse
|
32
|
Madry H, Cucchiarini M. Advances and challenges in gene-based approaches for osteoarthritis. J Gene Med 2014; 15:343-55. [PMID: 24006099 DOI: 10.1002/jgm.2741] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/06/2013] [Accepted: 08/30/2013] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA), a paramount cause of physical disability for which there is no definitive cure, is mainly characterized by the gradual loss of the articular cartilage. Current nonsurgical and reconstructive surgical therapies have not met success in reversing the OA phenotype so far. Gene transfer approaches allow for a long-term and site-specific presence of a therapeutic agent to re-equilibrate the metabolic balance in OA cartilage and may consequently be suited to treat this slow and irreversible disorder. The distinct stages of OA need to be respected in individual gene therapy strategies. In this context, molecular therapy appears to be most effective for early OA. A critical step forward has been made by directly transferring candidate sequences into human articular chondrocytes embedded within their native extracellular matrix via recombinant adeno-associated viral vectors. Although extensive studies in vitro attest to a growing interest in this approach, data from animal models of OA are sparse. A phase I dose-escalating trial was recently performed in patients with advanced knee OA to examine the safety and activity of chondrocytes modified to produce the transforming growth factor β1 via intra-articular injection, showing a dose-dependent trend toward efficacy. Proof-of-concept studies in patients prior to undergoing total knee replacement may be privileged in the future to identify the best mode of translating this approach to clinical application, followed by randomized controlled trials.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Saarland University, Homburg, Saar, Germany
| | | |
Collapse
|
33
|
Orth P, Rey-Rico A, Venkatesan JK, Madry H, Cucchiarini M. Current perspectives in stem cell research for knee cartilage repair. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2014; 7:1-17. [PMID: 24520197 PMCID: PMC3897321 DOI: 10.2147/sccaa.s42880] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Protocols based on the delivery of stem cells are currently applied in patients, showing encouraging results for the treatment of articular cartilage lesions (focal defects, osteoarthritis). Yet, restoration of a fully functional cartilage surface (native structural organization and mechanical functions) especially in the knee joint has not been reported to date, showing the need for improved designs of clinical trials. Various sources of progenitor cells are now available, originating from adult tissues but also from embryonic or reprogrammed tissues, most of which have already been evaluated for their chondrogenic potential in culture and for their reparative properties in vivo upon implantation in relevant animal models of cartilage lesions. Nevertheless, particular attention will be needed regarding their safe clinical use and their potential to form a cartilaginous repair tissue of proper quality and functionality in the patient. Possible improvements may reside in the use of biological supplements in accordance with regulations, while some challenges remain in establishing standardized, effective procedures in the clinics.
Collapse
Affiliation(s)
- Patrick Orth
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Henning Madry
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany ; Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
34
|
Cucchiarini M, Madry H. The potential of gene transfer for the treatment of osteoarthritis. Regen Med 2014; 9:5-8. [DOI: 10.2217/rme.13.70] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg/Saar, Germany and Department of Orthopaedic Surgery, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg/Saar, Germany
| |
Collapse
|
35
|
Madry H, Kaul G, Zurakowski D, Vunjak-Novakovic G, Cucchiarini M. Cartilage constructs engineered from chondrocytes overexpressing IGF-I improve the repair of osteochondral defects in a rabbit model. Eur Cell Mater 2013; 25:229-47. [PMID: 23588785 PMCID: PMC4476264 DOI: 10.22203/ecm.v025a17] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Tissue engineering combined with gene therapy is a promising approach for promoting articular cartilage repair. Here, we tested the hypothesis that engineered cartilage with chondrocytes overexpressing a human insulin-like growth factor I (IGF-I) gene can enhance the repair of osteochondral defects, in a manner dependent on the duration of cultivation. Genetically modified chondrocytes were cultured on biodegradable polyglycolic acid scaffolds in dynamic flow rotating bioreactors for either 10 or 28 d. The resulting cartilaginous constructs were implanted into osteochondral defects in rabbit knee joints. After 28 weeks of in vivo implantation, immunoreactivity to ß-gal was detectable in the repair tissue of defects that received lacZ constructs. Engineered cartilaginous constructs based on IGF-I-overexpressing chondrocytes markedly improved osteochondral repair compared with control (lacZ) constructs. Moreover, IGF-I constructs cultivated for 28 d in vitro significantly promoted osteochondral repair vis-à-vis similar constructs cultivated for 10 d, leading to significantly decreased osteoarthritic changes in the cartilage adjacent to the defects. Hence, the combination of spatially defined overexpression of human IGF-I within a tissue-engineered construct and prolonged bioreactor cultivation resulted in most enhanced articular cartilage repair and reduction of osteoarthritic changes in the cartilage adjacent to the defect. Such genetically enhanced tissue engineering provides a versatile tool to evaluate potential therapeutic genes in vivo and to improve our comprehension of the development of the repair tissue within articular cartilage defects. Insights gained with additional exploration using this model may lead to more effective treatment options for acute cartilage defects.
Collapse
Affiliation(s)
- Henning Madry
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany,Department of Orthopaedic Surgery, Saarland University, Homburg, Germany,Address for correspondence: Henning Madry Centre of Experimental Orthopaedics Medical Faculty Building 37 Saarland University D-66421 Homburg, Germany Telephone Number: +49-6841-1624515 FAX Number: +49-6841-1624988
| | - Gunter Kaul
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - David Zurakowski
- Departments of Anaesthesia and Surgery, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Magali Cucchiarini
- Centre of Experimental Orthopaedics, Saarland University, Homburg, Germany
| |
Collapse
|
36
|
Kiss A, Cucchiarini M, Menger MD, Kohn D, Hannig M, Madry H. Enamel matrix derivative inhibits proteoglycan production and articular cartilage repair, delays the restoration of the subchondral bone and induces changes of the synovial membrane in a lapine osteochondral defect modelin vivo. J Tissue Eng Regen Med 2012; 8:41-9. [DOI: 10.1002/term.1495] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 01/09/2012] [Accepted: 01/25/2012] [Indexed: 12/25/2022]
Affiliation(s)
- Alice Kiss
- Department of Operative Dentistry and Periodontology; Saarland University Medical Centre; Homburg/Saar Germany
| | - Magali Cucchiarini
- Centre of Experimental Orthopaedics; Saarland University; Homburg/Saar Germany
| | - Michael D. Menger
- Institute für Experimental Surgery, Department of Surgery; Saarland University Medical Centre; Homburg/Saar Germany
| | - Dieter Kohn
- Department of Orthopaedic Surgery; Saarland University Medical Centre; Homburg/Saar Germany
| | - Matthias Hannig
- Department of Operative Dentistry and Periodontology; Saarland University Medical Centre; Homburg/Saar Germany
| | - Henning Madry
- Centre of Experimental Orthopaedics; Saarland University; Homburg/Saar Germany
- Department of Orthopaedic Surgery; Saarland University Medical Centre; Homburg/Saar Germany
| |
Collapse
|
37
|
Madry H, Luyten FP, Facchini A. Biological aspects of early osteoarthritis. Knee Surg Sports Traumatol Arthrosc 2012; 20:407-22. [PMID: 22009557 DOI: 10.1007/s00167-011-1705-8] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 10/04/2011] [Indexed: 01/15/2023]
Abstract
PURPOSE Early OA primarily affects articular cartilage and involves the entire joint, including the subchondral bone, synovial membrane, menisci and periarticular structures. The aim of this review is to highlight the molecular basis and histopathological features of early OA. METHODS Selective review of literature. RESULTS Risk factors for developing early OA include, but are not limited to, a genetic predisposition, mechanical factors such as axial malalignment, and aging. In early OA, the articular cartilage surface is progressively becoming discontinuous, showing fibrillation and vertical fissures that extend not deeper than into the mid-zone of the articular cartilage, reflective of OARSI grades 1.0-3.0. Early changes in the subchondral bone comprise a progressive increase in subchondral plate and subarticular spongiosa thickness. Early OA affects not only the articular cartilage and the subchondral bone but also other structures of the joint, such as the menisci, the synovial membrane, the joint capsule, ligaments, muscles and the infrapatellar fat pad. Genetic markers or marker combinations may become useful in the future to identify early OA and patients at risk. CONCLUSION The high socioeconomic impact of OA suggests that a better insight into the mechanisms of early OA may be a key to develop more targeted reconstructive therapies at this first stage of the disease. LEVEL OF EVIDENCE Systematic review, Level II.
Collapse
Affiliation(s)
- Henning Madry
- Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Kirrbergerstrasse, Building 37, 66421, Homburg, Germany.
| | | | | |
Collapse
|
38
|
Orth P, Zurakowski D, Wincheringer D, Madry H. Reliability, reproducibility, and validation of five major histological scoring systems for experimental articular cartilage repair in the rabbit model. Tissue Eng Part C Methods 2011; 18:329-39. [PMID: 22081995 DOI: 10.1089/ten.tec.2011.0462] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Histological evaluation of the repair tissue is a main pillar in the advancing field of experimental articular cartilage repair. Despite their widespread use, the major histological scoring systems for cartilage repair have seldom been validated. We tested the hypotheses (1) that elementary scores have a better reproducibility compared with more complex systems and (2) that the data from these different histological scores correlate with the DNA and proteoglycan contents of the repair tissue. A total of 1,165 observations of cartilage repair based on histological sections (n=233) from an experimental investigation on the repair of standardized osteochondral defects in vivo were made by three investigators with different levels of experience in cartilage research to determine the inter- and intra-observer reproducibility of elementary (Pineda and Wakitani score) and complex (O'Driscoll, Sellers, Fortier score) histological grading systems. DNA and proteoglycan contents of the repair tissues from simultaneously created defects were determined and correlated with histological (a) overall score values, (b) matrix staining, and (c) cellular characteristics of the five scores. Finally, applying the proteoglycan content as validating test, sensitivity, and specificity of the grading systems were assessed. All histological scores provided high intra- (Pearson r=0.92-0.99) and inter-observer reliability (intra-class correlation=0.94-0.99), low numerical intra- and inter-observer differences, and high internal correlations (Spearman's ρ=0.63-0.91). No disparity in reliability and reproducibility was detected between elementary and complex scores or between investigators with different levels of experience (all p>0.05). Individual histological overall score values did not correlate with proteoglycan contents but with DNA contents of the repair tissue (O'Driscoll, Wakitani, Sellers score). In all systems, proteoglycan contents did not correlate with matrix staining (all p>0.05), but histological cellular characteristics correlated with total cell numbers (p<0.001). These data indicate that both elementary and comprehensive histological scores are suited to quantify cartilage repair. Histological and biochemical evaluations may serve as complementary tools to assess articular cartilage repair in vivo.
Collapse
Affiliation(s)
- Patrick Orth
- Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg/Saar, Germany
| | | | | | | |
Collapse
|
39
|
Madry H, Grün UW, Knutsen G. Cartilage repair and joint preservation: medical and surgical treatment options. DEUTSCHES ARZTEBLATT INTERNATIONAL 2011; 108:669-77. [PMID: 22114626 DOI: 10.3238/arztebl.2011.0669] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 05/31/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND Articular cartilage defects are most often caused by trauma and osteoarthritis and less commonly by metabolic disorders of the subchondral bone, such as osteonecrosis and osteochondritis dissecans. Such defects do not heal spontaneously in adults and can lead to secondary osteoarthritis. Medications are indicated for symptomatic relief. Slow-acting drugs in osteoarthritis (SADOA), such as glucosamine and chondroitin, are thought to prevent cartilage degeneration. Reconstructive surgical treatment strategies aim to form a repair tissue or to unload compartments of the joint with articular cartilage damage. METHODS In this article, we selectively review the pertinent literature, focusing on original publications of the past 5 years and older standard texts. Particular attention is paid to guidelines and clinical studies with a high level of evidence, along with review articles, clinical trials, and book chapters. RESULTS There have been only a few randomized trials of medical versus surgical treatments. Pharmacological therapies are now available that are intended to treat the cartilage defect per se, rather than the associated symptoms, yet none of them has yet been shown to slow or reverse the progression of cartilage destruction. Surgical débridement of cartilage does not prevent the progression of osteoarthritis and is thus not recommended as the sole treatment. Marrow-stimulating procedures and osteochondral grafts are indicated for small focal articular cartilage defects, while autologous chondrocyte implantationis mainly indicated for larger cartilage defects. These surgical reconstructive techniques play a lesser role in the treatment of osteoarthritis. Osteotomy near the knee joint is indicated for axial realignment when unilateral osteoarthritis of the knee causes axis deviation. CONCLUSION Surgical reconstructive techniques can improve joint function and thereby postpone the need for replacement of the articular surface with an artificial joint.
Collapse
Affiliation(s)
- Henning Madry
- Lehrstuhl für Experimentelle Orthopädie und Arthroseforschung, Universität des Saarlandes, 66421 Homburg, Germany.
| | | | | |
Collapse
|