1
|
Salehi S. A comprehensive review on using injectable chitosan microgels for osteochondral tissue repair. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:647-662. [PMID: 39460952 DOI: 10.1080/09205063.2024.2419715] [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: 04/10/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Restoring cartilage to healthy state is challenging due to low cell density and hence low regenerative capacity. The current platforms are not compatible with clinical translation and require dedicated handling of trained personnel. However, by engineering and implanting cell microaggregates in higher concentrations, efficient formation of new cartilage can be achieved, even in the absence of exogenous growth factors. Therefore, one-step surgeries are preferable for novel treatments and we need cell laden microgels allowing the formation of microaggregaets in vivo. Injectability is a key parameter for in situ forming the shape and minimally invasive clinical applications. Hydrogels as bioinks can restore damaged tissues to their primary shape. Chitosan is a polysaccharide derived from chitin with abundant usage in tissue engineering. This review highlights the use of chitosan as an injectable hydrogel for osteochondral defects. Several studies focused on encapsulating mesenchymal stem cells within chitosan hydrogels have been categorized and incorporating microfluidic devices has been identified in the forefront to form microgels. Additionally, the printability is another convenience of chitosan for using in 3D printing for cartilage tissue engineering which is described in this review.
Collapse
Affiliation(s)
- Sarah Salehi
- School of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran
| |
Collapse
|
2
|
Ghamrawi A, Basso R, Shakik N, Haddad L, Nasr Z, Harmouch C. Wharton's Jelly Mesenchymal Stem Cells: Shaping the Future of Osteoarthritis Therapy with Advancements in Chitosan-Hyaluronic Acid Scaffolds. Stem Cells Dev 2025; 34:1-16. [PMID: 39605205 DOI: 10.1089/scd.2024.0033] [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: 11/29/2024] Open
Abstract
This review explores the potential of Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) in cartilage regeneration and osteoarthritis treatment. It covers key factors influencing chondrogenesis, including growth factors, cytokines, and hypoxia, focusing on precise timing. The effectiveness of three-dimensional cultures and scaffold-based strategies in chondrogenic differentiation is discussed. Specific biomaterials such as chitosan and hyaluronic acid are highlighted for tissue engineering. The document reviews clinical applications, incorporating evidence from animal research and early trials and molecular and histological assessments of chondrogenic differentiation processes. It addresses challenges and strategies for optimizing MSC-derived chondrocyte therapy, emphasizing the immunomodulatory properties of these cells. The review concludes as a comprehensive road map for future research and clinical applications in regenerative medicine.
Collapse
Affiliation(s)
- Ahed Ghamrawi
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli, Lebanon
| | - Rasha Basso
- Department of Medical Laboratory Sciences, Faculty of Health Sciences University of Balamand, Beirut, Lebanon
| | - Nour Shakik
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli, Lebanon
| | - Lara Haddad
- Department of Medical Laboratory Sciences, Faculty of Health Sciences University of Balamand, Beirut, Lebanon
| | - Zeina Nasr
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli, Lebanon
| | - Chaza Harmouch
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli, Lebanon
| |
Collapse
|
3
|
Chen C, Xiong K, Li K, Zhou B, Cheng J, Zhu B, Zheng L, Zhao J. Identification of key genes involved in collagen hydrogel-induced chondrogenic differentiation of mesenchymal stem cells through transcriptome analysis: the role of m6A modification. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:43. [PMID: 39073623 PMCID: PMC11286723 DOI: 10.1007/s10856-024-06801-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 05/16/2024] [Indexed: 07/30/2024]
Abstract
Collagen hydrogel has been shown promise as an inducer for chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), contributing to the repair of cartilage defects. However, the precise molecular mechanism underlying this phenomenon remains poorly elucidated. Here, we induced chondrogenic differentiation of BMSCs using collagen hydrogel and identified 4451 differentially expressed genes (DEGs) through transcriptomic sequencing. Our analysis revealed that DEGs were enriched in the focal adhesion pathway, with a notable decrease in expression levels in the collagen hydrogel group compared to the control group. Protein-protein interaction network analysis suggested that actinin alpha 1 (ACTN1) and actinin alpha 4 (ACTN4), two proteins also involved in cytoskeletal recombination, may be crucial in collagen hydrogel-induced chondrogenic differentiation of BMSCs. Additionally, we found that N6-methyladenosine RNA methylation (m6A) modification was involved in collagen hydrogel-mediated chondrogenic differentiation, with fat mass and obesity-associated protein (FTO) implicated in regulating the expression of ACTN1 and ACTN4. These findings suggest that collagen hydrogel might regulate focal adhesion and actin cytoskeletal signaling pathways through down-regulation of ACTN1 and ACTN4 mRNA via FTO-mediated m6A modification, ultimately driving chondrogenic differentiation of BMSCs. In conclusion, our study provides valuable insights into the molecular mechanisms of collagen hydrogel-induced chondrogenic differentiation of BMSCs, which may aid in developing more effective strategies for cartilage regeneration.
Collapse
Affiliation(s)
- Chaotao Chen
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, 530021, China
- Department of Orthopaedics Trauma and HandSurgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| | - Kai Xiong
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, 530021, China
- Department of Orthopaedics Trauma and HandSurgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Department of Orthopaedics Trauma and HandSurgery, The Third Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530031, China
| | - Kanglu Li
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, 530021, China
- Department of Orthopaedics Trauma and HandSurgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| | - Bo Zhou
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, 530021, China
| | - Jianwen Cheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, 530021, China.
- Department of Orthopaedics Trauma and HandSurgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China.
| | - Bo Zhu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, 530021, China.
- International Joint Laboratory of Ministry of Education for Regeneration of Bone and Soft Tissues, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, 530021, China.
- International Joint Laboratory of Ministry of Education for Regeneration of Bone and Soft Tissues, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China.
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, 530021, China
- Department of Orthopaedics Trauma and HandSurgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- International Joint Laboratory of Ministry of Education for Regeneration of Bone and Soft Tissues, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
4
|
Yu X, Ma H, Wang Y, Hao J, Chen L, Gelinsky M, Wu C. Assembled/Disassembled Modular Scaffolds for Multicellular Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308126. [PMID: 38533956 DOI: 10.1002/adma.202308126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 02/28/2024] [Indexed: 03/28/2024]
Abstract
The behavior of tissue resident cells can be influenced by the spatial arrangement of cellular interactions. Therefore, it is of significance to precisely control the spatial organization of various cells within multicellular constructs. It remains challenging to construct a versatile multicellular scaffold with ordered spatial organization of multiple cell types. Herein, a modular multicellular tissue engineering scaffold with ordered spatial distribution of different cell types is constructed by assembling varying cell-laden modules. Interestingly, the modular scaffolds can be disassembled into individual modules to evaluate the specific contribution of each cell type in the system. Through assembling cell-laden modules, the macrophage-mesenchymal stem cell (MSC), endothelial cell-MSC, and chondrocyte-MSC co-culture models are successfully established. The in vitro results indicate that the intercellular cross-talk can promote the proliferation and differentiation of each cell type in the system. Moreover, MSCs in the modular scaffolds may regulate the behavior of chondrocytes through the nuclear factor of activated T-Cells (NFAT) signaling pathway. Furthermore, the modular scaffolds loaded with co-cultured chondrocyte-MSC exhibit enhanced regeneration ability of osteochondral tissue, compared with other groups. Overall, this work offers a promising strategy to construct a multicellular tissue engineering scaffold for the systematic investigation of intercellular cross-talk and complex tissue engineering.
Collapse
Affiliation(s)
- Xiaopeng Yu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hongshi Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yufeng Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Jianxin Hao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lei Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Michael Gelinsky
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine of Dresden University of Technology, Fetscherstr. 74, 01307, Dresden, Germany
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
5
|
Sulcanese L, Prencipe G, Canciello A, Cerveró-Varona A, Perugini M, Mauro A, Russo V, Barboni B. Stem-Cell-Driven Chondrogenesis: Perspectives on Amnion-Derived Cells. Cells 2024; 13:744. [PMID: 38727280 PMCID: PMC11083072 DOI: 10.3390/cells13090744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Regenerative medicine harnesses stem cells' capacity to restore damaged tissues and organs. In vitro methods employing specific bioactive molecules, such as growth factors, bio-inductive scaffolds, 3D cultures, co-cultures, and mechanical stimuli, steer stem cells toward the desired differentiation pathways, mimicking their natural development. Chondrogenesis presents a challenge for regenerative medicine. This intricate process involves precise modulation of chondro-related transcription factors and pathways, critical for generating cartilage. Cartilage damage disrupts this process, impeding proper tissue healing due to its unique mechanical and anatomical characteristics. Consequently, the resultant tissue often forms fibrocartilage, which lacks adequate mechanical properties, posing a significant hurdle for effective regeneration. This review comprehensively explores studies showcasing the potential of amniotic mesenchymal stem cells (AMSCs) and amniotic epithelial cells (AECs) in chondrogenic differentiation. These cells exhibit innate characteristics that position them as promising candidates for regenerative medicine. Their capacity to differentiate toward chondrocytes offers a pathway for developing effective regenerative protocols. Understanding and leveraging the innate properties of AMSCs and AECs hold promise in addressing the challenges associated with cartilage repair, potentially offering superior outcomes in tissue regeneration.
Collapse
Affiliation(s)
- Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Monia Perugini
- Department of Bioscience and Technology for Food, Agriculture, and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| |
Collapse
|
6
|
Banihashemian SA, Zamanlui Benisi S, Hosseinzadeh S, Shojaei S, Abbaszadeh HA. Chitosan/Hyaluronan and Alginate-Nanohydroxyapatite Biphasic Scaffold as a Promising Matrix for Osteoarthritis Disorders. Adv Pharm Bull 2024; 14:176-191. [PMID: 38585453 PMCID: PMC10997938 DOI: 10.34172/apb.2024.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/24/2023] [Accepted: 07/19/2023] [Indexed: 04/09/2024] Open
Abstract
Purpose Regenerative medicine offers new techniques for osteoarthritis (OA) disorders, especially while considering simultaneous chondral and subchondral regenerations. Methods Chitosan and hyaluronan were chemically bound as the chondral phase and the osteogenic layer was prepared with alginate and nano-hydroxyapatite (nHAP). These scaffolds were fixed by fibrin glue as a biphasic scaffold and then examined. Results Scanning electron microscopy (SEM) confirmed the porosity of 61.45±4.51 and 44.145±2.81 % for the subchondral and chondral layers, respectively. The composition analysis by energy dispersive X-ray (EDAX) indicated the various elements of both hydrogels. Also, their mechanical properties indicated that the highest modulus and resistance values corresponded to the biphasic hydrogel as 108.33±5.56 and 721.135±8.21 kPa, despite the same strain value as other groups. Their individual examinations demonstrated the proteoglycan synthesis of the chondral layer and also, the alkaline phosphatase (ALP) activity of the subchondral layer as 13.3±2.2 ng. After 21 days, the cells showed a mineralized surface and a polygonal phenotype, confirming their commitment to bone and cartilage tissues, respectively. Immunostaining of collagen I and II represented greater extracellular matrix (ECM) secretion in the biphasic composite group due to the paracrine effect of the two cell types on each other. Conclusion For the first time, the ability of this biphasic scaffold to regenerate both tissue types was evaluated and the results showed satisfactory cellular commitment to bone and cartilage tissues. Thus, this scaffold can be considered a new strategy for the preparation of implants for OA.
Collapse
Affiliation(s)
- Seyed Abdolvahab Banihashemian
- Advanced Medical Sciences and Technologies Department, Faculty of Biomedical Engineering, Central Tehran Branch Islamic Azad University, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Tehran Central Branch, Islamic Azad University, Tehran, Iran
| | - Simzar Hosseinzadeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahrokh Shojaei
- Islamic Azad University Central Tehran Branch, Department of Biomedical Engineering, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Zeng D, Chen Y, Liao Z, Wei G, Huang X, Liang R, Lu WW, Yi D, Chen Y. Cartilage organoids and osteoarthritis research: a narrative review. Front Bioeng Biotechnol 2023; 11:1278692. [PMID: 38026876 PMCID: PMC10666186 DOI: 10.3389/fbioe.2023.1278692] [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] [Received: 08/24/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common degenerative joint diseases, significantly impacting individuals and society. With the acceleration of global aging, the incidence of OA is increasing. The pathogenesis of osteoarthritis is not fully understood, and there is no effective way to alleviate the progression of osteoarthritis. Therefore, it is necessary to develop new disease models and seek new treatments for OA. Cartilage organoids are three-dimensional tissue masses that can simulate organ structure and physiological function and play an important role in disease modeling, drug screening, and regenerative medicine. This review will briefly analyze the research progress of OA, focusing on the construction and current development of cartilage organoids, and then describe the application of cartilage organoids in OA modeling, drug screening, and regeneration and repair of cartilage and bone defects. Finally, some challenges and prospects in the development of cartilaginous organoids are discussed.
Collapse
Affiliation(s)
- Daofu Zeng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yeping Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhidong Liao
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Guizheng Wei
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiajie Huang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Rongyuan Liang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - William W. Lu
- Department of Orthopedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Dan Yi
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yan Chen
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
8
|
Zhang W, Zha K, Hu W, Xiong Y, Knoedler S, Obed D, Panayi AC, Lin Z, Cao F, Mi B, Liu G. Multifunctional hydrogels: advanced therapeutic tools for osteochondral regeneration. Biomater Res 2023; 27:76. [PMID: 37542353 PMCID: PMC10403923 DOI: 10.1186/s40824-023-00411-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/05/2023] [Indexed: 08/06/2023] Open
Abstract
Various joint pathologies such as osteochondritis dissecans, osteonecrosis, rheumatic disease, and trauma, may result in severe damage of articular cartilage and other joint structures, ranging from focal defects to osteoarthritis (OA). The osteochondral unit is one of the critical actors in this pathophysiological process. New approaches and applications in tissue engineering and regenerative medicine continue to drive the development of OA treatment. Hydrogel scaffolds, a component of tissue engineering, play an indispensable role in osteochondral regeneration. In this review, tissue engineering strategies regarding osteochondral regeneration were highlighted and summarized. The application of hydrogels for osteochondral regeneration within the last five years was evaluated with an emphasis on functionalized physical and chemical properties of hydrogel scaffolds, functionalized delivery hydrogel scaffolds as well as functionalized intelligent response hydrogel scaffolds. Lastly, to serve as guidance for future efforts in the creation of bioinspired hydrogel scaffolds, a succinct summary and new views for specific mechanisms, applications, and existing limitations of the newly designed functionalized hydrogel scaffolds were offered.
Collapse
Affiliation(s)
- Wenqian Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Kangkang Zha
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Weixian Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
| | - Doha Obed
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Adriana C Panayi
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, 67071, Ludwigshafen/Rhine, Germany
| | - Ze Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Faqi Cao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore.
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
9
|
Decarli MC, Seijas‐Gamardo A, Morgan FLC, Wieringa P, Baker MB, Silva JVL, Moraes ÂM, Moroni L, Mota C. Bioprinting of Stem Cell Spheroids Followed by Post-Printing Chondrogenic Differentiation for Cartilage Tissue Engineering. Adv Healthc Mater 2023; 12:e2203021. [PMID: 37057819 PMCID: PMC11468754 DOI: 10.1002/adhm.202203021] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/13/2023] [Indexed: 04/15/2023]
Abstract
Cartilage tissue presents low self-repair capability and lesions often undergo irreversible progression. Structures obtained by tissue engineering, such as those based in extrusion bioprinting of constructs loaded with stem cell spheroids may offer valuable alternatives for research and therapeutic purposes. Human mesenchymal stromal cell (hMSC) spheroids can be chondrogenically differentiated faster and more efficiently than single cells. This approach allows obtaining larger tissues in a rapid, controlled and reproducible way. However, it is challenging to control tissue architecture, construct stability, and cell viability during maturation. Herein, this work reports a reproducible bioprinting process followed by a successful post-bioprinting chondrogenic differentiation procedure using large quantities of hMSC spheroids encapsulated in a xanthan gum-alginate hydrogel. Multi-layered constructs are bioprinted, ionically crosslinked, and post chondrogenically differentiated for 28 days. The expression of glycosaminoglycan, collagen II and IV are observed. After 56 days in culture, the bioprinted constructs are still stable and show satisfactory cell metabolic activity with profuse extracellular matrix production. These results show a promising procedure to obtain 3D models for cartilage research and ultimately, an in vitro proof-of-concept of their potential use as stable chondral tissue implants.
Collapse
Affiliation(s)
- Monize Caiado Decarli
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
- Department of Engineering of Biomaterials and of BioprocessesSchool of Chemical EngineeringUniversity of Campinas ‐ UNICAMPAv. Albert Einstein, 500, Cidade Universitária “Zeferino Vaz”CampinasSP13083‐852Brazil
| | - Adrián Seijas‐Gamardo
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Francis L. C. Morgan
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Paul Wieringa
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Matthew B. Baker
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Jorge Vicente L. Silva
- Three‐Dimensional Technologies Research GroupCTI Renato ArcherRodovia Dom Pedro I SP‐65, Km 143,6 ‐ AmaraisCampinasSP13069‐901Brazil
| | - Ângela Maria Moraes
- Department of Engineering of Biomaterials and of BioprocessesSchool of Chemical EngineeringUniversity of Campinas ‐ UNICAMPAv. Albert Einstein, 500, Cidade Universitária “Zeferino Vaz”CampinasSP13083‐852Brazil
| | - Lorenzo Moroni
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Carlos Mota
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| |
Collapse
|
10
|
Goh D, Yang Y, Lee EH, Hui JHP, Yang Z. Managing the Heterogeneity of Mesenchymal Stem Cells for Cartilage Regenerative Therapy: A Review. Bioengineering (Basel) 2023; 10:bioengineering10030355. [PMID: 36978745 PMCID: PMC10045936 DOI: 10.3390/bioengineering10030355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.
Collapse
Affiliation(s)
- Doreen Goh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Yanmeng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
- Correspondence: ; Tel.: +65-6516-5398
| |
Collapse
|
11
|
Zhang C, Wang G, Lin H, Shang Y, Liu N, Zhen Y, An Y. Cartilage 3D bioprinting for rhinoplasty using adipose-derived stem cells as seed cells: Review and recent advances. Cell Prolif 2023; 56:e13417. [PMID: 36775884 PMCID: PMC10068946 DOI: 10.1111/cpr.13417] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/10/2023] [Accepted: 01/18/2023] [Indexed: 02/14/2023] Open
Abstract
Nasal deformities due to various causes affect the aesthetics and use of the nose, in which case rhinoplasty is necessary. However, the lack of cartilage for grafting has been a major problem and tissue engineering seems to be a promising solution. 3D bioprinting has become one of the most advanced tissue engineering methods. To construct ideal cartilage, bio-ink, seed cells, growth factors and other methods to promote chondrogenesis should be considered and weighed carefully. With continuous progress in the field, bio-ink choices are becoming increasingly abundant, from a single hydrogel to a combination of hydrogels with various characteristics, and more 3D bioprinting methods are also emerging. Adipose-derived stem cells (ADSCs) have become one of the most popular seed cells in cartilage 3D bioprinting, owing to their abundance, excellent proliferative potential, minimal morbidity during harvest and lack of ethical considerations limitations. In addition, the co-culture of ADSCs and chondrocytes is commonly used to achieve better chondrogenesis. To promote chondrogenic differentiation of ADSCs and construct ideal highly bionic tissue-engineered cartilage, researchers have used a variety of methods, including adding appropriate growth factors, applying biomechanical stimuli and reducing oxygen tension. According to the process and sequence of cartilage 3D bioprinting, this review summarizes and discusses the selection of hydrogel and seed cells (centered on ADSCs), the design of printing, and methods for inducing the chondrogenesis of ADSCs.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Guanhuier Wang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Hongying Lin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Yujia Shang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China.,Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Na Liu
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China.,Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| |
Collapse
|
12
|
Aydin SM. Blood Products. Regen Med 2023. [DOI: 10.1007/978-3-030-75517-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
13
|
Liu Q, Dai W, Gao Y, Dong L, Jia H, Li S, Guo L, Fan Y, Zhang X. The synergistic regulation of chondrogenesis by collagen-based hydrogels and cell co-culture. Acta Biomater 2022; 154:194-211. [PMID: 36309191 DOI: 10.1016/j.actbio.2022.10.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/19/2022] [Accepted: 10/07/2022] [Indexed: 01/24/2023]
Abstract
The suitable seeding cells and scaffolds are very important for tissue engineering to create functional cartilage. Although the physicochemical properties of scaffold and co-culture system of mesenchymal stem cells (MSCs) and chondrocytes could affect functional properties of engineered cartilage tissues respectively, the combined effects of them on chondrogenesis is currently unknown. Herein, methacrylated collagen (CMA30 and CMA80) hydrogels with different degradation rate and stiffness were prepared. The MSCs and chondrocytes were co-cultured or monocultured in collagen, CMA30 and CMA80 hydrogels in vitro or in vivo. The results demonstrated that cell spreading and proliferation was regulated by degradation rate and stiffness of hydrogels. Compared to single MSCs culture, co-culture cells in all collagen-based hydrogels significantly improved chondrogenesis. CMA30 hydrogel with moderate degradation rate and low storage modulus was the most effective for co-culture system to promote chondrogenesis compared to Col and CMA80 hydrogel in vitro culture, while there was no obvious difference between CMA30 and CMA80 hydrogel in vivo. Furthermore, the intercellular substance exchange was very important for co-culture system to maintain the positive effect on chondrogenesis. Overall, the current study highlights the synergistic effects of the physicochemical properties of collagen-based hydrogel and co-culture system on cartilage formation. STATEMENT OF SIGNIFICANCE: Scaffolds and cells play a key role in cartilage tissue engineering. The combined effects of physicochemical properties of collagen hydrogels and co-culture system (MSCs and chondrocytes) on chondrogenesis is unknown. In contrast to the studies that investigated the effect of single factor (scaffolds or cells) on cartilage formation, this manuscript explored the synergistic regulation of both scaffold properties and biological factors on chondrogenesis, and provided a promising strategy for cartilage tissue engineering.
Collapse
Affiliation(s)
- Qingli Liu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Wenling Dai
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Yongli Gao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Longpeng Dong
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Hengxing Jia
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Shikui Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Likun Guo
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China.
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| |
Collapse
|
14
|
de Jong WC, Penn TW. Single-Surgery Co-Implantation of Autologous Primary Articular Chondrocytes with Bone Marrow Cells in the Treatment of Articular Cartilage Lesions: Observations from the Operating Room on Tissue Input and Cell Output. Cartilage 2022; 13:32-42. [PMID: 36278377 PMCID: PMC9924989 DOI: 10.1177/19476035221132259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Single-surgery variants of autologous chondrocyte implantation to repair cartilage are emerging, but practical data on such procedures are scarce. We set out to describe a 1-stage autologous chondrocyte co-implantation procedure and include quantitative characteristics of the biopsy tissues collected and of the cells obtained from those tissues in the operating room. DESIGN Data concerning patient age, articular cartilage lesion size and location, as well as measurements of cartilage biopsy mass, bone marrow aspirate volume, and the cell yields harvested from those biopsies were intraoperatively collected for 141 patients. RESULTS The mean patient age was 35.7 ± 9.8 years, and the mean total lesion size was 4.0 ± 3.1 cm2. Cartilage biopsy mass ranged from 0.19 to 2.0 gram and provided a mean yield of 1.23 × 106 chondrocytes/gram. Bone marrow aspirate volume ranged from 7.2 to 62 milliliters and provided a mean yield of 7.18 × 106 mononuclear cells/mL. The cell yields did not correlate with patient age, which ranged from 12 to 57 years. The mean primary chondrocyte supply was 272 thousand per cm2 of lesion, ranging from 10.4 thousand to 1.07 million per cm2. The total cell supply was kept at 9 million cells per cm2 of lesion by adding mononuclear cells to the chondrocytes. The mean tissue processing time was 100 ± 19 minutes, which was frequently used to perform concurrent interventions. CONCLUSION Single-surgery co-implantation of clinically relevant numbers of autologous primary articular chondrocytes and bone marrow cells is feasible for a wide range of ages and lesion sizes.
Collapse
Affiliation(s)
- Willem Cornelis de Jong
- Cartilage Repair Systems, LLC, New
York, NY, USA,Willem Cornelis de Jong, c/o Cartilage
Repair Systems B.V., Breullaan 1, 3971 NG Driebergen-Rijsenburg, The
Netherlands.
| | | |
Collapse
|
15
|
He Z, Liu M, Zhang Q, Tian Y, Wang L, Yan X, Ren D, Yuan X. Wnt/β-catenin signaling pathway is activated in the progress of mandibular condylar cartilage degeneration and subchondral bone loss induced by overloaded functional orthopedic force (OFOF). Heliyon 2022; 8:e10847. [PMID: 36262297 PMCID: PMC9573886 DOI: 10.1016/j.heliyon.2022.e10847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/11/2022] [Accepted: 09/26/2022] [Indexed: 11/24/2022] Open
Abstract
Objective To explore the role of Wnt/β-catenin signaling pathway in the pathogenesis and progression of temporomandibular joint osteoarthritis (TMJ OA) caused by overloaded force. Materials and methods We generated a rat model of forward mandibular extension device to induce TMJ OA by overloaded force. Condylar cartilage samples were collected at 2wk, 4wk, and 8wk after appliances were installed. Changes of the condylar cartilage and subchondral bone were evaluated by hematoxylin and eosin (HE), Safranin O and Fast Green staining (SO&FG), micro-CT, tartrate resistant acid phosphatase (TRAP) staining. The expression levels of β-catenin, COL-2, MMP3 and sclerostin (SOST) were detected by immunohistochemistry (IHC) and PCR. Results HE, SO&FG, micro-CT, OARSI and Mankin scores showed that the condyle cartilage layer was significantly thinner and proteoglycan loss in the overloded group. TRAP staining exhibited that the number of positive osteoclasts increased and OPG level decreased in the overload group. IHC, PCR showed that the expression of COL2 and SOST decreased, while MMP3 and β-catenin increased in the overload group. Conclusion Wnt/β-catenin signaling pathway is activated in the progress of mandibular condylar cartilage degeneration and subchondral bone loss induced by overloaded functional orthopedic force (OFOF)
Collapse
|
16
|
Zheng K, Ma Y, Chiu C, Pang Y, Gao J, Zhang C, Du D. Co-culture pellet of human Wharton's jelly mesenchymal stem cells and rat costal chondrocytes as a candidate for articular cartilage regeneration: in vitro and in vivo study. Stem Cell Res Ther 2022; 13:386. [PMID: 35907866 PMCID: PMC9338579 DOI: 10.1186/s13287-022-03094-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Seeding cells are key factors in cell-based cartilage tissue regeneration. Monoculture of either chondrocyte or mesenchymal stem cells has several limitations. In recent years, co-culture strategies have provided potential solutions. In this study, directly co-cultured rat costal chondrocytes (CCs) and human Wharton's jelly mesenchymal stem (hWJMSCs) cells were evaluated as a candidate to regenerate articular cartilage. METHODS Rat CCs are directly co-cultured with hWJMSCs in a pellet model at different ratios (3:1, 1:1, 1:3) for 21 days. The monoculture pellets were used as controls. RT-qPCR, biochemical assays, histological staining and evaluations were performed to analyze the chondrogenic differentiation of each group. The 1:1 ratio co-culture pellet group together with monoculture controls were implanted into the osteochondral defects made on the femoral grooves of the rats for 4, 8, 12 weeks. Then, macroscopic and histological evaluations were performed. RESULTS Compared to rat CCs pellet group, 3:1 and 1:1 ratio group demonstrated similar extracellular matrix production but less hypertrophy intendency. Immunochemistry staining found the consistent results. RT-PCR analysis indicated that chondrogenesis was promoted in co-cultured rat CCs, while expressions of hypertrophic genes were inhibited. However, hWJMSCs showed only slightly improved in chondrogenesis but not significantly different in hypertrophic expressions. In vivo experiments showed that all the pellets filled the defects but co-culture pellets demonstrated reduced hypertrophy, better surrounding cartilage integration and appropriate subchondral bone remodeling. CONCLUSION Co-culture of rat CCs and hWJMSCs demonstrated stable chondrogenic phenotype and decreased hypertrophic intendency in both vitro and vivo. These results suggest this co-culture combination as a promising candidate in articular cartilage regeneration.
Collapse
Affiliation(s)
- Kaiwen Zheng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Yiyang Ma
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Cheng Chiu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Yidan Pang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Junjie Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Dajiang Du
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
17
|
Kindlin-2 Promotes Chondrogenesis and Ameliorates IL-1beta-Induced Inflammation in Chondrocytes Cocultured with BMSCs in the Direct Contact Coculture System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3156245. [PMID: 35450413 PMCID: PMC9018182 DOI: 10.1155/2022/3156245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/17/2022]
Abstract
The osteoarthritis caused by trauma or inflammation is associated with severe patient morbidity and economic burden. Accumulating studies are focusing on the repair of articular cartilage defects by constructing tissue-engineered cartilage. Recent evidence suggests that optimizing the source and quality of seed cells is one of the key points of cartilage tissue engineering. In this study, we demonstrated that Kindlin-2 and its activated PI3K/AKT signaling played an essential role in promoting extracellular matrix (ECM) secretion and ameliorating IL-1beta-induced inflammation in chondrocytes cocultured with bone marrow stem cells (BMSCs). In vivo experiments revealed that coculture significantly promoted hyaline cartilage regeneration. In vitro studies further uncovered that chondrocytes cocultured with BMSCs in the direct contact coculture system upregulated Kindlin-2 expression and subsequently activated the PI3K/AKT signaling pathway, which not only increases Sox9 and Col2 expression but also restores mitochondrial membrane potential and reduces ROS levels and apoptosis under inflammatory conditions. Overall, our findings indicated that direct contact BMSC-chondrocyte coculture system could promote chondrogenesis, and identified Kindlin-2 represents a key regulator in this process.
Collapse
|
18
|
Posniak S, Chung JHY, Liu X, Mukherjee P, Gambhir S, Khansari A, Wallace GG. Bioprinting of Chondrocyte Stem Cell Co-Cultures for Auricular Cartilage Regeneration. ACS OMEGA 2022; 7:5908-5920. [PMID: 35224351 PMCID: PMC8867593 DOI: 10.1021/acsomega.1c06102] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/21/2022] [Indexed: 05/06/2023]
Abstract
Advances in 3D bioprinting allows not only controlled deposition of cells or cell-laden hydrogels but also flexibility in creating constructs that match the anatomical features of the patient. This is especially the case for reconstructing the pinna (ear), which is a large feature of the face and made from elastic cartilage that primarily relies on diffusion for nutrient transfer. The selection of cell lines for reconstructing this cartilage becomes a crucial step in clinical translation. Chondrocytes and mesenchymal stem cells are both studied extensively in the area of cartilage regeneration as they are capable of producing cartilage in vitro. However, such monoculture systems involve unfavorable processes and produce cartilage with suboptimal characteristics. Co-cultures of these cell types are known to alleviate these limitations to produce synergically active chondrocytes and cartilage. The current study utilized a 3D bioprinted scaffold made from combined gelatine methacryloyl and methacrylated hyaluronic acid (GelMA/HAMA) to interrogate monocultures and co-cultures of human septal chondrocytes (primary chondrocytes, PCs) and human bone marrow-derived mesenchymal stem cells (BM-hMSCs). This study is also the first to examine co-cultures of healthy human chondrocytes with human BM-hMSCs encapsulated in GelMA/HAMA bioprinted scaffolds. Findings revealed that the combination of MSCs and PCs not only yielded cell proliferation that mimicked MSCs but also produced chondrogenic expressions that mimicked PCs. These findings suggested that co-cultures of BM-hMSCs and healthy septal PCs can be employed to replace monocultures in chondrogenic studies for cartilage regeneration in this model. The opportunity for MSCs used to replace PCs alleviates the requirement of large cartilage biopsies that would otherwise be needed for sufficient cell numbers and therefore can be employed for clinical applications.
Collapse
Affiliation(s)
- Steven Posniak
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Johnson H. Y. Chung
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Xiao Liu
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Payal Mukherjee
- ENT
Research Lead, RPA Institute of Academic Surgery, Sydney Royal Prince Alfred Hospital, Camperdown, New South Wales 2050, Australia
| | - Sanjeev Gambhir
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Afsaneh Khansari
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Gordon G. Wallace
- ARC
Centre of Excellence for Electromaterials Science, Intelligent Polymer
Research Institute, University of Wollongong, Wollongong, New South Wales 2522, Australia
| |
Collapse
|
19
|
The essential anti-angiogenic strategies in cartilage engineering and osteoarthritic cartilage repair. Cell Mol Life Sci 2022; 79:71. [PMID: 35029764 PMCID: PMC9805356 DOI: 10.1007/s00018-021-04105-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/01/2021] [Accepted: 12/18/2021] [Indexed: 01/16/2023]
Abstract
In the cartilage matrix, complex interactions occur between angiogenic and anti-angiogenic components, growth factors, and environmental stressors to maintain a proper cartilage phenotype that allows for effective load bearing and force distribution. However, as seen in both degenerative disease and tissue engineering, cartilage can lose its vascular resistance. This vascularization then leads to matrix breakdown, chondrocyte apoptosis, and ossification. Research has shown that articular cartilage inflammation leads to compromised joint function and decreased clinical potential for regeneration. Unfortunately, few articles comprehensively summarize what we have learned from previous investigations. In this review, we summarize our current understanding of the factors that stabilize chondrocytes to prevent terminal differentiation and applications of these factors to rescue the cartilage phenotype during cartilage engineering and osteoarthritis treatment. Inhibiting vascularization will allow for enhanced phenotypic stability so that we are able to develop more stable implants for cartilage repair and regeneration.
Collapse
|
20
|
Cao L, Tong Y, Wang X, Zhang Q, Qi Y, Zhou C, Yu X, Wu Y, Miao X. Effect of Amniotic Membrane/Collagen-Based Scaffolds on the Chondrogenic Differentiation of Adipose-Derived Stem Cells and Cartilage Repair. Front Cell Dev Biol 2021; 9:647166. [PMID: 34900977 PMCID: PMC8657407 DOI: 10.3389/fcell.2021.647166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/20/2021] [Indexed: 11/24/2022] Open
Abstract
Objectives: Repairing articular cartilage damage is challenging. Clinically, tissue engineering technology is used to induce stem cell differentiation and proliferation on biological scaffolds to repair defective joints. However, no ideal biological scaffolds have been identified. This study investigated the effects of amniotic membrane/collagen scaffolds on the differentiation of adipose-derived stem cells (ADSCs) and articular cartilage repair. Methods: Adipose tissue of New Zealand rabbits was excised, and ADSCs were isolated and induced for differentiation. An articular cartilage defect model was constructed to identify the effect of amniotic membrane/collagen scaffolds on cartilage repair. Cartilage formation was analyzed by imaging and toluene blue staining. Knee joint recovery in rabbits was examined using hematoxylin and eosin, toluidine, safranine, and immunohistochemistry at 12 weeks post-operation. Gene expression was examined using ELISA, RT-PCR, Western blotting, and immunofluorescence. Results: The adipose tissue was effectively differentiated into ADSCs, which further differentiated into chondrogenic, osteogenic, and lipogenic lineages after 3 weeks’ culture in vitro. Compared with platelet-rich plasmon (PRP) scaffolds, the amniotic membrane scaffolds better promoted the growth and differentiation of ADSCs. Additionally, scaffolds containing the PRP and amniotic membrane efficiently enhanced the osteogenic differentiation of ADSCs. The levels of COL1A1, COL2A1, COL10A1, SOX9, and ACAN in ADSCs + amniotic membrane + PRP group were significantly higher than the other groups both in vitro and in vivo. The Wakitani scores of the ADSC + amniotic membrane + PRP group were lower than that in ADSC + PRP (4.4 ± 0.44**), ADSC + amniotic membrane (2.63 ± 0.38**), and control groups (6.733 ± 0.21) at week 12 post-operation. Osteogenesis in rabbits of the ADSC + amniotic membrane + PRP group was significantly upregulated when compared with other groups. Amniotic membranes significantly promoted the expression of cartilage regeneration-related factors (SOX6, SOX9, RUNX2, NKX3-2, MEF2C, and GATA4). The ADSC + PRP + amniotic membrane group exhibited the highest levels of TGF-β, PDGF, and FGF while exhibiting the lowest level of IL-1β, IL6, and TNF-α in articular cavity. Conclusion: Amniotic membrane/collagen combination-based scaffolds promoted the proliferation and cartilage differentiation of ADSCs, and may provide a new treatment paradigm for patients with cartilage injury.
Collapse
Affiliation(s)
- Le Cao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yuling Tong
- Department of General Practice, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Wang
- Shaoxing Shangyu Hospital of Traditional Chinese medicine, Shaoxing, China
| | - Qiang Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yiying Qi
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Chenhe Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Xinning Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yongping Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Xudong Miao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| |
Collapse
|
21
|
Maciulaitis J, Miskiniene M, Rekštytė S, Bratchikov M, Darinskas A, Simbelyte A, Daunoras G, Laurinaviciene A, Laurinavicius A, Gudas R, Malinauskas M, Maciulaitis R. Osteochondral Repair and Electromechanical Evaluation of Custom 3D Scaffold Microstructured by Direct Laser Writing Lithography. Cartilage 2021; 13:615S-625S. [PMID: 31072136 PMCID: PMC8804810 DOI: 10.1177/1947603519847745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE The objective of this study was to assess a novel 3D microstructured scaffold seeded with allogeneic chondrocytes (cells) in a rabbit osteochondral defect model. DESIGN Direct laser writing lithography in pre-polymers was employed to fabricate custom silicon-zirconium containing hybrid organic-inorganic (HOI) polymer SZ2080 scaffolds of a predefined morphology. Hexagon-pored HOI scaffolds were seeded with chondrocytes (cells), and tissue-engineered cartilage biocompatibility, potency, efficacy, and shelf-life in vitro was assessed by morphological, ELISA (enzyme-linked immunosorbent assay) and PCR (polymerase chain reaction) analysis. Osteochondral defect was created in the weight-bearing area of medial femoral condyle for in vivo study. Polymerized fibrin was added to every defect of 5 experimental groups. Cartilage repair was analyzed after 6 months using macroscopical (Oswestry Arthroscopy Score [OAS]), histological, and electromechanical quantitative potential (QP) scores. Collagen scaffold (CS) was used as a positive comparator for in vitro and in vivo studies. RESULTS Type II collagen gene upregulation and protein secretion was maintained up to 8 days in seeded HOI. In vivo analysis revealed improvement in all scaffold treatment groups. For the first time, electromechanical properties of a cellular-based scaffold were analyzed in a preclinical study. Cell addition did not enhance OAS but improved histological and QP scores in HOI groups. CONCLUSIONS HOI material is biocompatible for up to 8 days in vitro and is supportive of cartilage formation at 6 months in vivo. Electromechanical measurement offers a reliable quality assessment of repaired cartilage.
Collapse
Affiliation(s)
- Justinas Maciulaitis
- Institute of Sports, Lithuanian
University of Health Sciences, Kaunas, Lithuania,Justinas Maciulaitis, Institute of Sports,
Lithuanian University of Health Sciences, Tilzes st. 18, 9 House, Kaunas 47181,
Lithuania.
| | - Milda Miskiniene
- Laboratory of Immunology, National
Institute of Cancer, Vilnius, Lithuania
| | - Sima Rekštytė
- Laser Research Center, Faculty of
Physics, Vilnius University, Vilnius, Lithuania
| | - Maksim Bratchikov
- Department of Physiology, Biochemistry,
Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of
Medicine, Vilnius University, Vilnius, Lithuania
| | - Adas Darinskas
- Laboratory of Immunology, National
Institute of Cancer, Vilnius, Lithuania
| | - Agne Simbelyte
- National Center of Pathology, Affiliate
of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Gintaras Daunoras
- Non-infectious Disease Department,
Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Aida Laurinaviciene
- National Center of Pathology, Affiliate
of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Arvydas Laurinavicius
- National Center of Pathology, Affiliate
of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Rimtautas Gudas
- Institute of Sports, Lithuanian
University of Health Sciences, Kaunas, Lithuania
| | | | - Romaldas Maciulaitis
- Institute of Physiology and
Pharmacology, Medical Academy, Lithuanian University of Health Sciences, Kaunas,
Lithuania
| |
Collapse
|
22
|
Nogueira LFB, Maniglia BC, Buchet R, Millán JL, Ciancaglini P, Bottini M, Ramos AP. Three-dimensional cell-laden collagen scaffolds: From biochemistry to bone bioengineering. J Biomed Mater Res B Appl Biomater 2021; 110:967-983. [PMID: 34793621 DOI: 10.1002/jbm.b.34967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/05/2021] [Accepted: 10/30/2021] [Indexed: 12/22/2022]
Abstract
The bones can be viewed as both an organ and a material. As an organ, the bones give structure to the body, facilitate skeletal movement, and provide protection to internal organs. As a material, the bones consist of a hybrid organic/inorganic three-dimensional (3D) matrix, composed mainly of collagen, noncollagenous proteins, and a calcium phosphate mineral phase, which is formed and regulated by the orchestrated action of a complex array of cells including chondrocytes, osteoblasts, osteocytes, and osteoclasts. The interactions between cells, proteins, and minerals are essential for the bone functions under physiological loading conditions, trauma, and fractures. The organization of the bone's organic and inorganic phases stands out for its mechanical and biological properties and has inspired materials research. The objective of this review is to fill the gaps between the physical and biological characteristics that must be achieved to fabricate scaffolds for bone tissue engineering with enhanced performance. We describe the organization of bone tissue highlighting the characteristics that have inspired the development of 3D cell-laden collagenous scaffolds aimed at replicating the mechanical and biological properties of bone after implantation. The role of noncollagenous macromolecules in the organization of the collagenous matrix and mineralization ability of entrapped cells has also been reviewed. Understanding the modulation of cell activity by the extracellular matrix will ultimately help to improve the biological performance of 3D cell-laden collagenous scaffolds used for bone regeneration and repair as well as for in vitro studies aimed at unravelling physiological and pathological processes occurring in the bone.
Collapse
Affiliation(s)
- Lucas Fabricio Bahia Nogueira
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil.,Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Bianca C Maniglia
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| | - Rene Buchet
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Ana Paula Ramos
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), São Paulo, Brazil
| |
Collapse
|
23
|
Roberti Di Sarsisa T, Fiore M, Coco V, Govoni M, Vivarelli L, Rani N, Del Piccolo N, Dallari D. Fresh Osteochondral Allograft Transplantation in Osteochondritis Dissecans in the Knee Joint. Life (Basel) 2021; 11:life11111205. [PMID: 34833081 PMCID: PMC8622509 DOI: 10.3390/life11111205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
Osteochondritis dissecans (OCD) is a chronic and painful joint condition that can occur from childhood through to adult life. Microtrauma, vascular insufficiency, or abnormal endochondral ossification are the most common causes of OCD. Reconstructive techniques for OCD of the knee are typically necessary when either non-operative or reparative/regenerative operative treatments fail, or when the OCD is irreversible. To analyze the clinical outcomes and failure rates of fresh osteochondral allograft transplantation (FOCA) used as a reconstructive strategy in OCD patients, an in-depth search was carried out on the PubMed, Scopus, and Web of Science databases concerning the existing evidence related to the use of FOCA for OCD patients in the knee joint. A total of 646 studies were found through the search and 2 studies were added after a cross-referenced examination of the articles within the bibliography. Six studies with a total of 303 OCD lesions treated with FOCA, with a mean follow-up of 6.3 years, were included. Although a limited number of low-level evidence studies on this topic are available in previous research, satisfactory clinical results and survival rates of the reconstruction are reported. However, to better define the real advantages of FOCA in the healing process of OCD lesions, comparative studies with different techniques are needed.
Collapse
|
24
|
Liu H, Liu P. Kartogenin Promotes the BMSCs Chondrogenic Differentiation in Osteoarthritis by Down-Regulation of miR-145-5p Targeting Smad4 Pathway. Tissue Eng Regen Med 2021; 18:989-1000. [PMID: 34669172 DOI: 10.1007/s13770-021-00390-9] [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: 06/12/2021] [Revised: 08/02/2021] [Accepted: 08/19/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Transplantation of mesenchymal stem cells (MSCs) is a potential therapeutic strategy for cartilage degeneration of osteoarthritis (OA). But controlling chondrogenic differentiation of the implanted MSCs in the joints remains a challenge. The role of kartogenin (KGN) for chondrogenesis of MSCs has been widely reported, however, the mechanism of chondrogenesis has not been elucidated in OA. METHODS In this study, we investigated the miR-145-5p, TGF-β, Samd4, and p-stat3/stat3 expression in cartilage of OA patients and bone marrow mesenchymal stem cells (BMSCs) treated with KGN or miR-145-5p inhibitor. In addition, the cell proliferation and chondrogenic differentiation in vitro and in vivo of BMSCs treated with KGN was also detected. RESULTS In OA patients, the expression of miR-145-5p was up-regulated, and the expression of TGF-β, Samd4, and p-stat3/stat3 was inhibited. When the BMSCs treated with miR-145-5p inhibitor, the expression of TGF-β, Samd4, and p-stat3/stat3 was also significantly up-regulated. KGN-treated BMSCs had better proliferation and chondrogenic differentiation by up-regulating the expression of Sox 9, Col-2a1, aggrecan in vitro and in OA by down-regulation of miR-145-5p targeting Smad4 pathway. Moreover, intra-articular injection of KGN-treated BMSCs had a better pain relief effect in OA. CONCLUSION The double effect on cartilage protection and pain relief indicates a great potential of intra-articular injection of KGN-treated BMSCs for the treatment of OA.
Collapse
Affiliation(s)
- Huimin Liu
- Department of Paediatrics, Liyuan Hospital Affiliated To Tongji Medical College of Huazhong University of Science and Technology, 43006, Wuhan, People's Republic of China
| | - Ping Liu
- Department of Orthopaedics, Liyuan Hospital Affiliated To Tongji Medical College of Huazhong University of Science and Technology, 43006, Wuhan, People's Republic of China.
| |
Collapse
|
25
|
Li Q, Xu S, Feng Q, Dai Q, Yao L, Zhang Y, Gao H, Dong H, Chen D, Cao X. 3D printed silk-gelatin hydrogel scaffold with different porous structure and cell seeding strategy for cartilage regeneration. Bioact Mater 2021; 6:3396-3410. [PMID: 33842736 PMCID: PMC8010633 DOI: 10.1016/j.bioactmat.2021.03.013] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
Hydrogel scaffolds are attractive for tissue defect repair and reorganization because of their human tissue-like characteristics. However, most hydrogels offer limited cell growth and tissue formation ability due to their submicron- or nano-sized gel networks, which restrict the supply of oxygen, nutrients and inhibit the proliferation and differentiation of encapsulated cells. In recent years, 3D printed hydrogels have shown great potential to overcome this problem by introducing macro-pores within scaffolds. In this study, we fabricated a macroporous hydrogel scaffold through horseradish peroxidase (HRP)-mediated crosslinking of silk fibroin (SF) and tyramine-substituted gelatin (GT) by extrusion-based low-temperature 3D printing. Through physicochemical characterization, we found that this hydrogel has excellent structural stability, suitable mechanical properties, and an adjustable degradation rate, thus satisfying the requirements for cartilage reconstruction. Cell suspension and aggregate seeding methods were developed to assess the inoculation efficiency of the hydrogel. Moreover, the chondrogenic differentiation of stem cells was explored. Stem cells in the hydrogel differentiated into hyaline cartilage when the cell aggregate seeding method was used and into fibrocartilage when the cell suspension was used. Finally, the effect of the hydrogel and stem cells were investigated in a rabbit cartilage defect model. After implantation for 12 and 16 weeks, histological evaluation of the sections was performed. We found that the enzymatic cross-linked and methanol treatment SF5GT15 hydrogel combined with cell aggregates promoted articular cartilage regeneration. In summary, this 3D printed macroporous SF-GT hydrogel combined with stem cell aggregates possesses excellent potential for application in cartilage tissue repair and regeneration.
Collapse
Affiliation(s)
- Qingtao Li
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- School of Medicine, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Zhongshan Institute of Modern Industrial Technology of SCUT, Zhongshan, Guangdong, 528437, China
| | - Sheng Xu
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, GuangDong, 510641, China
| | - Qi Feng
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, GuangDong, 510641, China
| | - Qiyuan Dai
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, GuangDong, 510641, China
| | - Longtao Yao
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, GuangDong, 510641, China
| | - Yichen Zhang
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, GuangDong, 510641, China
| | - Huichang Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- School of Medicine, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, GuangDong, 510641, China
| | - Hua Dong
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, GuangDong, 510641, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing, 100035, China
| | - Xiaodong Cao
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, GuangDong, 510641, China
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, GuangDong, 510641, China
- Zhongshan Institute of Modern Industrial Technology of SCUT, Zhongshan, Guangdong, 528437, China
| |
Collapse
|
26
|
Wang Q, Chen Y, Shen X, Chen J, Li Y. Intra-Articular Injection of miR-29a-3p of BMSCs Promotes Cartilage Self-Repairing and Alleviates Pain in the Rat Osteoarthritis. Tissue Eng Regen Med 2021; 18:1045-1055. [PMID: 34542842 DOI: 10.1007/s13770-021-00384-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Stem cells intra-articular injection stagey indicated a potential therapeutic effect on improving the pathological progress of osteoarthritis (OA). However, the long-term effect of stem cells intra-articular injection on the cartilage regeneration remains unclear. miR-29a-3p is predicted to be a critical target for inhibiting insulin-like growth factor-1 expression and may aggravate the progression of OA. METHODS In this study, we investigated the therapeutic efficacy of intra-articular injection of bone marrow mesenchymal stem cells (BMSCs) transfected with miR-29a-3p inhibitor in OA. RESULTS miR-29a-3p inhibitor transfection did not influence cell viability of BMSCs, while the chondrogenic differentiation potential of BMSCs was significantly improved. Interestingly, intra-articular injection of BMSCs with miR-29a-3p inhibition significantly prevented articular cartilage degeneration by up-regulating the expression of Sox 9, Col-2a1, aggrecan and down-regulating the expression of matrix metalloproteinase, as well as relieved pain in OA. CONCLUSION The double effects on cartilage protection and pain relief indicated a great potential of intra-articular injection of miR-29a-3p inhibitor-transfected BMSCs for the treatment of OA.
Collapse
Affiliation(s)
- Qing Wang
- Department of Orthopedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, 215300, China.,Department of Orthopedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, China
| | - Yong Chen
- Department of Orthopedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, 215300, China.,Department of Orthopedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, China
| | - Xiaofeng Shen
- Department of Orthopedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China
| | - Ji Chen
- Department of Orthopedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, 215300, China.,Department of Orthopedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, China
| | - Yuwei Li
- Department of Orthopedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215000, China.
| |
Collapse
|
27
|
Sun J, Xing F, Zou M, Gong M, Li L, Xiang Z. Comparison of chondrogenesis-related biological behaviors between human urine-derived stem cells and human bone marrow mesenchymal stem cells from the same individual. Stem Cell Res Ther 2021; 12:366. [PMID: 34183056 PMCID: PMC8240221 DOI: 10.1186/s13287-021-02370-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Stem cells are the main choice for seed cells in tissue engineering, but using most traditional stem cells requires invasive and complicated procedures. Human urine-derived stem cells (hUSCs) are an alternative stem cell source with the advantages of being isolated noninvasively and repetitively from the same individual. The aim of this study was to compare chondrogenesis-related biological behaviors between hUSCs and human bone marrow mesenchymal stem cells (hBMSCs) from the same individual. METHODS hUSCs and hBMSCs were isolated from six patients who underwent iliac bone grafting. Cell morphology, proliferation, colony-forming, migration, and multidifferentiation analyses were performed in vitro. Then, acellular cartilage extracellular matrix (ACM) scaffolds were fabricated for in vivo implantation. The comparisons of cell viability, morphology, proliferation, and chondrogenesis between hUSCs and hBMSCs cultured on scaffolds were performed before implantation. The scaffolds loaded with hUSCs or hBMSCs were implanted into a rabbit knee model to repair cartilage defects. Magnetic resonance imaging (MRI) and micro-computed tomography (μCT) Analyses, inflammation and toxicity assays, gross observation, and histological evaluation were performed to evaluate the cartilage repair effects. RESULTS In in vitro experiments, hUSCs had better capacity for proliferation, colony-forming, and migration compared to hBMSCs in the same passage, while hBMSCs had greater osteogenic, adipogenic, and chondrogenic abilities compared to hUSCs in the same passage. Both hUSCs and hBMSCs at passage 3 had the strongest potential for proliferation, colony-forming, and multilineage differentiation compared to cells in other passages. The ACM scaffolds loaded with hUSCs or hBMSCs both significantly promoted the repair of cartilage defects in the rabbit knee model at 12 weeks' postimplantation, and the new tissue was mainly hyaline cartilage. However, there was no significant difference in cartilage repair effects between hUSCs and hBMSCs. CONCLUSIONS In in vitro experiments, hUSCs presented better capacity for proliferation, while hBMSCs had greater chondrogenic ability. However, hUSCs and hBMSCs had similar cartilage repair effects in vivo. Results indicated that hUSCs can be a stem cell alternative for cartilage regeneration and provide a powerful platform for cartilage tissue engineering and clinical transformation.
Collapse
Affiliation(s)
- Jiachen Sun
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Fei Xing
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Min Zou
- Department of Orthopedics, NO. 1 People's Hospital of Chengdu, Chengdu, Sichuan, 610016, People's Republic of China
| | - Min Gong
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, People's Republic of China
| | - Lang Li
- Department of Orthopaedics, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Zhou Xiang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
28
|
Abusharkh HA, Mallah AH, Amr MM, Mendenhall J, Gozen BA, Tingstad EM, Abu-Lail NI, Van Wie BJ. Enhanced matrix production by cocultivated human stem cells and chondrocytes under concurrent mechanical strain. In Vitro Cell Dev Biol Anim 2021; 57:631-640. [PMID: 34129185 DOI: 10.1007/s11626-021-00592-4] [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] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/10/2021] [Indexed: 11/28/2022]
Abstract
Conventional treatments of osteoarthritis have failed to re-build functional articular cartilage. Tissue engineering clinical treatments for osteoarthritis, including autologous chondrocyte implantation, provides an alternative approach by injecting a cell suspension to fill lesions within the cartilage in osteoarthritic knees. The success of chondrocyte implantation relies on the availability of chondrogenic cell lines, and their resilience to high mechanical loading. We hypothesize we can reduce the numbers of human articular chondrocytes necessary for a treatment by supplementing cultures with human adipose-derived stem cells, in which stem cells will have protective and stimulatory effects on mixed cultures when exposed to high mechanical loads, and in which coculture will enhance production of requisite extracellular matrix proteins over those produced by stretched chondrocytes alone. In this work, adipose-derived stem cells and articular chondrocytes were cultured separately or cocultivated at ratios of 3:1, 1:1, and 1:3 in static plates or under excessive cyclic tensile strain of 10% and results were compared to culturing of both cell types alone with and without cyclic strain. Results indicate 75% of chondrocytes in engineered articular cartilage can be replaced with stem cells with enhanced collagen over all culture conditions and glycosaminoglycan content over stretched cultures of chondrocytes. This can be done without observing adverse effects on cell viability. Collagen and glycosaminoglycan secretion, when compared to chondrocyte alone under 10% strain, was enhanced 6.1- and 2-fold, respectively, by chondrocytes cocultivated with stem cells at a ratio of 1:3.
Collapse
Affiliation(s)
- Haneen A Abusharkh
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, 1505 NE Stadium Way, Pullman, WA, 99164-6515, USA
| | - Alia H Mallah
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Mahmoud M Amr
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Juana Mendenhall
- Department of Chemistry, Morehouse College, Atlanta, GA, 30314, USA
| | - Bulent A Gozen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA, 99164-2920, USA
| | - Edwin M Tingstad
- Inland Orthopedic Surgery and Sports Medicine Clinic, Pullman, WA, 99163, USA
| | - Nehal I Abu-Lail
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Bernard J Van Wie
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, 1505 NE Stadium Way, Pullman, WA, 99164-6515, USA.
| |
Collapse
|
29
|
Improving In Vitro Cartilage Generation by Co-Culturing Adipose-Derived Stem Cells and Chondrocytes on an Allograft Adipose Matrix Framework. Plast Reconstr Surg 2021; 147:87-99. [PMID: 33002984 DOI: 10.1097/prs.0000000000007511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Microtia is an inherited condition that results in varying degrees of external ear deformities; the most extreme form is anotia. Effective surgical reconstruction techniques have been developed. However, these usually require multistage procedures and have other inherent disadvantages. Tissue engineering technologies offer new approaches in the field of external ear reconstruction. In this setting, chondrocytes are cultured in the laboratory with the aim of creating bioengineered cartilage matrices. However, cartilage engineering has many challenges, including difficulty in culturing sufficient chondrocytes. To overcome these hurdles, the authors propose a novel model of cartilage engineering that involves co-culturing chondrocytes and adipose-derived stem cells on an allograft adipose-derived extracellular matrix scaffold. METHODS Auricular chondrocytes from porcine ear were characterized. Adipose-derived stem cells were isolated and expanded from human lipoaspirate. Then, the auricular chondrocytes were cultured on the allograft adipose matrix either alone or with the adipose-derived stem cells at different ratios and examined histologically. RESULTS Cartilage induction was most prominent when the cells were co-cultured on the allograft adipose matrix at a ratio of 1:9 (auricular chondrocyte-to-adipose-derived stem cell ratio). Furthermore, because of the xenogeneic nature of the experiment, the authors were able to determine that the adipose-derived stem cells contributed to chondrogenesis by means of a paracrine stimulation of the chondrocytes. CONCLUSIONS In this situation, adipose-derived stem cells provide sufficient support to induce the formation of cartilage when the number of auricular chondrocytes available is limited. This novel model of cartilage engineering provides a setting for using the patient's own chondrocytes and adipose tissue to create a customized ear framework that could be further used for surgical reconstruction.
Collapse
|
30
|
Recent Updates of Diagnosis, Pathophysiology, and Treatment on Osteoarthritis of the Knee. Int J Mol Sci 2021; 22:ijms22052619. [PMID: 33807695 PMCID: PMC7961389 DOI: 10.3390/ijms22052619] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative and chronic joint disease characterized by clinical symptoms and distortion of joint tissues. It primarily damages joint cartilage, causing pain, swelling, and stiffness around the joint. It is the major cause of disability and pain. The prevalence of OA is expected to increase gradually with the aging population and increasing prevalence of obesity. Many potential therapeutic advances have been made in recent years due to the improved understanding of the underlying mechanisms, diagnosis, and management of OA. Embryonic stem cells and induced pluripotent stem cells differentiate into chondrocytes or mesenchymal stem cells (MSCs) and can be used as a source of injectable treatments in the OA joint cavity. MSCs are known to be the most studied cell therapy products in cell-based OA therapy owing to their ability to differentiate into chondrocytes and their immunomodulatory properties. They have the potential to improve cartilage recovery and ultimately restore healthy joints. However, despite currently available therapies and advances in research, unfulfilled medical needs persist for OA treatment. In this review, we focused on the contents of non-cellular and cellular therapies for OA, and briefly summarized the results of clinical trials for cell-based OA therapy to lay a solid application basis for clinical research.
Collapse
|
31
|
Horita M, Farquharson C, Stephen LA. The role of miR-29 family in disease. J Cell Biochem 2021; 122:696-715. [PMID: 33529442 PMCID: PMC8603934 DOI: 10.1002/jcb.29896] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 02/06/2023]
Abstract
MicroRNAs are small noncoding RNAs that can bind to the target sites in the 3’‐untranslated region of messenger RNA to regulate posttranscriptional gene expression. Increasing evidence has identified the miR‐29 family, consisting of miR‐29a, miR‐29b‐1, miR‐29b‐2, and miR‐29c, as key regulators of a number of biological processes. Moreover, their abnormal expression contributes to the etiology of numerous diseases. In the current review, we aimed to summarize the differential expression patterns and functional roles of the miR‐29 family in the etiology of diseases including osteoarthritis, osteoporosis, cardiorenal, and immune disease. Furthermore, we highlight the therapeutic potential of targeting members of miR‐29 family in these diseases. We present miR‐29s as promoters of osteoblast differentiation and apoptosis but suppressors of chondrogenic and osteoclast differentiation, fibrosis, and T cell differentiation, with clear avenues for therapeutic manipulation. Further research will be crucial to identify the precise mechanism of miR‐29 family in these diseases and their full potential in therapeutics.
Collapse
Affiliation(s)
- Masahiro Horita
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, Scotland, UK
| | - Colin Farquharson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, Scotland, UK
| | - Louise A Stephen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, Scotland, UK
| |
Collapse
|
32
|
Shamdani S, Chantepie S, Flageollet C, Henni-Chebra N, Jouan Y, Eymard F, Hay E, Cohen-Solal M, Papy-Garcia D, Chevalier X, Albanese P. Heparan sulfate functions are altered in the osteoarthritic cartilage. Arthritis Res Ther 2020; 22:283. [PMID: 33287871 PMCID: PMC7722421 DOI: 10.1186/s13075-020-02352-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/12/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Heparan sulfate (HS) proteoglycans (PG) may be found at the chondrocyte surface and in the pericellular cartilage matrix, and are involved in cell-cell and cell-matrix interactions. An important function of HS chains is to regulate cell fate through specific interactions with heparin-binding proteins (HBP) modulated by their complex sulfation pattern. Osteoarthritis (OA) is a joint disorder characterized by the degradation of articular cartilaginous extracellular matrix. The aim of this study was to investigate HS structure and functions in osteoarthritic cartilages compared to normal cartilages (controls). METHODS Glycosaminoglycans (GAG) were extracted from human macroscopically normal cartilages (controls, n = 7) and (OA cartilages n = 11). HS were isolated and quantified using the DMMB quantification method. Their structure and functions were then compared using respectively a HPLC analysis and HBP binding tests and their phenotypic effects on murine chondrocytes were studied by RQ-PCR. Statistical analyzes were performed using a one-way ANOVA followed by a Dunnett's test or a t test for pairwise comparisons. RESULTS In OA, HS were characterized by increased sulfation levels compared to controls. Moreover, the capacity of these HS to bind HBP involved in the OA pathophysiological process such as FGF2 and VEGF was reduced. Chondroitin sulfates and keratan sulfates regulated these binding properties. Finally, HS from OA cartilages induced the mRNA levels of catabolic markers such as MMP3, MMP13, and TS4 and inhibited the mRNA levels of anabolic markers such as COL2, ACAN, SOX9, and VEGF in murine articular chondrocytes. CONCLUSION The sulfation of HS chains was increased in OA cartilages with changes in HBP binding properties and biological effects on chondrocyte phenotypes. Thus, modified HS present in altered cartilages could be a novel therapeutic target in OA.
Collapse
Affiliation(s)
- Sara Shamdani
- Univ Paris Est Creteil, Gly-CRRET, Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Créteil, F-94010, France
| | - Sandrine Chantepie
- Univ Paris Est Creteil, Gly-CRRET, Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Créteil, F-94010, France
| | - Camille Flageollet
- Univ Paris Est Creteil, Gly-CRRET, Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Créteil, F-94010, France
| | - Nadia Henni-Chebra
- Univ Paris Est Creteil, Gly-CRRET, Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Créteil, F-94010, France
| | - Yohann Jouan
- INSERM, UMR-S 1132 Bioscar, Centre Viggo Petersen, Hôpital Lariboisière, 2, Rue Ambroise Paré,, Creteil, F-94010, France.,BIOSCAR Inserm U1132, Université de Paris, F-75010, Paris, France
| | - Florent Eymard
- Univ Paris Est Creteil, Gly-CRRET, Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Créteil, F-94010, France.,INSERM, UMR-S 1132 Bioscar, Centre Viggo Petersen, Hôpital Lariboisière, 2, Rue Ambroise Paré, Créteil, 75010, France
| | - Eric Hay
- INSERM, UMR-S 1132 Bioscar, Centre Viggo Petersen, Hôpital Lariboisière, 2, Rue Ambroise Paré,, Creteil, F-94010, France.,BIOSCAR Inserm U1132, Université de Paris, F-75010, Paris, France
| | - Martine Cohen-Solal
- INSERM, UMR-S 1132 Bioscar, Centre Viggo Petersen, Hôpital Lariboisière, 2, Rue Ambroise Paré,, Creteil, F-94010, France.,BIOSCAR Inserm U1132, Université de Paris, F-75010, Paris, France.,Université de Paris (UFR de Médecine), Paris, 75010, France
| | - Dulce Papy-Garcia
- Univ Paris Est Creteil, Gly-CRRET, Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Créteil, F-94010, France
| | - Xavier Chevalier
- Univ Paris Est Creteil, Gly-CRRET, Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Créteil, F-94010, France.,INSERM, UMR-S 1132 Bioscar, Centre Viggo Petersen, Hôpital Lariboisière, 2, Rue Ambroise Paré, Créteil, 75010, France
| | - Patricia Albanese
- Univ Paris Est Creteil, Gly-CRRET, Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Créteil, F-94010, France.
| |
Collapse
|
33
|
Chen Y, Ouyang X, Wu Y, Guo S, Xie Y, Wang G. Co-culture and Mechanical Stimulation on Mesenchymal Stem Cells and Chondrocytes for Cartilage Tissue Engineering. Curr Stem Cell Res Ther 2020; 15:54-60. [PMID: 31660820 DOI: 10.2174/1574888x14666191029104249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023]
Abstract
Defects in articular cartilage injury and chronic osteoarthritis are very widespread and common, and the ability of injured cartilage to repair itself is limited. Stem cell-based cartilage tissue engineering provides a promising therapeutic option for articular cartilage damage. However, the application of the technique is limited by the number, source, proliferation, and differentiation of stem cells. The co-culture of mesenchymal stem cells and chondrocytes is available for cartilage tissue engineering, and mechanical stimulation is an important factor that should not be ignored. A combination of these two approaches, i.e., co-culture of mesenchymal stem cells and chondrocytes under mechanical stimulation, can provide sufficient quantity and quality of cells for cartilage tissue engineering, and when combined with scaffold materials and cytokines, this approach ultimately achieves the purpose of cartilage repair and reconstruction. In this review, we focus on the effects of co-culture and mechanical stimulation on mesenchymal stem cells and chondrocytes for articular cartilage tissue engineering. An in-depth understanding of the impact of co-culture and mechanical stimulation of mesenchymal stem cells and chondrocytes can facilitate the development of additional strategies for articular cartilage tissue engineering.
Collapse
Affiliation(s)
- Yawen Chen
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Xinli Ouyang
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Yide Wu
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Shaojia Guo
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Yongfang Xie
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Guohui Wang
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| |
Collapse
|
34
|
Liu T, Li X, Wang T, Chen X, Zhang S, Liao J, Wang W, Zou X, Zhou G. Kartogenin mediates cartilage regeneration by stimulating the IL-6/Stat3-dependent proliferation of cartilage stem/progenitor cells. Biochem Biophys Res Commun 2020; 532:385-392. [PMID: 32888652 DOI: 10.1016/j.bbrc.2020.08.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/14/2020] [Indexed: 01/20/2023]
Abstract
A decrease in the number of endogenous stem cells in cartilage is regarded as the cause of cartilage degeneration. Kartogenin (KGN) is known to induce chondrogenesis of cartilage stem/progenitor cells (CSPCs). Using CSPCs isolated from rat cartilage, we analysed changes in the transcriptome after treatment with KGN in vitro. An animal model of destabilization of the medial meniscus (DMM) was then used to identify the effect of intra-articular (IA) KGN injection on CSPC proliferation in vivo. Here, we demonstrated that KGN promoted the proliferation of CSPCs isolated from cartilage. The percentage of G2-M phase cells in the KGN-treated group reached over 10%, nearly twice that in the control group. Transcriptomic profiling of rat CSPCs revealed significant changes in KGN-treated samples compared to control samples. The gene expression levels of IL-6 and its coreceptor Gp130 were much higher in the KGN-treated group than in the control group. Phosphorylation of the IL-6 downstream molecule Stat3 was enhanced via KGN stimulation. The DMM animal model showed increased articular cartilage thickness after IA KGN injection. IHC staining also demonstrated upregulation of Stat3 phosphorylation and enhanced distribution of CD44+/CD105+ cells in cartilage following IA KGN injection. Thus, our data suggested that KGN promoted cartilage regeneration at least partially by stimulating IL-6/Stat3-dependent proliferation.
Collapse
Affiliation(s)
- Tao Liu
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, 518060, China; Department of Oncology Rehabilitation, Shenzhen Luohu People's Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen, Guangdong, 518001, China.
| | - Xiaolin Li
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, 518060, China; Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Ting Wang
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Xuemei Chen
- Department of Oncology Rehabilitation, Shenzhen Luohu People's Hospital, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen, Guangdong, 518001, China
| | - Shuai Zhang
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jinqi Liao
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Xuenong Zou
- Department of Spine Surgery, Orthopedic Research Institute, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Science Center, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
35
|
Wang M, Chen G, Li G, Wang B, Lei C. Creating Cartilage in Tissue-Engineered Chamber Using Platelet-Rich Plasma Without Cell Culture. Tissue Eng Part C Methods 2020; 26:375-383. [PMID: 32539669 DOI: 10.1089/ten.tec.2020.0049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Clinically available cartilage, such as large-volume tissue-engineered cartilage, is urgently required for various clinical applications. Tissue engineering chamber (TEC) models are a promising organ-level strategy for efficient enlargement of cells or tissues within the chamber. The conventional TEC technology is not suitable for cartilage culture, because it lacks the necessary chondrogenic growth factor, which is present in platelet-rich plasma (PRP). In this study, we added autogenous auricular cartilage fragments mixed with PRP in a TEC to obtain a large amount of engineered cartilage. Experiment: To prove the efficacy of this method, 48 New Zealand white rabbits were randomly divided into 4 groups: PRP, vascularized (Ves), PRP, PRP+Ves, and control. Auricular cartilage was harvested from the rabbits, cut into fragments (2 mm), and then injected into TECs. Cartilage constructs were harvested at week 8, and construct volumes were measured. Histological morphology, immunochemical staining, and mechanical strength were evaluated. Results: At week 8, PRP+Ves constructs developed a white, cartilage-like appearance. The volume of cartilage increased by 600% the original volume from 0.30 to 1.8 ± 0.1789 mL. Histological staining showed proliferation of edge chondrocytes in the embedded cartilage in the PRP and PRP+Ves groups. Furthermore, the cartilage constructs in the PRP+Ves group show mechanical characteristics similar to those of normal cartilage. Conclusions: Auricular cartilage fragments mixed with PRP and vascularization of the TEC showed a significantly increased cartilage tissue volume after 8 weeks of incubation in rabbits. Impact Statement Repair of defects of ear cartilage tissue has always been a huge challenge to plastic surgeons. In this article, a new method is presented to produce within 8 weeks auricular cartilage in a tissue engineering chamber without cell culture. Having such a method is a valuable step toward creating a large volume of functional cartilage tissue, which may lead to successful construction of normal auricular structure with minimal donor-site morbidity.
Collapse
Affiliation(s)
- Meishui Wang
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| | - Guojie Chen
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China.,Department of Burn and Plastic Surgery, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Guanmin Li
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| | - Biao Wang
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| | - Chen Lei
- Department of Plastic and Cosmetic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, P.R. China
| |
Collapse
|
36
|
Zhang Y, Hao C, Guo W, Peng X, Wang M, Yang Z, Li X, Zhang X, Chen M, Sui X, Peng J, Lu S, Liu S, Guo Q, Jiang Q. Co-culture of hWJMSCs and pACs in double biomimetic ACECM oriented scaffold enhances mechanical properties and accelerates articular cartilage regeneration in a caprine model. Stem Cell Res Ther 2020; 11:180. [PMID: 32430067 PMCID: PMC7238567 DOI: 10.1186/s13287-020-01670-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/20/2020] [Accepted: 04/06/2020] [Indexed: 12/24/2022] Open
Abstract
Background The dedifferentiation of chondrocytes and the unstable chondrogenic differentiation status of pluripotent mesenchymal stem cells (MSCs) are immense issues in cell-based articular cartilage repair and regenerative strategies. Here, to improve the cartilage characteristics of seed cells, a double biomimetic acellular cartilage extracellular matrix (ACECM)-oriented scaffold was used to mimic the cartilage microenvironment for human umbilical cord Wharton’s jelly-derived MSCs (hWJMSCs) and primary cartilage cells (pACs) to regenerate hyaline cartilage. Methods A double biomimetic ACECM-oriented scaffold was created from the cartilage extracellular matrix of pig articular cartilage using pulverization decellularization freeze-drying procedures. hWJMSCs and pACs were co-cultured at ratios of 50:50 (co-culture group, ACCC), 0:100 (ACAC group) and 100:0 (ACWJ group) in the ACECM-oriented scaffold, and the co-culture system was implanted in a caprine model for 6 months or 9 months to repair full-thickness articular cartilage defects. The control groups, which had no cells, comprised the blank control (BC) group and the ACECM-oriented scaffold (AC) group. Gross morphology and magnetic resonance imaging (MRI) as well as histological and biomechanical evaluations were used to characterize the cartilage of the repair area. Results Relative to the control groups, both the gross morphology and histological staining results demonstrated that the neotissue of the ACCC group was more similar to native cartilage and better integrated with the surrounding tissue. Measurements of glycosaminoglycan content and Young’s modulus showed that the repair areas had more abundant cartilage-specific content and significantly higher mechanical strength in the ACCC group than in the control groups, especially at 9 months. On MRI, the T2-weighted signal of the repair area was homogeneous, and the oedema signal disappeared almost completely in the ACCC group at 9 months. HLA-ABC immunofluorescence staining demonstrated that hWJMSCs participated in the repair and regeneration of articular cartilage and escaped surveillance and clearance by the caprine immune system. Conclusion The structure and components of double biomimetic ACECM-oriented scaffolds provided a cartilage-like microenvironment for co-cultured seed cells and enhanced the biomechanics and compositions of neotissue. This co-culture system has the potential to overcome the dedifferentiation of passage chondrocytes and the unstable chondrogenic differentiation status of MSCs.
Collapse
Affiliation(s)
- Yu Zhang
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Gulou District, Nanjing, 210008, China
| | - Chunxiang Hao
- Institute of Anesthesia, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Weimin Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xiaoyu Peng
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Gulou District, Nanjing, 210008, China
| | - Mingjie Wang
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Zhen Yang
- School of Medicine, Naikai University, Tianjin, 300071, China
| | - Xu Li
- School of Medicine, Naikai University, Tianjin, 300071, China
| | - Xueliang Zhang
- Shanxi Traditional Chinese, No. 46 Binzhou west Street, YingZe District, Taiyuan, 030001, China
| | - Mingxue Chen
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xiang Sui
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Jiang Peng
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shibi Lu
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China.,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shuyun Liu
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China. .,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China. .,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Quanyi Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China. .,Beijing Key Lab of Regenerative Medicine in Orthopaedics, 28 Fuxing Road, Haidian District, Beijing, 100853, China. .,Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Gulou District, Nanjing, 210008, China.
| |
Collapse
|
37
|
McCreery KP, Calve S, Neu CP. Ontogeny informs regeneration: explant models to investigate the role of the extracellular matrix in cartilage tissue assembly and development. Connect Tissue Res 2020; 61:278-291. [PMID: 32186210 PMCID: PMC7190409 DOI: 10.1080/03008207.2019.1698556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/22/2019] [Indexed: 02/03/2023]
Abstract
Osteoarthritis (OA) is typically managed in late stages by replacement of the articular cartilage surface with a prosthesis as an effective, though undesirable outcome. As an alternative, hydrogel implants or growth factor treatments are currently of great interest in the tissue engineering community, and scaffold materials are often designed to emulate the mechanical and chemical composition of mature extracellular matrix (ECM) tissue. However, scaffolds frequently fail to capture the structure and organization of cartilage. Additionally, many current scaffold designs do not mimic processes by which structurally sound cartilage is formed during musculoskeletal development. The objective of this review is to highlight methods that investigate cartilage ontogenesis with native and model systems in the context of regenerative medicine. Specific emphasis is placed on the use of cartilage explant cultures that provide a physiologically relevant microenvironment to study tissue assembly and development. Ex vivo cartilage has proven to be a cost-effective and accessible model system that allows researchers to control the culture conditions and stimuli and perform proteomics and imaging studies that are not easily possible using in vivo experiments, while preserving native cell-matrix interactions. We anticipate our review will promote a developmental biology approach using explanted tissues to guide cartilage tissue engineering and inform new treatment methods for OA and joint damage.
Collapse
Affiliation(s)
| | - Sarah Calve
- Department of Mechanical Engineering, University of Colorado, Boulder, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, USA
| | - Corey P. Neu
- Department of Mechanical Engineering, University of Colorado, Boulder, USA
| |
Collapse
|
38
|
The Optimal Regimen for the Treatment of Temporomandibular Joint Injury Using Low-Intensity Pulsed Ultrasound in Rats with Chronic Sleep Deprivation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5468173. [PMID: 32462002 PMCID: PMC7212287 DOI: 10.1155/2020/5468173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/16/2020] [Accepted: 04/18/2020] [Indexed: 11/19/2022]
Abstract
Low-intensity pulsed ultrasound (LIPUS) is an emerging physical therapy for the treatment of early temporomandibular joint injury and has a good effect on promoting cartilage and subchondral bone tissue repair. However, the best LIPUS intensity and treatment duration remain unclear. This study is aimed at observing the preventive and therapeutic effects of different modes of LIPUS and at identifying the optimal LIPUS treatment regimen for temporomandibular joint injury. In the present study, rat models of temporomandibular joint injury were established using a chronic sleep deprivation (CSD) method, and the effect of LIPUS as intensities of 30, 45, and 60 mW/cm2 was observed at 7, 14, and 21 days. After CSD, the condylar cartilage of the rats demonstrated variable degrees of surface roughening, collagen fiber disarrangement or even partial exfoliation, decreased proteoglycan synthesis and cartilage thickness, decreased chondrocyte proliferation, decreased type 2 collagen (COL-2) expression, and increased matrix metalloproteinase- (MMP-) 3 expression at all three time points. When the rats with CSD received different intensities of LIPUS treatment, the pathological changes were alleviated to various extents. The groups receiving 45 mW/cm2 LIPUS showed the most significant relief of cartilage damage, and this significant effect was observed on days 14 and 21. These results demonstrated that LIPUS can effectively inhibit CSD-induced condylar cartilage damage in rats, and LIPUS treatment at an intensity of 45 mW/cm2 for at least 2 weeks is the optimal regimen for temporomandibular joint injury.
Collapse
|
39
|
Kunze KN, Burnett RA, Wright-Chisem J, Frank RM, Chahla J. Adipose-Derived Mesenchymal Stem Cell Treatments and Available Formulations. Curr Rev Musculoskelet Med 2020; 13:264-280. [PMID: 32328959 DOI: 10.1007/s12178-020-09624-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The use of human adipose-derived mesenchymal stem cells (ADSCs) has gained attention due to its potential to expedite healing and the ease of harvesting; however, clinical evidence is limited, and questions concerning optimal method of delivery and long-term outcomes remain unanswered. RECENT FINDINGS Administration of ADSCs in animal models has been reported to aid in improved healing benefits with enhanced repair biomechanics, superior gross histological appearance of injury sites, and higher concentrations of growth factors associated with healing compared to controls. Recently, an increasing body of research has sought to examine the effects of ADSCs in humans. Several available processing techniques and formulations for ADSCs exist with evidence to suggest benefits with the use of ADSCs, but the superiority of any one method is not clear. Evidence from the most recent clinical studies available demonstrates promising outcomes following treatment of select musculoskeletal pathologies with ADSCs despite reporting variability among ADSCs harvesting and processing; these include (1) healing benefits and pain improvement for rotator cuff and Achilles tendinopathies, (2) improvements in pain and function in those with knee and hip osteoarthritis, and (3) improved cartilage regeneration for osteochondral focal defects of the knee and talus. The limitation to most of this literature is the use of other therapeutic biologics in combination with ADSCs. Additionally, many studies lack control groups, making establishment of causation inappropriate. It is imperative to perform higher-quality studies using consistent, predictable control populations and to standardize formulations of ADSCs in these trials.
Collapse
Affiliation(s)
- Kyle N Kunze
- Department of Orthopaedic Surgery, Division of Sports Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Robert A Burnett
- Department of Orthopaedic Surgery, Division of Sports Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Joshua Wright-Chisem
- Department of Orthopaedic Surgery, Hospital for Special Surgery, New York, NY, USA
| | - Rachel M Frank
- Department of Orthopaedic Surgery, Division of Sports Medicine, University of Colorado School of Medicine, Boulder, CO, USA
| | - Jorge Chahla
- Department of Orthopaedic Surgery, Division of Sports Medicine, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
40
|
Comparative Study of Electrospun Scaffolds Containing Native GAGs and a GAG Mimetic for Human Mesenchymal Stem Cell Chondrogenesis. Ann Biomed Eng 2020; 48:2040-2052. [DOI: 10.1007/s10439-020-02499-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/24/2020] [Indexed: 12/20/2022]
|
41
|
Multifactor dimensionality reduction reveals a strong gene-gene interaction between STC1 and COL11A1 genes as a possible risk factor of knee osteoarthritis. Mol Biol Rep 2020; 47:2627-2634. [PMID: 32140959 DOI: 10.1007/s11033-020-05351-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/25/2020] [Indexed: 12/31/2022]
Abstract
Articular cartilage is an avascular tissue with a structure that allows it to support and cushion the overload of the surfaces in contact. It maintains its metabolic functions due to the contribution of different signaling pathways. However, several factors play a role in its deterioration, allowing to the development of osteoarthritis (OA), and one of the major factors is genetic. Our goal was to identify gene-gene interactions (epistasis) between five signaling pathways involved in the articular cartilage metabolism as possible indicators of OA risk. We applied the Multifactor-Dimensionality Reduction (MDR) method to identify and characterize the epistasis between 115 SNPs located in 73 genes related to HIF-1α, Wnt/β-catenin, cartilage extracellular matrix metabolism, oxidative stress, and uric acid transporters. Ninety three patients diagnosed with primary knee OA and 150 healthy controls were included in the study. Genotyping was performed with the OpenArray system, the statistical analysis was carried out with the STATA software v14, and epistasis was analyzed with the MDR software v3.0.2. The MDR analysis revealed that the best interaction model was between polymorphisms rs17786744 of the STC1 gene and rs2615977 of the COL11A1 gene, with an entropy value of 4.44%, CVC 8/10, OR 5.60, 95% CI 3.27-9.59, p < 0.0001. Under this interaction model, we identified high and low risk genotypes involved in OA development. Our results suggest complex interactions between STC1 and COL11A1 genes that might have an impact on genetic susceptibility to develop OA. Further studies are required to confirm it.
Collapse
|
42
|
Yilmaz EN, Zeugolis DI. Electrospun Polymers in Cartilage Engineering-State of Play. Front Bioeng Biotechnol 2020; 8:77. [PMID: 32133352 PMCID: PMC7039817 DOI: 10.3389/fbioe.2020.00077] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Articular cartilage defects remain a clinical challenge. Articular cartilage defects progress to osteoarthritis, which negatively (e.g., remarkable pain, decreased mobility, distress) affects millions of people worldwide and is associated with excessive healthcare costs. Surgical procedures and cell-based therapies have failed to deliver a functional therapy. To this end, tissue engineering therapies provide a promise to deliver a functional cartilage substitute. Among the various scaffold fabrication technologies available, electrospinning is continuously gaining pace, as it can produce nano- to micro- fibrous scaffolds that imitate architectural features of native extracellular matrix supramolecular assemblies and can deliver variable cell populations and bioactive molecules. Herein, we comprehensively review advancements and shortfalls of various electrospun scaffolds in cartilage engineering.
Collapse
Affiliation(s)
- Elif Nur Yilmaz
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
43
|
Zhang S, Hu B, Liu W, Wang P, Lv X, Chen S, Liu H, Shao Z. Articular cartilage regeneration: The role of endogenous mesenchymal stem/progenitor cell recruitment and migration. Semin Arthritis Rheum 2019; 50:198-208. [PMID: 31767195 DOI: 10.1016/j.semarthrit.2019.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/04/2019] [Accepted: 11/01/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Trauma- or osteoarthritis-related cartilage damage resulted in functional decline of joints and heavy burden of public health. Recently, the reparative role of mesenchymal stem/progenitor cells (MSCs) in articular cartilage (AC) reconstruction is drawing more and more attention. OBJECTIVE To provide a review on (1) the locations and categories of joint-resident MSCs, (2) the regulation of chondrogenic capacities of MSCs, (3) the migratory approaches of MSCs to diseased AC and regulatory mechanisms. METHODS PubMed and Web of Science were searched for English-language articles related to MSC recruitment and migration for AC repair until June 2019. The presence of various MSCs in or around joints, the potential approaches to diseased AC` and the regenerative capacities of MSCs were reviewed. RESULTS Various intra- and peri-articular MSCs, with inherent migratory potentials, are present in multiple stem cell niches in or around joints. The recruitment and migration of joint-resident MSCs play crucial roles in endogenous AC repair. Multiple recruiting signals, such as chemokines, growth factors, etc., emerge during the development of AC diseases and participate in the regulation of MSC mobilization. Motivated MSCs could migrate into cartilage lesions and then exert multiple reparative potentials, including extracellular matrix (ECM) reconstruction and microenvironment modulation. CONCLUSION In general, AC repair based on endogenous MSC recruitment and migration is a feasible strategy, and a promising research field. Furthermore, endogenous AC repair mediated by native MSCs would provide new opportunities to efficient preventative or therapeutic options for AC diseases.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Binwu Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Peng Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Songfeng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| | - Hongjian Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| |
Collapse
|
44
|
Scalzone A, Ferreira AM, Tonda-Turo C, Ciardelli G, Dalgarno K, Gentile P. The interplay between chondrocyte spheroids and mesenchymal stem cells boosts cartilage regeneration within a 3D natural-based hydrogel. Sci Rep 2019; 9:14630. [PMID: 31601910 PMCID: PMC6787336 DOI: 10.1038/s41598-019-51070-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/25/2019] [Indexed: 12/30/2022] Open
Abstract
Articular cartilage (AC) lacks the ability to self-repair and cell-based approaches, primarily based on using chondrocytes and mesenchymal stem cells (MSCs), are emerging as effective technology to restore cartilage functionality, because cells synergic functionality may support the maintenance of chondrogenic phenotype and promote extracellular matrix regeneration. This work aims to develop a more physiologically representative co-culture system to investigate the influence of MSCs on the activity of chondrocytes. A thermo-sensitive chitosan-based hydrogel, ionically crosslinked with β-glycerophosphate, is optimised to obtain sol/gel transition at physiological conditions within 5 minutes, high porosity with pores diameter <30 µm, and in vitro mechanical integrity with compressive and equilibrium Young's moduli of 37 kPa and 17 kPa, respectively. Live/dead staining showed that after 1 and 3 days in culture, the encapsulated MSCs into the hydrogels are viable and characterised by round-like morphology. Furthermore chondrocyte spheroids, seeded on top of gels that contained either MSCs or no cells, show that the encapsulated MSCs stimulate chondrocyte activity within a gel co-culture, both in terms of maintaining the coherence of chondrocyte spheroids, leading to a larger quantity of CD44 (by immunofluorescence) and a higher production of collagen and glycosaminoglycans (by histology) compared with the mono-culture.
Collapse
Affiliation(s)
- Annachiara Scalzone
- School of Engineering, Newcastle University, Claremont Road, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Ana M Ferreira
- School of Engineering, Newcastle University, Claremont Road, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Chiara Tonda-Turo
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino Corso Duca degli Abruzzi 29, Turin, 10129, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino Corso Duca degli Abruzzi 29, Turin, 10129, Italy
| | - Kenny Dalgarno
- School of Engineering, Newcastle University, Claremont Road, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Claremont Road, Newcastle upon Tyne, NE1 7RU, United Kingdom.
| |
Collapse
|
45
|
Mechanical stimulation promotes the proliferation and the cartilage phenotype of mesenchymal stem cells and chondrocytes co-cultured in vitro. Biomed Pharmacother 2019; 117:109146. [PMID: 31387186 DOI: 10.1016/j.biopha.2019.109146] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 01/29/2023] Open
Abstract
Mesenchymal stem cells and chondrocytes are an important source of the cells for cartilage tissue engineering. Therefore, the culture and expansion methods of these cells need to be improved to overcome the aging of chondrocytes and induced chondrogenic differentiation of mesenchymal stem cells. The aim of this study was to expand the cells for cartilage tissue engineering by combining the advantages of growing cells in co-culture and under a mechanically-stimulated environment. Rabbit chondrocytes and co-cultured cells (bone mesenchymal stem cells and chondrocytes) were subjected to cyclic sinusoidal dynamic tensile mechanical stimulationusing the FX-4000 tension system. Chondrocyte proliferation was assayed by flow cytometry and CFSE labeling. The cell cartilage phenotype was determined by detecting GAG, collagen II and TGF-β1 protein expression by ELISA and the Col2α1, TGF-β1 and Sox9 gene expression by RT-PCR. The results show that the co-culture improved both the proliferation ability of chondrocytes and the cartilage phenotype of co-cultured cells. A proper cyclic sinusoidal dynamic tensile mechanical stimulation improved the proliferation ability and cartilage phenotype of chondrocytes and co-cultured cells. These results suggest that the co-culture of mesenchymal stem cells with chondrocytes and proper mechanical stimulation may be an appropriate way to rapidly expand the cells that have an improved cartilage phenotype for cartilage tissue engineering.
Collapse
|
46
|
Hyaluronic Acid (HA), Platelet-Rich Plasm and Extracorporeal Shock Wave Therapy (ESWT) promote human chondrocyte regeneration in vitro and ESWT-mediated increase of CD44 expression enhances their susceptibility to HA treatment. PLoS One 2019; 14:e0218740. [PMID: 31251756 PMCID: PMC6599220 DOI: 10.1371/journal.pone.0218740] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/07/2019] [Indexed: 12/27/2022] Open
Abstract
Novel strategies have been proposed for articular cartilage damage occurring during osteoarthritis (OA) and -among these- Extracorporeal Shock Wave Therapy (ESWT), intra-articular injections of Platelet-Rich Plasma (PRP) or Hyaluronic Acid (HA) revealed encouraging results. To investigate the possible mechanisms responsible for those clinical benefits, we established primary cultures of human chondrocytes derived from cartilage explants and measured the in vitro effects of ESW, PRP and HA therapies. After molecular/morphological cell characterization, we assessed those effects on the functional activities of the chondrocyte cell cultures, at the protein and molecular levels. ESWT significantly prevented the progressive dedifferentiation that spontaneously occurs during prolonged chondrocyte culture. We then attested the efficiency of all such treatments to stimulate the expression of markers of chondrogenic potential such as SOX9 and COL2A, to increase the Ki67 proliferation index as well as to antagonize the traditional marker of chondrosenescence p16INK4a (known as Cdkn2a). Furthermore, all our samples showed an ESW- and HA-mediated enhancement of migratory and anti-inflammatory activity onto the cytokine-rich environment characterizing OA. Taken together, those results suggest a regenerative effect of such therapies on primary human chondrocytes in vitro. Moreover, we also show for the first time that ESW treatment induces the surface expression of major hyaluronan cell receptor CD44 allowing the increase of COL2A/COL1A ratio upon HA administration. Therefore, this work suggests that ESW-induced CD44 overexpression enhances the in vitro cell susceptibility of human chondrocytes to HA, presumably favouring the repair of degenerated cartilage.
Collapse
|
47
|
Affiliation(s)
- Ameer Elbuluk
- Department of Orthopaedic Surgery, NYU Langone Medical Center, Hospital for Joint Diseases, New York, NY
| | | | | |
Collapse
|
48
|
Coculture of hWJMSCs and pACs in Oriented Scaffold Enhances Hyaline Cartilage Regeneration In Vitro. Stem Cells Int 2019; 2019:5130152. [PMID: 30881462 PMCID: PMC6383394 DOI: 10.1155/2019/5130152] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
Seed cells of articular cartilage tissue engineering face many obstacles in their application because of the dedifferentiation of chondrocytes or unstable chondrogenic differentiation status of pluripotent stem cells. To overcome mentioned dilemmas, a simulation of the articular cartilage microenvironment was constructed by primary articular cartilage cells (pACs) and acellular cartilage extracellular matrix- (ACECM-) oriented scaffold cocultured with human umbilical cord Wharton's jelly-derived mesenchymal stem cells (hWJMSCs) in vitro. The coculture groups showed more affluent cartilage special matrix ingredients including collagen II and aggrecan based on the results of histological staining and western blotting and cut down as many pACs as possible. The RT-PCR and cell viability experiments also demonstrated that hWJMSCs were successfully induced to differentiate into chondrocytes when cultured in the simulated cartilage microenvironment, as confirmed by the significant upregulation of collagen II and aggrecan, while the cell proliferation activity of pACs was significantly improved by cell-cell interactions. Therefore, compared with monoculture and chondrogenic induction of inducers, coculture providing a simulated native articular microenvironment was a potential and temperate way to regulate the biological behaviors of pACs and hWJMSCs to regenerate the hyaline articular cartilage.
Collapse
|
49
|
Mesenchymal stem cell-based therapy of osteoarthritis: Current knowledge and future perspectives. Biomed Pharmacother 2018; 109:2318-2326. [PMID: 30551490 DOI: 10.1016/j.biopha.2018.11.099] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/30/2018] [Accepted: 11/25/2018] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a chronic, prevalent, debilitating joint disease characterized by progressive cartilage degradation, subchondral bone remodeling, bone marrow lesions, meniscal damage, and synovitis. Innate immune cells (natural killer cells, macrophages, and mast cells) play the most important pathogenic role in the early inflammatory response, while cells of adaptive immunity (CD4 + Th1 lymphocytes and antibody producing B cells) significantly contribute to the development of chronic, relapsing course of inflammation in OA patients. Conventional therapy for OA is directed toward symptomatic treatment, mainly pain management, and is not able to promote regeneration of degenerated cartilage or to attenuate joint inflammation. Since articular cartilage, intra-articular ligaments, and menisci have no ability to heal, regeneration of these tissues remains one of the most important goals of new therapeutic approaches used for OA treatment. Due to their capacity for differentiation into chondrocytes and due to their immunomodulatory properties, mesenchymal stem cells (MSCs) have been the most extensively explored as new therapeutic agents in the cell-based therapy of OA. Simple acquisition, rapid proliferation, maintenance of differentiation potential after repeated passages in vitro, minor immunological rejection due to the low surface expression of major histocompatibility complex antigens, efficient engraftment and long-term coexistence in the host are the main characteristics of MSCs that enable their therapeutic use in OA. In this review article, we emphasized current knowledge and future perspectives regarding molecular and cellular mechanisms responsible for beneficial effects of autologous and allogeneic MSCs in the treatment of OA.
Collapse
|
50
|
Huang J, Liang Y, Jia Z, Chen J, Duan L, Liu W, Zhu F, Liang Q, Zhu W, You W, Xiong J, Wang D. Development of Magnetic Nanocomposite Hydrogel with Potential Cartilage Tissue Engineering. ACS OMEGA 2018; 3:6182-6189. [PMID: 30023943 PMCID: PMC6044747 DOI: 10.1021/acsomega.8b00291] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/19/2018] [Indexed: 05/26/2023]
Abstract
Magnetic nanocomposite hydrogels show high potential to improve tissue engineering. In this study, a magnetic nanocomposite hydrogel was prepared from poly(vinyl alcohol), nano-hydroxyapatite (n-HA), and magnetic nanoparticles (Fe2O3) using the ultrasonic dispersion method and freeze-thaw cross-linking molding. The water content and crystallinity of the magnetic nanocomposite hydrogel were tested. Microscopic morphology assessment, mechanical testing, and characterization were performed. Additionally, the magnetic nanocomposite hydrogel was co-cultured with bone mesenchymal stem cells (BMSCs) to determine its cell compatibility. We found that the magnetic nanocomposite hydrogel had good mechanical properties and that its mechanical properties were enhanced by the addition of n-HA. The BMSCs showed uniform growth on the surface of the magnetic nanocomposite hydrogel and high rates of proliferation. BMSC growth was also enhanced by the addition of Fe2O3 and also significant stimulated chondrocyte-related gene expression. Thus, the magnetic nanocomposite hydrogel scaffold material we describe here could have broad applications in cartilage tissue engineering.
Collapse
Affiliation(s)
- Jianghong Huang
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Yujie Liang
- Shenzhen
Institute of Mental Health, Shenzhen Mental Health Center, Shenzhen Kangning Hospital, Shenzhen 518020, Guangdong Province, China
- Departments
of Chemistry, The Chinese University of
Hong Kong, Shatin 999077, Hong Kong SAR, China
| | - ZhaoFeng Jia
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Jielin Chen
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Li Duan
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Wei Liu
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Feiyan Zhu
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Qian Liang
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Weimin Zhu
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Wei You
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Jianyi Xiong
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| | - Daping Wang
- Shenzhen
National Key Department of Orthopedics, Shenzhen Key Laboratory of Tissue
Engineering, and Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First
Hospital Affiliated to Shenzhen University), Shenzhen 518035, China
| |
Collapse
|