1
|
Wu Y, Jia Z, Sun K, Zhou G, Tao K. A multi-gradient organoid of articular cartilage with bionic matrix microenvironment. Biomaterials 2025; 322:123393. [PMID: 40339197 DOI: 10.1016/j.biomaterials.2025.123393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/26/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Reconstructing the zonal organization of articular cartilage, including the heterogeneity in matrix distribution and chondrocyte status, remains a significant challenge. In this study, we developed a compression technique to engineer artificial cartilage architecture. By controlling the orientation of fibers within a collagen hydrogel, we obtained a gradient from parallel alignment in the surface layer to random distribution in deeper layers. Simultaneously, we established a diverse concentration gradient of chondroitin sulfate to mimic cartilage composition. Encapsulating chondrocytes within this construct yielded a "cartilage organoid." In vitro culture demonstrated that the plastic compression achieved an increased density, parallel alignment, and a flattened morphology of cells in the surface layer. Especially, type II collagen and superficial zone protein (SZP), which are crucial for the functional durability of articular cartilage, were specifically excreted by the regulated cells within the surface region. Subcutaneous implantation of the cartilage organoid confirmed the stable retention of these specific features of the organoid in vivo, accompanied by further tissue maturation. Following implantation into articular cartilage defects, successful regeneration of well-integrated cartilage tissue with region-specific characteristics was achieved. These findings suggest a biomimetic cartilage organoid fully mimicking the factors in the structure and composition of natural cartilages, which may be a promising candidate for cartilage reconstruction and functional regeneration.
Collapse
Affiliation(s)
- Yongjie Wu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Zenghui Jia
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200011, PR China; Research Institute of Plastic Surgery, Shandong Second Medical University, Weifang, Shandong, 261053, PR China
| | - Kang Sun
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200011, PR China; Research Institute of Plastic Surgery, Shandong Second Medical University, Weifang, Shandong, 261053, PR China.
| | - Ke Tao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| |
Collapse
|
2
|
Tong L, Shi G, Liu Q, Qian Z, Li J, Zhang K, Zhu Y, Fang Y, Sha L, Bai L, Li Y, Wang X, Ma Y, Jirigala E, Wang H, Li X. Fabrication and evaluation of 3D printed PLGA/nHA/GO scaffold for bone tissue engineering. Sci Rep 2025; 15:12446. [PMID: 40216829 PMCID: PMC11992093 DOI: 10.1038/s41598-025-96099-z] [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: 01/26/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
The study aimed to fabricate and evaluate a bone tissue engineering scaffold made from a composite of polylactic-co-glycolic acid (PLGA), nano-hydroxyapatite (nHA), and graphene oxide (GO) using low-temperature 3D printing and freeze-drying techniques. The scaffolds were produced with varying compositions: PLGA alone and in combination with nHA and GO. The macro and microstructure, pore size, porosity, mechanical properties, and in vitro biocompatibility were assessed. Bone marrow mesenchymal stem cells (BMSCs) were co-cultured with the scaffolds to evaluate cell adhesion, proliferation, and cytotoxicity. The PLGA/nHA/GO composite scaffolds exhibited optimal pore size and microtopography, enhanced mechanical properties, excellent water absorption, and appropriate degradability. The co-culture with BMSCs demonstrated improved cell adhesion and proliferation, indicating good biocompatibility. The PLGA/nHA/GO composite scaffolds show potential as a bone tissue engineering material due to their favorable properties and biocompatibility, suggesting their suitability for bone defect repair applications.
Collapse
Affiliation(s)
- Ling Tong
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China
| | - Guopeng Shi
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China
| | - Qinghua Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China
| | - Zhiyong Qian
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China
| | - Jing Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Kai Zhang
- Department of Orthopaedics, the Second Hospital of Ulanqab City, Ulanqab, 012000, Inner Mongolia, China
| | - Yong Zhu
- Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Inner Mongolia Cancer Center, Hohhot, 010110, Inner Mongolia, China
| | - Yuan Fang
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China
| | - Lirong Sha
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China
| | - Lin Bai
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China
| | - Yumo Li
- School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China
| | - Xing Wang
- Digital Medical Center, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China
| | - Yuan Ma
- Digital Medical Center, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China
| | - Enhe Jirigala
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China.
| | - Haiyan Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China.
| | - Xiaohe Li
- Department of Human Anatomy, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia, China.
| |
Collapse
|
3
|
Zheng Z, Yu D, Wang H, Wu H, Tang Z, Wu Q, Cao P, Chen Z, Huang H, Li X, Liu C, Guo Z. Advancement of 3D biofabrication in repairing and regeneration of cartilage defects. Biofabrication 2025; 17:022003. [PMID: 39793203 DOI: 10.1088/1758-5090/ada8e1] [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: 08/13/2024] [Accepted: 01/10/2025] [Indexed: 01/13/2025]
Abstract
Three-dimensional (3D) bioprinting, an additive manufacturing technology, fabricates biomimetic tissues that possess natural structure and function. It involves precise deposition of bioinks, including cells, and bioactive factors, on basis of computer-aided 3D models. Articular cartilage injuries, a common orthopedic issue. Current repair methods, for instance microfracture procedure (MF), autologous chondrocyte implantation (ACI), and osteochondral autologous transfer surgery have been applied in clinical practice. However, each procedure has inherent limitation. For instance, MF surgery associates with increased subchondral cyst formation and brittle subchondral bone. ACI procedure involves two surgeries, and associate with potential risks infection and delamination of the regenerated cartilage. In addition, chondrocyte implantation's efficacy depends on the patient's weight, joint pathology, gender-related histological changes of cartilage, and hormonal influences that affect treatment and prognosis. So far, it is a still a grand challenge for achieving a clinical satisfactory in repairing and regeneration of cartilage defects using conditional strategies. 3D biofabrication provide a potential to fabricate biomimetic articular cartilage construct that has shown promise in specific cartilage repair and regeneration of patients. This review reported the techniques of 3D bioprinting applied for cartilage repair, and analyzed their respective merits and demerits, and limitations in clinical application. A summary of commonly used bioinks has been provided, along with an outlook on the challenges and prospects faced by 3D bioprinting in the application of cartilage tissue repair. It provided an overall review of current development and promising application of 3D biofabrication technology in articular cartilage repair.
Collapse
Affiliation(s)
- Zenghui Zheng
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
- School of Clinical Medicine, Xi'an Medical University, Xi 'an 710021, People's Republic of China
| | - Dongmei Yu
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
- Institute of Orthopaedics and Musculoskeletal Science, University College London, The Royal National Orthopaedic Hospital, Stanmore HA7 4LP, United Kingdom
| | - Haoyu Wang
- Institute of Orthopaedics and Musculoskeletal Science, University College London, The Royal National Orthopaedic Hospital, Stanmore HA7 4LP, United Kingdom
| | - Hao Wu
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
| | - Zhen Tang
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
| | - Qi Wu
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
| | - Pengfei Cao
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
- School of Clinical Medicine, Xi'an Medical University, Xi 'an 710021, People's Republic of China
| | - Zhiyuan Chen
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
- School of Clinical Medicine, Xi'an Medical University, Xi 'an 710021, People's Republic of China
| | - Hai Huang
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
| | - Xiaokang Li
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
| | - Chaozong Liu
- Institute of Orthopaedics and Musculoskeletal Science, University College London, The Royal National Orthopaedic Hospital, Stanmore HA7 4LP, United Kingdom
| | - Zheng Guo
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, People's Republic of China
| |
Collapse
|
4
|
Fan H, Xue B, Lu J, Sun T, Zhao Q, Liu Y, Niu M, Yu S, Yang Y, Zhang L. Recent advances of bioaerogels in medicine: Preparation, property and application. Int J Biol Macromol 2025; 291:139144. [PMID: 39722377 DOI: 10.1016/j.ijbiomac.2024.139144] [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: 08/29/2024] [Revised: 12/14/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Bioaerogels represent a type of three-dimensional porous materials fabricated from natural biopolymers, and show a significant potential for medical application due to their characteristics of extremely low density, high specific surface area, excellent biocompatibility and biodegradability. The preparation method and parameters of bioaerogels are focused on, and their influence on the structure and properties of bioaerogels are discussed in detail. Then, to match the properties of bioaerogels with the medical applications, this work emphasizes the main properties (including biocompatibility, degradability, and mechanical properties), structural parameters (such as suitable porosity, pore size and high specific surface area), and further summarizes the influence of single-component and composite bioaerogels on their properties. Moreover, according to the different applications (wound healing, drug delivery, and tissue engineering and other fields), the function method, mechanism and practical effect of bioaerogels are comprehensively analyzed. Finally, the challenges, future research directions, and solutions for the practical application of bioaerogels in medicine are discussed.
Collapse
Affiliation(s)
- Haoyong Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Baoxia Xue
- College of Textile Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jiaxin Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Tao Sun
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Qinke Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yong Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Mei Niu
- College of Textile Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Shiping Yu
- Department of Interventional Therapy, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China
| | - Yongzhen Yang
- Key Laboratory of Interface Science and Engineering in Advanced Materials, Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China.
| | - Li Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China.
| |
Collapse
|
5
|
Metzler NF, Kondo M, Matsukura K, Ford AJ, Grainger DW, Okano T. Differentiated and Untreated Juvenile Chondrocyte Sheets Regenerate Cartilage Similarly In Vivo. Tissue Eng Part A 2025; 31:184-194. [PMID: 39556329 DOI: 10.1089/ten.tea.2024.0208] [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] [Indexed: 11/19/2024] Open
Abstract
Osteoarthritis, a degenerative disease of articular cartilage and the leading cause of disability, is preceded by acute cartilage injury in a significant proportion of cases. Current auto- and allograft interventions are limited by supply and variability in therapeutic efficacy, prompting interest in tissue engineering solutions. Cell sheet tissue engineering, a scaffold-free regenerative technique, has shown promise in preclinical and clinical trials across various cell types and diseases. Polydactyly-derived juvenile cartilage-derived chondrocyte (JCC) sheets from juvenile patients are a potent cell source for developing allogeneic therapies. JCC sheets have proven safe and effective in animal models and as an add-on therapy in a recent clinical cartilage repair study. However, JCC ex vivo expansion leads to de-differentiation, contributing to long healing times. This study hypothesized that in vitro differentiation of JCC sheets into hyaline-like cartilage constructs could accelerate cartilage regeneration without compromising implant integration. To this end, sheet integration, maturation, and healing of conventionally prepared vs. differentiated JCC sheets were compared in an established nude rat focal chondral defect model. Differentiated JCC sheets exhibit mature cartilage phenotypes prior to transplant. Both conventional and differentiated JCC sheets are reliably transplanted without additional fixation. Histological evaluation reveals that both transplant groups produced equivalent neocartilage regeneration, filling defects with mature hyaline cartilage at 2- and 4-weeks post-transplant. Notably, differentiated JCC sheets respond to in vivo signals, undergoing matrix remodeling and integration with adjacent and subchondral tissue. Given equivalent healing outcomes, the future utility of in vitro JCC sheet predifferentiation from other JCC donors with different healing capacities should be balanced against their increased culture costs over conventional sheets.
Collapse
Affiliation(s)
- Nicolás F Metzler
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Makoto Kondo
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
| | - Keisuke Matsukura
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Orthopaedic Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Adam J Ford
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
| | - David W Grainger
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Teruo Okano
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
6
|
Toktarov T, Raimagambetov Y, Balbossynov B, Saginova D, Abilmazhinov M, Ogay V. Implantation of Heparin-Conjugated Fibrin Hydrogel for Local Defects of Cartilage in Knee Osteoarthritis: A Case Report. Int Med Case Rep J 2025; 18:151-156. [PMID: 39871859 PMCID: PMC11769846 DOI: 10.2147/imcrj.s483485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/08/2025] [Indexed: 01/29/2025] Open
Abstract
Background Cartilage defects in the knee joint are areas of damage and wear to the cartilage that normally covers and protects the ends of bones. These defects occur due to sudden injuries, such as trauma or sports accidents, or due to chronic conditions, such as osteoarthritis. Cartilage acts as a shock absorber (cushion absorber), reducing the impact of mechanical stress on the joints, which helps prevent bone damage during movement. Cartilage also serves as a gliding surface for the joints, allowing them to move smoothly, which minimizes friction between the bones. Its damage can cause pain, swelling, and decreased joint function. Treatment of localized cartilage defects is important to prevent further damage to the joint and maintain good knee function. Identifying problems early and treating them correctly can help improve outcomes and reduce the likelihood of more serious joint problems. Case Description We describe the case of a 46-year-old man with a localized cartilage defect in the knee joint who was followed for one year after the application of heparin-conjugated fibrin hydrogel for the treatment. We watched the patient for a year, doing functional tests, checking MRI results after the procedure, and watching for side effects. Results This case demonstrates that implantation of hydrogel successfully engraft and lead to remodeling of hyaline-like cartilage, thereby improving the condition of damaged knee cartilage. Comparison of MRI images before and 1 year after surgery showed the effectiveness of this technology.
Collapse
Affiliation(s)
- Tusipkhan Toktarov
- National Scientific Center of Traumatology and Orthopedics Named After Academician Batpenov N.D., Astana, Kazakhstan
- Astana Medical University, Astana, Kazakhstan
| | - Yerik Raimagambetov
- National Scientific Center of Traumatology and Orthopedics Named After Academician Batpenov N.D., Astana, Kazakhstan
| | - Bagdat Balbossynov
- National Scientific Center of Traumatology and Orthopedics Named After Academician Batpenov N.D., Astana, Kazakhstan
| | - Dina Saginova
- National Scientific Center of Traumatology and Orthopedics Named After Academician Batpenov N.D., Astana, Kazakhstan
| | | | - Vyacheslav Ogay
- National Scientific Center of Traumatology and Orthopedics Named After Academician Batpenov N.D., Astana, Kazakhstan
- Stem Cell Laboratory, National Center for Biotechnology, Astana, Kazakhstan
| |
Collapse
|
7
|
Parasuraman G, Amirtham SM, Francis DV, Livingston A, Ramasamy B, Sathishkumar S, Vinod E. Evaluation of Chondral Defect Repair Using Human Fibronectin Adhesion Assay-Derived Chondroprogenitors Suspended in Lyophilized Fetal Collagen Scaffold: An Ex Vivo Osteochondral Unit Model Study. Indian J Orthop 2024; 58:991-1000. [PMID: 39087036 PMCID: PMC11286923 DOI: 10.1007/s43465-024-01192-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/22/2024] [Indexed: 08/02/2024]
Abstract
Introduction Chondral defect repair is challenging due to a scarcity of reparative cells and the need to fill a large surface area, compounded by the absence of self-healing mechanisms. Fibronectin adhesion assay-derived chondroprogenitors (FAA-CPs) have emerged as a promising alternative with enhanced chondrogenic ability and reduced hypertrophy. De-cellularized bio-scaffolds are reported to act as extracellular matrix, mimicking the structural and functional characteristics of native tissue, thereby facilitating cell attachment and differentiation. This study primarily assessed the synergistic effect of FAA-CPs suspended in fetal cartilage-derived collagen-containing scaffolds in repairing chondral defects. Methodology The de-cellularized and lyophilized fetal collagen was prepared from the tibio-femoral joint of a 36 + 4-week gestational age fetus. FAA-CPs were isolated from osteoarthritic cartilage samples (n = 3) and characterized. In ex vivo analysis, FAA-CPs at a density of 1 × 106 cells were suspended in the lyophilized scaffold and placed into the chondral defects created in the Osteochondral Units and harvested on the 35th day for histological examination. Results The lyophilized scaffold of de-cellularized fetal cartilage with FAA-CPs demonstrated effective healing of the critical size chondral defect. This was evidenced by a uniform distribution of cells, a well-organized collagen-fibrillar network, complete filling of the defect with alignment to the surface, and favorable integration with the adjacent cartilage. However, these effects were less pronounced in the plain scaffold control group and no demonstrable repair observed in the empty defect group. Conclusion This study suggests the synergistic potential of FAA-CPs and collagen scaffold for chondral repair which needs to be further explored for clinical therapy. Supplementary Information The online version contains supplementary material available at 10.1007/s43465-024-01192-6.
Collapse
Affiliation(s)
- Ganesh Parasuraman
- Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India
| | - Soosai Manickam Amirtham
- Department of Physiology/Centre for Stem Cell Research, Christian Medical College, Vellore, India
| | | | - Abel Livingston
- Department of Orthopaedics, Christian Medical College, Vellore, India
| | - Boopalan Ramasamy
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, Australia
| | - Solomon Sathishkumar
- Department of Physiology/Centre for Stem Cell Research, Christian Medical College, Vellore, India
| | - Elizabeth Vinod
- Centre for Stem Cell Research, (A Unit of InStem, Bengaluru), Christian Medical College, Vellore, India
- Department of Physiology/Centre for Stem Cell Research, Christian Medical College, Vellore, India
| |
Collapse
|
8
|
Eldeen GN, Elkhooly TA, El Bassyouni GT, Hamdy TM, Hawash AR, Aly RM. Enhancement of the chondrogenic differentiation capacity of human dental pulp stem cells via chondroitin sulfate-coated polycaprolactone-MWCNT nanofibers. Sci Rep 2024; 14:16396. [PMID: 39013921 PMCID: PMC11252133 DOI: 10.1038/s41598-024-66497-w] [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: 04/13/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024] Open
Abstract
Most of the conditions involving cartilaginous tissues are irreversible and involve degenerative processes. The aim of the present study was to fabricate a biocompatible fibrous and film scaffolds using electrospinning and casting techniques to induce chondrogenic differentiation for possible application in cartilaginous tissue regeneration. Polycaprolactone (PCL) electrospun nanofibrous scaffolds and PCL film were fabricated and incorporated with multi-walled carbon nanotubes (MWCNTs). Thereafter, coating of chondroitin sulfate (CS) on the fibrous and film structures was applied to promote chondrogenic differentiation of human dental pulp stem cells (hDPSCs). First, the morphology, hydrophilicity and mechanical properties of the scaffolds were characterized by scanning electron microscopy (SEM), spectroscopic characterization, water contact angle measurements and tensile strength testing. Subsequently, the effects of the fabricated scaffolds on stimulating the proliferation of human dental pulp stem cells (hDPSCs) and inducing their chondrogenic differentiation were evaluated via electron microscopy, flow cytometry and RT‒PCR. The results of the study demonstrated that the different forms of the fabricated PCL-MWCNTs scaffolds analyzed demonstrated biocompatibility. The nanofilm structures demonstrated a higher rate of cellular proliferation, while the nanofibrous architecture of the scaffolds supported the cellular attachment and differentiation capacity of hDPSCs and was further enhanced with CS addition. In conclusion, the results of the present investigation highlighted the significance of this combination of parameters on the viability, proliferation and chondrogenic differentiation capacity of hDPSCs seeded on PCL-MWCNT scaffolds. This approach may be applied when designing PCL-based scaffolds for future cell-based therapeutic approaches developed for chondrogenic diseases.
Collapse
Affiliation(s)
- Ghada Nour Eldeen
- Human Genetics and Genome Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Tarek A Elkhooly
- Refractories, Ceramics, and Building Materials Department, National Research Centre, Dokki, Giza, 12622, Egypt
- Nanomedicine Research Unit, Faculty of Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Gehan T El Bassyouni
- Refractories, Ceramics, and Building Materials Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Tamer M Hamdy
- Restorative and Dental Materials Department, Oral and Dental Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Ahmed R Hawash
- Faculty of Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Riham M Aly
- Basic Dental Science Department, Oral and Dental Research Institute, National Research Centre, 33 El Bohouth St., Dokki, Giza, 12622, Egypt.
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, 12622, Egypt.
| |
Collapse
|
9
|
Velasquillo C, Melgarejo-Ramírez Y, García-López J, Gutiérrez-Gómez C, Lecona H, González-Torres M, Sánchez-Betancourt JI, Ibarra C, Lee SJ, Yoo JJ. Remaining microtia tissue as a source for 3D bioprinted elastic cartilage tissue constructs, potential use for surgical microtia reconstruction. Cell Tissue Bank 2024; 25:571-582. [PMID: 38038782 DOI: 10.1007/s10561-023-10118-9] [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: 05/29/2022] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
The absence of ears in children is a global problem. An implant made of costal cartilage is the standard procedure for ear reconstruction; however, side effects such as pneumothorax, loss of thoracic cage shape, and respiratory complications have been documented. Three-dimensional (3D) printing allows the generation of biocompatible scaffolds that mimic the shape, mechanical strength, and architecture of the native extracellular matrix necessary to promote new elastic cartilage formation. We report the potential use of a 3D-bioprinted poly-ε-caprolactone (3D-PCL) auricle-shaped framework seeded with remaining human microtia chondrocytes for the development of elastic cartilage for autologous microtia ear reconstruction. An in vivo assay of the neo-tissue formed revealed the generation of a 3D pinna-shaped neo-tissue, and confirmed the formation of elastic cartilage by the presence of type II collagen and elastin with histological features and a protein composition consistent with normal elastic cartilage. According to our results, a combination of 3D-PCL auricle frameworks and autologous microtia remnant tissue generates a suitable pinna structure for autologous ear reconstruction.
Collapse
Affiliation(s)
- Cristina Velasquillo
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación LGII, Mexico City, Mexico.
| | - Yaaziel Melgarejo-Ramírez
- Laboratorio de Biotecnología, Unidad de Gerociencias, Instituto Nacional de Rehabilitación LGII, Mexico City, Mexico.
| | - Julieta García-López
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación LGII, Mexico City, Mexico
| | - Claudia Gutiérrez-Gómez
- División de Cirugía Plástica y Reconstructiva, Hospital General Dr. Manuel Gea González, Mexico City, Mexico
| | - Hugo Lecona
- Bioterio y Cirugía Experimental, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | - Maykel González-Torres
- Laboratorio de Biotecnología, Unidad de Gerociencias, Instituto Nacional de Rehabilitación LGII, Mexico City, Mexico
| | - José Iván Sánchez-Betancourt
- Departamento de Producción Animal. Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Clemente Ibarra
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación LGII, Mexico City, Mexico
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
10
|
Wei B, Xu Y, Tang C, Liu NQ, Li X, Yao Q, Wang L. An injectable active hydrogel based on BMSC-derived extracellular matrix for cartilage regeneration enhancement. BIOMATERIALS ADVANCES 2024; 160:213857. [PMID: 38657287 DOI: 10.1016/j.bioadv.2024.213857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/20/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
Articular cartilage injury impairs joint function and necessitates orthopedic intervention to restore the structure and function of the cartilage. Extracellular matrix (ECM) scaffolds derived from bone marrow mesenchymal stem cells (BMSCs) can effectively promote cell adhesion, proliferation, and chondrogenesis. However, pre-shaped ECM scaffolds have limited applicability due to their poor fit with the irregular surface of most articular cartilage defects. In this study, we fabricated an injectable active ECM hydrogel from autologous BMSCs-derived ECM by freeze-drying, liquid nitrogen milling, and enzymatic digestion. Moreover, our in vitro and in vivo results demonstrated that the prepared hydrogel enhanced chondrocyte adhesion and proliferation, chondrogenesis, cartilage regeneration, and integration with host tissue, respectively. These findings indicate that active ECM components can provide trophic support for cell proliferation and differentiation, restoring the structure and function of damaged cartilage.
Collapse
Affiliation(s)
- Bo Wei
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| | - Yan Xu
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Cheng Tang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Nancy Q Liu
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA 90007, USA
| | - Xuxiang Li
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Liming Wang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| |
Collapse
|
11
|
Hoveidaei AH, Sadat-Shojai M, Mosalamiaghili S, Salarikia SR, Roghani-Shahraki H, Ghaderpanah R, Ersi MH, Conway JD. Nano-hydroxyapatite structures for bone regenerative medicine: Cell-material interaction. Bone 2024; 179:116956. [PMID: 37951520 DOI: 10.1016/j.bone.2023.116956] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
Bone tissue engineering holds great promise for the regeneration of damaged or severe bone defects. However, several challenges hinder its translation into clinical practice. To address these challenges, interdisciplinary efforts and advances in biomaterials, cell biology, and bioengineering are required. In recent years, nano-hydroxyapatite (nHA)-based scaffolds have emerged as a promising approach for the development of bone regenerative agents. The unique similarity of nHA with minerals found in natural bones promotes remineralization and stimulates bone growth, which are crucial factors for efficient bone regeneration. Moreover, nHA exhibits desirable properties, such as strong chemical interactions with bone and facilitation of tissue growth, without inducing inflammation or toxicity. It also promotes osteoblast survival, adhesion, and proliferation, as well as increasing alkaline phosphatase activity, osteogenic differentiation, and bone-specific gene expression. However, it is important to note that the effect of nHA on osteoblast behavior is dose-dependent, with cytotoxic effects observed at higher doses. Additionally, the particle size of nHA plays a crucial role, with smaller particles having a more significant impact. Therefore, in this review, we highlighted the potential of nHA for improving bone regeneration processes and summarized the available data on bone cell response to nHA-based scaffolds. In addition, an attempt is made to portray the current status of bone tissue engineering using nHA/polymer hybrids and some recent scientific research in the field.
Collapse
Affiliation(s)
- Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Mehdi Sadat-Shojai
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran
| | - Seyedarad Mosalamiaghili
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Rezvan Ghaderpanah
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hamed Ersi
- Evidence Based Medicine Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Clinical Research Development Center of Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Janet D Conway
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA.
| |
Collapse
|
12
|
Zhu J, Zhang S, Jin S, Huang C, Shi B, Chen Z, Ji W. Endochondral Repair of Jawbone Defects Using Periosteal Cell Spheroids. J Dent Res 2024; 103:31-41. [PMID: 37968792 DOI: 10.1177/00220345231205273] [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] [Indexed: 11/17/2023] Open
Abstract
Recapitulation of the natural healing process is receiving increasing recognition as a strategy to induce robust tissue regeneration. Endochondral ossification has been recognized as an essential reparative approach in natural jawbone defect healing. However, such an approach has been overlooked in the recent development of cell-based therapeutics for jawbone repair. Therefore, this study aimed to explore a bioinspired stem cell-based strategy for jawbone repair by mimicking the mesenchymal condensation of progenitor cells during the early endochondral ossification process. For this purpose, passage 3 of jawbone periosteum-derived cells (jb-PDCs) was cultured in our previously reported nonadherent microwells (200 µm in diameter, 148 µm in depth, and 100 µm space in between) and self-assembled into spheroids with a diameter of 96.4 ± 5.8 µm after 48 h. Compared to monolayer culture, the jb-PDC spheroids showed a significant reduction of stemness marker expression evidenced by flow cytometry. Furthermore, a significant upregulation of chondrogenic transcription factor SOX9 in both gene and protein levels was observed in the jb-PDC spheroids after 48 h of chondrogenic induction. RNA sequencing and Western blotting analysis further suggested that the enhanced SOX9-mediated chondrogenic differentiation in jb-PDC spheroids was attributed to the activation of the p38 MAPK pathway. Impressively, inhibition of p38 kinase activity significantly attenuated chondrogenic differentiation jb-PDC spheroids, evidenced by a significant decline of SOX9 in both gene and protein levels. Strikingly, the jb-PDC spheroids implanted in 6- to 8-wk-old male C57BL/6 mice with critical-size jawbone defects (1.8 mm in diameter) showed an evident contribution to cartilaginous callus formation after 1 wk, evidenced by histological analysis. Furthermore, micro-computed tomography analysis showed that the jb-PDC spheroids significantly accelerated bone healing after 2 wk in the absence of exogenous growth factors. In sum, the presented findings represent the successful development of cell-based therapeutics to reengineer the endochondral bone repair process and illustrate the potential application to improve bone repair and regeneration in the craniofacial skeleton.
Collapse
Affiliation(s)
- J Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - S Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - S Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - C Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - B Shi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
- Department of Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Z Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - W Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
- Department of Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
13
|
Huang YH, Chen HA, Chen CH, Liao HT, Kuo CY, Chen JP. Injectable gelatin/glucosamine cryogel microbeads as scaffolds for chondrocyte delivery in cartilage tissue engineering. Int J Biol Macromol 2023; 253:126528. [PMID: 37633562 DOI: 10.1016/j.ijbiomac.2023.126528] [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: 07/05/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
In this study, we fabricate squeezable cryogel microbeads as injectable scaffolds for minimum invasive delivery of chondrocytes for cartilage tissue engineering applications. The microbeads with different glucosamine concentrations were prepared by combining the water-in-oil emulsion and cryogelation through crosslinking of gelatin with glutaraldehyde in the presence of glucosamine. The physicochemical characterization results show the successful preparation of cryogel microbeads with uniform shape and size, high porosity, large pore size, high water uptake capacity, and good injectability. In vitro analysis indicates proliferation, migration, and differentiated phenotype of rabbit chondrocytes in the cryogel scaffolds. The seeded chondrocytes in the cryogel scaffold can be delivered by injecting through an 18G needle to fully retain the cell viability. Furthermore, the incorporation of glucosamine in the cryogel promoted the differentiated phenotype of chondrocytes in a dose-dependent manner, from cartilage-specific gene expression and protein production. The in vivo study by injecting the cryogel microbeads into the subcutaneous pockets of nude mice indicates good retention ability as well as good biocompatibility and suitable biodegradability of the cryogel scaffold. Furthermore, the injected chondrocyte/cryogel microbead constructs can form ectopic functional neocartilage tissues following subcutaneous implantation in 21 days, as evidenced by histological and immunohistochemical analysis.
Collapse
Affiliation(s)
- Yen-Hsiang Huang
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Huai-An Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Chih-Hao Chen
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Keelung, Keelung 20401, Taiwan; Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan
| | - Han-Tsung Liao
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan
| | - Chang-Yi Kuo
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan; Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33305, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
14
|
Wang XH, Liu N, Zhang H, Yin ZS, Zha ZG. From cells to organs: progress and potential in cartilaginous organoids research. J Transl Med 2023; 21:926. [PMID: 38129833 PMCID: PMC10740223 DOI: 10.1186/s12967-023-04591-9] [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: 08/10/2023] [Accepted: 10/04/2023] [Indexed: 12/23/2023] Open
Abstract
While cartilage tissue engineering has significantly improved the speed and quality of cartilage regeneration, the underlying metabolic mechanisms are complex, making research in this area lengthy and challenging. In the past decade, organoids have evolved rapidly as valuable research tools. Methods to create these advanced human cell models range from simple tissue culture techniques to complex bioengineering approaches. Cartilaginous organoids in part mimic the microphysiology of human cartilage and fill a gap in high-fidelity cartilage disease models to a certain extent. They hold great promise to elucidate the pathogenic mechanism of a diversity of cartilage diseases and prove crucial in the development of new drugs. This review will focus on the research progress of cartilaginous organoids and propose strategies for cartilaginous organoid construction, study directions, and future perspectives.
Collapse
Affiliation(s)
- Xiao-He Wang
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Ning Liu
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Hui Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Zong-Sheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Zhen-Gang Zha
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
15
|
Nogoceke R, Josino R, Robert AW, Stimamiglio MA. Evaluation of a Peptide Hydrogel as a Chondro-Instructive Three-Dimensional Microenvironment. Polymers (Basel) 2023; 15:4630. [PMID: 38139882 PMCID: PMC10747086 DOI: 10.3390/polym15244630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Articular cartilage injuries are inherently irreversible, even with the advancement in current therapeutic options. Alternative approaches, such as the use of mesenchymal stem/stromal cells (MSCs) and tissue engineering techniques, have gained prominence. MSCs represent an ideal source of cells due to their low immunogenicity, paracrine activity, and ability to differentiate. Among biomaterials, self-assembling peptide hydrogels (SAPH) are interesting given their characteristics such as good biocompatibility and tunable properties. Herein we associate human adipose-derived stem cells (hASCs) with a commercial SAPH, Puramatrix™, to evaluate how this three-dimensional microenvironment affects cell behavior and its ability to undergo chondrogenic differentiation. We demonstrate that the Puramatrix™ hydrogel comprises a highly porous matrix permissible for hASC adhesion and in vitro expansion. The morphology and cell growth dynamics of hASCs were affected when cultured on the hydrogel but had minimal alteration in their immunophenotype. Interestingly, hASCs spontaneously formed cell aggregates throughout culturing. Analysis of glycosaminoglycan production and gene expression revealed a noteworthy and donor-dependent trend suggesting that Puramatrix™ hydrogel may have a natural capacity to support the chondrogenic differentiation of hASCs. Altogether, the results provide a more comprehensive understanding of the potential applications and limitations of the Puramatrix™ hydrogel in developing functional cartilage tissue constructs.
Collapse
Affiliation(s)
| | | | - Anny Waloski Robert
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas—ICC-FIOCRUZ/PR, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba 81350-010, Brazil; (R.N.); (R.J.)
| | - Marco Augusto Stimamiglio
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas—ICC-FIOCRUZ/PR, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba 81350-010, Brazil; (R.N.); (R.J.)
| |
Collapse
|
16
|
El-Wakil N, Kamel R, Mahmoud AA, Dufresne A, Abouzeid RE, Abo El-Fadl M, Maged A. Risedronate-loaded aerogel scaffolds for bone regeneration. Drug Deliv 2023; 30:51-63. [PMID: 36474425 PMCID: PMC9937015 DOI: 10.1080/10717544.2022.2152135] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sugarcane bagasse-derived nanofibrillated cellulose (NFC), a type of cellulose with a fibrous structure, is potentially used in the pharmaceutical field. Regeneration of this cellulose using a green process offers a more accessible and less ordered cellulose II structure (amorphous cellulose; AmC). Furthermore, the preparation of cross-linked cellulose (NFC/AmC) provides a dual advantage by building a structural block that could exhibit distinct mechanical properties. 3D aerogel scaffolds loaded with risedronate were prepared in our study using NFC or cross-linked cellulose (NFC/AmC), then combined with different concentrations of chitosan. Results proved that the aerogel scaffolds composed of NFC and chitosan had significantly improved the mechanical properties and retarded drug release compared to all other fabricated aerogel scaffolds. The aerogel scaffolds containing the highest concentration of chitosan (SC-T3) attained the highest compressive strength and mean release time values (415 ± 41.80 kPa and 2.61 ± 0.23 h, respectively). Scanning electron microscope images proved the uniform highly porous microstructure of SC-T3 with interconnectedness. All the tested medicated as well as unmedicated aerogel scaffolds had the ability to regenerate bone as assessed using the MG-63 cell line, with the former attaining a higher effect than the latter. However, SC-T3 aerogel scaffolds possessed a lower regenerative effect than those composed of NFC only. This study highlights the promising approach of the use of biopolymers derived from agro-wastes for tissue engineering.
Collapse
Affiliation(s)
- Nahla El-Wakil
- Cellulose and Paper Department, National Research Centre, Giza, Egypt
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre, Giza, Egypt
| | - Azza A. Mahmoud
- Pharmaceutics and Pharmaceutical Technology Department, Faculty of Pharmacy, Future University in Egypt, New Cairo, Egypt,CONTACT Azza A. Mahmoud Pharmaceutics and Pharmaceutical Technology Department, Faculty of Pharmacy, Future University in Egypt, New Cairo, Egypt
| | - Alain Dufresne
- CNRS, Grenoble INP, LGP2, Université Grenoble Alpes, Grenoble, France
| | - Ragab E. Abouzeid
- Cellulose and Paper Department, National Research Centre, Giza, Egypt
| | - Mahmoud T. Abo El-Fadl
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Giza, Egypt,Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | - Amr Maged
- Pharmaceutics and Pharmaceutical Technology Department, Faculty of Pharmacy, Future University in Egypt, New Cairo, Egypt,Pharmaceutical Factory, Faculty of Pharmacy, Future University in Egypt, New Cairo, Egypt
| |
Collapse
|
17
|
Ma C, Wang T, Jin X, Zhang W, Lv Q. Lineage-specific multifunctional double-layer scaffold accelerates the integrated regeneration of cartilage and subchondral bone. Mater Today Bio 2023; 23:100800. [PMID: 37766897 PMCID: PMC10520449 DOI: 10.1016/j.mtbio.2023.100800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/04/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Repairing cartilage/subchondral bone defects that involve subchondral bone is a major challenge in clinical practice. Overall, the integrated repair of the structure and function of the osteochondral (OC) unit is very important. Some studies have demonstrated that the differentiation of cartilage is significantly enhanced by reducing the intake of nutrients such as lipids. This study demonstrates that using starvation can effectively optimize the therapeutic effect of bone marrow mesenchymal stem cells (BMSCs)-derived extracellular vesicles (EVs). A hyaluronic acid (HA)-based hydrogel containing starved BMSCs-EVs displayed continuous release for more than 3 weeks and significantly promoted the proliferation and biosynthesis of chondrocytes around the defect regulated by the forkhead-box class O (FOXO) pathway. When combined with vascular inhibitors, the hydrogel inhibited cartilage hypertrophy and facilitated the regeneration of hyaline cartilage. A porous methacrylate gelatine (GelMA)-based hydrogel containing calcium salt loaded with thrombin rapidly promoted haematoma formation upon contact with the bone marrow cavity to quickly block the pores and prevent the blood vessels in the bone marrow cavity from invading the cartilage layer. Furthermore, the haematoma could be used as nutrients to accelerate bone survival. The in vivo experiments demonstrated that the multifunctional lineage-specific hydrogel promoted the integrated regeneration of cartilage/subchondral bone. Thus, this hydrogel may represent a new strategy for osteochondral regeneration and repair.
Collapse
Affiliation(s)
- Chunhui Ma
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Tao Wang
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Xinmeng Jin
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Wanglin Zhang
- Department of Orthopaedics, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Lv
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
18
|
Huang Y, Zhao H, Wang Y, Bi S, Zhou K, Li H, Zhou C, Wang Y, Wu W, Peng B, Tang J, Pan B, Wang B, Chen Z, Li Z, Zhang Z. The application and progress of tissue engineering and biomaterial scaffolds for total auricular reconstruction in microtia. Front Bioeng Biotechnol 2023; 11:1089031. [PMID: 37811379 PMCID: PMC10556751 DOI: 10.3389/fbioe.2023.1089031] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/21/2023] [Indexed: 10/10/2023] Open
Abstract
Microtia is a congenital deformity of the ear with an incidence of about 0.8-4.2 per 10,000 births. Total auricular reconstruction is the preferred treatment of microtia at present, and one of the core technologies is the preparation of cartilage scaffolds. Autologous costal cartilage is recognized as the best material source for constructing scaffold platforms. However, costal cartilage harvest can lead to donor-site injuries such as pneumothorax, postoperative pain, chest wall scar and deformity. Therefore, with the need of alternative to autologous cartilage, in vitro and in vivo studies of biomaterial scaffolds and cartilage tissue engineering have gradually become novel research hot points in auricular reconstruction research. Tissue-engineered cartilage possesses obvious advantages including non-rejection, minimally invasive or non-invasive, the potential of large-scale production to ensure sufficient donors and controllable morphology. Exploration and advancements of tissue-engineered cartilaginous framework are also emerging in aspects including three-dimensional biomaterial scaffolds, acquisition of seed cells and chondrocytes, 3D printing techniques, inducing factors for chondrogenesis and so on, which has greatly promoted the research process of biomaterial substitute. This review discussed the development, current application and research progress of cartilage tissue engineering in auricular reconstruction, particularly the usage and creation of biomaterial scaffolds. The development and selection of various types of seed cells and inducing factors to stimulate chondrogenic differentiation in auricular cartilage were also highlighted. There are still confronted challenges before the clinical application becomes widely available for patients, and its long-term effect remains to be evaluated. We hope to provide guidance for future research directions of biomaterials as an alternative to autologous cartilage in ear reconstruction, and finally benefit the transformation and clinical application of cartilage tissue engineering and biomaterials in microtia treatment.
Collapse
Affiliation(s)
- Yeqian Huang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hanxing Zhao
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Yixi Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Siwei Bi
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Zhou
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Hairui Li
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Yudong Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqing Wu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Peng
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Jun Tang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Bo Pan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baoyun Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhixing Chen
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhenyu Zhang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Chen CH, Kao HH, Lee YC, Chen JP. Injectable Thermosensitive Hyaluronic Acid Hydrogels for Chondrocyte Delivery in Cartilage Tissue Engineering. Pharmaceuticals (Basel) 2023; 16:1293. [PMID: 37765101 PMCID: PMC10535600 DOI: 10.3390/ph16091293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
In this study, we synthesize a hyaluronic acid-g-poly(N-isopropylacrylamide) (HPN) copolymer by grafting the amine-terminated poly(N-isopropylacrylamide) (PNIPAM-NH2) to hyaluronic acid (HA). The 5% PNIPAM-NH2 and HPN polymer solution is responsive to temperature changes with sol-to-gel phase transition temperatures around 32 °C. Compared with the PNIPAM-NH2 hydrogel, the HPN hydrogel shows higher water content and mechanical strength, as well as lower volume contraction, making it a better choice as a scaffold for chondrocyte delivery. From an in vitro cell culture, we see that cells can proliferate in an HPN hydrogel with full retention of cell viability and show the phenotypic morphology of chondrocytes. In the HPN hydrogel, chondrocytes demonstrate a differentiated phenotype with the upregulated expression of cartilage-specific genes and the enhanced secretion of extracellular matrix components, when compared with the monolayer culture on tissue culture polystyrene. In vivo studies confirm the ectopic cartilage formation when HPN was used as a cell delivery vehicle after implanting chondrocyte/HPN in nude mice subcutaneously, which is shown from a histological and gene expression analysis. Taken together, the HPN thermosensitive hydrogel will be a promising injectable scaffold with which to deliver chondrocytes in cartilage-tissue-engineering applications.
Collapse
Affiliation(s)
- Chih-Hao Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Keelung, Chang Gung University College of Medicine, Keelung 20401, Taiwan
| | - Hao-Hsi Kao
- Division of Nephrology, Chang Gung Memorial Hospital at Keelung, Chang Gung University College of Medicine, Keelung 20401, Taiwan
| | - Yen-Chen Lee
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-San, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
20
|
Sanapalli BKR, Yele V, Singh MK, Thumbooru SN, Parvathaneni M, Karri VVSR. Human beta defensin-2 loaded PLGA nanoparticles impregnated in collagen-chitosan composite scaffold for the management of diabetic wounds. Biomed Pharmacother 2023; 161:114540. [PMID: 36934557 DOI: 10.1016/j.biopha.2023.114540] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/26/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Diabetic wound (DW) is the most devastating complication resulting in significant mortality and morbidity in diabetic patients. The standard treatment of DW care fails to address the prerequisites of treating DW owing to its multifactorial pathophysiology. Henceforth, developing a single treatment strategy to handle all the loopholes may effectively manage DW. The objective of the current study was to formulate Human beta defensin-2 (HBD-2) loaded Poly (lactic-co-glycolic acid) (PLGA) nanoparticle impregnated in collagen/chitosan (COL-CS) composite scaffolds for the accelerated healing of DW. Upon investigation, the developed biodegradable crosslinked scaffold possesses low matrix degradation, optimum porosity, and sustained drug release than the non-crosslinked scaffold. In vitro studies revealed that the HBD-2 COL-CS scaffold was biocompatible and accelerated cell migration and angiogenesis. The HBD-2 COL-CS scaffold showed significant antimicrobial activity in S. aureus, E. coli, and P. aeruginosa. The in vivo studies revealed that the HBD-2 COL-CS treated group accelerated healing compared to those in COL-CS and control groups. The ELISA results indicated a significant decrease in MMP-9, TNF-α, MPO, NAG, and NO with an increase in IL-10 in HBD-2 COL-CS treated group. The accelerated healing in HBD-2 COL-CS treated group might be due to the synergistic effects of PLGA (collagen synthesis and deposition and positive angiogenic effect), HBD-2 (anti-inflammatory, antibacterial, positive angiogenic effect, cell proliferation, and migration), COL (established wound healer and stabilizer) and CS (antibacterial, controlled drug release).
Collapse
Affiliation(s)
- Bharat Kumar Reddy Sanapalli
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu 643001, India.
| | - Vidyasrilekha Yele
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu 643001, India.
| | - Mantosh Kumar Singh
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu 643001, India.
| | - Shilpa N Thumbooru
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu 643001, India.
| | - Madhukiran Parvathaneni
- Department of Biotechnology, Harrisburg University of Science & Technology, 326 Market Street, Harrisburg, PA 17101, USA; Arni Medica, 4475 South Clinton Ave, Suite 230, South Plainfield, NJ 07080, USA; CRC Pharma LLC, 333 Littleton Road, Parsippany, NJ 07054, USA.
| | | |
Collapse
|
21
|
Nie K, Zhou S, Li H, Tian J, Shen W, Huang W. Advanced silk materials for musculoskeletal tissue regeneration. Front Bioeng Biotechnol 2023; 11:1199507. [PMID: 37200844 PMCID: PMC10185897 DOI: 10.3389/fbioe.2023.1199507] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/19/2023] [Indexed: 05/20/2023] Open
Abstract
Musculoskeletal diseases are the leading causes of chronic pain and physical disability, affecting millions of individuals worldwide. Over the past two decades, significant progress has been made in the field of bone and cartilage tissue engineering to combat the limitations of conventional treatments. Among various materials used in musculoskeletal tissue regeneration, silk biomaterials exhibit unique mechanical robustness, versatility, favorable biocompatibility, and tunable biodegradation rate. As silk is an easy-to-process biopolymer, silks have been reformed into various materials formats using advanced bio-fabrication technology for the design of cell niches. Silk proteins also offer active sites for chemical modifications to facilitate musculoskeletal system regeneration. With the emergence of genetic engineering techniques, silk proteins have been further optimized from the molecular level with other functional motifs to introduce new advantageous biological properties. In this review, we highlight the frontiers in engineering natural and recombinant silk biomaterials, as well as recent progress in the applications of these new silks in the field of bone and cartilage regeneration. The future potentials and challenges of silk biomaterials in musculoskeletal tissue engineering are also discussed. This review brings together perspectives from different fields and provides insight into improved musculoskeletal engineering.
Collapse
Affiliation(s)
- Kexin Nie
- Centre for Regeneration and Cell Therapy, The Zhejiang University—University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Sicheng Zhou
- Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Hu Li
- Centre for Regeneration and Cell Therapy, The Zhejiang University—University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingyi Tian
- Centre for Regeneration and Cell Therapy, The Zhejiang University—University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Weiliang Shen
- Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenwen Huang
- Centre for Regeneration and Cell Therapy, The Zhejiang University—University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Wenwen Huang,
| |
Collapse
|
22
|
Dehghan-Baniani D, Mehrjou B, Chu PK, Lee WYW, Wu H. Recent Advances in "Functional Engineering of Articular Cartilage Zones by Polymeric Biomaterials Mediated with Physical, Mechanical, and Biological/Chemical Cues". Adv Healthc Mater 2022; 12:e2202581. [PMID: 36571465 DOI: 10.1002/adhm.202202581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/19/2022] [Indexed: 12/27/2022]
Abstract
Articular cartilage (AC) plays an unquestionable role in joint movements but unfortunately the healing capacity is restricted due to its avascular and acellular nature. While cartilage tissue engineering has been lifesaving, it is very challenging to remodel the complex cartilage composition and architecture with gradient physio-mechanical properties vital to proper tissue functions. To address these issues, a better understanding of the intrinsic AC properties and how cells respond to stimuli from the external microenvironment must be better understood. This is essential in order to take one step closer to producing functional cartilaginous constructs for clinical use. Recently, biopolymers have aroused much attention due to their versatility, processability, and flexibility because the properties can be tailored to match the requirements of AC. This review highlights polymeric scaffolds developed in the past decade for reconstruction of zonal AC layers including the superficial zone, middle zone, and deep zone by means of exogenous stimuli such as physical, mechanical, and biological/chemical signals. The mimicked properties are reviewed in terms of the biochemical composition and organization, cell fate (morphology, orientation, and differentiation), as well as mechanical properties and finally, the challenges and potential ways to tackle them are discussed.
Collapse
Affiliation(s)
- Dorsa Dehghan-Baniani
- Department of Chemical and Biological Engineering Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.,Musculoskeletal Research Laboratory, SH Ho Scoliosis Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Babak Mehrjou
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Wayne Yuk Wai Lee
- Musculoskeletal Research Laboratory, SH Ho Scoliosis Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China.,Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong SAR, China.,Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong SAR, China
| | - Hongkai Wu
- Department of Chemical and Biological Engineering Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.,Department of Chemistry and the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong SAR, China
| |
Collapse
|
23
|
Regeneration of Osteochondral Defects by Combined Delivery of Synovium-Derived Mesenchymal Stem Cells, TGF-β1 and BMP-4 in Heparin-Conjugated Fibrin Hydrogel. Polymers (Basel) 2022; 14:polym14245343. [PMID: 36559710 PMCID: PMC9780905 DOI: 10.3390/polym14245343] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/08/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The regeneration of cartilage and osteochondral defects remains one of the most challenging clinical problems in orthopedic surgery. Currently, tissue-engineering techniques based on the delivery of appropriate growth factors and mesenchymal stem cells (MSCs) in hydrogel scaffolds are considered as the most promising therapeutic strategy for osteochondral defects regeneration. In this study, we fabricated a heparin-conjugated fibrin (HCF) hydrogel with synovium-derived mesenchymal stem cells (SDMSCs), transforming growth factor-β1 (TGF-β1) and bone morphogenetic protein-4 (BMP-4) to repair osteochondral defects in a rabbit model. An in vitro study showed that HCF hydrogel exhibited good biocompatibility, a slow degradation rate and sustained release of TGF-β1 and BMP-4 over 4 weeks. Macroscopic and histological evaluations revealed that implantation of HCF hydrogel with SDMSCs, TGF-β1 and BMP-4 significantly enhanced the regeneration of hyaline cartilage and the subchondral bone plate in osteochondral defects within 12 weeks compared to hydrogels with SDMSCs or growth factors alone. Thus, these data suggest that combined delivery of SDMSCs with TGF-β1 and BMP-4 in HCF hydrogel may synergistically enhance the therapeutic efficacy of osteochondral defect repair of the knee joints.
Collapse
|
24
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
25
|
Yaylaci S, Guler MO, Tekinay AB. Sulfated GAG mimetic peptide nanofibers enhance chondrogenic differentiation of mesenchymal stem cells in 3D in vitro models. Regen Biomater 2022; 10:rbac084. [PMID: 36683737 PMCID: PMC9847523 DOI: 10.1093/rb/rbac084] [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: 07/29/2022] [Revised: 09/27/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022] Open
Abstract
Articular cartilage, which is exposed to continuous repetitive compressive stress, has limited self-healing capacity in the case of trauma. Thus, it is crucial to develop new treatment options for the effective regeneration of the cartilage tissue. Current cellular therapy treatment options are microfracture and autologous chondrocyte implantation; however, these treatments induce the formation of fibrous cartilage, which degenerates over time, rather than functional hyaline cartilage tissue. Tissue engineering studies using biodegradable scaffolds and autologous cells are vital for developing an effective long-term treatment option. 3D scaffolds composed of glycosaminoglycan-like peptide nanofibers are synthetic, bioactive, biocompatible, and biodegradable and trigger cell-cell interactions that enhance chondrogenic differentiation of cells without using any growth factors. We showed differentiation of mesenchymal stem cells into chondrocytes in both 2D and 3D culture, which produce a functional cartilage extracellular matrix, employing bioactive cues integrated into the peptide nanofiber scaffold without adding exogenous growth factors.
Collapse
Affiliation(s)
| | - Mustafa O Guler
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
26
|
He Y, Wang W, Luo P, Wang Y, He Z, Dong W, Jia M, Yu X, Yang B, Wang J. Mettl3 regulates hypertrophic differentiation of chondrocytes through modulating Dmp1 mRNA via Ythdf1-mediated m 6A modification. Bone 2022; 164:116522. [PMID: 35981698 DOI: 10.1016/j.bone.2022.116522] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 11/02/2022]
Abstract
As the main cells in endochondral osteogenesis, chondrocytes have limited self-repair ability due to weak proliferation activity, low density, and dedifferentiation tendency. Here, a thorough inquiry about the effect and underlying mechanisms of methyltransferase like-3 (Mettl3) on chondrocytes was made. Functionally, it was indicated that Mettl3 promoted the proliferation and hypertrophic differentiation of chondrocytes. Mechanically, Dmp1 (dentin matrix protein 1) was proved to be the downstream direct target of Mettl3 for m6A modification to regulate the differentiation of chondrocytes through bioinformatics analysis and correlated experiments. The Reader protein Ythdf1 mediated Dmp1 mRNA catalyzed by Mettl3. In vivo, the formation of subcutaneous ectopic cartilage-like tissue further supported the in vitro results. In conclusion, the gene regulation of Mettl3/m6A/Ythdf1/Dmp1 axis in hypertrophic differentiation of chondrocytes for the development of endochondral osteogenesis may supply a promising treatment strategy for the repair and regeneration of bone defects.
Collapse
Affiliation(s)
- Ying He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Wei Wang
- Department of Hepatobiliary Surgery in East Hospital, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ping Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Yan Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Zhenru He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Wei Dong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Meie Jia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Xijie Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Beining Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Jiawei Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School &Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| |
Collapse
|
27
|
Wang C, Liu A, Liao Q, Zhong D. Umbilical Cord Wharton’s Jelly Could be the Potential Precursor of Cartilage Tissue Engineering Complex. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We aimed to investigate the structure and composition of human umbilical cord Wharton’s jelly. Umbilical cord was obtained from the newborn in the operating room of our hospital. The transverse and longitudinal sections of umbilical cord were analyzed by hematoxylin-eosin (H&E)
staining. The ultrastructure of umbilical cord Wharton’s jelly was observed by scanning electron microscope (SEM). H&E stain and SEM observation indicate that the collagen fiber closing to the blood vessels is consistent with the direction of the blood vessels. At the peripheral
of the umbilical cord, the mainly direction of the collagen fiber surrounds vessels. At the same time, the density of collagen fiber including Collagen I, Collagen II, and Collagen III from outside to inside gradually becomes dense. Furthermore, Wharton’s jelly is enriched with Collagen
fiber, glycosaminoglycans (GAGs), water and cells. The mean density of cells in Wharton’s jelly was 2.04×106 cell/g, and the mean percentage of MSCs was 54.67% of all separated cells. The structure and composition of the Wharton’s jelly are similar with cartilage.
Therefore, Wharton’s jelly is supposed to be a suitable biological material for cartilage tissue engineering.
Collapse
Affiliation(s)
- Chenggong Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Ansong Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Qiande Liao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Da Zhong
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| |
Collapse
|
28
|
Zhao M, Gao X, Wei J, Tu C, Zheng H, Jing K, Chu J, Ye W, Groth T. Chondrogenic differentiation of mesenchymal stem cells through cartilage matrix-inspired surface coatings. Front Bioeng Biotechnol 2022; 10:991855. [PMID: 36246378 PMCID: PMC9557131 DOI: 10.3389/fbioe.2022.991855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
The stem cell niche comprises soluble molecules and extracellular matrix components which provide chemical and mechanical cues that determine the differentiation of stem cells. Here, the effect of polyelectrolyte multilayer (PEM) composition and terminal layer fabricated with layer-by-layer technique (LBL) pairing either hyaluronan [in its native (nHA) and oxidized form (oHA)] or chondroitin sulfate (CS) with type I collagen (Col I) is investigated on chondrogenic differentiation of human umbilical mesenchymal stem cells (hUC-MSCs). Physical studies performed to investigate the establishment and structure of the surface coatings show that PEM composed of HA and Col I show a dominance of nHA or oHA with considerably lesser organization of Col I fibrils. In contrast, distinguished fibrilized Col I is found in nCS-containing PEM. Generally, Col I-terminated PEM promote the adhesion, migration, and growth of hUC-MSCs more than GAG-terminated surfaces due to the presence of fibrillar Col I but show a lower degree of differentiation towards the chondrogenic lineage. Notably, the Col I/nHA PEM not only supports adhesion and growth of hUC-MSCs but also significantly promotes cartilage-associated gene and protein expression as found by histochemical and molecular biology studies, which is not seen on the Col I/oHA PEM. This is related to ligation of HA to the cell receptor CD44 followed by activation of ERK/Sox9 and noncanonical TGF-β signaling-p38 pathways that depends on the molecular weight of HA as found by immune histochemical and western blotting. Hence, surface coatings on scaffolds and other implants by PEM composed of nHA and Col I may be useful for programming MSC towards cartilage regeneration.
Collapse
Affiliation(s)
- Mingyan Zhao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- *Correspondence: Mingyan Zhao, ; Thomas Groth,
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jinsong Wei
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chenlin Tu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hong Zheng
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Kaipeng Jing
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wei Ye
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle Wittenberg, Halle (Saale), Germany
- *Correspondence: Mingyan Zhao, ; Thomas Groth,
| |
Collapse
|
29
|
Salvador J, Berthelot J, Bony C, Robin B, Him JLK, Noël D, Belamie E, Morille M. Size-tunable lipid vectors for controlled local delivery of siRNA from gene activated matrix. Acta Biomater 2022; 153:97-107. [PMID: 36113724 DOI: 10.1016/j.actbio.2022.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022]
Abstract
Tissue engineering aims to restore or replace different types of biological tissues through the association of cells, biologic factors and biomaterials. Currently, stem cells arise as a major cell source for many therapeutic indications, and their association with 3D scaffolds allow increasing regenerative medicine efficiency. In this context, the use of RNA interference to enhance or control stem cell differentiation into the desired phenotype appears as a promising strategy. However, achieving high transfection efficiency of cells in a 3D structure requires the use of a vector allowing for the spatiotemporally controlled release of the genetic material from these scaffolds. In this study, we report a new siRNA nanovector, called solvent exchange lipoplexe formulation (SELF), which has a tunable size, is stable over time in cell culture conditions and possess a high efficiency to transfect primary human mesenchymal stromal cells (hMSC). We associated SELFs with porous 3D collagen microspheres and demonstrated that the loading capacity and release kinetics were different depending on the size of the associated SELF. Interestingly, these different release profiles resulted in differences in the transfection kinetics of hMSCs. This original and unique type of gene activated matrix, with adaptable release kinetics, could be of interest for long-term and/or sequential transfection profiles of stem cells in 3D culture. STATEMENT OF SIGNIFICANCE: This work combines the use of human mesenchymal stromal cell (hMSC) and gene therapy for tissue engineering. Here, a gene-activated matrix was elaborated with collagen microspheres supporting hMSCs and acting as a reservoir for transfection vectors. This injectable GAM allows for the local and sustained delivery of nucleic acids, hence long-lasting transfection of the supported cells. With the original synthesis protocol presented herein, the size of the nanocarriers can be easily adapted, resulting in different siRNA release profiles from the microspheres. Most interestingly, different siRNA release profiles gave rise to different cell transfection profiles as assessed by the downregulation of a target gene. This highlights the versatility of the system and its suitability for various pathophysiological needs in regenerative medicine.
Collapse
Affiliation(s)
- Jeremy Salvador
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; IRMB, University of Montpellier, INSERM, Montpellier, France; EPHE, PSL Research University, 75014 Paris, France
| | - Jade Berthelot
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; EPHE, PSL Research University, 75014 Paris, France
| | - Claire Bony
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Baptiste Robin
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Josephine Lai Kee Him
- Centre de Biologie Structurale (CBS), Univ Montpellier, INSERM, CNRS, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France.
| | - Emmanuel Belamie
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; EPHE, PSL Research University, 75014 Paris, France.
| | - Marie Morille
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France.
| |
Collapse
|
30
|
Kaewkong P, Kosorn W, Sonthithai P, Lertwimol T, Thavornyutikarn B, Chantaweroad S, Janvikul W. Chondrogenic Differentiation of Human Mesenchymal Stem Cells and Macrophage Polarization on 3D-Printed Poly(ε-caprolactone)/Poly(3-hydroxybutyrate- co-3-hydroxyvalerate) Blended Scaffolds with Different Secondary Porous Structures. ACS APPLIED BIO MATERIALS 2022; 5:2689-2702. [PMID: 35594556 DOI: 10.1021/acsabm.2c00161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study was aimed to evaluate the chondrogenic differentiation of human mesenchymal stem cells (hMSCs) and polarization of THP-1-derived macrophages cultured on poly(ε-caprolactone) (PC)/poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PH) blended scaffolds with dual primary (PP) and secondary (SP) pores, which were fabricated via a 3D printing technique, i.e., fused deposition modeling, followed by a salt-leaching process at 50 °C for varied times, i.e., 15, 30, and 60 min. Sodium chloride (SC), a porogen, was initially incorporated in the blend at varied weight percentages, i.e., 0, 25, and 50%, whereas 1 M NaOH solution and deionized water were used as salt-leaching agents. To elucidate the surface properties of the developed scaffolds, directly governed by the amount of the salt originally mixed and the salt-leaching efficiency, several characterization techniques, e.g., scanning electron microscopy, X-ray microcomputed tomography, mercury intrusion porosimetry, atomic force microscopy, and contact angle measurement, were used. Meanwhile, the salt-leaching efficiency was determined by means of weight loss measurement and thermogravimetric analysis. It was found that the alkaline solution could satisfactorily leach out the salt particles in 60 min with a mild etching of the polymer framework. The most immensely and homogeneously pitted filament surface was observed in the NaOH-treated scaffold initially integrated with 50% salt, i.e., 60B_PC/PH/50SC; the SP structure was mostly open and interconnected. The size of most of micropores was about 0.14 μm. With its suitable microsurface roughness and hydrophilicity, 60B_PC/PH/50SC could properly support the initial attachment and lamellipodia formation of hMSCs, which was favorable for chondrogenesis. Consequently, a significantly increased ratio of glycosaminoglycans/deoxyribonucleic acid and a superior expression of the COL2A1 gene were detected when cells were grown on this material. Although 60B_PC/PH/50SC induced the macrophages to secrete a slightly high level of IL-1β during the first few days of culture, the polarized M1 cells could return to a nearly normal stage at Day7, suggesting no unfavorable chronic inflammation caused by the material.
Collapse
Affiliation(s)
- Pakkanun Kaewkong
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park, Phahonyothin Road, Klong Luang, Pathum Thani 12120, Thailand
| | - Wasana Kosorn
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park, Phahonyothin Road, Klong Luang, Pathum Thani 12120, Thailand
| | - Pacharapan Sonthithai
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park, Phahonyothin Road, Klong Luang, Pathum Thani 12120, Thailand
| | - Tareerat Lertwimol
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park, Phahonyothin Road, Klong Luang, Pathum Thani 12120, Thailand
| | - Boonlom Thavornyutikarn
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park, Phahonyothin Road, Klong Luang, Pathum Thani 12120, Thailand
| | - Surapol Chantaweroad
- Assistive Technology and Medical Devices Research Center, Central Office, 111 Thailand Science Park, Phahonyothin Road, Klong Luang, Pathum Thani 12120, Thailand
| | - Wanida Janvikul
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, 114 Thailand Science Park, Phahonyothin Road, Klong Luang, Pathum Thani 12120, Thailand
| |
Collapse
|
31
|
Lesage C, Lafont M, Guihard P, Weiss P, Guicheux J, Delplace V. Material-Assisted Strategies for Osteochondral Defect Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200050. [PMID: 35322596 PMCID: PMC9165504 DOI: 10.1002/advs.202200050] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/25/2022] [Indexed: 05/08/2023]
Abstract
The osteochondral (OC) unit plays a pivotal role in joint lubrication and in the transmission of constraints to bones during movement. The OC unit does not spontaneously heal; therefore, OC defects are considered to be one of the major risk factors for developing long-term degenerative joint diseases such as osteoarthritis. Yet, there is currently no curative treatment for OC defects, and OC regeneration remains an unmet medical challenge. In this context, a plethora of tissue engineering strategies have been envisioned over the last two decades, such as combining cells, biological molecules, and/or biomaterials, yet with little evidence of successful clinical transfer to date. This striking observation must be put into perspective with the difficulty in comparing studies to identify overall key elements for success. This systematic review aims to provide a deeper insight into the field of material-assisted strategies for OC regeneration, with particular considerations for the therapeutic potential of the different approaches (with or without cells or biological molecules), and current OC regeneration evaluation methods. After a brief description of the biological complexity of the OC unit, the recent literature is thoroughly analyzed, and the major pitfalls, emerging key elements, and new paths to success are identified and discussed.
Collapse
Affiliation(s)
- Constance Lesage
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
- HTL Biotechnology7 Rue Alfred KastlerJavené35133France
| | - Marianne Lafont
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| | - Pierre Guihard
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| | - Pierre Weiss
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| | - Jérôme Guicheux
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| | - Vianney Delplace
- Université de NantesOnirisCHU NantesINSERMRegenerative Medicine and SkeletonRMeSUMR 1229NantesF‐44000France
| |
Collapse
|
32
|
Mairpady A, Mourad AHI, Mozumder MS. Accelerated Discovery of the Polymer Blends for Cartilage Repair through Data-Mining Tools and Machine-Learning Algorithm. Polymers (Basel) 2022; 14:polym14091802. [PMID: 35566970 PMCID: PMC9104973 DOI: 10.3390/polym14091802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
In designing successful cartilage substitutes, the selection of scaffold materials plays a central role, among several other important factors. In an empirical approach, the selection of the most appropriate polymer(s) for cartilage repair is an expensive and time-consuming affair, as traditionally it requires numerous trials. Moreover, it is humanly impossible to go through the huge library of literature available on the potential polymer(s) and to correlate the physical, mechanical, and biological properties that might be suitable for cartilage tissue engineering. Hence, the objective of this study is to implement an inverse design approach to predict the best polymer(s)/blend(s) for cartilage repair by using a machine-learning algorithm (i.e., multinomial logistic regression (MNLR)). Initially, a systematic bibliometric analysis on cartilage repair has been performed by using the bibliometrix package in the R program. Then, the database was created by extracting the mechanical properties of the most frequently used polymers/blends from the PoLyInfo library by using data-mining tools. Then, an MNLR algorithm was run by using the mechanical properties of the polymers, which are similar to the cartilages, as the input and the polymer(s)/blends as the predicted output. The MNLR algorithm used in this study predicts polyethylene/polyethylene-graftpoly(maleic anhydride) blend as the best candidate for cartilage repair.
Collapse
Affiliation(s)
- Anusha Mairpady
- Chemical and Petroleum Engineering Department, UAE University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Abdel-Hamid I. Mourad
- Mechanical and Aerospace Engineering Department, UAE University, Al Ain P.O. Box 15551, United Arab Emirates;
- National Water and Energy Center, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Mohammad Sayem Mozumder
- Chemical and Petroleum Engineering Department, UAE University, Al Ain P.O. Box 15551, United Arab Emirates;
- Correspondence:
| |
Collapse
|
33
|
Shen K, Duan A, Cheng J, Yuan T, Zhou J, Song H, Chen Z, Wan B, Liu J, Zhang X, Zhang Y, Xie R, Liu F, Fan W, Zuo Q. Exosomes derived from hypoxia preconditioned mesenchymal stem cells laden in a silk hydrogel promote cartilage regeneration via the miR-205-5p/PTEN/AKT pathway. Acta Biomater 2022; 143:173-188. [PMID: 35202856 DOI: 10.1016/j.actbio.2022.02.026] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022]
Abstract
Tissue engineering has promising prospects for cartilage regeneration. However, there remains an urgent need to harvest high quality seed cells. Bone marrow mesenchymal cells (BMSCs), and in particular their exosomes, might promote the function of articular chondrocytes (ACs) via paracrine mechanisms. Furthermore, preconditioned BMSCs could provide an enhanced therapeutic effect. BMSCs naturally exist in a relatively hypoxic environment (1%-5% O2); however, they are usually cultured under higher oxygen concentrations (21% O2). Herein, we hypothesized that hypoxia preconditioned exosomes (H-Exos) could improve the quality of ACs and be more conducive to cartilage repair. In our study, we compared the effects of exosomes derived from BMSCs preconditioned with hypoxia and normoxia (N-Exos) on ACs, demonstrating that H-Exos significantly promoted the proliferation, migration, anabolism and anti-inflammation effects of ACs. Furthermore, we confirmed that hypoxia preconditioning upregulated the expression of miR-205-5p in H-Exos, suggesting that ACs were promoted via the miR-205-5p/PTEN/AKT pathway. Finally, an injectable silk fibroin (SF) hydrogel containing ACs and H-Exos (SF/ACs/H-Exos) was utilized to repair cartilage defects and effectively promote cartilage regeneration in vivo. The application of SF/ACs/H-Exos hydrogel in cartilage regeneration therefore has promising prospects. STATEMENT OF SIGNIFICANCE: Cartilage tissue engineering (CTE) has presented a promising prospect. However, the quality of seed cells is an important factor affecting the repair efficiency. Our study demonstrates for the first time that the exosomes derived from hypoxia preconditioned BMSCs (H-Exos) effectively promote the proliferation, migration and anabolism of chondrocytes and inhibit inflammation through miR-205-5p/PTEN/AKT pathway. Furthermore, we fabricated an injectable silk fibrion (SF) hydrogel to preserve and sustained release H-Exos. A complex composed of SF hydrogel, H-Exos and chondrocytes can effectively promote the regeneration of cartilage defects. Therefore, this study demonstrates that hypoxia pretreatment could optimize the therapeutic effects of BMSCs-derived exosomes, and the combination of exosomes and SF hydrogel could be a promising therapeutic method for cartilage regeneration.
Collapse
|
34
|
Kumar Reddy Sanapalli B, Tyagi R, Shaik AB, Ranakishor P, Bhandare RR, Annadurai S, Venkata Satyanarayana Reddy Karri V. L-Glutamic acid loaded collagen chitosan composite scaffold as regenerative medicine for the accelerated healing of diabetic wounds. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
35
|
Kamel R, Mabrouk M, El-Sayed SAM, Beherei HH, Abouzeid RE, Abo El-Fadl MT, Mahmoud AA, Maged A. Nanofibrillated cellulose/glucosamine 3D aerogel implants loaded with rosuvastatin and bioactive ceramic for dental socket preservation. Int J Pharm 2022; 616:121549. [PMID: 35131357 DOI: 10.1016/j.ijpharm.2022.121549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/20/2022] [Accepted: 02/01/2022] [Indexed: 11/19/2022]
Abstract
Recycling of agro-wastes presents a great economic and ecologic value. In this study, TEMPO-oxidized nanofibrillated cellulose (TONFC) originating from sugarcane bagasse pulp was exploited in regenerative medicine. TONFC in combination with glucosamine HCl (G) were used to prepare a 3D aerogel implant loaded with rosuvastatin as an integrative approach for extraction-socket healing. Comparing the prepared devices, aerogel composed of TONFC: G (4:1 wt ratio) had the best mechanical properties and integrity. Strontium borate-based bioactive ceramic particles were prepared and characterized for crystal structure, shape, porosity, and zeta potential. The particles had a crystalline diffraction pattern relative to Sr3B2O6, and they were rod in shape with nanopores with a zeta potential value of -16 mV. The prepared bioactive ceramic (BC) was then added in different concentrations (3 or 6% w/w) to the selected aerogel implant. The BC had a concentration-dependent effect on the aerogel properties as it ameliorated its mechanical performance (compressive strength = 90 and 150 kPa for 3 and 6%, respectively) and retarded drug release (mean release time = 2.34 and 3.4 h for 3 and 6%, respectively) (p < 0.05). The microphotograph of the selected aerogel implant loaded with BC showed a rough surface with an interconnective porous structure. During cell biology testing, the selected implant loaded with the lower BC concentration had the highest ability to increase MG-63 cells proliferation. In conclusion, TONFC is a promising material to formulate rosuvastatin-loaded aerogel implant with the aid of glucosamine and bioactive ceramic for dental socket preservation.
Collapse
Affiliation(s)
- Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre, Dokki, 12622 Cairo, Egypt
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, National Research Centre, Dokki, 12622 Cairo, Egypt
| | - Sara A M El-Sayed
- Refractories, Ceramics and Building Materials Department, National Research Centre, Dokki, 12622 Cairo, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, National Research Centre, Dokki, 12622 Cairo, Egypt
| | - Ragab E Abouzeid
- Cellulose and Paper Department, National Research Centre, Dokki, 12622 Cairo, Egypt
| | - Mahmoud T Abo El-Fadl
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki, 12622 Cairo, Egypt; Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, 12622 Cairo, Egypt
| | - Azza A Mahmoud
- Pharmaceutics and Pharmaceutical Technology Department, Faculty of Pharmacy, Future University in Egypt, Egypt.
| | - Amr Maged
- Pharmaceutics and Pharmaceutical Technology Department, Faculty of Pharmacy, Future University in Egypt, Egypt; Pharmaceutical Factory, Faculty of Pharmacy, Future University in Egypt, Egypt.
| |
Collapse
|
36
|
The Induced Pluripotent Stem Cells in Articular Cartilage Regeneration and Disease Modelling: Are We Ready for Their Clinical Use? Cells 2022; 11:cells11030529. [PMID: 35159338 PMCID: PMC8834349 DOI: 10.3390/cells11030529] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
The development of induced pluripotent stem cells has brought unlimited possibilities to the field of regenerative medicine. This could be ideal for treating osteoarthritis and other skeletal diseases, because the current procedures tend to be short-term solutions. The usage of induced pluripotent stem cells in the cell-based regeneration of cartilage damages could replace or improve on the current techniques. The patient’s specific non-invasive collection of tissue for reprogramming purposes could also create a platform for drug screening and disease modelling for an overview of distinct skeletal abnormalities. In this review, we seek to summarise the latest achievements in the chondrogenic differentiation of pluripotent stem cells for regenerative purposes and disease modelling.
Collapse
|
37
|
BRUNI D, PANTALONE A, MASTROIANNI F, DEL GUSTO F, BUDA R. The role of mesenchimal stem cells in the treatment of osteochondral lesions and osteoarthritis of the ankle. MINERVA ORTHOPEDICS 2022; 72. [DOI: 10.23736/s2784-8469.20.04068-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
38
|
Thampi P, Dubey R, Lowney R, Adam EN, Janse S, Wood CL, MacLeod JN. Effect of Skeletal Paracrine Signals on the Proliferation of Interzone Cells. Cartilage 2021; 13:82S-94S. [PMID: 31023058 PMCID: PMC8804777 DOI: 10.1177/1947603519841680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Articular cartilage in mammals has limited intrinsic capacity to repair structural defects, a fact that contributes to the chronic and progressive nature of osteoarthritis. In contrast, Mexican axolotl salamanders have demonstrated the remarkable ability to spontaneously and completely repair large joint cartilage lesions, a healing process that involves interzone cells in the intraarticular space. Furthermore, interzone tissue transplanted into skeletal defects in the axolotl salamander demonstrates a multi-differentiation potential. Cellular and molecular mechanisms of this repair process remain unclear. The objective of this study was to examine whether paracrine mitogenic signals are an important variable in the interaction between interzone cells and the skeletal microenvironment. DESIGN The paracrine regulation of the proliferation of equine interzone cells was evaluated in an in vitro co-culture system. Cell viability and proliferation were measured in equine fetal interzone cells after exposure to conditioned medium from skeletal and nonskeletal primary cell lines. Steady-state expression was determined for genes encoding 37 putative mitogens secreted by cells that generated the conditioned medium. RESULTS All experimental groups of conditioned media elicited a mitogenic response in interzone cells. Fetal anlage chondrocytes (P < 0.0001) and dermal fibroblasts (P < 0.0001) conditioned medium showed a significantly higher mitogenic potential compared with interzone cells. Conditioned medium from bone marrow-derived cells elicited a significantly higher proliferative response relative to that from young adult articular chondrocytes (P < 0.0001) or dermal fibroblasts (P < 0.0001). Sixteen genes had expression patterns consistent with the functional proliferation assays. CONCLUSIONS The results indicate a mitogenic effect of skeletal paracrine signals on interzone cells.
Collapse
Affiliation(s)
- Parvathy Thampi
- Maxwell H. Gluck Equine Research Center,
Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Rashmi Dubey
- Maxwell H. Gluck Equine Research Center,
Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Rachael Lowney
- Maxwell H. Gluck Equine Research Center,
Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Emma N. Adam
- Maxwell H. Gluck Equine Research Center,
Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Sarah Janse
- Department of Statistics, University of
Kentucky, Lexington, KY, USA
| | - Constance L. Wood
- Department of Statistics, University of
Kentucky, Lexington, KY, USA
| | - James N. MacLeod
- Maxwell H. Gluck Equine Research Center,
Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
39
|
Wei W, Dai H. Articular cartilage and osteochondral tissue engineering techniques: Recent advances and challenges. Bioact Mater 2021; 6:4830-4855. [PMID: 34136726 PMCID: PMC8175243 DOI: 10.1016/j.bioactmat.2021.05.011] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
In spite of the considerable achievements in the field of regenerative medicine in the past several decades, osteochondral defect regeneration remains a challenging issue among diseases in the musculoskeletal system because of the spatial complexity of osteochondral units in composition, structure and functions. In order to repair the hierarchical tissue involving different layers of articular cartilage, cartilage-bone interface and subchondral bone, traditional clinical treatments including palliative and reparative methods have showed certain improvement in pain relief and defect filling. It is the development of tissue engineering that has provided more promising results in regenerating neo-tissues with comparable compositional, structural and functional characteristics to the native osteochondral tissues. Here in this review, some basic knowledge of the osteochondral units including the anatomical structure and composition, the defect classification and clinical treatments will be first introduced. Then we will highlight the recent progress in osteochondral tissue engineering from perspectives of scaffold design, cell encapsulation and signaling factor incorporation including bioreactor application. Clinical products for osteochondral defect repair will be analyzed and summarized later. Moreover, we will discuss the current obstacles and future directions to regenerate the damaged osteochondral tissues.
Collapse
Affiliation(s)
- Wenying Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan, 528200, China
| |
Collapse
|
40
|
Choi S, Ahn H, Kim S. Tyrosinase‐mediated hydrogel crosslinking for tissue engineering. J Appl Polym Sci 2021. [DOI: 10.1002/app.51887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sumi Choi
- Department of Chemical Engineering (BK 21 FOUR) Dong‐A University Busan Republic of Korea
| | - Hyerin Ahn
- Department of Chemical Engineering (BK 21 FOUR) Dong‐A University Busan Republic of Korea
| | - Su‐Hwan Kim
- Department of Chemical Engineering (BK 21 FOUR) Dong‐A University Busan Republic of Korea
| |
Collapse
|
41
|
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
|
42
|
Nguyen HTN, Vu NB. A Simple Method to Produce Engineered Cartilage from Human Adipose-Derived Mesenchymal Stem Cells and Poly ε-Caprolactone Scaffolds. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021:181-191. [PMID: 34739719 DOI: 10.1007/5584_2021_669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION The damaged articular cartilage has limited self-regeneration capacity because of the absence of blood vessels, lymphatics, and nerves. Cartilage transplantation is, hence, a popular method used to treat this disease. However, sources of autograft and allogenic cartilage for transplantation are limited. Therefore, this study aims to suggest a simple method to produce engineered cartilage from human adipose-derived mesenchymal stem cells (ADSCs) and poly (ε-caprolactone) (PCL) scaffolds. METHODS ADSCs were isolated and expanded from fat tissues according to published protocols. PCL-porous scaffolds were produced from PCL with 5 × 5 × 0.6 mm3 with 200-400 μ m pore sizes. ADSCs were seeded on the PCL scaffolds at three different densities (104, 105, 106 cells per scaffold). The adherence of ADSCs on the surface of PCL scaffolds was evaluated based on an immunostaining assay to determine the presence of ADSCs. The cell proliferation on PCL scaffolds was determined by MTT assay. The complexity in ADSCs and PCL scaffolds was induced to cartilage using a chondrogenesis medium. The engineered cartilage was characterized by the accumulation of proteoglycan and aggrecan by Safranin O staining assay. Their structures were evaluated using an H-E staining assay. Finally, these engineered cartilage tissues were transplanted into mice to assess cartilage maturation when compared to natural cartilage. RESULTS The results showed that the engineered cartilage tissues could be successfully produced by cultures of ADSCs on poly ε-caprolactone scaffolds in combination with chondrogenesis medium. The suitable density of ADSCs was 106 cells/per scaffold of 5 × 6 × 0.6 mm3 with pore size from 200 to 400 μ m. CONCLUSION The results showed that an in vitro cartilage tissue was created from ADSCs and PCL scaffold. The cartilage tissue exists in the mice for 6 months.
Collapse
Affiliation(s)
- Hue Thi-Ngoc Nguyen
- Stem Cell Institute, University of Science, Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ngoc Bich Vu
- Stem Cell Institute, University of Science, Ho Chi Minh City, Vietnam.
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| |
Collapse
|
43
|
Nguyen TPT, Li F, Shrestha S, Tuan RS, Thissen H, Forsythe JS, Frith JE. Cell-laden injectable microgels: Current status and future prospects for cartilage regeneration. Biomaterials 2021; 279:121214. [PMID: 34736147 DOI: 10.1016/j.biomaterials.2021.121214] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/19/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022]
Abstract
Injectable hydrogels have been employed extensively as versatile materials for cartilage regeneration due to their excellent biocompatibility, tunable structure, and ability to accommodate bioactive factors, as well as their ability to be locally delivered via minimally invasive injection to fill irregular defects. More recently, in vitro and in vivo studies have revealed that processing these materials to produce cell-laden microgels can enhance cell-cell and cell-matrix interactions and boost nutrient and metabolite exchange. Moreover, these studies have demonstrated gene expression profiles and matrix regeneration that are superior compared to conventional injectable bulk hydrogels. As cell-laden microgels and their application in cartilage repair are moving closer to clinical translation, this review aims to present an overview of the recent developments in this field. Here we focus on the currently used biomaterials and crosslinking strategies, the innovative fabrication techniques being used for the production of microgels, the cell sources used, the signals used for induction of chondrogenic differentiation and the resultant biological responses, and the ability to create three-dimensional, functional cartilage tissues. In addition, this review also covers the current clinical approaches for repairing cartilage as well as specific challenges faced when attempting the regeneration of damaged cartilage tissue. New findings related to the macroporous nature of the structures formed by the assembled microgel building blocks and the novel use of microgels in 3D printing for cartilage tissue engineering are also highlighted. Finally, we outline the challenges and future opportunities for employing cell-laden microgels in clinical applications.
Collapse
Affiliation(s)
- Thuy P T Nguyen
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Fanyi Li
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Surakshya Shrestha
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Helmut Thissen
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Clayton, VIC 3800, Australia.
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Clayton, VIC 3800, Australia.
| |
Collapse
|
44
|
Chen D, Zhang Y, Lin Q, Chen D, Li X, Dai J, Sun Y. The effect of cartilage decellularized extracellular matrix-chitosan compound on treating knee osteoarthritis in rats. PeerJ 2021; 9:e12188. [PMID: 34721961 PMCID: PMC8519179 DOI: 10.7717/peerj.12188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/30/2021] [Indexed: 11/20/2022] Open
Abstract
Knee osteoarthritis (KOA) refers to a common disease in orthopaedics, whereas effective treatments have been rarely developed. As indicated from existing studies, chondrocyte death, extracellular matrix degradation and subchondral bone injury are recognized as the pathological basis of KOA. The present study aimed to determine the therapeutic effect of decellularized extracellular matrix-chitosan (dECM-CS) compound on KOA. In this study, rat knee cartilage was decellularized, and a satisfactory decellularized extracellular matrix was developed. As suggested from the in vitro experiments, the rat chondrocytes co-cultured with allogeneic dECM grew effectively. According to the results of the alamar blue detection, dECM did not adversely affect the viability of rat chondrocytes, and dECM could up-regulate the genes related to the cartilage synthesis and metabolism. As reported from the animal experiments, dECM-CS compound could protect cartilage, alleviate knee joint pain in rats, significantly delay the progress of KOA in rats, and achieve high drug safety. In brief, dECM-CS compound shows a good therapeutic effect on KOA.
Collapse
Affiliation(s)
- Deng Chen
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Yaxin Zhang
- Dalian Medical University, Dalian, Liaoning, China
| | - Qun Lin
- Dalian Medical University, Dalian, Liaoning, China
| | - Duoyun Chen
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaolei Li
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Yu Sun
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
45
|
Maumus M, Fonteneau G, Ruiz M, Assou S, Boukhaddaoui H, Pastoureau P, De Ceuninck F, Jorgensen C, Noel D. Neuromedin B promotes chondrocyte differentiation of mesenchymal stromal cells via calcineurin and calcium signaling. Cell Biosci 2021; 11:183. [PMID: 34663442 PMCID: PMC8525028 DOI: 10.1186/s13578-021-00695-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 09/30/2021] [Indexed: 11/11/2022] Open
Abstract
Background Articular cartilage is a complex tissue with poor healing capacities. Current approaches for cartilage repair based on mesenchymal stromal cells (MSCs) are often disappointing because of the lack of relevant differentiation factors that could drive MSC differentiation towards a stable mature chondrocyte phenotype. Results We used a large-scale transcriptomic approach to identify genes that are modulated at early stages of chondrogenic differentiation using the reference cartilage micropellet model. We identified several modulated genes and selected neuromedin B (NMB) as one of the early and transiently modulated genes. We found that the timely regulated increase of NMB was specific for chondrogenesis and not observed during osteogenesis or adipogenesis. Furthermore, NMB expression levels correlated with the differentiation capacity of MSCs and its inhibition resulted in impaired chondrogenic differentiation indicating that NMB is required for chondrogenesis. We further showed that NMB activated the calcineurin activity through a Ca2+-dependent signaling pathway. Conclusion NMB is a newly described chondroinductive bioactive factor that upregulates the key chondrogenic transcription factor Sox9 through the modulation of Ca2+ signaling pathway and calcineurin activity. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00695-1.
Collapse
Affiliation(s)
- Marie Maumus
- IRMB, Univ Montpellier, INSERM, Montpellier, France
| | | | - Maxime Ruiz
- IRMB, Univ Montpellier, INSERM, Montpellier, France
| | - Said Assou
- IRMB, Univ Montpellier, INSERM, Montpellier, France
| | - Hassan Boukhaddaoui
- INM, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Philippe Pastoureau
- Institut de Recherches Servier, Center for Therapeutic Innovation, Immuno-Inflammatory Disease, Croissy-sur-Seine, France
| | - Frédéric De Ceuninck
- Institut de Recherches Servier, Center for Therapeutic Innovation, Immuno-Inflammatory Disease, Croissy-sur-Seine, France
| | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, Montpellier, France.,Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France
| | - Danièle Noel
- IRMB, Univ Montpellier, INSERM, Montpellier, France. .,Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France.
| |
Collapse
|
46
|
Zhong Y, Caplan AI, Welter JF, Baskaran H. Glucose Availability Affects Extracellular Matrix Synthesis During Chondrogenesis In Vitro. Tissue Eng Part A 2021; 27:1321-1332. [PMID: 33499734 PMCID: PMC8610032 DOI: 10.1089/ten.tea.2020.0144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/22/2021] [Indexed: 01/09/2023] Open
Abstract
Understanding in vitro chondrogenesis of human mesenchymal stem cells (hMSCs) is important as it holds great promise for cartilage tissue engineering and other applications. The current technology produces the end tissue quality that is highly variable and dependent on culture conditions. We investigated the effect of nutrient availability on hMSC chondrogenesis in a static aggregate culture system by varying the medium-change frequency together with starting glucose levels. Glucose uptake and lactate secretion profiles were obtained to monitor the metabolism change during hMSC chondrogenesis with different culture conditions. Higher medium-change frequency led to increases in cumulative glucose uptake for all starting glucose levels. Furthermore, increase in glucose uptake by aggregates led to increased end tissue glycosaminoglycan (GAG) and hydroxyproline (HYP) content. The results suggest that increased glucose availability either through increased medium-change frequency or higher initial glucose levels lead to improved chondrogenesis. Also, cumulative glucose uptake and lactate secretion were found to correlate well with GAG and HYP content, indicating both molecules are promising biomarkers for noninvasive assessment of hMSC chondrogenesis. Collectively, our results can be used to design optimal culture conditions and develop dynamic assessment strategies for cartilage tissue engineering applications. Impact statement In this study, we investigated how culture conditions, medium-change frequency and glucose levels, affect chondrogenesis of human mesenchymal stem cells in an aggregate culture model. Doubling the medium-change frequency significantly increased the biochemical quality of the resultant tissue aggregates, as measured by their glycosaminoglycan and hydroxyproline content. We attribute this to increased glucose uptake through the glycolysis pathway, as secretion of lactate, a key endpoint product of the glycolysis pathway, increased concurrently. These findings can be used to design optimal culture conditions for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- CWRU Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, Ohio, USA
| | - Arnold I. Caplan
- CWRU Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biology and Case Western Reserve University, Cleveland, Ohio, USA
| | - Jean F. Welter
- CWRU Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biology and Case Western Reserve University, Cleveland, Ohio, USA
| | - Harihara Baskaran
- CWRU Center for Multimodal Evaluation of Engineered Cartilage, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
47
|
Kuhlmann C, Schenck TL, Aszodi A, Giunta RE, Wiggenhauser PS. Zone-Dependent Architecture and Biochemical Composition of Decellularized Porcine Nasal Cartilage Modulate the Activity of Adipose Tissue-Derived Stem Cells in Cartilage Regeneration. Int J Mol Sci 2021; 22:ijms22189917. [PMID: 34576079 PMCID: PMC8470846 DOI: 10.3390/ijms22189917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 01/22/2023] Open
Abstract
Previous anatomical studies have shown different functional zones in human nasal septal cartilage (NC). These zones differ in respect to histological architecture and biochemical composition. The aim of this study was to investigate the influence of these zones on the fate of stem cells from a regenerative perspective. Therefore, decellularized porcine septal cartilage was prepared and subjected to histological assessment to demonstrate its equivalence to human cartilage. Decellularized porcine NC (DPNC) exposed distinct surfaces depending on two different histological zones: the outer surface (OS), which is equivalent to the superficial zone, and the inner surface (IS), which is equivalent to the central zone. Human adipose tissue-derived stem cells (ASCs) were isolated from the abdominal fat tissue of five female patients and were seeded on the IS and OS of DPNC, respectively. Cell seeding efficiency (CSE), vitality, proliferation, migration, the production of sulfated glycosaminoglycans (sGAG) and chondrogenic differentiation capacity were evaluated by histological staining (DAPI, Phalloidin, Live-Dead), biochemical assays (alamarBlue®, PicoGreen®, DMMB) and the quantification of gene expression (qPCR). Results show that cell vitality and CSE were not influenced by DPNC zones. ASCs, however, showed a significantly higher proliferation and elevated expression of early chondrogenic differentiation, as well as fibrocartilage markers, on the OS. On the contrary, there was a significantly higher upregulation of hypertrophy marker MMP13 (p < 0.0001) and GAG production (p = 0.0105) on the IS, whereas cell invasion into the three-dimensional DPNC was higher in comparison to the OS. We conclude that the zonal-dependent distinct architecture and composition of NC modulates activities of ASCs seeded on DPNC. These findings might be used for engineering of cartilage substitutes needed in facial reconstructive surgery that yield an equivalent histological and functional structure, such as native NC.
Collapse
Affiliation(s)
- Constanze Kuhlmann
- Department of Hand, Plastic and Aesthetic Surgery, LMU Klinikum, University Hospital, LMU Munich, Ziemsenstrasse 5, 80336 Munich, Germany; (C.K.); (T.L.S.); (R.E.G.)
- Laboratory of Cartilage Development, Diseases and Regeneration, Department for Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Fraunhoferstrasse 20, 82152 Planegg, Germany;
| | - Thilo L. Schenck
- Department of Hand, Plastic and Aesthetic Surgery, LMU Klinikum, University Hospital, LMU Munich, Ziemsenstrasse 5, 80336 Munich, Germany; (C.K.); (T.L.S.); (R.E.G.)
| | - Attila Aszodi
- Laboratory of Cartilage Development, Diseases and Regeneration, Department for Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Fraunhoferstrasse 20, 82152 Planegg, Germany;
| | - Riccardo E. Giunta
- Department of Hand, Plastic and Aesthetic Surgery, LMU Klinikum, University Hospital, LMU Munich, Ziemsenstrasse 5, 80336 Munich, Germany; (C.K.); (T.L.S.); (R.E.G.)
| | - Paul Severin Wiggenhauser
- Department of Hand, Plastic and Aesthetic Surgery, LMU Klinikum, University Hospital, LMU Munich, Ziemsenstrasse 5, 80336 Munich, Germany; (C.K.); (T.L.S.); (R.E.G.)
- Correspondence:
| |
Collapse
|
48
|
Uzieliene I, Bironaite D, Bernotas P, Sobolev A, Bernotiene E. Mechanotransducive Biomimetic Systems for Chondrogenic Differentiation In Vitro. Int J Mol Sci 2021; 22:9690. [PMID: 34575847 PMCID: PMC8469886 DOI: 10.3390/ijms22189690] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is a long-term chronic joint disease characterized by the deterioration of bones and cartilage, which results in rubbing of bones which causes joint stiffness, pain, and restriction of movement. Tissue engineering strategies for repairing damaged and diseased cartilage tissue have been widely studied with various types of stem cells, chondrocytes, and extracellular matrices being on the lead of new discoveries. The application of natural or synthetic compound-based scaffolds for the improvement of chondrogenic differentiation efficiency and cartilage tissue engineering is of great interest in regenerative medicine. However, the properties of such constructs under conditions of mechanical load, which is one of the most important factors for the successful cartilage regeneration and functioning in vivo is poorly understood. In this review, we have primarily focused on natural compounds, particularly extracellular matrix macromolecule-based scaffolds and their combinations for the chondrogenic differentiation of stem cells and chondrocytes. We also discuss different mechanical forces and compression models that are used for In Vitro studies to improve chondrogenic differentiation. Summary of provided mechanical stimulation models In Vitro reviews the current state of the cartilage tissue regeneration technologies and to the potential for more efficient application of cell- and scaffold-based technologies for osteoarthritis or other cartilage disorders.
Collapse
Affiliation(s)
- Ilona Uzieliene
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, LT-08406 Vilnius, Lithuania; (I.U.); (D.B.); (P.B.)
| | - Daiva Bironaite
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, LT-08406 Vilnius, Lithuania; (I.U.); (D.B.); (P.B.)
| | - Paulius Bernotas
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, LT-08406 Vilnius, Lithuania; (I.U.); (D.B.); (P.B.)
| | - Arkadij Sobolev
- Latvian Institute of Organic Synthesis, 21 Aizkraukles Str., LV-1006 Riga, Latvia;
| | - Eiva Bernotiene
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, LT-08406 Vilnius, Lithuania; (I.U.); (D.B.); (P.B.)
| |
Collapse
|
49
|
Dehghani Nazhvani F, Mohammadi Amirabad L, Azari A, Namazi H, Hosseinzadeh S, Samanipour R, Khojasteh A, Golchin A, Hashemi S. Effects of in vitro low oxygen tension preconditioning of buccal fat pad stem cells on in Vivo articular cartilage tissue repair. Life Sci 2021; 280:119728. [PMID: 34144057 DOI: 10.1016/j.lfs.2021.119728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 01/20/2023]
Abstract
AIMS Progenitor cells-based regenerative strategy has shown promise to repair cartilage, an avascular tissue in which cells experience hypoxia. Hypoxia is known to improve the early chondrogenic differentiation of stem cells. Therefore, this study aimed to determine whether hypoxia preconditioning could be used to enhance the regenerative potential of the combination of buccal fat pad stem cells (BFPSCs) and bilayer chitosan-based hydrogel scaffold for articular cartilage repair. MATERIALS AND METHODS Human BFPSCs were seeded on the bilayer chitosan-based hydrogel scaffolds in the culture medium. The viability and proliferation of cells on the scaffolds were monitored using scanning electron microscopy (SEM), MTT assay, and DAPI staining. Hypoxia preconditioned BFPSCs-seeded scaffolds were transplanted into rabbit articular cartilage knee defects for 12 weeks. The newly formed tissue was evaluated by cartilage-specific immunohistological analysis and histological staining. KEY FINDINGS It was found that the chondrogenic differentiation and osteochondral conjunction in articular cartilage defect via BFPSCs-seeded bilayer scaffolds was enhanced by hypoxic preconditioning compared to a normoxic environment. SIGNIFICANCE Based on our study, the integrity with subchondral bone in osteochondral defect was enhanced by BFPSCs on bilayer scaffold. Thus, this study provides evidence on the design of preconditioned cell-seeded bilayer hydrogels for articular cartilage regeneration.
Collapse
Affiliation(s)
| | | | - Arezo Azari
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Namazi
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Samanipour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Khojasteh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Golchin
- Department of Clinical Biochemistry and Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Sheida Hashemi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Amsar RM, Barlian A, Judawisastra H, Wibowo UA, Karina K. Cell penetration and chondrogenic differentiation of human adipose derived stem cells on 3D scaffold. Future Sci OA 2021; 7:FSO734. [PMID: 34295538 PMCID: PMC8288224 DOI: 10.2144/fsoa-2021-0040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/19/2021] [Indexed: 11/25/2022] Open
Abstract
The ability of cells to penetrate the scaffold and differentiate into chondrocyte is important in cartilage engineering. The aim of this research was to evaluate the use of silk fibroin 3D scaffold in facilitating the growth of stem cell and to study the role of L-ascorbic acid and platelet rich plasma (PRP) in proliferation and differentiation genes. Cell penetration and type II collagen content in the silk fibroin scaffold was analyzed by confocal microscopy. Relative expressions of CDH2, CCND1, CTNNB1 and COL2A1 were analyzed by reverse transcription-quantitative PCR (RT-qPCR). The silk fibroin 3D scaffold could facilitate cell penetration. L-ascorbic acid and PRP increased the expression of CDH2 and COL2A1 on the 21st day of treatment while PRP inhibited CTNNB1 and CCND1.
Collapse
Affiliation(s)
- Rizka Musdalifah Amsar
- School of Life Science & Technology, Institute of Technology Bandung, Bandung, West Java, Indonesia
| | - Anggraini Barlian
- School of Life Science & Technology, Institute of Technology Bandung, Bandung, West Java, Indonesia
| | - Hermawan Judawisastra
- Faculty of Mechanical & Aerospace of Engineering, Institute of Technology Bandung, Bandung, West Java, Indonesia
| | - Untung Ari Wibowo
- Faculty of Mechanical & Aerospace of Engineering, Institute of Technology Bandung, Bandung, West Java, Indonesia
| | | |
Collapse
|