1
|
Lyu X, Wang J, Su J. Intelligent Manufacturing for Osteoarthritis Organoids. Cell Prolif 2025:e70043. [PMID: 40285592 DOI: 10.1111/cpr.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/22/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease worldwide, imposing a substantial global disease burden. However, its pathogenesis remains incompletely understood, and effective treatment strategies are still lacking. Organoid technology, in which stem cells or progenitor cells self-organise into miniature tissue structures under three-dimensional (3D) culture conditions, provides a promising in vitro platform for simulating the pathological microenvironment of OA. This approach can be employed to investigate disease mechanisms, carry out high-throughput drug screening and facilitate personalised therapies. This review summarises joint structure, OA pathogenesis and pathological manifestations, thereby establishing the disease context for the application of organoid technology. It then examines the components of the arthrosis organoid system, specifically addressing cartilage, subchondral bone, synovium, skeletal muscle and ligament organoids. Furthermore, it details various strategies for constructing OA organoids, including considerations of cell selection, pathological classification and fabrication techniques. Notably, this review introduces the concept of intelligent manufacturing of OA organoids by incorporating emerging engineering technologies such as artificial intelligence (AI) into the organoid fabrication process, thereby forming an innovative software and hardware cluster. Lastly, this review discusses the challenges currently facing intelligent OA organoid manufacturing and highlights future directions for this rapidly evolving field. By offering a comprehensive overview of state-of-the-art methodologies and challenges, this review anticipates that intelligent, automated fabrication of OA organoids will expedite fundamental research, drug discovery and personalised translational applications in the orthopaedic field.
Collapse
Affiliation(s)
- Xukun Lyu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
2
|
Wang Y, Wu Q, You Y, Jiang W, Fu P, Dai K, Sun Y. ABCA6 Regulates Chondrogenesis and Inhibits Joint Degeneration via Orchestrated Cholesterol Efflux and Cellular Senescence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410414. [PMID: 39823538 PMCID: PMC11904997 DOI: 10.1002/advs.202410414] [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: 08/28/2024] [Revised: 01/06/2025] [Indexed: 01/19/2025]
Abstract
Patellar dysplasia (PD) can cause patellar dislocation and subsequent osteoarthritis (OA) development. Herein, a novel ABCA6 mutation contributing to a four-generation family with familiar patellar dysplasia (FPD) is identified. In this study, whole exome sequencing (WES) and genetic linkage analysis across a four-generation lineage presenting with six cases of FPD are conducted. A disease-causing mutation in ABCA6 is identified for FPD. Further analyses reveal a consistent correlation between ABCA6 expression downregulation and PD occurrence, chondrocyte degeneration, and OA onset. Moreover, ABCA6-KO mice demonstrate severe knee joint degeneration and accelerated OA progression. Besides, synovial mesenchymal stem cells (SMSCs) are extracted from WT, ABCA6-/+, and ABCA6-/- mice to create chondrogenic organoids in vitro, confirming ABCA6 deficiency can lead to chondrocyte degeneration via modulating cell cycle and activating cellular senescence. Moreover, transcriptome and metabolomic sequencing analysis on ABCA6-KO chondrocytes unveils that the ABCA6 deficiency inhibits cholesterol efflux, leading to intracellular cholesterol accumulation and subsequent cellular senescence and impaired chondrogenesis.A disease-causing mutation of ABCA6 is identified for FPD. ABCA6 is correlated with PD occurrence and subsequent OA progression. ABCA6 can serve as a potential target in chondrogenesis and OA treatment by orchestrated intracellular cholesterol efflux and delayed cellular senescence.
Collapse
Affiliation(s)
- Yi Wang
- Department of OrthopaedicsThe First Affiliated Hospital of Nanjing Medical UniversityJiangsu210029China
| | - Qiang Wu
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Yongqing You
- Renal DivisionAffiliated Hospital of Nanjing University of Chinese MedicineNanjing210008China
| | - Wenbo Jiang
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Peiliang Fu
- Department of OrthopaedicsShanghai Changzheng hospitalNaval Medical UniversityShanghai200003China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| | - Ye Sun
- Department of OrthopaedicsThe First Affiliated Hospital of Nanjing Medical UniversityJiangsu210029China
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
| |
Collapse
|
3
|
Wang J, Chen X, Li R, Wang S, Geng Z, Shi Z, Jing Y, Xu K, Wei Y, Wang G, He C, Dong S, Liu G, Hou Z, Xia Z, Wang X, Ye Z, Zhou F, Bai L, Tan H, Su J. Standardization and consensus in the development and application of bone organoids. Theranostics 2025; 15:682-706. [PMID: 39744680 PMCID: PMC11671374 DOI: 10.7150/thno.105840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
Organoids, self-organized structures derived from stem cells cultured in a specific three-dimensional (3D) in vitro microenvironment, have emerged as innovative platforms that closely mimic in vivo cellular behavior, tissue architecture, and organ function. Bone organoids, a frontier in organoid research, can replicate the complex structures and functional characteristics of bone tissue. Recent advancements have led to the successful development of bone organoids, including models of callus, woven bone, cartilage, trabecular bone, and bone marrow. These organoids are widely utilized in establishing bone-related disease models, bone injury repair, and drug screening. However, significant discrepancies remain between current bone organoids and human skeletal tissues in terms of morphology and functionality, limiting their ability to accurately model human bone physiology and pathology. To address these challenges and promote standardization in the construction, evaluation, and application of bone organoids, we have convened experts and research teams with substantial expertise in the field. By integrating existing research findings, this consortium aims to establish a consensus to guide future research and application of bone organoids.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Ruiyang Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Zhongmin Shi
- Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Chongru He
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China
| | - Guohui Liu
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Zhidao Xia
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, UK
| | - Xinglong Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, 85721, USA
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, 999077, China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hongbo Tan
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force of Chinese PLA, Kunming, 650032, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
4
|
Du C, Liu J, Liu S, Xiao P, Chen Z, Chen H, Huang W, Lei Y. Bone and Joint-on-Chip Platforms: Construction Strategies and Applications. SMALL METHODS 2024; 8:e2400436. [PMID: 38763918 DOI: 10.1002/smtd.202400436] [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: 03/25/2024] [Revised: 04/28/2024] [Indexed: 05/21/2024]
Abstract
Organ-on-a-chip, also known as "tissue chip," is an advanced platform based on microfluidic systems for constructing miniature organ models in vitro. They can replicate the complex physiological and pathological responses of human organs. In recent years, the development of bone and joint-on-chip platforms aims to simulate the complex physiological and pathological processes occurring in human bones and joints, including cell-cell interactions, the interplay of various biochemical factors, the effects of mechanical stimuli, and the intricate connections between multiple organs. In the future, bone and joint-on-chip platforms will integrate the advantages of multiple disciplines, bringing more possibilities for exploring disease mechanisms, drug screening, and personalized medicine. This review explores the construction and application of Organ-on-a-chip technology in bone and joint disease research, proposes a modular construction concept, and discusses the new opportunities and future challenges in the construction and application of bone and joint-on-chip platforms.
Collapse
Affiliation(s)
- Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Xiao
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
5
|
Bai L, Zhou D, Li G, Liu J, Chen X, Su J. Engineering bone/cartilage organoids: strategy, progress, and application. Bone Res 2024; 12:66. [PMID: 39567500 PMCID: PMC11579019 DOI: 10.1038/s41413-024-00376-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/19/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
The concept and development of bone/cartilage organoids are rapidly gaining momentum, providing opportunities for both fundamental and translational research in bone biology. Bone/cartilage organoids, essentially miniature bone/cartilage tissues grown in vitro, enable the study of complex cellular interactions, biological processes, and disease pathology in a representative and controlled environment. This review provides a comprehensive and up-to-date overview of the field, focusing on the strategies for bone/cartilage organoid construction strategies, progresses in the research, and potential applications. We delve into the significance of selecting appropriate cells, matrix gels, cytokines/inducers, and construction techniques. Moreover, we explore the role of bone/cartilage organoids in advancing our understanding of bone/cartilage reconstruction, disease modeling, drug screening, disease prevention, and treatment strategies. While acknowledging the potential of these organoids, we discuss the inherent challenges and limitations in the field and propose potential solutions, including the use of bioprinting for organoid induction, AI for improved screening processes, and the exploration of assembloids for more complex, multicellular bone/cartilage organoids models. We believe that with continuous refinement and standardization, bone/cartilage organoids can profoundly impact patient-specific therapeutic interventions and lead the way in regenerative medicine.
Collapse
Affiliation(s)
- Long Bai
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, China
| | - Dongyang Zhou
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Guangfeng Li
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Jinlong Liu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
| |
Collapse
|
6
|
Huang J, Li A, Liang R, Wu X, Jia S, Chen J, Jiao Z, Li C, Zhang X, Lin J. Future perspectives: advances in bone/cartilage organoid technology and clinical potential. BIOMATERIALS TRANSLATIONAL 2024; 5:425-443. [PMID: 39872930 PMCID: PMC11764185 DOI: 10.12336/biomatertransl.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 01/30/2025]
Abstract
Bone and cartilage tissues are essential for movement and structure, yet diseases like osteoarthritis affect millions. Traditional therapies have limitations, necessitating innovative approaches. Organoid technology, leveraging stem cells' regenerative potential, offers a novel platform for disease modelling and therapy. This review focuses on advancements in bone/cartilage organoid technology, highlighting the role of stem cells, biomaterials, and external factors in organoid development. We discuss the implications of these organoids for regenerative medicine, disease research, and personalised treatment strategies, presenting organoids as a promising avenue for enhancing cartilage repair and bone regeneration. Bone/cartilage organoids will play a greater role in the treatment of bone/cartilage diseases in the future, and promote the progress of biological tissue engineering.
Collapse
Affiliation(s)
- Jingtao Huang
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, Gunagdong Province, China
| | - Aikang Li
- Department of Clinical Medicine, Shantou University Medical College, Shantou, Gunagdong Province, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
| | - Rongji Liang
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, Gunagdong Province, China
| | - Xiaohao Wu
- Immunology and Rheumatology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Shicheng Jia
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, Gunagdong Province, China
| | - Jiayou Chen
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, Gunagdong Province, China
| | - Zilu Jiao
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
| | - Canfeng Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
| | - Xintao Zhang
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Gunagdong Province, China
| |
Collapse
|
7
|
Zhang C, Jing Y, Wang J, Xia Z, Lai Y, Bai L, Su J. Skeletal organoids. BIOMATERIALS TRANSLATIONAL 2024; 5:390-410. [PMID: 39872931 PMCID: PMC11764188 DOI: 10.12336/biomatertransl.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 01/30/2025]
Abstract
The skeletal system, composed of bones, muscles, joints, ligaments, and tendons, serves as the foundation for maintaining human posture, mobility, and overall biomechanical functionality. However, with ageing, chronic overuse, and acute injuries, conditions such as osteoarthritis, intervertebral disc degeneration, muscle atrophy, and ligament or tendon tears have become increasingly prevalent and pose serious clinical challenges. These disorders not only result in pain, functional loss, and a marked reduction in patients' quality of life but also impose substantial social and economic burdens. Current treatment modalities, including surgical intervention, pharmacotherapy, and physical rehabilitation, often do not effectively restore the functionality of damaged tissues and are associated with high recurrence rates and long-term complications, highlighting significant limitations in their efficacy. Thus, there is a strong demand to develop novel and more effective therapeutic and reparative strategies. Organoid technology, as a three-dimensional micro-tissue model, can replicate the structural and functional properties of native tissues in vitro, providing a novel platform for in-depth studies of disease mechanisms, optimisation of drug screening, and promotion of tissue regeneration. In recent years, substantial advancements have been made in the research of bone, muscle, and joint organoids, demonstrating their broad application potential in personalised and regenerative medicine. Nonetheless, a comprehensive review of current research on skeletal organoids is still lacking. Therefore, this article aims to present an overview of the definition and technological foundation of organoids, systematically summarise the progress in the construction and application of skeletal organoids, and explore future opportunities and challenges in this field, offering valuable insights and references for researchers.
Collapse
Affiliation(s)
- Chen Zhang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Yingying Jing
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Jianhua Wang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhidao Xia
- Centre for Nanohealth, Swansea University Medical School, Swansea University, Swansea, UK
| | - Yuxiao Lai
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang Province, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Skoracka J, Bajewska K, Kulawik M, Suchorska W, Kulcenty K. Advances in cartilage tissue regeneration: a review of stem cell therapies, tissue engineering, biomaterials, and clinical trials. EXCLI JOURNAL 2024; 23:1170-1182. [PMID: 39391058 PMCID: PMC11464958 DOI: 10.17179/excli2024-7088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/22/2024] [Indexed: 10/12/2024]
Abstract
Cartilage tissue, characterized by its limited regenerative capacity, presents significant challenges in clinical therapy. Recent advancements in cartilage regeneration have focused on integrating stem cell therapies, tissue engineering strategies, and advanced modeling techniques to overcome existing limitations. Stem cells, particularly Mesenchymal Stem Cells (MSCs) and induced pluripotent stem cells (iPSCs), hold promise for cartilage repair due to their ability to differentiate into chondrocytes, the key cells responsible for cartilage formation. Tissue engineering approaches, including 3D models, organ-on-a-chip systems, and organoids, offer innovative methods to mimic natural tissue microenvironments and evaluate potential treatments. MSC-based techniques, such as cell sheet tissue engineering, address challenges associated with traditional therapies, including cell availability and culture difficulties. Furthermore, advancements in 3D bioprinting enable the fabrication of complex tissue structures, while organ-on-a-chip systems provide microfluidic platforms for disease modeling and physiological mimicry. Organoids serve as simplified models of organs, capturing some complexity and enabling the monitoring of pathophysiological aspects of cartilage diseases. This comprehensive review underscores the transformative potential of integrating stem cell therapies, tissue engineering strategies, and advanced modeling techniques to improve cartilage regeneration and pave the way for more effective clinical treatments.
Collapse
Affiliation(s)
- Julia Skoracka
- Poznan University of Medical Sciences, Poznan, Poland, Fredry 10 Street, 61-701 Poznan, Poland
| | - Kaja Bajewska
- Poznan University of Medical Sciences, Poznan, Poland, Fredry 10 Street, 61-701 Poznan, Poland
| | - Maciej Kulawik
- Poznan University of Medical Sciences, Poznan, Poland, Fredry 10 Street, 61-701 Poznan, Poland
| | - Wiktoria Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences,Garbary 15 Street, 61-866 Poznan, Poland
- Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866 Poznan, Poland
| | - Katarzyna Kulcenty
- Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866 Poznan, Poland
| |
Collapse
|
9
|
Li X, Sheng S, Li G, Hu Y, Zhou F, Geng Z, Su J. Research Progress in Hydrogels for Cartilage Organoids. Adv Healthc Mater 2024; 13:e2400431. [PMID: 38768997 DOI: 10.1002/adhm.202400431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/29/2024] [Indexed: 05/22/2024]
Abstract
The repair and regeneration of cartilage has always been a hot topic in medical research. Cartilage organoids (CORGs) are special cartilage tissue created using tissue engineering techniques outside the body. These engineered organoids tissues provide models that simulate the complex biological functions of cartilage, opening new possibilities for cartilage regenerative medicine and treatment strategies. However, it is crucial to establish suitable matrix scaffolds for the cultivation of CORGs. In recent years, utilizing hydrogel to culture stem cells and induce their differentiation into chondrocytes has emerged as a promising method for the in vitro construction of CORGs. In this review, the methods for establishing CORGs are summarized and an overview of the advantages and limitations of using matrigel in the cultivation of such organoids is provided. Furthermore, the importance of cartilage tissue ECM and alternative hydrogel substitutes for Matrigel, such as alginate, peptides, silk fibroin, and DNA derivatives is discussed, and the pros and cons of using these hydrogels for the cultivation of CORGs are outlined. Finally, the challenges and future directions in hydrogel research for CORGs are discussed. It is hoped that this article provides valuable references for the design and development of hydrogels for CORGs.
Collapse
Affiliation(s)
- Xiaolong Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics and Traumatology, Nanning Hospital of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530000, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
10
|
Hall GN, Fan Y, Viellerobe B, Iazzolino A, Dimopoulos A, Poiron C, Clapies A, Luyten FP, Guillemot F, Papantoniou I. Laser-assisted bioprinting of targeted cartilaginous spheroids for high density bottom-up tissue engineering. Biofabrication 2024; 16:045029. [PMID: 39136309 DOI: 10.1088/1758-5090/ad6e1a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Multicellular spheroids such as microtissues and organoids have demonstrated great potential for tissue engineering applications in recent years as these 3D cellular units enable improved cell-cell and cell-matrix interactions. Current bioprinting processes that use multicellular spheroids as building blocks have demonstrated limited control on post printing distribution of cell spheroids or moderate throughput and printing efficiency. In this work, we presented a laser-assisted bioprinting approach able to transfer multicellular spheroids as building blocks for larger tissue structures. Cartilaginous multicellular spheroids formed by human periosteum derived cells (hPDCs) were successfully bioprinted possessing high viability and the capacity to undergo chondrogenic differentiation post printing. Smaller hPDC spheroids with diameters ranging from ∼100 to 150µm were successfully bioprinted through the use of laser-induced forward transfer method (LIFT) however larger spheroids constituted a challenge. For this reason a novel alternative approach was developed termed as laser induced propulsion of mesoscopic objects (LIPMO) whereby we were able to bioprint spheroids of up to 300µm. Moreover, we combined the bioprinting process with computer aided image analysis demonstrating the capacity to 'target and shoot', through automated selection, multiple large spheroids in a single sequence. By taking advantage of target and shoot system, multilayered constructs containing high density cell spheroids were fabricated.
Collapse
Affiliation(s)
- Gabriella Nilsson Hall
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Yuchao Fan
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Bertrand Viellerobe
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Antonio Iazzolino
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Andreas Dimopoulos
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Claire Poiron
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Aude Clapies
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Frank P Luyten
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Fabien Guillemot
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Ioannis Papantoniou
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas, Stadiou 26504, Platani, Patras, Greece
| |
Collapse
|
11
|
Eldeen GN, Elkhooly TA, El Bassyouni GT, Hamdy TM, Hawash AR, Aly RM. Enhancement of the chondrogenic differentiation capacity of human dental pulp stem cells via chondroitin sulfate-coated polycaprolactone-MWCNT nanofibers. Sci Rep 2024; 14:16396. [PMID: 39013921 PMCID: PMC11252133 DOI: 10.1038/s41598-024-66497-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024] Open
Abstract
Most of the conditions involving cartilaginous tissues are irreversible and involve degenerative processes. The aim of the present study was to fabricate a biocompatible fibrous and film scaffolds using electrospinning and casting techniques to induce chondrogenic differentiation for possible application in cartilaginous tissue regeneration. Polycaprolactone (PCL) electrospun nanofibrous scaffolds and PCL film were fabricated and incorporated with multi-walled carbon nanotubes (MWCNTs). Thereafter, coating of chondroitin sulfate (CS) on the fibrous and film structures was applied to promote chondrogenic differentiation of human dental pulp stem cells (hDPSCs). First, the morphology, hydrophilicity and mechanical properties of the scaffolds were characterized by scanning electron microscopy (SEM), spectroscopic characterization, water contact angle measurements and tensile strength testing. Subsequently, the effects of the fabricated scaffolds on stimulating the proliferation of human dental pulp stem cells (hDPSCs) and inducing their chondrogenic differentiation were evaluated via electron microscopy, flow cytometry and RT‒PCR. The results of the study demonstrated that the different forms of the fabricated PCL-MWCNTs scaffolds analyzed demonstrated biocompatibility. The nanofilm structures demonstrated a higher rate of cellular proliferation, while the nanofibrous architecture of the scaffolds supported the cellular attachment and differentiation capacity of hDPSCs and was further enhanced with CS addition. In conclusion, the results of the present investigation highlighted the significance of this combination of parameters on the viability, proliferation and chondrogenic differentiation capacity of hDPSCs seeded on PCL-MWCNT scaffolds. This approach may be applied when designing PCL-based scaffolds for future cell-based therapeutic approaches developed for chondrogenic diseases.
Collapse
Affiliation(s)
- Ghada Nour Eldeen
- Human Genetics and Genome Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Tarek A Elkhooly
- Refractories, Ceramics, and Building Materials Department, National Research Centre, Dokki, Giza, 12622, Egypt
- Nanomedicine Research Unit, Faculty of Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Gehan T El Bassyouni
- Refractories, Ceramics, and Building Materials Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Tamer M Hamdy
- Restorative and Dental Materials Department, Oral and Dental Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Ahmed R Hawash
- Faculty of Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Riham M Aly
- Basic Dental Science Department, Oral and Dental Research Institute, National Research Centre, 33 El Bohouth St., Dokki, Giza, 12622, Egypt.
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, 12622, Egypt.
| |
Collapse
|
12
|
Wu X, Fu Y, Ma J, Li C, He A, Zhang T. LGR5 Modulates Differentiated Phenotypes of Chondrocytes Through PI3K/AKT Signaling Pathway. Tissue Eng Regen Med 2024; 21:791-807. [PMID: 38771465 PMCID: PMC11187034 DOI: 10.1007/s13770-024-00645-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Tissue engineering is increasingly viewed as a promising avenue for functional cartilage reconstruction. However, chondrocyte dedifferentiation during in vitro culture remains an obstacle for clinical translation of tissue engineered cartilage. Re-differentiated induction have been employed to induce dedifferentiated chondrocytes back to their original phenotype. Regrettably, these strategies have been proven to be only moderately effective. METHODS To explore underlying mechanism, RNA transcriptome sequencing was conducted on primary chondrocytes (P0), dedifferentiated chondrocytes (P5), and redifferentiated chondrocytes (redifferentiation-induction of P5, P5.R). Based on multiple bioinformatics analysis, LGR5 was identified as a target gene. Subsequently, stable cell lines with LGR5 knocking-down and overexpression were established using P0 chondrocytes. The phenotypic changes in P1 and P5 chondrocytes with either LGR5 knockdown or overexpression were assessed to ascertain the potential influence of LGR5 dysregulation on chondrocyte phenotypes. Regulatory mechanism was then investigated using bioinformatic analysis, protein-protein docking, immunofluorescence co-localization and immunoprecipitation. RESULTS The current study found that dysregulation of LGR5 can significantly impact the dedifferentiated phenotypes of chondrocytes (P5). Upregulation of LGR5 appears to activate the PI3K/AKT signal via increasing the phosphorylation levels of AKT (p-AKT1). Moreover, the increase of p-AKT1 may stabilize β-catenin and enhance the intensity of Wnt/β-catenin signal, and help to restore the dedifferentated phenotype of chondrocytes. CONCLUSION LGR5 can modulate the phenotypes of chondrocytes in P5 passage through PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xu Wu
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China
| | - Yaoyao Fu
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China
| | - Jing Ma
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, China
| | - Chenlong Li
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China
| | - Aijuan He
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Tianyu Zhang
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China.
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
13
|
Faeed M, Ghiasvand M, Fareghzadeh B, Taghiyar L. Osteochondral organoids: current advances, applications, and upcoming challenges. Stem Cell Res Ther 2024; 15:183. [PMID: 38902814 PMCID: PMC11191177 DOI: 10.1186/s13287-024-03790-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024] Open
Abstract
In the realm of studying joint-related diseases, there is a continuous quest for more accurate and representative models. Recently, regenerative medicine and tissue engineering have seen a growing interest in utilizing organoids as powerful tools for studying complex biological systems in vitro. Organoids, three-dimensional structures replicating the architecture and function of organs, provide a unique platform for investigating disease mechanisms, drug responses, and tissue regeneration. The surge in organoid research is fueled by the need for physiologically relevant models to bridge the gap between traditional cell cultures and in vivo studies. Osteochondral organoids have emerged as a promising avenue in this pursuit, offering a better platform to mimic the intricate biological interactions within bone and cartilage. This review explores the significance of osteochondral organoids and the need for their development in advancing our understanding and treatment of bone and cartilage-related diseases. It summarizes osteochondral organoids' insights and research progress, focusing on their composition, materials, cell sources, and cultivation methods, as well as the concept of organoids on chips and application scenarios. Additionally, we address the limitations and challenges these organoids face, emphasizing the necessity for further research to overcome these obstacles and facilitate orthopedic regeneration.
Collapse
Affiliation(s)
- Maryam Faeed
- Cell and Molecular School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mahsa Ghiasvand
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahar Fareghzadeh
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Leila Taghiyar
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
14
|
Zhang Y, Li G, Wang J, Zhou F, Ren X, Su J. Small Joint Organoids 3D Bioprinting: Construction Strategy and Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302506. [PMID: 37814373 DOI: 10.1002/smll.202302506] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 09/28/2023] [Indexed: 10/11/2023]
Abstract
Osteoarthritis (OA) is a chronic disease that causes pain and disability in adults, affecting ≈300 million people worldwide. It is caused by damage to cartilage, including cellular inflammation and destruction of the extracellular matrix (ECM), leading to limited self-repairing ability due to the lack of blood vessels and nerves in the cartilage tissue. Organoid technology has emerged as a promising approach for cartilage repair, but constructing joint organoids with their complex structures and special mechanisms is still challenging. To overcome these boundaries, 3D bioprinting technology allows for the precise design of physiologically relevant joint organoids, including shape, structure, mechanical properties, cellular arrangement, and biological cues to mimic natural joint tissue. In this review, the authors will introduce the biological structure of joint tissues, summarize key procedures in 3D bioprinting for cartilage repair, and propose strategies for constructing joint organoids using 3D bioprinting. The authors also discuss the challenges of using joint organoids' approaches and perspectives on their future applications, opening opportunities to model joint tissues and response to joint disease treatment.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Trauma Orthopedics, Zhongye Hospital, Shanghai, 200941, China
| | - Jian Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Fengjin Zhou
- Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
15
|
Wang XH, Liu N, Zhang H, Yin ZS, Zha ZG. From cells to organs: progress and potential in cartilaginous organoids research. J Transl Med 2023; 21:926. [PMID: 38129833 PMCID: PMC10740223 DOI: 10.1186/s12967-023-04591-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/04/2023] [Indexed: 12/23/2023] Open
Abstract
While cartilage tissue engineering has significantly improved the speed and quality of cartilage regeneration, the underlying metabolic mechanisms are complex, making research in this area lengthy and challenging. In the past decade, organoids have evolved rapidly as valuable research tools. Methods to create these advanced human cell models range from simple tissue culture techniques to complex bioengineering approaches. Cartilaginous organoids in part mimic the microphysiology of human cartilage and fill a gap in high-fidelity cartilage disease models to a certain extent. They hold great promise to elucidate the pathogenic mechanism of a diversity of cartilage diseases and prove crucial in the development of new drugs. This review will focus on the research progress of cartilaginous organoids and propose strategies for cartilaginous organoid construction, study directions, and future perspectives.
Collapse
Affiliation(s)
- Xiao-He Wang
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Ning Liu
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Hui Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Zong-Sheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Zhen-Gang Zha
- Department of Bone and Joint Surgery, the First Affliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
16
|
Reina-Mahecha A, Beers MJ, van der Veen HC, Zuhorn IS, van Kooten TG, Sharma PK. A Review of the Role of Bioreactors for iPSCs-Based Tissue-Engineered Articular Cartilage. Tissue Eng Regen Med 2023; 20:1041-1052. [PMID: 37861960 PMCID: PMC10645985 DOI: 10.1007/s13770-023-00573-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common degenerative joint disease without an ultimate treatment. In a search for novel approaches, tissue engineering (TE) has shown great potential to be an effective way for hyaline cartilage regeneration and repair in advanced stages of OA. Recently, induced pluripotent stem cells (iPSCs) have been appointed to be essential stem cells for degenerative disease treatment because they allow a personalized medicine approach. For clinical translation, bioreactors in combination with iPSCs-engineerd cartilage could match patients needs, serve as platform for large-scale patient specific cartilage production, and be a tool for patient OA modelling and drug screening. Furthermore, to minimize in vivo experiments and improve cell differentiation and cartilage extracellular matrix (ECM) deposition, TE combines existing approaches with bioreactors. METHODS This review summarizes the current understanding of bioreactors and the necessary parameters when they are intended for cartilage TE, focusing on the potential use of iPSCs. RESULTS Bioreactors intended for cartilage TE must resemble the joint cavity niche. However, recreating human synovial joints is not trivial because the interactions between various stimuli are not entirely understood. CONCLUSION The use of mechanical and electrical stimulation to differentiate iPSCs, and maintain and test chondrocytes are key stimuli influencing hyaline cartilage homeostasis. Incorporating these stimuli to bioreactors can positively impact cartilage TE approaches and their possibility for posterior translation into the clinics.
Collapse
Affiliation(s)
- Alejandro Reina-Mahecha
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Martine J Beers
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Hugo C van der Veen
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge S Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Theo G van Kooten
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Prashant K Sharma
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands.
| |
Collapse
|
17
|
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
|