1
|
Wu H, Wang X, Wang G, Yuan G, Jia W, Tian L, Zheng Y, Ding W, Pei J. Advancing Scaffold-Assisted Modality for In Situ Osteochondral Regeneration: A Shift From Biodegradable to Bioadaptable. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2407040. [PMID: 39104283 DOI: 10.1002/adma.202407040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/10/2024] [Indexed: 08/07/2024]
Abstract
Over the decades, the management of osteochondral lesions remains a significant yet unmet medical challenge without curative solutions to date. Owing to the complex nature of osteochondral units with multi-tissues and multicellularity, and inherently divergent cellular turnover capacities, current clinical practices often fall short of robust and satisfactory repair efficacy. Alternative strategies, particularly tissue engineering assisted with biomaterial scaffolds, achieve considerable advances, with the emerging pursuit of a more cost-effective approach of in situ osteochondral regeneration, as evolving toward cell-free modalities. By leveraging endogenous cell sources and innate regenerative potential facilitated with instructive scaffolds, promising results are anticipated and being evidenced. Accordingly, a paradigm shift is occurring in scaffold development, from biodegradable and biocompatible to bioadaptable in spatiotemporal control. Hence, this review summarizes the ongoing progress in deploying bioadaptable criteria for scaffold-based engineering in endogenous osteochondral repair, with emphases on precise control over the scaffolding material, degradation, structure and biomechanics, and surface and biointerfacial characteristics, alongside their distinguished impact on the outcomes. Future outlooks of a highlight on advanced, frontier materials, technologies, and tools tailoring precision medicine and smart healthcare are provided, which potentially paves the path toward the ultimate goal of complete osteochondral regeneration with function restoration.
Collapse
Affiliation(s)
- Han Wu
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite & Center of Hydrogen Science, School of Materials Science & Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xuejing Wang
- Interdisciplinary Research Center of Biology & Catalysis, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Guocheng Wang
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong, 518055, China
| | - Guangyin Yuan
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite & Center of Hydrogen Science, School of Materials Science & Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weitao Jia
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Liangfei Tian
- Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Wenjiang Ding
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite & Center of Hydrogen Science, School of Materials Science & Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jia Pei
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite & Center of Hydrogen Science, School of Materials Science & Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Institute of Medical Robotics & National Engineering Research Center for Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
2
|
Zhang Y, Dong Q, Zhao X, Sun Y, Lin X, Zhang X, Wang T, Yang T, Jiang X, Li J, Cao Z, Cai T, Liu W, Zhang H, Bai J, Yao Q. Honeycomb-like biomimetic scaffold by functionalized antibacterial hydrogel and biodegradable porous Mg alloy for osteochondral regeneration. Front Bioeng Biotechnol 2024; 12:1417742. [PMID: 39070169 PMCID: PMC11273084 DOI: 10.3389/fbioe.2024.1417742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction: Osteochondral repair poses a significant challenge due to its unique pathological mechanisms and complex repair processes, particularly in bacterial tissue conditions resulting from open injuries, infections, and surgical contamination. This study introduces a biomimetic honeycomb-like scaffold (Zn-AlgMA@Mg) designed for osteochondral repair. The scaffold consists of a dicalcium phosphate dihydrate (DCPD)-coated porous magnesium scaffold (DCPD Mg) embedded within a dual crosslinked sodium alginate hydrogel (Zn-AlgMA). This combination aims to synergistically exert antibacterial and osteochondral integrated repair properties. Methods: The Zn-AlgMA@Mg scaffold was fabricated by coating porous magnesium scaffolds with DCPD and embedding them within a dual crosslinked sodium alginate hydrogel. The structural and mechanical properties of the DCPD Mg scaffold were characterized using scanning electron microscopy (SEM) and mechanical testing. The microstructural features and hydrophilicity of Zn-AlgMA were assessed. In vitro studies were conducted to evaluate the controlled release of magnesium and zinc ions, as well as the scaffold's osteogenic, chondrogenic, and antibacterial properties. Proteomic analysis was performed to elucidate the mechanism of osteochondral integrated repair. In vivo efficacy was evaluated using a rabbit full-thickness osteochondral defect model, with micro-CT evaluation, quantitative analysis, and histological staining (hematoxylin-eosin, Safranin-O, and Masson's trichrome). Results: The DCPD Mg scaffold exhibited a uniform porous structure and superior mechanical properties. The Zn-AlgMA hydrogel displayed consistent microstructural features and enhanced hydrophilicity. The Zn-AlgMA@Mg scaffold provided controlled release of magnesium and zinc ions, promoting cell proliferation and vitality. In vitro studies demonstrated significant osteogenic and chondrogenic properties, as well as antibacterial efficacy. Proteomic analysis revealed the underlying mechanism of osteochondral integrated repair facilitated by the scaffold. Micro-CT evaluation and histological analysis confirmed successful osteochondral integration in the rabbit model. Discussion: The biomimetic honeycomb-like scaffold (Zn-AlgMA@Mg) demonstrated promising results for osteochondral repair, effectively addressing the challenges posed by bacterial tissue conditions. The scaffold's ability to release magnesium and zinc ions in a controlled manner contributed to its significant osteogenic, chondrogenic, and antibacterial properties. Proteomic analysis provided insights into the scaffold's mechanism of action, supporting its potential for integrated osteochondral regeneration. The successful in vivo results highlight the scaffold's efficacy, making it a promising biomaterial for future applications in osteochondral repair.
Collapse
Affiliation(s)
- Yongqiang Zhang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Qiangsheng Dong
- School of Materials Science and Engineering, Jiangsu Key Laboratory of Advanced Structural Materials and Application Technology, Nanjing Institute of Technology, Nanjing, China
| | - Xiao Zhao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Yuzhi Sun
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Xin Lin
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Zhang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Tianming Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Tianxiao Yang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Xiao Jiang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Jiaxiang Li
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Zhicheng Cao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Tingwen Cai
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Wanshun Liu
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Hongjing Zhang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| | - Jing Bai
- School of Materials Science and Engineering, Southeast University, Nanjing, Jiangsu, China
- Institute of Medical Devices (Suzhou), Southeast University, Suzhou, China
| | - Qingqiang Yao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Key Lab of Additive Manufacturing Technology, Institute of Digital Medicine, Nanjing Medical University, Nanjing, China
- Research Center of Digital Medicine and 3D Printing Technology of Jiangsu Province, Nanjing, China
| |
Collapse
|
3
|
Sangiorgio A, Andriolo L, Gersoff W, Kon E, Nakamura N, Nehrer S, Vannini F, Filardo G. Subchondral bone: An emerging target for the treatment of articular surface lesions of the knee. J Exp Orthop 2024; 11:e12098. [PMID: 39040436 PMCID: PMC11260998 DOI: 10.1002/jeo2.12098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Purpose When dealing with the health status of the knee articular surface, the entire osteochondral unit has gained increasing attention, and in particular the subchondral bone, which plays a key role in the integrity of the osteochondral unit. The aim of this article was to discuss the current evidence on the role of the subchondral bone. Methods Experts from different geographical regions were involved in performing a review on highly discussed topics about the subchondral bone, ranging from its etiopathogenetic role in joint degeneration processes to its prognostic role in chondral and osteochondral defects, up to treatment strategies to address both the subchondral bone and the articular surface. Discussion Subchondral bone has a central role both from an aetiologic point of view and as a diagnostic tool, and its status was found to be relevant also as a prognostic factor in the follow-up of chondral treatment. Finally, the recognition of its importance in the natural history of these lesions led to consider subchondral bone as a treatment target, with the development of osteochondral scaffolds and procedures to specifically address osteochondral lesions. Conclusion Subchondral bone plays a central role in articular surface lesions from different points of view. Several aspects still need to be understood, but a growing interest in subchondral bone is to be expected in the upcoming future towards the optimization of joint preservation strategies. Level of Evidence Level V, expert opinion.
Collapse
Affiliation(s)
| | - Luca Andriolo
- Clinica Ortopedica e Traumatologica 2IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Wayne Gersoff
- Orthopedic Centers of Colorado Joint Preservation Institute, Clinical InstructorUniversity of Colorado Health Sciences CenterAuroraColoradoUSA
| | - Elizaveta Kon
- IRCCS Humanitas Research HospitalRozzanoItaly
- Department of Biomedical SciencesHumanitas University, Pieve EmanueleMilanItaly
- Department of Traumatology, Orthopaedics and Disaster SurgerySechenov First Moscow State Medical University (Sechenov University)MoscowRussia
| | - Norimasa Nakamura
- Institute for Medical Science in SportsOsaka Health Science UniversityOsakaJapan
- Center for Advanced Medical Engineering and InformaticsOsaka UniversitySuitaJapan
| | - Stefan Nehrer
- Faculty Health & MedicineUniversity for Continuing EducationKremsAustria
- Department of Orthopaedics and TraumatologyUniversity Hospital Krems, Karl Landsteiner University of Health SciencesKremsAustria
| | - Francesca Vannini
- Clinica Ortopedica e Traumatologica1 IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Giuseppe Filardo
- Service of Orthopaedics and Traumatology, Department of SurgeryEOCLuganoSwitzerland
- Faculty of Biomedical SciencesUniversità della Svizzera ItalianaLuganoSwitzerland
- Applied and Translational Research (ATR) CenterIRCCS Istituto Ortopedico RizzoliBolognaItaly
| |
Collapse
|
4
|
Zadegan S, Vahidi B, Nourmohammadi J, Shojaee A, Haghighipour N. Evaluation of rabbit adipose derived stem cells fate in perfused multilayered silk fibroin composite scaffold for Osteochondral repair. J Biomed Mater Res B Appl Biomater 2024; 112:e35396. [PMID: 38433653 DOI: 10.1002/jbm.b.35396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/30/2023] [Accepted: 02/18/2024] [Indexed: 03/05/2024]
Abstract
Development of osteochondral tissue engineering approaches using scaffolds seeded with stem cells in association with mechanical stimulations has been recently considered as a promising technique for the repair of this tissue. In this study, an integrated and biomimetic trilayered silk fibroin (SF) scaffold containing SF nanofibers in each layer was fabricated. The osteogenesis and chondrogenesis of stem cells seeded on the fabricated scaffolds were investigated under a perfusion flow. 3-Dimethylthiazol-2,5-diphenyltetrazolium bromide assay showed that the perfusion flow significantly enhanced cell viability and proliferation. Analysis of gene expression by stem cells revealed that perfusion flow had significantly upregulated the expression of osteogenic and chondrogenic genes in the bone and cartilage layers and downregulated the hypertrophic gene expression in the intermediate layer of the scaffold. In conclusion, applying flow perfusion on the prepared integrated trilayered SF-based scaffold can support osteogenic and chondrogenic differentiation for repairing osteochondral defects.
Collapse
Affiliation(s)
- Sara Zadegan
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Bahman Vahidi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Jhamak Nourmohammadi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Asiyeh Shojaee
- Division of Physiology, Department of Basic Science, Faculty of Veterinary, Amol University of Special Modern Technologies, Amol, Iran
| | | |
Collapse
|
5
|
Banihashemian SA, Zamanlui Benisi S, Hosseinzadeh S, Shojaei S, Abbaszadeh HA. Chitosan/Hyaluronan and Alginate-Nanohydroxyapatite Biphasic Scaffold as a Promising Matrix for Osteoarthritis Disorders. Adv Pharm Bull 2024; 14:176-191. [PMID: 38585453 PMCID: PMC10997938 DOI: 10.34172/apb.2024.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/24/2023] [Accepted: 07/19/2023] [Indexed: 04/09/2024] Open
Abstract
Purpose Regenerative medicine offers new techniques for osteoarthritis (OA) disorders, especially while considering simultaneous chondral and subchondral regenerations. Methods Chitosan and hyaluronan were chemically bound as the chondral phase and the osteogenic layer was prepared with alginate and nano-hydroxyapatite (nHAP). These scaffolds were fixed by fibrin glue as a biphasic scaffold and then examined. Results Scanning electron microscopy (SEM) confirmed the porosity of 61.45±4.51 and 44.145±2.81 % for the subchondral and chondral layers, respectively. The composition analysis by energy dispersive X-ray (EDAX) indicated the various elements of both hydrogels. Also, their mechanical properties indicated that the highest modulus and resistance values corresponded to the biphasic hydrogel as 108.33±5.56 and 721.135±8.21 kPa, despite the same strain value as other groups. Their individual examinations demonstrated the proteoglycan synthesis of the chondral layer and also, the alkaline phosphatase (ALP) activity of the subchondral layer as 13.3±2.2 ng. After 21 days, the cells showed a mineralized surface and a polygonal phenotype, confirming their commitment to bone and cartilage tissues, respectively. Immunostaining of collagen I and II represented greater extracellular matrix (ECM) secretion in the biphasic composite group due to the paracrine effect of the two cell types on each other. Conclusion For the first time, the ability of this biphasic scaffold to regenerate both tissue types was evaluated and the results showed satisfactory cellular commitment to bone and cartilage tissues. Thus, this scaffold can be considered a new strategy for the preparation of implants for OA.
Collapse
Affiliation(s)
- Seyed Abdolvahab Banihashemian
- Advanced Medical Sciences and Technologies Department, Faculty of Biomedical Engineering, Central Tehran Branch Islamic Azad University, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Tehran Central Branch, Islamic Azad University, Tehran, Iran
| | - Simzar Hosseinzadeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahrokh Shojaei
- Islamic Azad University Central Tehran Branch, Department of Biomedical Engineering, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Stocco TD, Zhang T, Dimitrov E, Ghosh A, da Silva AMH, Melo WCMA, Tsumura WG, Silva ADR, Sousa GF, Viana BC, Terrones M, Lobo AO. Carbon Nanomaterial-Based Hydrogels as Scaffolds in Tissue Engineering: A Comprehensive Review. Int J Nanomedicine 2023; 18:6153-6183. [PMID: 37915750 PMCID: PMC10616695 DOI: 10.2147/ijn.s436867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/12/2023] [Indexed: 11/03/2023] Open
Abstract
Carbon-based nanomaterials (CBNs) are a category of nanomaterials with various systems based on combinations of sp2 and sp3 hybridized carbon bonds, morphologies, and functional groups. CBNs can exhibit distinguished properties such as high mechanical strength, chemical stability, high electrical conductivity, and biocompatibility. These desirable physicochemical properties have triggered their uses in many fields, including biomedical applications. In this review, we specifically focus on applying CBNs as scaffolds in tissue engineering, a therapeutic approach whereby CBNs can act for the regeneration or replacement of damaged tissue. Here, an overview of the structures and properties of different CBNs will first be provided. We will then discuss state-of-the-art advancements of CBNs and hydrogels as scaffolds for regenerating various types of human tissues. Finally, a perspective of future potentials and challenges in this field will be presented. Since this is a very rapidly growing field, we expect that this review will promote interdisciplinary efforts in developing effective tissue regeneration scaffolds for clinical applications.
Collapse
Affiliation(s)
- Thiago Domingues Stocco
- Bioengineering Program, Scientific and Technological Institute, Brazil University, São Paulo, SP, Brazil
| | - Tianyi Zhang
- Pennsylvania State University, University Park, PA, USA
| | | | - Anupama Ghosh
- Department of Chemical and Materials Engineering (DEQM), Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Wanessa C M A Melo
- FTMC, State Research institute Center for Physical Sciences and Technology, Department of Functional Materials and Electronics, Vilnius, Lithuanian
| | - Willian Gonçalves Tsumura
- Bioengineering Program, Scientific and Technological Institute, Brazil University, São Paulo, SP, Brazil
| | - André Diniz Rosa Silva
- FATEC, Ribeirão Preto, SP, Brazil
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), BioMatLab Group, Materials Science and Engineering Graduate Program, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Gustavo F Sousa
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), BioMatLab Group, Materials Science and Engineering Graduate Program, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | - Bartolomeu C Viana
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), BioMatLab Group, Materials Science and Engineering Graduate Program, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| | | | - Anderson Oliveira Lobo
- Interdisciplinary Laboratory for Advanced Materials (LIMAV), BioMatLab Group, Materials Science and Engineering Graduate Program, Federal University of Piauí (UFPI), Teresina, PI, Brazil
| |
Collapse
|
7
|
Jarecki J, Waśko MK, Widuchowski W, Tomczyk-Warunek A, Wójciak M, Sowa I, Blicharski T. Knee Cartilage Lesion Management-Current Trends in Clinical Practice. J Clin Med 2023; 12:6434. [PMID: 37892577 PMCID: PMC10607427 DOI: 10.3390/jcm12206434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/26/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Many patients, particularly those aged above 40, experience knee joint pain, which hampers both sports activities and daily living. Treating isolated chondral and osteochondral defects in the knee poses a significant clinical challenge, particularly in younger patients who are not typically recommended partial or total knee arthroplasty as alternatives. Several surgical approaches have been developed to address focal cartilage defects. The treatment strategies are characterized as palliation (e.g., chondroplasty and debridement), repair (e.g., drilling and microfracture), or restoration (e.g., autologous chondrocyte implantation, osteochondral autograft, and osteochondral allograft). This review offers an overview of the commonly employed clinical methods for treating articular cartilage defects, with a specific focus on the clinical trials conducted in the last decade. Our study reveals that, currently, no single technology fully meets the essential requirements for effective cartilage healing while remaining easily applicable during surgical procedures. Nevertheless, numerous methods are available, and the choice of treatment should consider factors such as the location and size of the cartilage lesion, patient preferences, and whether it is chondral or osteochondral in nature. Promising directions for the future include tissue engineering, stem cell therapies, and the development of pre-formed scaffolds from hyaline cartilage, offering hope for improved outcomes.
Collapse
Affiliation(s)
- Jaromir Jarecki
- Department of Orthopaedics and Rehabilitation, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Marcin Krzysztof Waśko
- Department of Radiology and Imaging, The Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland;
| | - Wojciech Widuchowski
- Department of Physiotherapy, The College of Physiotherapy, 50-038 Wrocław, Poland;
| | - Agnieszka Tomczyk-Warunek
- Laboratory of Locomotor Systems Research, Department of Rehabilitation and Physiotherapy, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Magdalena Wójciak
- Department of Analytical Chemistry, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (M.W.); (I.S.)
| | - Ireneusz Sowa
- Department of Analytical Chemistry, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (M.W.); (I.S.)
| | - Tomasz Blicharski
- Department of Orthopaedics and Rehabilitation, Medical University of Lublin, 20-059 Lublin, Poland;
| |
Collapse
|
8
|
Jeyachandran D, Murshed M, Haglund L, Cerruti M. A Bioglass-Poly(lactic-co-glycolic Acid) Scaffold@Fibrin Hydrogel Construct to Support Endochondral Bone Formation. Adv Healthc Mater 2023; 12:e2300211. [PMID: 37462089 PMCID: PMC11468889 DOI: 10.1002/adhm.202300211] [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: 01/19/2023] [Revised: 06/21/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023]
Abstract
Bone tissue engineering using stem cells to build bone directly on a scaffold matrix often fails due to lack of oxygen at the injury site. This may be avoided by following the endochondral ossification route; herein, a cartilage template is promoted first, which can survive hypoxic environments, followed by its hypertrophy and ossification. However, hypertrophy is so far only achieved using biological factors. This work introduces a Bioglass-Poly(lactic-co-glycolic acid@fibrin (Bg-PLGA@fibrin) construct where a fibrin hydrogel infiltrates and encapsulates a porous Bg-PLGA. The hypothesis is that mesenchymal stem cells (MSCs) loaded in the fibrin gel and induced into chondrogenesis degrade the gel and become hypertrophic upon reaching the stiffer, bioactive Bg-PLGA core, without external induction factors. Results show that Bg-PLGA@fibrin induces hypertrophy, as well as matrix mineralization and osteogenesis; it also promotes a change in morphology of the MSCs at the gel/scaffold interface, possibly a sign of osteoblast-like differentiation of hypertrophic chondrocytes. Thus, the Bg-PLGA@fibrin construct can sequentially support the different phases of endochondral ossification purely based on material cues. This may facilitate clinical translation by decreasing in-vitro cell culture time pre-implantation and the complexity associated with the use of external induction factors.
Collapse
Affiliation(s)
| | - Monzur Murshed
- Faculty of DentistryDepartment of Medicineand Shriners Hospital for ChildrenMcGill UniversityMontrealQuebecH4A 0A9Canada
| | - Lisbet Haglund
- Experimental SurgeryMcGill UniversityMontrealH3G 2M1Canada
| | - Marta Cerruti
- Department of Mining and Materials EngineeringMcGill UniversityMontrealH3A 0C1Canada
| |
Collapse
|
9
|
Yao H, Wang C, Zhang Y, Wan Y, Min Q. Manufacture of Bilayered Composite Hydrogels with Strong, Elastic, and Tough Properties for Osteochondral Repair Applications. Biomimetics (Basel) 2023; 8:biomimetics8020203. [PMID: 37218789 DOI: 10.3390/biomimetics8020203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Layered composite hydrogels have been considered attractive materials for use in osteochondral repair and regeneration. These hydrogel materials should be mechanically strong, elastic, and tough besides fulfilling some basic requirements such as biocompatibility and biodegradability. A novel type of bilayered composite hydrogel with multi-network structures and well-defined injectability was thus developed for osteochondral tissue engineering using chitosan (CH), hyaluronic acid (HA), silk fibroin (SF), CH nanoparticles (NPs), and amino-functionalized mesoporous bioglass (ABG) NPs. CH was combined with HA and CH NPs to build the chondral phase of the bilayered hydrogel, and CH, SF, and ABG NPs were used together to construct the subchondral phase of the bilayer hydrogel. Rheological measurements showed that the optimally achieved gels assigned to the chondral and subchondral layers had their elastic moduli of around 6.5 and 9.9 kPa, respectively, with elastic modulus/viscous modulus ratios higher than 36, indicating that they behaved like strong gels. Compressive measurements further demonstrated that the bilayered hydrogel with an optimally formulated composition had strong, elastic, and tough characteristics. Cell culture revealed that the bilayered hydrogel had the capacity to support the in-growth of chondrocytes in the chondral phase and osteoblasts in the subchondral phase. Results suggest that the bilayered composite hydrogel can act as an injective biomaterial for osteochondral repair applications.
Collapse
Affiliation(s)
- Hui Yao
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, China
| | - Congcong Wang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuchen Zhang
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, China
| | - Ying Wan
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qing Min
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, China
| |
Collapse
|
10
|
Barui S, Ghosh D, Laurencin CT. Osteochondral regenerative engineering: challenges, state-of-the-art and translational perspectives. Regen Biomater 2022; 10:rbac109. [PMID: 36683736 PMCID: PMC9845524 DOI: 10.1093/rb/rbac109] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/22/2022] [Accepted: 12/09/2022] [Indexed: 12/27/2022] Open
Abstract
Despite quantum leaps, the biomimetic regeneration of cartilage and osteochondral regeneration remains a major challenge, owing to the complex and hierarchical nature of compositional, structural and functional properties. In this review, an account of the prevailing challenges in biomimicking the gradients in porous microstructure, cells and extracellular matrix (ECM) orientation is presented. Further, the spatial arrangement of the cues in inducing vascularization in the subchondral bone region while maintaining the avascular nature of the adjacent cartilage layer is highlighted. With rapid advancement in biomaterials science, biofabrication tools and strategies, the state-of-the-art in osteochondral regeneration since the last decade has expansively elaborated. This includes conventional and additive manufacturing of synthetic/natural/ECM-based biomaterials, tissue-specific/mesenchymal/progenitor cells, growth factors and/or signaling biomolecules. Beyond the laboratory-based research and development, the underlying challenges in translational research are also provided in a dedicated section. A new generation of biomaterial-based acellular scaffold systems with uncompromised biocompatibility and osteochondral regenerative capability is necessary to bridge the clinical demand and commercial supply. Encompassing the basic elements of osteochondral research, this review is believed to serve as a standalone guide for early career researchers, in expanding the research horizon to improve the quality of life of osteoarthritic patients affordably.
Collapse
Affiliation(s)
- Srimanta Barui
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Debolina Ghosh
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | |
Collapse
|
11
|
A Macroporous Cryogel with Enhanced Mechanical Properties for Osteochondral Regeneration In vivo. CHINESE JOURNAL OF POLYMER SCIENCE 2022. [DOI: 10.1007/s10118-022-2835-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
12
|
Deng Z, Zhu W, Lu B, Li M, Xu D. A Slotted Decellularized Osteochondral Scaffold With Layer-Specific Release of Stem Cell Differentiation Stimulators Enhances Cartilage and Bone Regeneration in Osteochondral Defects in a Rabbit Model. Am J Sports Med 2022; 50:3390-3405. [PMID: 36122351 DOI: 10.1177/03635465221114412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Owing to the disappointing regenerative ability of osteochondral tissue, without treatment an osteochondral defect would progress to osteoarthritis. This situation motivates the need for new strategies to enhance the regeneration of osteochondral defects. PURPOSE To develop a tissue-engineering scaffold by tethering bone morphogenetic protein 2 (BMP2) and transforming growth factor beta 3 (TGFβ3) in a layer-specific manner on a slotted decellularized osteochondral matrix (SDOM) and to evaluate the efficacy of this scaffold for osteochondral regeneration. STUDY DESIGN Controlled laboratory study. METHODS Normal osteochondral tissue from the rabbit patellofemoral groove was sectioned into a slot shape and decellularized for fabricating an SDOM. The collagen-binding domain (CBD) was fused into the N-terminus of BMP2 or TGFβ3 to synthesize 2 recombinant growth factors (GFs) (CBD-BMP2 or CBD-TGFβ3), which were tethered to the bone layer and cartilage layer, respectively, of the SDOM to prepare a tissue-engineering scaffold (namely, CBD-GFs/SDOM). After examining the influence of the CBD-GFs/SDOM on the viability and layer-specific differentiation of bone marrow mesenchymal stem cells in vitro, we determined the regeneration potential of the CBD-GFs/SDOM on osteochondral regeneration in a rabbit model. A total of 72 New Zealand White rabbits with a cylindrical osteochondral defect in the patellofemoral groove were randomly assigned to 3 groups: defect only (control [CTL] group), defect patched with an SDOM (SDOM group), and defect patched with the CBD-GFs/SDOM (CBD-GFs/SDOM group). At 6 or 12 weeks postoperatively, the rabbits were euthanized to harvest the knee joint, which was then evaluated via gross observation, micro-computed tomography, histological staining, and mechanical testing. RESULTS In vitro, the CBD-GFs/SDOM was noncytotoxic, showed high biomimetics with normal osteochondral tissue, was suitable for cell adhesion and growth, and had good layer-specific ability in inducing stem cell differentiation. Macroscopic images showed that the CBD-GFs/SDOM group had significantly better osteochondral regeneration than the CTL and SDOM groups had. Micro-computed tomography demonstrated that much more bony tissue was formed at the defect sites in the CBD-GFs/SDOM group compared with the defect sites in the CTL or SDOM group. Histological analysis showed that the CBD-GFs/SDOM group had a significant enhancement in osteochondral regeneration at 6 and 12 weeks postoperatively in comparison with the CTL or SDOM group. At 12 weeks postoperatively, the mechanical properties of reparative tissue were significantly better in the CBD-GFs/SDOM group than in the other groups. CONCLUSION The CBD-GFs/SDOM is a promising scaffold for osteochondral regeneration. CLINICAL RELEVANCE The findings of this study indicated that the CBD-GFs/SDOM is an excellent candidate for reconstructing osteochondral defects, which may be translated for clinical use in the future.
Collapse
Affiliation(s)
- Zhenhan Deng
- Department of Sports Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Weimin Zhu
- Department of Sports Medicine, First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Muzhi Li
- Department of Rehabilitation, Second Xiangya Hospital, Central South University, Changsha, China
| | - Daqi Xu
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, China.,Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Progress in Osteochondral Regeneration with Engineering Strategies. Ann Biomed Eng 2022; 50:1232-1242. [PMID: 35994165 DOI: 10.1007/s10439-022-03060-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/01/2022]
Abstract
Osteoarthritis, the main cause of disability worldwide, involves not only cartilage injury but also subchondral bone injury, which brings challenges to clinical repair. Tissue engineering strategies provide a promising solution to this degenerative disease. Articular cartilage connects to subchondral bone through the osteochondral interfacial tissue, which has a complex anatomical architecture, distinct cell distribution and unique biomechanical properties. Forming a continuous and stable osteochondral interface between cartilage tissue and subchondral bone is challenging. Thus, successful osteochondral regeneration with engineering strategies requires intricately coordinated interplay between cells, materials, biological factors, and physical/chemical factors. This review provides an overview of the anatomical composition, microstructure, and biomechanical properties of the osteochondral interface. Additionally, the latest research on the progress related to osteochondral regeneration is reviewed, especially discussing the fabrication of biomimetic scaffolds and the regulation of biological factors for osteochondral defects.
Collapse
|
14
|
Zhou Z, Cui J, Wu S, Geng Z, Su J. Silk fibroin-based biomaterials for cartilage/osteochondral repair. Am J Cancer Res 2022; 12:5103-5124. [PMID: 35836802 PMCID: PMC9274741 DOI: 10.7150/thno.74548] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/18/2022] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a common joint disease with a high disability rate. In addition, OA not only causes great physiological and psychological harm to patients, but also puts great pressure on the social healthcare system. Pathologically, the disintegration of cartilage and the lesions of subchondral bone are related to OA. Currently, tissue engineering, which is expected to overcome the defects of existing treatment methods, had a lot of research in the field of cartilage/osteochondral repair. Silk fibroin (SF), as a natural macromolecular material with good biocompatibility, unique mechanical properties, excellent processability and degradability, holds great potential in the field of tissue engineering. Nowadays, SF had been prepared into various materials to adapt to the demands of cartilage/osteochondral repair. SF-based biomaterials can also be functionally modified to enhance repair performance further. In this review, the preparation methods, types, structures, mechanical properties, and functional modifications of SF-based biomaterials used for cartilage/osteochondral repair are summarized and discussed. We hope that this review will provide a reference for the design and development of SF-based biomaterials in cartilage/osteochondral repair field.
Collapse
Affiliation(s)
- Ziyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China,School of Medicine, Shanghai University, Shanghai 200444, China,School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jin Cui
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China,Department of Orthopedics Trauma, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Shunli Wu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China,School of Medicine, Shanghai University, Shanghai 200444, China,School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China,✉ Corresponding authors: Zhen Geng, ; Jiacan Su,
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China,✉ Corresponding authors: Zhen Geng, ; Jiacan Su,
| |
Collapse
|
15
|
Steele JAM, Moore AC, St-Pierre JP, McCullen SD, Gormley AJ, Horgan CC, Black CR, Meinert C, Klein T, Saifzadeh S, Steck R, Ren J, Woodruff MA, Stevens MM. In vitro and in vivo investigation of a zonal microstructured scaffold for osteochondral defect repair. Biomaterials 2022; 286:121548. [PMID: 35588688 PMCID: PMC7615488 DOI: 10.1016/j.biomaterials.2022.121548] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/28/2022] [Accepted: 04/23/2022] [Indexed: 01/15/2023]
Abstract
Articular cartilage is comprised of zones that vary in architecture, extracellular matrix composition, and mechanical properties. Here, we designed and engineered a porous zonal microstructured scaffold from a single biocompatible polymer (poly [ϵ-caprolactone]) using multiple fabrication strategies: electrospinning, spherical porogen leaching, directional freezing, and melt electrowriting. With this approach we mimicked the zonal structure of articular cartilage and produced a stiffness gradient through the scaffold which aligns with the mechanics of the native tissue. Chondrocyte-seeded scaffolds accumulated extracellular matrix including glycosaminoglycans and collagen II over four weeks in vitro. This prompted us to further study the repair efficacy in a skeletally mature porcine model. Two osteochondral lesions were produced in the trochlear groove of 12 animals and repaired using four treatment conditions: (1) microstructured scaffold, (2) chondrocyte seeded microstructured scaffold, (3) MaioRegen™, and (4) empty defect. After 6 months the defect sites were harvested and analyzed using histology, micro computed tomography, and Raman microspectroscopy mapping. Overall, the scaffolds were retained in the defect space, repair quality was repeatable, and there was clear evidence of osteointegration. The repair quality of the microstructured scaffolds was not superior to the control based on histological scoring; however, the lower score was biased by the lack of histological staining due to the limited degradation of the implant at 6 months. Longer follow up studies (e.g., 1 yr) will be required to fully evaluate the efficacy of the microstructured scaffold. In conclusion, we found consistent scaffold retention, osteointegration, and prolonged degradation of the microstructured scaffold, which we propose may have beneficial effects for the long-term repair of osteochondral defects.
Collapse
Affiliation(s)
- Joseph A M Steele
- Department of Materials, Imperial College London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, SW7 2AZ, UK; Division of Biomaterials and Regenerative Medicine, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, SE 171 77, Sweden
| | - Axel C Moore
- Department of Materials, Imperial College London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, SW7 2AZ, UK
| | - Jean-Philippe St-Pierre
- Department of Materials, Imperial College London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, SW7 2AZ, UK
| | - Seth D McCullen
- Department of Materials, Imperial College London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, SW7 2AZ, UK
| | - Adam J Gormley
- Department of Materials, Imperial College London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, SW7 2AZ, UK; Division of Biomaterials and Regenerative Medicine, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, SE 171 77, Sweden
| | - Conor C Horgan
- Department of Materials, Imperial College London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, SW7 2AZ, UK
| | - Cameron Rm Black
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Developmental Origins of Health and Disease, Institute of Developmental Sciences, University of Southampton Medical School, Southampton, SO16 6YD, UK; Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia; Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Australia
| | - Christoph Meinert
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Travis Klein
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia; ARC Training Centre in Additive Biomanufacturing, Brisbane, Australia
| | - Siamak Saifzadeh
- Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Australia
| | - Roland Steck
- Medical Engineering Research Facility, Queensland University of Technology, Brisbane, Australia
| | - Jiongyu Ren
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia; ARC Training Centre in Additive Biomanufacturing, Brisbane, Australia
| | - Maria A Woodruff
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia; ARC Training Centre in Additive Biomanufacturing, Brisbane, Australia.
| | - Molly M Stevens
- Department of Materials, Imperial College London, SW7 2AZ, UK; Department of Bioengineering, Imperial College London, SW7 2AZ, UK; Institute of Biomedical Engineering, Imperial College London, SW7 2AZ, UK; Division of Biomaterials and Regenerative Medicine, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, SE 171 77, Sweden.
| |
Collapse
|
16
|
Begines B, Arevalo C, Romero C, Hadzhieva Z, Boccaccini AR, Torres Y. Fabrication and Characterization of Bioactive Gelatin-Alginate-Bioactive Glass Composite Coatings on Porous Titanium Substrates. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15008-15020. [PMID: 35316017 PMCID: PMC8990524 DOI: 10.1021/acsami.2c01241] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/10/2022] [Indexed: 05/10/2023]
Abstract
In this research work, the fabrication of biphasic composite implants has been investigated. Porous, commercially available pure Ti (50 vol % porosity and pore distributions of 100-200, 250-355, and 355-500 μm) has been used as a cortical bone replacement, while different composites based on a polymer blend (gelatin and alginate) and bioactive glass (BG) 45S5 have been applied as a soft layer for cartilage tissues. The microstructure, degradation rates, biofunctionality, and wear behavior of the different composites were analyzed to find the best possible coating. Experiments demonstrated the best micromechanical balance for the substrate containing 200-355 μm size range distribution. In addition, although the coating prepared from alginate presented a lower mass loss, the composite containing 50% alginate and 50% gelatin showed a higher elastic recovery, which entails that this type of coating could replicate the functions of the soft tissue in areas of the joints. Therefore, results revealed that the combinations of porous commercially pure Ti and composites prepared from alginate/gelatin/45S5 BG are candidates for the fabrication of biphasic implants not only for the treatment of osteochondral defects but also potentially for any other diseases affecting simultaneously hard and soft tissues.
Collapse
Affiliation(s)
- Belen Begines
- Departamento
de Química Orgánica y Farmacéutica, Facultad
de Farmacia, Universidad de Sevilla, c/ Profesor García González
2, Seville 41012, Spain
| | - Cristina Arevalo
- Departamento
de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, c/ Virgen de África 7, Seville 41011, Spain
| | - Carlos Romero
- Departamento
de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, c/ Virgen de África 7, Seville 41011, Spain
- Department
of Materials Science and Engineering and Chemical Engineering, Universidad Carlos III de Madrid, Av. de la Universidad 30, Leganés, Madrid 28911, Spain
| | - Zoya Hadzhieva
- Institute
of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Aldo R. Boccaccini
- Institute
of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Yadir Torres
- Departamento
de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, c/ Virgen de África 7, Seville 41011, Spain
| |
Collapse
|
17
|
Bakhtiary N, Liu C, Ghorbani F. Bioactive Inks Development for Osteochondral Tissue Engineering: A Mini-Review. Gels 2021; 7:274. [PMID: 34940334 PMCID: PMC8700778 DOI: 10.3390/gels7040274] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 01/02/2023] Open
Abstract
Nowadays, a prevalent joint disease affecting both cartilage and subchondral bone is osteoarthritis. Osteochondral tissue, a complex tissue unit, exhibited limited self-renewal potential. Furthermore, its gradient properties, including mechanical property, bio-compositions, and cellular behaviors, present a challenge in repairing and regenerating damaged osteochondral tissues. Here, tissue engineering and translational medicine development using bioprinting technology provided a promising strategy for osteochondral tissue repair. In this regard, personalized stratified scaffolds, which play an influential role in osteochondral regeneration, can provide potential treatment options in early-stage osteoarthritis to delay or avoid the use of joint replacements. Accordingly, bioactive scaffolds with possible integration with surrounding tissue and controlling inflammatory responses have promising future tissue engineering perspectives. This minireview focuses on introducing biologically active inks for bioprinting the hierarchical scaffolds, containing growth factors and bioactive materials for 3D printing of regenerative osteochondral substitutes.
Collapse
Affiliation(s)
- Negar Bakhtiary
- Department of Biomaterials, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran 14115-114, Iran;
| | - Chaozong Liu
- Institute of Orthopaedic & Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK;
| | - Farnaz Ghorbani
- Institute of Biomaterials, Department of Material Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany
| |
Collapse
|
18
|
Deng X, Chen X, Geng F, Tang X, Li Z, Zhang J, Wang Y, Wang F, Zheng N, Wang P, Yu X, Hou S, Zhang W. Precision 3D printed meniscus scaffolds to facilitate hMSCs proliferation and chondrogenic differentiation for tissue regeneration. J Nanobiotechnology 2021; 19:400. [PMID: 34856996 PMCID: PMC8641190 DOI: 10.1186/s12951-021-01141-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022] Open
Abstract
Background The poor regenerative capability and structural complexity make the reconstruction of meniscus particularly challenging in clinic. 3D printing of polymer scaffolds holds the promise of precisely constructing complex tissue architecture, however the resultant scaffolds usually lack of sufficient bioactivity to effectively generate new tissue. Results Herein, 3D printing-based strategy via the cryo-printing technology was employed to fabricate customized polyurethane (PU) porous scaffolds that mimic native meniscus. In order to enhance scaffold bioactivity for human mesenchymal stem cells (hMSCs) culture, scaffold surface modification through the physical absorption of collagen I and fibronectin (FN) were investigated by cell live/dead staining and cell viability assays. The results indicated that coating with fibronectin outperformed coating with collagen I in promoting multiple-aspect stem cell functions, and fibronectin favors long-term culture required for chondrogenesis on scaffolds. In situ chondrogenic differentiation of hMSCs resulted in a time-dependent upregulation of SOX9 and extracellular matrix (ECM) assessed by qRT-PCR analysis, and enhanced deposition of collagen II and aggrecan confirmed by immunostaining and western blot analysis. Gene expression data also revealed 3D porous scaffolds coupled with surface functionalization greatly facilitated chondrogenesis of hMSCs. In addition, the subcutaneous implantation of 3D porous PU scaffolds on SD rats did not induce local inflammation and integrated well with surrounding tissues, suggesting good in vivo biocompatibility. Conclusions Overall, this study presents an approach to fabricate biocompatible meniscus constructs that not only recapitulate the architecture and mechanical property of native meniscus, but also have desired bioactivity for hMSCs culture and cartilage regeneration. The generated 3D meniscus-mimicking scaffolds incorporated with hMSCs offer great promise in tissue engineering strategies for meniscus regeneration. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Xingyu Deng
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Fang Geng
- Medtronic Technology Center, Shanghai, 201114, China
| | - Xin Tang
- Medtronic Technology Center, Shanghai, 201114, China
| | - Zhenzhen Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Jie Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yikai Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Hangzhou, Zhejiang Province, China
| | - Fangqian Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Hangzhou, Zhejiang Province, China
| | - Na Zheng
- State Key Laboratory of Chemical Engineering, School of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Peng Wang
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Nanjing, 210042, China
| | - Xiaohua Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China. .,Zhejiang Provincial Key Laboratory of Orthopaedics, Hangzhou, Zhejiang Province, China. .,Orthopedics Research Institute of Zhejiang University, Hangzhou, 310009, Zhejiang Province, China.
| | - Shurong Hou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Wei Zhang
- Medtronic Technology Center, Shanghai, 201114, China.
| |
Collapse
|
19
|
Filardo G, Andriolo L, Angele P, Berruto M, Brittberg M, Condello V, Chubinskaya S, de Girolamo L, Di Martino A, Di Matteo B, Gille J, Gobbi A, Lattermann C, Nakamura N, Nehrer S, Peretti GM, Shabshin N, Verdonk P, Zaslav K, Kon E. Scaffolds for Knee Chondral and Osteochondral Defects: Indications for Different Clinical Scenarios. A Consensus Statement. Cartilage 2021; 13:1036S-1046S. [PMID: 31941355 PMCID: PMC8808892 DOI: 10.1177/1947603519894729] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To develop patient-focused consensus guidelines on the indications for the use of scaffolds to address chondral and osteochondral femoral condyle lesions. DESIGN The RAND/UCLA Appropriateness Method (RAM) was used to develop patient-specific recommendations by combining the best available scientific evidence with the collective judgement of a panel of experts guided by a core panel and multidisciplinary discussers. A list of specific clinical scenarios was produced regarding adult patients with symptomatic lesions without instability, malalignment, or meniscal deficiency. Each scenario underwent discussion and a 2-round vote on a 9-point Likert-type scale (range 1-3 "inappropriate," 4-6 "uncertain," 7-9 "appropriate"). Scores were pooled to generate expert recommendations. RESULTS Scaffold (chondral vs. osteochondral), patient characteristics (age and sport activity level), and lesion characteristics (etiology, size, and the presence of osteoarthritis [OA]) were considered to define 144 scenarios. The use of scaffold-based procedures was considered appropriate in all cases of chondral or osteochondral lesions when joints are not affected by OA, while OA joints presented more controversial results. The analysis of the evaluated factors showed a different weight in influencing treatment appropriateness: the presence of OA influenced 58.3% of the indications, while etiology, size, and age were discriminating factors in 54.2%, 29.2%, and 16.7% of recommendations, respectively. CONCLUSIONS The consensus identified indications still requiring investigation, but also the convergence of the experts in several scenarios defined appropriate or inappropriate, which could support decision making in the daily clinical practice, guiding the use of scaffold-based procedures for the treatment of chondral and osteochondral knee defects.
Collapse
Affiliation(s)
- Giuseppe Filardo
- Applied and Translational Research (ATR)
Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luca Andriolo
- Clinica Ortopedica e Traumatologica 2,
IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy,Luca Andriolo, Clinica Ortopedica e
Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano, 1/10,
Bologna 40136, Italy.
| | - Peter Angele
- Clinic for Trauma and Reconstructive
Surgery, University Hospital Regensburg, Regensburg, Bayern, Germany,Sporthopaedicum Regensburg, Regensburg,
Germany
| | - Massimo Berruto
- UOS Knee SURGERY-1st University Clinic
of Orthopaedics, ASST Pini-CTO, Milan, Italy
| | - Mats Brittberg
- Cartilage Research Unit, University of
Gothenburg, Gothenburg, Sweden,Region Halland Orthopaedics, Kungsbacka
Hospital, Kungsbacka, Sweden
| | - Vincenzo Condello
- Joint Preservation and Reconstructive
Surgery and Sports Medicine Unit, Humanitas Castelli Clinic, Bergamo, Lombardy,
Italy
| | - Susan Chubinskaya
- Department of Pediatrics, Orthopedic
Surgery & Medicine (Section of Rheumatology), Rush University Medical Center,
Chicago, IL, USA
| | - Laura de Girolamo
- Orthopaedic Biotechnology Laboratory,
IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Alessandro Di Martino
- Clinica Ortopedica e Traumatologica 2,
IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Berardo Di Matteo
- Department of Biomedical Sciences,
Humanitas University, Rozzano, Milan, Italy,Humanitas Clinical and Research
Center- IRCCS, Via Manzoni 56, 20089, Rozzano - Milan, Italy
| | - Justus Gille
- Department of Trauma and Orthopaedic
Surgery, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck,
Germany
| | - Alberto Gobbi
- Orthopaedic Arthroscopic Surgery
International (OASI) Bioresearch Foundation, Milan, Italy
| | - Christian Lattermann
- Department of Orthopaedic Surgery,
Division of Sports Medicine, Center for Cartilage Repair, Brigham and Women’s
Hospital, Harvard Medical School, Boston, MA, USA
| | - Norimasa Nakamura
- Institute for Medical Science in
Sports, Osaka Health Science University, Osaka, Japan
| | - Stefan Nehrer
- Center for Regenerative Medicine,
Danube University, Krems an der Donau, Austria
| | - Giuseppe M. Peretti
- IRCCS Istituto Ortopedico Galeazzi,
Milan, Italy,Department of Biomedical Sciences for
Health, University of Milan, Milan, Italy
| | - Nogah Shabshin
- Department of Radiology, Emek Medical
Center, Clalit Healthcare Services, Afula, Israel,Department of Radiology, PennMedicine,
Philadelphia, PA, USA
| | - Peter Verdonk
- ORTHOCA, AZ Monica Hospitals, Antwerp,
Belgium,Aspetar Hospital, Doha, Qatar
| | - Kenneth Zaslav
- Ortho Virginia, Virginia Commonwealth
University, Richmond, VA, USA
| | - Elizaveta Kon
- Department of Biomedical Sciences,
Humanitas University, Rozzano, Milan, Italy,Humanitas Clinical and Research
Center- IRCCS, Via Manzoni 56, 20089, Rozzano - Milan, Italy,Department of Traumatology,
Orthopedics and Disaster Surgery, First Moscow State Medical University of the
Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russian
Federation
| |
Collapse
|
20
|
Van Genechten W, Vuylsteke K, Struijk C, Swinnen L, Verdonk P. Joint Surface Lesions in the Knee Treated with an Acellular Aragonite-Based Scaffold: A 3-Year Follow-Up Case Series. Cartilage 2021; 13:1217S-1227S. [PMID: 33448238 PMCID: PMC8808874 DOI: 10.1177/1947603520988164] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The study aimed to evaluate the clinical outcome and repair capacity of a cell-free aragonite-based scaffold in patients with an isolated symptomatic joint surface lesion (JSL) of the knee. DESIGN Thirteen patients (age 33.5 ± 8.9; female 23%; body mass index 25.3 ± 3.4, K/L [Kellgren-Lawrence] 1.8) with a JSL (2.6 ± 1.7 cm2 [1.0-7.5 cm2]) of the distal femur were enrolled in a single-center prospective case series. Safety and clinical outcome was assessed by the KOOS (Knee Injury and Osteoarthritis Outcome Score), IKDC (International Knee Documentation Committee), Lysholm, and Tegner activity scale at baseline and 6, 12, 18, 24, and 36 months follow-up. The MOCART 2.0 and scaffold integration were evaluated on magnetic resonance imaging at 12, 24, and 36 months postoperatively. RESULTS Primary outcome (KOOS pain) improved with 36.5 ± 14.7 points at 12 months (P = 0.002) and 41.2 ± 14.7 points at 36 months (P = 0.002) follow-up. Similar increasing trends were observed for the other KOOS subscales, IKDC, and Lysholm score, which were significantly better at each follow-up time point relative to baseline (P < 0.05). Activity level increased from 2.75 ± 1.6 to 4.6 ± 2.2 points at final follow-up (P = 0.07). The MOCART was 61.7 ± 12.6 at 12 months and 72.9 ± 13.0 at 36 months postoperatively. Sixty-six to 100% implant integration and remodeling was observed in 73.3% cases at 36 months. No serious adverse events were reported. CONCLUSION The study demonstrated that the biphasic aragonite-based scaffold is a safe and clinically effective implant for treating small-medium sized JSLs of the distal femur in a young and active patient cohort. The implant showed satisfying osteointegration and restoration of the osteochondral unit up to 3 years postimplantation.
Collapse
Affiliation(s)
- Wouter Van Genechten
- MoRe Foundation, Antwerp, Belgium,Antwerp University, Antwerp,
Belgium,Wouter Van Genechten, MoRe Foundation,
Stevenslei 20, Antwerp 2100, Belgium.
| | | | | | - Linus Swinnen
- Department of Radiology, AZ Monica,
Antwerp-Deurne, Antwerp, Belgium
| | - Peter Verdonk
- MoRe Foundation, Antwerp, Belgium,Antwerp University, Antwerp,
Belgium,ORTHOCA, Antwerp, Belgium
| |
Collapse
|
21
|
Shimomura K, Hamada H, Hart DA, Ando W, Nishii T, Trattnig S, Nehrer S, Nakamura N. Histological Analysis of Cartilage Defects Repaired with an Autologous Human Stem Cell Construct 48 Weeks Postimplantation Reveals Structural Details Not Detected by T2-Mapping MRI. Cartilage 2021; 13:694S-706S. [PMID: 33511856 PMCID: PMC8808920 DOI: 10.1177/1947603521989423] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE The aim of this study was to elucidate the efficacy of T2-mapping MRI and correlation with histology for the evaluation of tissue repair quality following the first-in-human implantation of an autologous tissue engineered construct. DESIGN We directly compared the results of T2-mapping MRI of cartilage repair tissue with the histology of a biopsy specimen from the corresponding area at 48 weeks postoperatively in 5 patients who underwent the implantation of a scaffold-free tissue-engineered construct generated from autologous synovial mesenchymal stem cells to repair an isolated cartilage lesion. T2 values and histological scores were compared at each of 2 layers of equally divided halves of the repair tissue (upper and lower zones). RESULTS Histology showed that the repair tissue in the upper zone was dominated by fibrous tissue and the ratio of hyaline-like matrix increased with the depth of the repair tissue. There were significant differences between upper and lower zones in histological scores. Conversely, there were no detectable statistically significant differences in T2 value detected among zones of the repair tissue, but zonal differences were detected in corresponding healthy cartilage. Accordingly, there were no correlations detected between histological scores and T2 values for each repair cartilage zone. CONCLUSION Discrepancies in the findings between T2 mapping and histology suggest that T2 mapping was limited in ability to detect details in the architecture and composition of the repair cartilage.
Collapse
Affiliation(s)
- Kazunori Shimomura
- Department of Orthopaedic Surgery, Osaka
University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Hamada
- Department of Orthopaedic Medical
Engineering, Osaka University Graduate School of Medicine, Osaka, Japan
| | - David A. Hart
- McCaig Institute for Bone & Joint
Health, University of Calgary, Calgary, Alberta, Canada
| | - Wataru Ando
- Department of Orthopaedic Medical
Engineering, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takashi Nishii
- Department of Orthopaedic Surgery, Osaka
General Medical Center, Osaka, Japan
| | - Siegfried Trattnig
- High Field MR Center, Department of
Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna,
Austria,Christian Doppler Laboratory for
Clinical Molecular MR Imaging (MOLIMA), Department of Biomedical Imaging and
Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Stefan Nehrer
- Faculty of Health and Medicine,
Department for Health Sciences, Medicine and Research, Center for Regenerative
Medicine, Danube University Krems, Krems, Austria
| | - Norimasa Nakamura
- Department of Orthopaedic Surgery, Osaka
University Graduate School of Medicine, Osaka, Japan,Institute for Medical Science in Sports,
Osaka Health Science University, Osaka, Japan,Global Center for Medical Engineering
and Informatics, Osaka University, Osaka, Japan,Norimasa Nakamura, Institute for Medical
Science in Sports, Osaka Health Science University, 1-9-27, Tenma, Kita-ku,
Osaka City, Osaka, 530-0043, Japan.
| |
Collapse
|
22
|
Combinations of Hydrogels and Mesenchymal Stromal Cells (MSCs) for Cartilage Tissue Engineering-A Review of the Literature. Gels 2021; 7:gels7040217. [PMID: 34842678 PMCID: PMC8628761 DOI: 10.3390/gels7040217] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 01/17/2023] Open
Abstract
Cartilage offers limited regenerative capacity. Cell-based approaches have emerged as a promising alternative in the treatment of cartilage defects and osteoarthritis. Due to their easy accessibility, abundancy, and chondrogenic potential mesenchymal stromal cells (MSCs) offer an attractive cell source. MSCs are often combined with natural or synthetic hydrogels providing tunable biocompatibility, biodegradability, and enhanced cell functionality. In this review, we focused on the different advantages and disadvantages of various natural, synthetic, and modified hydrogels. We examined the different combinations of MSC-subpopulations and hydrogels used for cartilage engineering in preclinical and clinical studies and reviewed the effects of added growth factors or gene transfer on chondrogenesis in MSC-laden hydrogels. The aim of this review is to add to the understanding of the disadvantages and advantages of various combinations of MSC-subpopulations, growth factors, gene transfers, and hydrogels in cartilage engineering.
Collapse
|
23
|
Chen YW, Shie MY, Chang WC, Shen YF. Approximate Optimization Study of Light Curing Waterborne Polyurethane Materials for the Construction of 3D Printed Cytocompatible Cartilage Scaffolds. MATERIALS 2021; 14:ma14226804. [PMID: 34832205 PMCID: PMC8626041 DOI: 10.3390/ma14226804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 11/18/2022]
Abstract
Articular cartilage, which is a white transparent tissue with 1–2 mm thickness, is located in the interface between the two hard bones. The main functions of articular cartilage are stress transmission, absorption, and friction reduction. The cartilage cannot be repaired and regenerated once it has been damaged, and it needs to be replaced by artificial joints. Many approaches, such as artificial joint replacement, hyaluronic acid injection, microfracture surgery and cartilage tissue engineering have been applied in clinical treatment. Basically, some of these approaches are foreign material implantation for joint replacement to reach the goal of pain reduction and mechanism support. This study demonstrated another frontier in the research of cartilage reconstruction by applying regeneration medicine additive manufacturing (3D Printing) and stem cell technology. Light curing materials have been modified and tested to be printable and cytocompatible for stem cells in this research. Design of experiments (DOE) is adapted in this investigation to search for the optimal manufacturing parameter for biocompatible scaffold fabrication and stem cell attachment and growth. Based on the results, an optimal working process of biocompatible and printable scaffolds for cartilage regeneration is reported. We expect this study will facilitate the development of cartilage tissue engineering.
Collapse
Affiliation(s)
- Yi-Wen Chen
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 40447, Taiwan; (Y.-W.C.); (M.-Y.S.); (W.-C.C.)
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City 40447, Taiwan
| | - Ming-You Shie
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 40447, Taiwan; (Y.-W.C.); (M.-Y.S.); (W.-C.C.)
- School of Dentistry, China Medical University, Taichung City 40447, Taiwan
| | - Wen-Ching Chang
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 40447, Taiwan; (Y.-W.C.); (M.-Y.S.); (W.-C.C.)
| | - Yu-Fang Shen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 41354, Taiwan
- High Performance Materials Institute for xD Printing, Asia University, Taichung City 41354, Taiwan
- Correspondence:
| |
Collapse
|
24
|
Wang X, Han X, Li C, Chen Z, Huang H, Chen J, Wu C, Fan T, Li T, Huang W, Al-Hartomy OA, Al-Ghamdi A, Wageh S, Zheng F, Al-Sehemi AG, Wang G, Xie Z, Zhang H. 2D materials for bone therapy. Adv Drug Deliv Rev 2021; 178:113970. [PMID: 34509576 DOI: 10.1016/j.addr.2021.113970] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/24/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Due to their prominent physicochemical properties, 2D materials are broadly applied in biomedicine. Currently, 2D materials have achieved great success in treating many diseases such as cancer and tissue engineering as well as bone therapy. Based on their different characteristics, 2D materials could function in various ways in different bone diseases. Herein, the application of 2D materials in bone tissue engineering, joint lubrication, infection of orthopedic implants, bone tumors, and osteoarthritis are firstly reviewed comprehensively together. Meanwhile, different mechanisms by which 2D materials function in each disease reviewed below are also reviewed in detail, which in turn reveals the versatile functions and application of 2D materials. At last, the outlook on how to further broaden applications of 2D materials in bone therapies based on their excellent properties is also discussed.
Collapse
Affiliation(s)
- Xiangjiang Wang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, Guangdong, China
| | - Xianjing Han
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, Guangdong, China
| | - Chaozhou Li
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Zhi Chen
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Hao Huang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jindong Chen
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, Guangdong, China
| | - Chenshuo Wu
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Taojian Fan
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China
| | - Tianzhong Li
- Shenzhen International Institute for Biomedical Research, Shenzhen 518116, Guangdong, China
| | - Weichun Huang
- Nantong Key Lab of Intelligent and New Energy Materials, School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, Jiangsu, PR China
| | - Omar A Al-Hartomy
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed Al-Ghamdi
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Swelm Wageh
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fei Zheng
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Abdullah G Al-Sehemi
- Department of Chemistry, Faculty of Science, Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, P.O. Box 9004, Saudi Arabia
| | - Guiqing Wang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, Guangdong, China
| | - Zhongjian Xie
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen 518038, Guangdong, PR China; Shenzhen International Institute for Biomedical Research, Shenzhen 518116, Guangdong, China
| | - Han Zhang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
25
|
Schoonraad SA, Fischenich KM, Eckstein KN, Crespo-Cuevas V, Savard LM, Muralidharan A, Tomaschke AA, Uzcategui AC, Randolph MA, McLeod RR, Ferguson VL, Bryant SJ. Biomimetic and mechanically supportive 3D printed scaffolds for cartilage and osteochondral tissue engineering using photopolymers and digital light processing. Biofabrication 2021; 13. [PMID: 34479218 DOI: 10.1088/1758-5090/ac23ab] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 09/03/2021] [Indexed: 02/08/2023]
Abstract
Successful 3D scaffold designs for musculoskeletal tissue engineering necessitate full consideration of the form and function of the tissues of interest. When designing structures for engineering cartilage and osteochondral tissues, one must reconcile the need to develop a mechanically robust system that maintains the health of cells embedded in the scaffold. In this work, we present an approach that decouples the mechanical and biochemical needs and allows for the independent development of the structural and cellular niches in a scaffold. Using the highly tuned capabilities of digital light processing-based stereolithography, structures with complex architectures are achieved over a range of effective porosities and moduli. The 3D printed structure is infilled with mesenchymal stem cells and soft biomimetic hydrogels, which are specifically formulated with extracellular matrix analogs and tethered growth factors to provide selected biochemical cues for the guided differentiation towards chondrogenesis and osteogenesis. We demonstrate the ability to utilize these structures to (a) infill a focal chondral defect and mitigate macroscopic and cellular level changes in the cartilage surrounding the defect, and (b) support the development of a stratified multi-tissue scaffold for osteochondral tissue engineering.
Collapse
Affiliation(s)
- Sarah A Schoonraad
- Materials Science and Engineering Program, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Kristine M Fischenich
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Kevin N Eckstein
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Victor Crespo-Cuevas
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Lea M Savard
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Andrew A Tomaschke
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Asais Camila Uzcategui
- Materials Science and Engineering Program, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Mark A Randolph
- Department of Orthopaedic Surgery, Laboratory for Musculoskeletal Tissue Engineering, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America
| | - Robert R McLeod
- Materials Science and Engineering Program, University of Colorado at Boulder, Boulder, CO 80309, United States of America.,Department of Electrical, Computer and Energy Engineering, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Virginia L Ferguson
- Materials Science and Engineering Program, University of Colorado at Boulder, Boulder, CO 80309, United States of America.,Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, CO 80309, United States of America.,BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, Boulder, CO 80309, United States of America.,BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309, United States of America.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, United States of America
| |
Collapse
|
26
|
Ricci M, Tradati D, Maione A, Uboldi FM, Usellini E, Berruto M. Cell-free osteochondral scaffolds provide a substantial clinical benefit in the treatment of osteochondral defects at a minimum follow-up of 5 years. J Exp Orthop 2021; 8:62. [PMID: 34398364 PMCID: PMC8368912 DOI: 10.1186/s40634-021-00381-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/29/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose The treatment of osteochondral lesions is challenging and no consensus has been established about the best option for restoring both cartilage and subchondral bone. Multilayer collagen-hydroxyapatite scaffolds have shown promising clinical results, but the outcome at a follow-up longer than 5 years still has to be proved. The aim was to evaluate the clinical outcome of patients with a knee isolated osteochondral lesion treated with a biomimetic three-layered scaffold at a minimum 5 years of follow-up. Methods Twenty-nine patients (23 males and 6 females, mean age 31.5 ± 11.4 years) were evaluated retrospectively before surgery, at 1 and 2 years and at last follow-up (FU). Visual Analog Scale (VAS) for pain, International Knee Documentation Committee (IKDC) Subjective Score, Tegner-Lysholm Knee Scoring Scale and Tegner Activity Level Scale were collected. Mean FU was 7.8 ± 2.0 years (min 5.1 - max 11.3). The etiology of the defect was Osteochondritis Dissecans or osteonecrosis (17 vs 12 cases). Results At 12 months FU the IKDC score improved from 51.1 ± 21.7 to 80.1 ± 17.9 (p < 0.01), Tegner Lysholm Score from 59.9 ± 17.3 to 92.5 ± 9.0 (p < 0.01), VAS from 6.1 ± 2.1 to 1.7 ± 2.3 (p < 0.01) and Tegner Activity Level Scale from 1.6 ± 0.5 to 4.9 ± 1.7 (p < 0.01). The results remained stable at 24 months, while at last FU a statistically significant decrease in IKDC, Tegner Lysholm and Tegner Activity Scale was recorded, though not clinically relevant. Patients under 35 achieved statistically better outcomes. Conclusions The use of a cell-free collagen-hydroxyapatite osteochondral scaffold provides substantial clinical benefits in the treatment of knee osteochondral lesions at a minimum follow-up of 5 years, especially in patients younger than 35 years. Level of evidence Level IV.
Collapse
Affiliation(s)
- Martina Ricci
- UOS Chirurgia Articolare del Ginocchio, I Clinica Ortopedica, ASST Gaetano Pini-CTO, Piazza Cardinal Ferrari 1, 20122, Milan, Italy.
| | - Daniele Tradati
- UOS Chirurgia Articolare del Ginocchio, I Clinica Ortopedica, ASST Gaetano Pini-CTO, Piazza Cardinal Ferrari 1, 20122, Milan, Italy
| | - Alessio Maione
- UOS Chirurgia Articolare del Ginocchio, I Clinica Ortopedica, ASST Gaetano Pini-CTO, Piazza Cardinal Ferrari 1, 20122, Milan, Italy
| | - Francesco Mattia Uboldi
- UOS Chirurgia Articolare del Ginocchio, I Clinica Ortopedica, ASST Gaetano Pini-CTO, Piazza Cardinal Ferrari 1, 20122, Milan, Italy
| | - Eva Usellini
- UOS Chirurgia Articolare del Ginocchio, I Clinica Ortopedica, ASST Gaetano Pini-CTO, Piazza Cardinal Ferrari 1, 20122, Milan, Italy
| | - Massimo Berruto
- UOS Chirurgia Articolare del Ginocchio, I Clinica Ortopedica, ASST Gaetano Pini-CTO, Piazza Cardinal Ferrari 1, 20122, Milan, Italy
| |
Collapse
|
27
|
Deng C, Yang J, He H, Ma Z, Wang W, Zhang Y, Li T, He C, Wang J. 3D bio-printed biphasic scaffolds with dual modification of silk fibroin for the integrated repair of osteochondral defects. Biomater Sci 2021; 9:4891-4903. [PMID: 34047307 DOI: 10.1039/d1bm00535a] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Repair of osteochondral defects is still a challenge, especially the regeneration of hyaline cartilage. Parathyroid hormone (PTH) can inhibit the hypertrophy of chondrocytes to maintain the phenotype of hyaline cartilage. Here, we aimed to construct a bio-printed biphasic scaffold with a mechanical gradient based on dual modification of silk fibroin (SF) for the integrated repair of osteochondral defects. Briefly, SF was grafted with PTH (SF-PTH) and covalently immobilized with methacrylic anhydride (SF-MA), respectively. Next, gelatin methacryloyl (GM) mixed with SF-PTH or SF-MA was used as a bio-ink for articular cartilage and subchondral bone regeneration. Finally, the GM + SF-PTH/GM + SF-MA osteochondral biphasic scaffold was constructed using 3D bioprinting technology, and implanted in a rabbit osteochondral defect model. In this study, the SF-PTH bio-ink was synthesized for the first time. In vitro results indicated that the GM + SF-MA bio-ink had good mechanical properties, while the GM + SF-PTH bio-ink inhibited the hypertrophy of chondrocytes and was beneficial for the production of hyaline cartilage extracellular matrix. Importantly, an integrated GM + SF-PTH/GM + SF-MA biphasic scaffold with a mechanical gradient was successfully constructed. The results in vivo demonstrated that the GM + SF-PTH/GM + SF-MA scaffold could promote the regeneration of osteochondral defects and maintain the phenotype of hyaline cartilage to a large extent. Collectively, our results indicate that the integrated GM + SF-PTH/GM + SF-MA biphasic scaffold constructed by 3D bioprinting is expected to become a new strategy for the treatment of osteochondral defects.
Collapse
Affiliation(s)
- Changxu Deng
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Jin Yang
- Key Laboratory of Science and Technology of Eco-Textiles, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, No. 2999, People North Road, Shanghai 201620, China.
| | - Hongtao He
- The Third Ward of Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, No. 467, Zhongshan Road, Shahekou District, Dalian 116000, Liaoning Province, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Wenhao Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Yuxin Zhang
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Tao Li
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Chuanglong He
- Key Laboratory of Science and Technology of Eco-Textiles, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, No. 2999, People North Road, Shanghai 201620, China.
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China. and Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1956 Huashan Road, Shanghai, 200030, China
| |
Collapse
|
28
|
Recent advances in bioprinting technologies for engineering different cartilage-based tissues. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112005. [PMID: 33812625 DOI: 10.1016/j.msec.2021.112005] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023]
Abstract
Inadequate self-repair and regenerative efficiency of the cartilage tissues has motivated the researchers to devise advanced and effective strategies to resolve this issue. Introduction of bioprinting to tissue engineering has paved the way for fabricating complex biomimetic engineered constructs. In this context, the current review gears off with the discussion of standard and advanced 3D/4D printing technologies and their implications for the repair of different cartilage tissues, namely, articular, meniscal, nasoseptal, auricular, costal, and tracheal cartilage. The review is then directed towards highlighting the current stem cell opportunities. On a concluding note, associated critical issues and prospects for future developments, particularly in this sphere of personalized medicines have been discussed.
Collapse
|
29
|
Zhou L, Gjvm VO, Malda J, Stoddart MJ, Lai Y, Richards RG, Ki-Wai Ho K, Qin L. Innovative Tissue-Engineered Strategies for Osteochondral Defect Repair and Regeneration: Current Progress and Challenges. Adv Healthc Mater 2020; 9:e2001008. [PMID: 33103381 DOI: 10.1002/adhm.202001008] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/19/2020] [Indexed: 12/20/2022]
Abstract
Clinical treatments for the repair of osteochondral defects (OCD) are merely palliative, not completely curative, and thus enormously unfulfilled challenges. With the in-depth studies of biology, medicine, materials, and engineering technology, the conception of OCD repair and regeneration should be renewed. During the past decades, many innovative tissue-engineered approaches for repairing and regenerating damaged osteochondral units have been widely explored. Various scaffold-free and scaffold-based strategies, such as monophasic, biphasic, and currently fabricated multiphasic and gradient architectures have been proposed and evaluated. Meanwhile, progenitor cells and tissue-specific cells have also been intensively investigated in vivo as well as ex vivo. Concerning bioactive factors and drugs, they have been combined with scaffolds and/or living cells, and even released in a spatiotemporally controlled manner. Although tremendous progress has been achieved, further research and development (R&D) is needed to convert preclinical outcomes into clinical applications. Here, the osteochondral unit structure, its defect classifications, and diagnosis are summarized. Commonly used clinical reparative techniques, tissue-engineered strategies, emerging 3D-bioprinting technologies, and the status of their clinical applications are discussed. Existing challenges to translation are also discussed and potential solutions for future R&D directions are proposed.
Collapse
Affiliation(s)
- Liangbin Zhou
- Musculoskeletal Research Laboratory of Department of Orthopedics & Traumatology, and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Van Osch Gjvm
- Department of Orthopedics and Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, 3000 CA, The Netherlands
- Department of Biomechanical Engineering, Delft University of Technology (TU Delft), Delft, 2600 AA, The Netherlands
| | - Jos Malda
- Department of Orthopaedics of University Medical Center Utrecht, and Department of Clinical Sciences of Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CS, The Netherlands
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, Davos, CH 7270, Switzerland
| | - Yuxiao Lai
- Centre for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, The Chinese Academy of Sciences, Shenzhen, 518000, China
| | - R Geoff Richards
- AO Research Institute Davos, Clavadelerstrasse 8, Davos, CH 7270, Switzerland
| | - Kevin Ki-Wai Ho
- Musculoskeletal Research Laboratory of Department of Orthopedics & Traumatology, and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopedics & Traumatology, and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
- Centre for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, The Chinese Academy of Sciences, Shenzhen, 518000, China
| |
Collapse
|
30
|
Jacob G, Shimomura K, Nakamura N. Osteochondral Injury, Management and Tissue Engineering Approaches. Front Cell Dev Biol 2020; 8:580868. [PMID: 33251212 PMCID: PMC7673409 DOI: 10.3389/fcell.2020.580868] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Osteochondral lesions (OL) are a common clinical problem for orthopedic surgeons worldwide and are associated with multiple clinical scenarios ranging from trauma to osteonecrosis. OL vary from chondral lesions in that they involve the subchondral bone and chondral surface, making their management more complex than an isolated chondral injury. Subchondral bone involvement allows for a natural healing response from the body as marrow elements are able to come into contact with the defect site. However, this repair is inadequate resulting in fibrous scar tissue. The second differentiating feature of OL is that damage to the subchondral bone has deleterious effects on the mechanical strength and nutritive capabilities to the chondral joint surface. The clinical solution must, therefore, address both the articular cartilage as well as the subchondral bone beneath it to restore and preserve joint health. Both cartilage and subchondral bone have distinctive functional requirements and therefore their physical and biological characteristics are very much dissimilar, yet they must work together as one unit for ideal joint functioning. In the past, the obvious solution was autologous graft transfer, where an osteochondral bone plug was harvested from a non-weight bearing portion of the joint and implanted into the defect site. Allografts have been utilized similarly to eliminate the donor site morbidity associated with autologous techniques and overall results have been good but both techniques have their drawbacks and limitations. Tissue engineering has thus been an attractive option to create multiphasic scaffolds and implants. Biphasic and triphasic implants have been under explored and have both a chondral and subchondral component with an interface between the two to deliver an implant which is biocompatible and emulates the osteochondral unit as a whole. It has been a challenge to develop such implants and many manufacturing techniques have been utilized to bring together two unalike materials and combine them with cellular therapies. We summarize the functions of the osteochondral unit and describe the currently available management techniques under study.
Collapse
Affiliation(s)
- George Jacob
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Orthopedics, Tejasvini Hospital, Mangalore, India
| | - Kazunori Shimomura
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norimasa Nakamura
- Institute for Medical Science in Sports, Osaka Health Science University, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| |
Collapse
|
31
|
Wang E, Shi H, Sun Y, Politis C, Lan L, Chen X. Computer‐aided porous implant design for cranio‐maxillofacial defect restoration. Int J Med Robot 2020; 16:1-10. [DOI: 10.1002/rcs.2134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Enpeng Wang
- School of Mechanical Engineering Shanghai Jiao Tong University Shanghai China
| | - Haochen Shi
- School of Mechanical Engineering Shanghai Jiao Tong University Shanghai China
| | - Yi Sun
- Department of Oral and Maxillofacial Surgery/Faculty of Medicine KU Leuven University Hospitals Leuven, Campus Sint‐Rafaël and Department of Imaging Leuven Belgium
| | - Constantinus Politis
- Department of Oral and Maxillofacial Surgery/Faculty of Medicine KU Leuven University Hospitals Leuven, Campus Sint‐Rafaël and Department of Imaging Leuven Belgium
| | - Lin Lan
- Department of Oral and Maxillofacial Surgery Peking University School and Hospital of Stomatology Beijing China
| | - Xiaojun Chen
- School of Mechanical Engineering Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
32
|
Yan X, Chen YR, Song YF, Yang M, Ye J, Zhou G, Yu JK. Scaffold-Based Gene Therapeutics for Osteochondral Tissue Engineering. Front Pharmacol 2020; 10:1534. [PMID: 31992984 PMCID: PMC6970981 DOI: 10.3389/fphar.2019.01534] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022] Open
Abstract
Significant progress in osteochondral tissue engineering has been made for biomaterials designed to deliver growth factors that promote tissue regeneration. However, due to diffusion characteristics of hydrogels, the accurate delivery of signaling molecules remains a challenge. In comparison to the direct delivery of growth factors, gene therapy can overcome these challenges by allowing the simultaneous delivery of growth factors and transcription factors, thereby enhancing the multifactorial processes of tissue formation. Scaffold-based gene therapy provides a promising approach for tissue engineering through transfecting cells to enhance the sustained expression of the protein of interest or through silencing target genes associated with bone and joint disease. Reports of the efficacy of gene therapy to regenerate bone/cartilage tissue regeneration are widespread, but reviews on osteochondral tissue engineering using scaffold-based gene therapy are sparse. Herein, we review the recent advances in gene therapy with a focus on tissue engineering scaffolds for osteochondral regeneration.
Collapse
Affiliation(s)
- Xin Yan
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - You-Rong Chen
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Yi-Fan Song
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Meng Yang
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Jing Ye
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Gang Zhou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jia-Kuo Yu
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
33
|
Zhao Y, Ding X, Dong Y, Sun X, Wang L, Ma X, Zhu M, Xu B, Yang Q. Role of the Calcified Cartilage Layer of an Integrated Trilayered Silk Fibroin Scaffold Used to Regenerate Osteochondral Defects in Rabbit Knees. ACS Biomater Sci Eng 2020; 6:1208-1216. [PMID: 33464868 DOI: 10.1021/acsbiomaterials.9b01661] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The repair of osteochondral defects remains challenging, given the complexity of native osteochondral tissue and the limited self-repair capacity of cartilage. Osteochondral tissue engineering is a promising strategy. Here, we fabricated a biomimetic osteochondral scaffold using silk fibroin and hydroxyapatite, including a calcified cartilage layer (CCL). We studied the role played by the CCL in terms of cell viability in vivo. We established osteochondral defects in rabbit knees to investigate the effects of CCL-containing scaffolds with or without adipose tissue-derived stem cells (ADSCs). We evaluated osteochondral tissue regeneration by calculating gross observational scores, via histological and immunohistochemical assessments, by performing quantitative biochemical and biomechanical analyses of new osteochondral tissue, and via microcomputed tomography of new bone at 4, 8, and 12 weeks after surgery. In terms of surface roughness and integrity, the CCL + ADSCs group was better than the CCL and the non-CCL + ADSCs groups at all time points tested; the glycosaminoglycan and collagen type II levels of the CCL + ADSCs group were highest, reflecting the important role played by the CCL in cartilage tissue repair. Subchondral bone smoothness was better in the CCL + ADSCs group than in the non-CCL + ADSCs and CCL groups. The CCL promoted smooth subchondral bone regeneration but did not obviously affect bone strength or quality. In conclusion, a biomimetic osteochondral scaffold with a CCL, combined with autologous ADSCs, satisfactorily regenerated a rabbit osteochondral defect. The CCL enhances cartilage and subchondral bone regeneration.
Collapse
Affiliation(s)
- Yanhong Zhao
- Stomatological Hospital of Tianjin Medical University, 12 Qixiangtai Road, Heping District, Tianjin 300070, People's Republic of China
| | - Xiaoming Ding
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China.,Department of Orthopedics, Rizhao Traditional Chinese Medicine Hospital, 35 Haiwang Road, Donggang District, Rizhao, Shandong 276800, People's Republic of China
| | - Yunsheng Dong
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Lianyong Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Xinlong Ma
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Meifeng Zhu
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Baoshan Xu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| |
Collapse
|
34
|
Mellor LF, Nordberg RC, Huebner P, Mohiti-Asli M, Taylor MA, Efird W, Oxford JT, Spang JT, Shirwaiker RA, Loboa EG. Investigation of multiphasic 3D-bioplotted scaffolds for site-specific chondrogenic and osteogenic differentiation of human adipose-derived stem cells for osteochondral tissue engineering applications. J Biomed Mater Res B Appl Biomater 2019; 108:2017-2030. [PMID: 31880408 PMCID: PMC7217039 DOI: 10.1002/jbm.b.34542] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 08/20/2019] [Accepted: 11/29/2019] [Indexed: 12/16/2022]
Abstract
Osteoarthritis is a degenerative joint disease that limits mobility of the affected joint due to the degradation of articular cartilage and subchondral bone. The limited regenerative capacity of cartilage presents significant challenges when attempting to repair or reverse the effects of cartilage degradation. Tissue engineered medical products are a promising alternative to treat osteochondral degeneration due to their potential to integrate into the patient's existing tissue. The goal of this study was to create a scaffold that would induce site-specific osteogenic and chondrogenic differentiation of human adipose-derived stem cells (hASC) to generate a full osteochondral implant. Scaffolds were fabricated using 3D-bioplotting of biodegradable polycraprolactone (PCL) with either β-tricalcium phosphate (TCP) or decellularized bovine cartilage extracellular matrix (dECM) to drive site-specific hASC osteogenesis and chondrogenesis, respectively. PCL-dECM scaffolds demonstrated elevated matrix deposition and organization in scaffolds seeded with hASC as well as a reduction in collagen I gene expression. 3D-bioplotted PCL scaffolds with 20% TCP demonstrated elevated calcium deposition, endogenous alkaline phosphatase activity, and osteopontin gene expression. Osteochondral scaffolds comprised of hASC-seeded 3D-bioplotted PCL-TCP, electrospun PCL, and 3D-bioplotted PCL-dECM phases were evaluated and demonstrated site-specific osteochondral tissue characteristics. This technique holds great promise as cartilage morbidity is minimized since autologous cartilage harvest is not required, tissue rejection is minimized via use of an abundant and accessible source of autologous stem cells, and biofabrication techniques allow for a precise, customizable methodology to rapidly produce the scaffold.
Collapse
Affiliation(s)
- Liliana F Mellor
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina
| | - Rachel C Nordberg
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina.,Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri
| | - Pedro Huebner
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina
| | - Mahsa Mohiti-Asli
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina
| | - Michael A Taylor
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina
| | - William Efird
- Department of Orthopaedics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Julia T Oxford
- Biomolecular Research Center, Boise State University, Boise, Idaho
| | - Jeffrey T Spang
- Department of Orthopaedics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Rohan A Shirwaiker
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina.,Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina
| | - Elizabeth G Loboa
- Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri
| |
Collapse
|
35
|
Pérez-Silos V, Moncada-Saucedo NK, Peña-Martínez V, Lara-Arias J, Marino-Martínez IA, Camacho A, Romero-Díaz VJ, Lara Banda M, García-Ruiz A, Soto-Dominguez A, Rodriguez-Rocha H, López-Serna N, Tuan RS, Lin H, Fuentes-Mera L. A Cellularized Biphasic Implant Based on a Bioactive Silk Fibroin Promotes Integration and Tissue Organization during Osteochondral Defect Repair in a Porcine Model. Int J Mol Sci 2019; 20:E5145. [PMID: 31627374 PMCID: PMC6834127 DOI: 10.3390/ijms20205145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 01/16/2023] Open
Abstract
In cartilage tissue engineering, biphasic scaffolds (BSs) have been designed not only to influence the recapitulation of the osteochondral architecture but also to take advantage of the healing ability of bone, promoting the implant's integration with the surrounding tissue and then bone restoration and cartilage regeneration. This study reports the development and characterization of a BS based on the assembly of a cartilage phase constituted by fibroin biofunctionalyzed with a bovine cartilage matrix, cellularized with differentiated autologous pre-chondrocytes and well attached to a bone phase (decellularized bovine bone) to promote cartilage regeneration in a model of joint damage in pigs. BSs were assembled by fibroin crystallization with methanol, and the mechanical features and histological architectures were evaluated. The scaffolds were cellularized and matured for 12 days, then implanted into an osteochondral defect in a porcine model (n = 4). Three treatments were applied per knee: Group I, monophasic cellular scaffold (single chondral phase); group II (BS), cellularized only in the chondral phase; and in order to study the influence of the cellularization of the bone phase, Group III was cellularized in chondral phases and a bone phase, with autologous osteoblasts being included. After 8 weeks of surgery, the integration and regeneration tissues were analyzed via a histology and immunohistochemistry evaluation. The mechanical assessment showed that the acellular BSs reached a Young's modulus of 805.01 kPa, similar to native cartilage. In vitro biological studies revealed the chondroinductive ability of the BSs, evidenced by an increase in sulfated glycosaminoglycans and type II collagen, both secreted by the chondrocytes cultured on the scaffold during 28 days. No evidence of adverse or inflammatory reactions was observed in the in vivo trial; however, in Group I, the defects were not reconstructed. In Groups II and III, a good integration of the implant with the surrounding tissue was observed. Defects in group II were fulfilled via hyaline cartilage and normal bone. Group III defects showed fibrous repair tissue. In conclusion, our findings demonstrated the efficacy of a biphasic and bioactive scaffold based on silk fibroin and cellularized only in the chondral phase, which entwined chondroinductive features and a biomechanical capability with an appropriate integration with the surrounding tissue, representing a promising alternative for osteochondral tissue-engineering applications.
Collapse
Affiliation(s)
- Vanessa Pérez-Silos
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León. Madero y Dr. Aguirre Pequeño S/N, Mitras Centro, Monterrey 64460, Mexico.
| | - Nidia K Moncada-Saucedo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León. Madero y Dr. Aguirre Pequeño S/N, Mitras Centro, Monterrey 64460, Mexico.
| | - Víctor Peña-Martínez
- Universidad Autónoma de Nuevo León (UANL), Servicio de Ortopedia y Traumatología, Hospital Universitario "Dr. José E. González", Monterrey 64460, Mexico.
| | - Jorge Lara-Arias
- Universidad Autónoma de Nuevo León (UANL), Servicio de Ortopedia y Traumatología, Hospital Universitario "Dr. José E. González", Monterrey 64460, Mexico.
| | - Iván A Marino-Martínez
- Universidad Autónoma de Nuevo León (UANL), Unidad de Terapias Experimentales, Centro de Investigación y Desarrollo en Ciencias de la Salud, Monterrey 64460, Mexico.
- Universidad Autónoma de Nuevo León (UANL), Departamento de Patología, Facultad de Medicina, Monterrey 64460, Mexico.
| | - Alberto Camacho
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León. Madero y Dr. Aguirre Pequeño S/N, Mitras Centro, Monterrey 64460, Mexico.
- Universidad Autónoma de Nuevo León (UANL), Unidad de Neurometabolismo, Centro de Investigación y Desarrollo en Ciencias de la Salud, Monterrey 64460, Mexico.
| | - Víktor J Romero-Díaz
- Universidad Autónoma de Nuevo León (UANL), Departamento de Histología, Facultad de Medicina, UANL, Monterrey 64460, Mexico.
| | - María Lara Banda
- Universidad Autónoma de Nuevo León, Facultad de Ingeniería Mecánica y Eléctrica, Monterrey 66451, Mexico.
| | - Alejandro García-Ruiz
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León. Madero y Dr. Aguirre Pequeño S/N, Mitras Centro, Monterrey 64460, Mexico.
| | - Adolfo Soto-Dominguez
- Universidad Autónoma de Nuevo León (UANL), Departamento de Histología, Facultad de Medicina, UANL, Monterrey 64460, Mexico.
| | - Humberto Rodriguez-Rocha
- Universidad Autónoma de Nuevo León (UANL), Departamento de Histología, Facultad de Medicina, UANL, Monterrey 64460, Mexico.
| | - Norberto López-Serna
- Universidad Autónoma de Nuevo León (UANL), Departamento de Embriología, Facultad de Medicina, Monterrey 64460, Mexico.
| | - Rocky S Tuan
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219-3143, USA.
| | - Hang Lin
- Department of Orthopaedic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15260, USA.
| | - Lizeth Fuentes-Mera
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Nuevo León. Madero y Dr. Aguirre Pequeño S/N, Mitras Centro, Monterrey 64460, Mexico.
| |
Collapse
|
36
|
Lepage SIM, Robson N, Gilmore H, Davis O, Hooper A, St John S, Kamesan V, Gelis P, Carvajal D, Hurtig M, Koch TG. Beyond Cartilage Repair: The Role of the Osteochondral Unit in Joint Health and Disease. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:114-125. [PMID: 30638141 PMCID: PMC6486663 DOI: 10.1089/ten.teb.2018.0122] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Once believed to be limited to articular cartilage, osteoarthritis is now considered to be an organ disease of the “whole joint.” Damage to the articular surface can lead to, be caused by, or occur in parallel with, damage to other tissues in the joint. The relationship between cartilage and the underlying subchondral bone has particular importance when assessing joint health and determining treatment strategies. The articular cartilage is anchored to the subchondral bone through an interface of calcified cartilage, which as a whole makes up the osteochondral unit. This unit functions primarily by transferring load-bearing weight over the joint to allow for normal joint articulation and movement. Unfortunately, irreversible damage and degeneration of the osteochondral unit can severely limit joint function. Our understanding of joint pain, the primary complaint of patients, is poorly understood and past efforts toward structural cartilage restoration have often not been associated with a reduction in pain. Continued research focusing on the contribution of subchondral bone and restoration of the entire osteochondral unit are therefore needed, with the hope that this will lead to curative, and not merely palliative, treatment options. The purpose of this narrative review is to investigate the role of the osteochondral unit in joint health and disease. Topics of discussion include the crosstalk between cartilage and bone, the efficacy of diagnostic procedures, the origins of joint pain, current and emerging treatment paradigms, and suitable preclinical animal models for safety and efficacy assessment of novel osteochondral therapies. The goal of the review is to facilitate an appreciation of the important role played by the subchondral bone in joint pain and why the osteochondral unit as a whole should be considered in many cases of joint restoration strategies.
Collapse
Affiliation(s)
- Sarah I M Lepage
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Naomi Robson
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Hillary Gilmore
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Ola Davis
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Allyssa Hooper
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Stephanie St John
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Vashine Kamesan
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Paul Gelis
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Diana Carvajal
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Mark Hurtig
- 2 Department of Clinical Studies, University of Guelph, Guelph, Canada
| | - Thomas G Koch
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| |
Collapse
|
37
|
Deng C, Xu C, Zhou Q, Cheng Y. Advances of nanotechnology in osteochondral regeneration. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1576. [PMID: 31329375 DOI: 10.1002/wnan.1576] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/28/2022]
Abstract
In the past few decades, nanotechnology has proven to be one of the most powerful engineering strategies. The nanotechnologies for osteochondral tissue engineering aim to restore the anatomical structures and physiological functions of cartilage, subchondral bone, and osteochondral interface. As subchondral bone and articular cartilage have different anatomical structures and the physiological functions, complete healing of osteochondral defects remains a great challenge. Considering the limitation of articular cartilage to self-healing and the complexity of osteochondral tissue, osteochondral defects are in urgently need for new therapeutic strategies. This review article will concentrate on the most recent advancements of nanotechnologies, which facilitates chondrogenic and osteogenic differentiation for osteochondral regeneration. Moreover, this review will also discuss the current strategies and physiological challenges for the regeneration of osteochondral tissue. Specifically, we will summarize the latest developments of nanobased scaffolds for simultaneously regenerating subchondral bone and articular cartilage tissues. Additionally, perspectives of nanotechnology in osteochondral tissue engineering will be highlighted. This review article provides a comprehensive summary of the latest trends in cartilage and subchondral bone regeneration, paving the way for nanotechnologies in osteochondral tissue engineering. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Cuijun Deng
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Chang Xu
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Quan Zhou
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Yu Cheng
- Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Kosik-Kozioł A, Costantini M, Mróz A, Idaszek J, Heljak M, Jaroszewicz J, Kijeńska E, Szöke K, Frerker N, Barbetta A, Brinchmann JE, Święszkowski W. 3D bioprinted hydrogel model incorporating β-tricalcium phosphate for calcified cartilage tissue engineering. Biofabrication 2019; 11:035016. [PMID: 30943457 DOI: 10.1088/1758-5090/ab15cb] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
One promising strategy to reconstruct osteochondral defects relies on 3D bioprinted three-zonal structures comprised of hyaline cartilage, calcified cartilage, and subchondral bone. So far, several studies have pursued the regeneration of either hyaline cartilage or bone in vitro while-despite its key role in the osteochondral region-only few of them have targeted the calcified layer. In this work, we present a 3D biomimetic hydrogel scaffold containing β-tricalcium phosphate (TCP) for engineering calcified cartilage through a co-axial needle system implemented in extrusion-based bioprinting process. After a thorough bioink optimization, we showed that 0.5% w/v TCP is the optimal concentration forming stable scaffolds with high shape fidelity and endowed with biological properties relevant for the development of calcified cartilage. In particular, we investigate the effect induced by ceramic nano-particles over the differentiation capacity of bioprinted bone marrow-derived human mesenchymal stem cells in hydrogel scaffolds cultured up to 21 d in chondrogenic media. To confirm the potential of the presented approach to generate a functional in vitro model of calcified cartilage tissue, we evaluated quantitatively gene expression of relevant chondrogenic (COL1, COL2, COL10A1, ACAN) and osteogenic (ALPL, BGLAP) gene markers by means of RT-qPCR and qualitatively by means of fluorescence immunocytochemistry.
Collapse
Affiliation(s)
- Alicja Kosik-Kozioł
- Warsaw University of Technology, Faculty of Materials Science and Engineering, 02-507 Warsaw, Poland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Deng C, Chang J, Wu C. Bioactive scaffolds for osteochondral regeneration. J Orthop Translat 2019; 17:15-25. [PMID: 31194079 PMCID: PMC6551354 DOI: 10.1016/j.jot.2018.11.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/13/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022] Open
Abstract
Treatment for osteochondral defects remains a great challenge. Although several clinical strategies have been developed for management of osteochondral defects, the reconstruction of both cartilage and subchondral bone has proved to be difficult due to their different physiological structures and functions. Considering the restriction of cartilage to self-healing and the different biological properties in osteochondral tissue, new therapy strategies are essential to be developed. This review will focus on the latest developments of bioactive scaffolds, which facilitate the osteogenic and chondrogenic differentiation for the regeneration of bone and cartilage. Besides, the topic will also review the basic anatomy, strategies and challenges for osteochondral reconstruction, the selection of cells, biochemical factors and bioactive materials, as well as the design and preparation of bioactive scaffolds. Specifically, we summarize the most recent developments of single-type bioactive scaffolds for simultaneously regenerating cartilage and subchondral bone. Moreover, the future outlook of bioactive scaffolds in osteochondral tissue engineering will be discussed. This review offers a comprehensive summary of the most recent trend in osteochondral defect reconstruction, paving the way for the bioactive scaffolds in clinical therapy. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE This review summaries the latest developments of single-type bioactive scaffolds for regeneration of osteochondral defects. We also highlight a new possible translational direction for cartilage formation by harnessing bioactive ions and propose novel paradigms for subchondral bone regeneration in application of bioceramic scaffolds.
Collapse
Affiliation(s)
| | | | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Biomaterials and Tissue Engineering Research Center, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, PR China
| |
Collapse
|
40
|
Xiao H, Huang W, Xiong K, Ruan S, Yuan C, Mo G, Tian R, Zhou S, She R, Ye P, Liu B, Deng J. Osteochondral repair using scaffolds with gradient pore sizes constructed with silk fibroin, chitosan, and nano-hydroxyapatite. Int J Nanomedicine 2019; 14:2011-2027. [PMID: 30962685 PMCID: PMC6435123 DOI: 10.2147/ijn.s191627] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background One of the main problems associated with the development of osteochondral reparative materials is that the accurate imitation of the structure of the natural osteochondral tissue and fabrication of a suitable scaffold material for osteochondral repair are difficult. The long-term outcomes of single- or bilayered scaffolds are often unsatisfactory because of the absence of a progressive osteochondral structure. Therefore, only scaffolds with gradient pore sizes are suitable for osteochondral repair to achieve better proliferation and differentiation of the stem cells into osteochondral tissues to complete the repair of defects. Methods A silk fibroin (SF) solution, chitosan (CS) solution, and nano-hydroxyapatite (nHA) suspension were mixed at the same weight fraction to obtain osteochondral scaffolds with gradient pore diameters by centrifugation, freeze-drying, and chemical cross-linking. Results The scaffolds prepared in this study are confirmed to have a progressive structure starting from the cartilage layer to bone layer, similar to that of the normal osteochondral tissues. The prepared scaffolds are cylindrical in shape and have high internal porosity. The structure consists of regular and highly interconnected pores with a progressively increasing pore distribution as well as a progressively changing pore diameter. The scaffold strongly absorbs water, and has a suitable degradation rate, sufficient space for cell growth and proliferation, and good resistance to compression. Thus, the scaffold can provide sufficient nutrients and space for cell growth, proliferation, and migration. Further, bone marrow mesenchymal stem cells seeded onto the scaffold closely attach to the scaffold and stably grow and proliferate, indicating that the scaffold has good biocompatibility with no cytotoxicity. Conclusion In brief, the physical properties and biocompatibility of our scaffolds fully comply with the requirements of scaffold materials required for osteochondral tissue engineering, and they are expected to become a new type of scaffolds with gradient pore sizes for osteochondral repair.
Collapse
Affiliation(s)
- Hongli Xiao
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| | - Wenliang Huang
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| | - Kun Xiong
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| | - Shiqiang Ruan
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| | - Cheng Yuan
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| | - Gang Mo
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| | - Renyuan Tian
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| | - Sirui Zhou
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| | - Rongfeng She
- Department of Orthopedics, Guizhou Province People's Hospital, Guiyang 550002, Guizhou Province, People's Republic of China
| | - Peng Ye
- Emergency and Trauma Ward, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China
| | - Bin Liu
- Surgical Laboratory, Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China
| | - Jiang Deng
- Department of Orthopedics, Third Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou Province, People's Republic of China,
| |
Collapse
|
41
|
Stocco TD, Antonioli E, Elias CDMV, Rodrigues BVM, Siqueira IAWDB, Ferretti M, Marciano FR, Lobo AO. Cell Viability of Porous Poly(d,l-lactic acid)/Vertically Aligned Carbon Nanotubes/Nanohydroxyapatite Scaffolds for Osteochondral Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E849. [PMID: 30871217 PMCID: PMC6471978 DOI: 10.3390/ma12060849] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 02/07/2023]
Abstract
Treatment of articular cartilage lesions remains an important challenge. Frequently the bone located below the cartilage is also damaged, resulting in defects known as osteochondral lesions. Tissue engineering has emerged as a potential approach to treat cartilage and osteochondral defects. The principal challenge of osteochondral tissue engineering is to create a scaffold with potential to regenerate both cartilage and the subchondral bone involved, considering the intrinsic properties of each tissue. Recent nanocomposites based on the incorporation of nanoscale fillers into polymer matrix have shown promising results for the treatment of osteochondral defects. In this present study, it was performed using the recently developed methodologies (electrodeposition and immersion in simulated body fluid) to obtain porous superhydrophilic poly(d,l-lactic acid)/vertically aligned carbon nanotubes/nanohydroxyapatite (PDLLA/VACNT-O:nHAp) nanocomposite scaffolds, to analyze cell behavior and gene expression of chondrocytes, and then assess the applicability of this nanobiomaterial for osteochondral regenerative medicine. The results demonstrate that PDLLA/VACNT-O:nHAp nanocomposite supports chondrocytes adhesion and decreases type I Collagen mRNA expression. Therefore, these findings suggest the possibility of novel nanobiomaterial as a scaffold for osteochondral tissue engineering applications.
Collapse
Affiliation(s)
- Thiago Domingues Stocco
- Faculty of Medical Sciences, State University of Campinas, São Paulo 13083-887, Brazil.
- Faculty of Physiotherapy, University of Santo Amaro, São Paulo 04829-300, Brazil.
| | - Eliane Antonioli
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil.
| | | | | | | | - Mario Ferretti
- Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil.
| | | | - Anderson Oliveira Lobo
- LIMAV-Interdisciplinary Laboratory for Advanced Materials, UFPI-Federal University of Piauí, Teresina 64049-550, Piauí, Brazil.
| |
Collapse
|
42
|
Daley EL, Kuttig J, Stegemann JP. Development of Modular, Dual-Perfused Osteochondral Constructs for Cartilage Repair. Tissue Eng Part C Methods 2019; 25:127-136. [PMID: 30724134 PMCID: PMC6457327 DOI: 10.1089/ten.tec.2018.0356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/25/2019] [Indexed: 11/13/2022] Open
Abstract
IMPACT STATEMENT This study describes methods for fabricating, culturing, and characterizing modular microbeads containing progenitor cells that can be used to create osteochondral tissue constructs. Such biphasic engineered tissues were cultured in a low flow rate perfusion bioreactor chamber to maintain tissue-specific differentiation while allowing development of the osteochondral interface.
Collapse
Affiliation(s)
- Ethan L.H. Daley
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Jochen Kuttig
- Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
43
|
Drobnic M, Perdisa F, Kon E, Cefalì F, Marcacci M, Filardo G. Implant strategy affects scaffold stability and integrity in cartilage treatment. Knee Surg Sports Traumatol Arthrosc 2018; 26:2774-2783. [PMID: 29022056 DOI: 10.1007/s00167-017-4737-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 09/28/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE To identify the most appropriate implantation strategy for a novel chondral scaffold in a model simulating the early post-operative phase, in order to optimize the implant procedure and reduce the risk of early failure. METHODS Eight human cadaveric limbs were strapped to a continuous passive motion device and exposed to extension-flexion cycles (0°-90°). Chondral lesions (1.8 cm diameter) were prepared on condyles, patella and trochlea for the implant of a bi-layer collagen-hydroxyapatite scaffold. The first set-up compared four fixation techniques: press-fit (PF) vs. fibrin glue (FG) vs. pins vs. sutures; the second compared circular and square implants; the third investigated stability in a weight-bearing simulation. The scaffolds were evaluated using semi-quantitative Drobnic and modified Bekkers scores. RESULTS FG presented higher total Drobnic and Bekkers scores compared to PF (both p = 0.002), pins (p = 0.013 and 0.001) and sutures (p = 0.001 and < 0.0005). Pins offered better total Drobnic and Bekkers scores than PF in the anterior femoral condyles (p = 0.007 and 0.065), similar to FG. The comparison of round and square implants applied by FG showed worst results for square lesions (Drobnic score p = 0.049, Bekkers score p = 0.037). Finally, load caused worst overall results (Drobnic p = 0.018). CONCLUSIONS FG improves the fixation of this collagen-HA scaffold regardless of lesion location, improving implant stability while preserving its integrity. Pins represent a suitable option only for lesions of the anterior condyles. Square scaffolds present weak corners, therefore, round implants should be preferred. Finally, partial weight-bearing simulation significantly affected the scaffold. These findings may be useful to improve surgical technique and post-operative management of patients, to optimize the outcome of chondral scaffold implantation.
Collapse
Affiliation(s)
- M Drobnic
- Orthopaedic Clinic, Medical Faculty, University of Ljubjana, Ljubljana, Slovenia
| | - Francesco Perdisa
- Nano-Biotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, Bologna, Italy. .,II Orthopaedic Clinic, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136, Bologna, Italy.
| | - E Kon
- Humanitas University, Department of Biomedical Science, Rozzano (Milan), Italy
| | - F Cefalì
- Finceramica S.p.A., Faenza, Italy
| | - M Marcacci
- Nano-Biotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, Bologna, Italy.,Humanitas University, Department of Biomedical Science, Rozzano (Milan), Italy
| | - G Filardo
- Nano-Biotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, Bologna, Italy
| |
Collapse
|
44
|
Controlled Non-Viral Gene Delivery in Cartilage and Bone Repair: Current Strategies and Future Directions. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
45
|
Tamaddon M, Liu C. Enhancing Biological and Biomechanical Fixation of Osteochondral Scaffold: A Grand Challenge. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:255-298. [PMID: 29736578 DOI: 10.1007/978-3-319-76735-2_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease, typified by degradation of cartilage and changes in the subchondral bone, resulting in pain, stiffness and reduced mobility. Current surgical treatments often fail to regenerate hyaline cartilage and result in the formation of fibrocartilage. Tissue engineering approaches have emerged for the repair of cartilage defects and damages to the subchondral bones in the early stage of OA and have shown potential in restoring the joint's function. In this approach, the use of three-dimensional scaffolds (with or without cells) provides support for tissue growth. Commercially available osteochondral (OC) scaffolds have been studied in OA patients for repair and regeneration of OC defects. However, some controversial results are often reported from both clinical trials and animal studies. The objective of this chapter is to report the scaffolds clinical requirements and performance of the currently available OC scaffolds that have been investigated both in animal studies and in clinical trials. The findings have demonstrated the importance of biological and biomechanical fixation of the OC scaffolds in achieving good cartilage fill and improved hyaline cartilage formation. It is concluded that improving cartilage fill, enhancing its integration with host tissues and achieving a strong and stable subchondral bone support for overlying cartilage are still grand challenges for the early treatment of OA.
Collapse
Affiliation(s)
- Maryam Tamaddon
- Institute of Orthopaedics & Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, Stanmore, UK
| | - Chaozong Liu
- Institute of Orthopaedics & Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, Stanmore, UK.
| |
Collapse
|
46
|
Chen L, Deng C, Li J, Yao Q, Chang J, Wang L, Wu C. 3D printing of a lithium-calcium-silicate crystal bioscaffold with dual bioactivities for osteochondral interface reconstruction. Biomaterials 2018; 196:138-150. [PMID: 29643002 DOI: 10.1016/j.biomaterials.2018.04.005] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/21/2018] [Accepted: 04/02/2018] [Indexed: 01/21/2023]
Abstract
It is difficult to achieve self-healing outcoming for the osteochondral defects caused by degenerative diseases. The simultaneous regeneration of both cartilage and subchondral bone tissues is an effective therapeutic strategy for osteochondral defects. However, it is challenging to design a single type of bioscaffold with suitable ionic components and beneficial osteo/chondral-stimulation ability for regeneration of osteochondral defects. In this study, we successfully synthesized a pure-phase lithium calcium silicate (Li2Ca4Si4O13, L2C4S4) bioceramic by a sol-gel method, and further prepared L2C4S4 scaffolds by using a 3D-printing method. The compressive strength of L2C4S4 scaffolds could be well controlled in the range of 15-40 MPa when pore size varied from 170 to 400 μm. L2C4S4 scaffolds have been demonstrated to possess controlled biodegradability and good apatite-mineralization ability. At a certain concentration range, the ionic products from L2C4S4 significantly stimulated the proliferation and maturation of chondrocytes, as well as promoted the osteogenic differentiation of rBMSCs. L2C4S4 scaffolds simultaneously promoted the regeneration of both cartilage and subchondral bone as compared to pure β-TCP scaffolds in rabbit osteochondral defects. These findings suggest that 3D-printed L2C4S4 scaffolds with such specific ionic combination, high mechanical strength and good degradability as well as dual bioactivities, represent a promising biomaterial for osteochondral interface reconstruction.
Collapse
Affiliation(s)
- Lei Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Cuijun Deng
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jiayi Li
- Department of Orthopaedic Surgery, Digital Medicine Institute, Nanjing Medical University Nanjing Hospital, No. 68 Changle Road, Nanjing, 210006, PR China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Digital Medicine Institute, Nanjing Medical University Nanjing Hospital, No. 68 Changle Road, Nanjing, 210006, PR China.
| | - Jiang Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China.
| | - Liming Wang
- Department of Orthopaedic Surgery, Digital Medicine Institute, Nanjing Medical University Nanjing Hospital, No. 68 Changle Road, Nanjing, 210006, PR China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China.
| |
Collapse
|
47
|
Abstract
Background: Collagens of marine origin are applied increasingly as alternatives to mammalian collagens in tissue engineering. The aim of the present study was to develop a biphasic scaffold from exclusively marine collagens supporting both osteogenic and chondrogenic differentiation and to find a suitable setup for in vitro chondrogenic and osteogenic differentiation of human mesenchymal stroma cells (hMSC). Methods: Biphasic scaffolds from biomimetically mineralized salmon collagen and fibrillized jellyfish collagen were fabricated by joint freeze-drying and crosslinking. Different experiments were performed to analyze the influence of cell density and TGF-β on osteogenic differentiation of the cells in the scaffolds. Gene expression analysis and analysis of cartilage extracellular matrix components were performed and activity of alkaline phosphatase was determined. Furthermore, histological sections of differentiated cells in the biphasic scaffolds were analyzed. Results: Stable biphasic scaffolds from two different marine collagens were prepared. An in vitro setup for osteochondral differentiation was developed involving (1) different seeding densities in the phases; (2) additional application of alginate hydrogel in the chondral part; (3) pre-differentiation and sequential seeding of the scaffolds and (4) osteochondral medium. Spatially separated osteogenic and chondrogenic differentiation of hMSC was achieved in this setup, while osteochondral medium in combination with the biphasic scaffolds alone was not sufficient to reach this ambition. Conclusions: Biphasic, but monolithic scaffolds from exclusively marine collagens are suitable for the development of osteochondral constructs.
Collapse
Affiliation(s)
- Anne Bernhardt
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Birgit Paul
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| |
Collapse
|
48
|
Deng C, Zhu H, Li J, Feng C, Yao Q, Wang L, Chang J, Wu C. Bioactive Scaffolds for Regeneration of Cartilage and Subchondral Bone Interface. Theranostics 2018; 8:1940-1955. [PMID: 29556366 PMCID: PMC5858510 DOI: 10.7150/thno.23674] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 01/17/2018] [Indexed: 01/15/2023] Open
Abstract
The cartilage lesion resulting from osteoarthritis (OA) always extends into subchondral bone. It is of great importance for simultaneous regeneration of two tissues of cartilage and subchondral bone. 3D-printed Sr5(PO4)2SiO4 (SPS) bioactive ceramic scaffolds may achieve the aim of regenerating both of cartilage and subchondral bone. We hypothesized that strontium (Sr) and silicon (Si) ions released from SPS scaffolds play a crucial role in osteochondral defect reconstruction. Methods: SPS bioactive ceramic scaffolds were fabricated by a 3D-printing method. The SEM and ICPAES were used to investigate the physicochemical properties of SPS scaffolds. The proliferation and maturation of rabbit chondrocytes stimulated by SPS bioactive ceramics were measured in vitro. The stimulatory effect of SPS scaffolds for cartilage and subchondral bone regeneration was investigated in vivo. Results: SPS scaffolds significantly stimulated chondrocyte proliferation, and SPS extracts distinctly enhanced the maturation of chondrocytes and preserved chondrocytes from OA. SPS scaffolds markedly promoted the regeneration of osteochondral defects. The complex interface microstructure between cartilage and subchondral bone was obviously reconstructed. The underlying mechanism may be related to Sr and Si ions stimulating cartilage regeneration by activating HIF pathway and promoting subchondral bone reconstruction through activating Wnt pathway, as well as preserving chondrocytes from OA via inducing autophagy and inhibiting hedgehog pathway. Conclusion: Our findings suggest that SPS scaffolds can help osteochondral defect reconstruction and well reconstruct the complex interface between cartilage and subchondral bone, which represents a promising strategy for osteochondral defect regeneration.
Collapse
|
49
|
Shimomura K, Ando W, Fujie H, Hart DA, Yoshikawa H, Nakamura N. Scaffold-free tissue engineering for injured joint surface restoration. J Exp Orthop 2018; 5:2. [PMID: 29330730 PMCID: PMC5768574 DOI: 10.1186/s40634-017-0118-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/20/2017] [Indexed: 12/31/2022] Open
Abstract
Articular cartilage does not heal spontaneously due to its limited healing capacity, and thus effective treatments for cartilage injuries has remained challenging. Since the first report by Brittberg et al. in 1994, autologous chondrocyte implantation (ACI) has been introduced into the clinic. Recently, as an alternative for chondrocyte-based therapy, mesenchymal stem cell (MSC)-based therapy has received considerable research attention because of the relative ease in handling for tissue harvest, and subsequent cell expansion and differentiation. In this review, we discuss the latest developments regarding stem cell-based therapies for cartilage repair, with special focus on recent scaffold-free approaches.
Collapse
Affiliation(s)
- Kazunori Shimomura
- Medicine for Sports and Performing Arts, Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan.,Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Wataru Ando
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Hiromichi Fujie
- Division of Human Mechatronics Systems, Faculty of System Design, Tokyo Metropolitan University, 6-6 Asahigaoka, Hino City, Tokyo, 191-0065, Japan
| | - David A Hart
- McCaig Institute for Bone & Joint Health, University of Calgary, 3330 Hospital Drive Northwest, Calgary, AB, T2N 4N1, Canada
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Norimasa Nakamura
- Institute for Medical Science in Sports, Osaka Health Science University, 1-9-27 Tenma, Kita-ku, Osaka City, Osaka, 530-0043, Japan. .,Center for Advanced Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan.
| |
Collapse
|
50
|
Kon E, Filardo G, Brittberg M, Busacca M, Condello V, Engebretsen L, Marlovits S, Niemeyer P, Platzer P, Posthumus M, Verdonk P, Verdonk R, Victor J, van der Merwe W, Widuchowski W, Zorzi C, Marcacci M. A multilayer biomaterial for osteochondral regeneration shows superiority vs microfractures for the treatment of osteochondral lesions in a multicentre randomized trial at 2 years. Knee Surg Sports Traumatol Arthrosc 2018; 26:2704-2715. [PMID: 28913600 PMCID: PMC6105149 DOI: 10.1007/s00167-017-4707-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/04/2017] [Indexed: 12/03/2022]
Abstract
PURPOSE The increasing awareness on the role of subchondral bone in the etiopathology of articular surface lesions led to the development of osteochondral scaffolds. While safety and promising results have been suggested, there are no trials proving the real potential of the osteochondral regenerative approach. Aim was to assess the benefit provided by a nanostructured collagen-hydroxyapatite (coll-HA) multilayer scaffold for the treatment of chondral and osteochondral knee lesions. METHODS In this multicentre randomized controlled clinical trial, 100 patients affected by symptomatic chondral and osteochondral lesions were treated and evaluated for up to 2 years (51 study group and 49 control group). A biomimetic coll-HA scaffold was studied, and bone marrow stimulation (BMS) was used as reference intervention. Primary efficacy measurement was IKDC subjective score at 2 years. Secondary efficacy measurements were: KOOS, IKDC Knee Examination Form, Tegner and VAS Pain scores evaluated at 6, 12 and 24 months. Tissue regeneration was evaluated with MRI MOCART scoring system at 6, 12 and 24 months. An external independent agency was involved to ensure data correctness and objectiveness. RESULTS A statistically significant improvement of all clinical scores was obtained from basal evaluation to 2-year follow-up in both groups, although no overall statistically significant differences were detected between the two treatments. Conversely, the subgroup of patients affected by deep osteochondral lesions (i.e. Outerbridge grade IV and OCD) showed a statistically significant better IKDC subjective outcome (+12.4 points, p = 0.036) in the coll-HA group. Statistically significant better results were also found for another challenging group: sport active patients (+16.0, p = 0.027). Severe adverse events related to treatment were documented only in three patients in the coll-HA group and in one in the BMS group. The MOCART score showed no statistical difference between the two groups. CONCLUSIONS This study highlighted the safety and potential of a biomimetic implant. While no statistically significant differences were found compared to BMS for chondral lesions, this procedure can be considered a suitable option for the treatment of osteochondral lesions. LEVEL OF EVIDENCE I.
Collapse
Affiliation(s)
- Elizaveta Kon
- Humanitas University Department of Biomedical Sciences - Humanitas Clinical and Research Center, Milan, Italy
| | - Giuseppe Filardo
- NABI Laboratory, Rizzoli Orthopaedic Institute, Via Di Barbiano 1/10, 40136, Bologna, Italy.
| | - Mats Brittberg
- Department of Orthopaedics, Cartilaginous research unit, Goteborg University, Kungsbacka Hospital, Kungsbacka, Sweden
| | | | - Vincenzo Condello
- Dipartimento di Ortopedia, Ospedale Sacro Cuore Don Calabria di Negrar, Verona, Italy
| | - Lars Engebretsen
- Department of orthopaedic surgery, Ullevål Hospital, Oslo University, Oslo, Norway
| | | | - Philipp Niemeyer
- Department of orthopaedic surgery and traumatology, Freiburg University Hospital, Freiburg Im Breisgau, Germany
| | - Patrik Platzer
- Department of traumatology, Medical University of Vienna, Vienna, Austria
| | - Michael Posthumus
- Division of Exercise Science and Sports Medicine, Faculty of Health Sciences, The University of Cape Town, Cape Town, South Africa
| | - Peter Verdonk
- Antwerp Orthopaedic Center, Monica Hospitals, Stevenslei, Deurne, Belgium
| | - Renè Verdonk
- Université Libre de Bruxelles, Brussels, Belgium
| | - Jan Victor
- Department of orthopaedic surgery, Ghent University Hospital, Ghent, Belgium
| | - Willem van der Merwe
- Sport Science Orthopaedic Clinic, Sport Science Institute of South Africa Newlands, Cape Town, South Africa
| | - Wojciech Widuchowski
- Wojewódzki Szpital Chirurgii Urazowej, II Oddział Urazowo-Ortopedyczny, Piekary Śląskie, Polen
| | - Claudio Zorzi
- Dipartimento di Ortopedia, Ospedale Sacro Cuore Don Calabria di Negrar, Verona, Italy
| | - Maurilio Marcacci
- Humanitas University Department of Biomedical Sciences - Humanitas Clinical and Research Center, Milan, Italy
| |
Collapse
|