1
|
Gai T, Zhang H, Hu Y, Li R, Wang J, Chen X, Wang J, Chen Z, Jing Y, Wang C, Bai L, Wang X, Su J. Sequential construction of vascularized and mineralized bone organoids using engineered ECM-DNA-CPO-based bionic matrix for efficient bone regeneration. Bioact Mater 2025; 49:362-377. [PMID: 40144795 PMCID: PMC11937690 DOI: 10.1016/j.bioactmat.2025.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Given the limitations of allogeneic and artificial bone grafts, bone organoids have attracted extensive attention for their physiological properties that closely resemble natural bone, offering great potential to bone reconstruction for critical-sized bone defects. Although early-stage bone organoids such as osteo-callus organoids and woven bone organoids have been reported, functional bone organoids with vascularization and mineralization are currently unavailable due to the lack of bone-mimicking matrix and dynamic culture systems suitable for the long-term cultivation of mature bone organoids. Herein, a novel engineered bionic matrix hydrogels with multifunctional components and double network structure are developed by incorporating calcium phosphate oligomers (CPO) into a combination of bone-derived decellularized extracellular matrix (ECM) and salmon-derived deoxyribonucleic acid (DNA) via photo-crosslinking and dynamic self-assembly strategies. This kind of bionic matrix hydrogels facilitate recruitment, proliferation, osteogenesis and angiogenesis of bone marrow mesenchymal stromal cells (BMSCs). More importantly, vascularized and mineralized bone organoids are sequentially constructed using BMSCs-loaded engineered bionic matrix hydrogels via in vitro dynamic culture and in vivo heterotopic ossification. Meanwhile, this kind of engineered bionic matrix are capable of achieving efficient bone repair for cranial defect. These findings suggest that engineered bionic matrix hydrogels combined with such dynamic culture system, providing a promising strategy for functional bone organoids construction.
Collapse
Affiliation(s)
- Tingting Gai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hao Zhang
- 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
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ruiyang Li
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jian Wang
- 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
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jianhua Wang
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhenhua Chen
- Yantai Zhenghai Bio-tech Co., Ltd, Yantai, 264006, China
| | - Yingying Jing
- 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
| | - Chenglong Wang
- Yantai Zhenghai Bio-tech Co., Ltd, Yantai, 264006, China
| | - Long Bai
- 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
| | - Xiuhui Wang
- 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
|
2
|
Feng Y, He D, An X. Hydrogel innovations for 3D organoid culture. Biomed Mater 2025; 20:042001. [PMID: 40359965 DOI: 10.1088/1748-605x/add82d] [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: 12/11/2024] [Accepted: 05/13/2025] [Indexed: 05/15/2025]
Abstract
Organoids are functional cell-tissue complexes that mimic structural and functional characteristics of organsin vitroin three dimensions (3D). Mimicking the natural extracellular matrix (ECM) environment is critical for guiding stem cell fate within organoid cultures. Current organoid cultures predominantly utilize animal- or tumor-derived ECMs such as dECMs and Matrigel. However, these materials introduce batch variability and uncertainty in composition, which hinders reproducibility. In contrast, naturally derived and synthetic hydrogels with excellent biocompatibility offer precise and adjustable compositions, along with tunable mechanical properties, thereby providing robust support for organoid development and maturation. We explore innovative hydrogel designs tailored specifically for organoid cultures, emphasizing the influence and meticulous control of functional hydrogels on organoid formation, differentiation, and maturation processes. Furthermore, the review highlights the potential of functionalized hydrogel scaffolds to advance both research and industrial applications in tissue and organ engineering. As research progresses, investigations will further concentrate on improving the adjustable properties, expanding their scope of application, and more biologically compatible gelation strategies of hydrogels.
Collapse
Affiliation(s)
- Yicheng Feng
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, People's Republic of China
| | - Dongyang He
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, People's Republic of China
| | - Xiao An
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, People's Republic of China
| |
Collapse
|
3
|
Liu Y, Zhou Z, Lu G, Zhang X, Shi D, Tong L, Chen D, Tuan RS, Li ZA. Musculoskeletal organoids: An emerging toolkit for establishing personalized models of musculoskeletal disorders and developing regenerative therapies. Acta Biomater 2025:S1742-7061(25)00362-9. [PMID: 40381929 DOI: 10.1016/j.actbio.2025.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Musculoskeletal (MSK) conditions are the primary cause of physical disability globally. These disorders are physically and mentally debilitating and severely impact the patients' quality of life. As the median age of the world's population increases, there has been an intensifying urgency of developing efficacious therapies for various orthopaedic conditions. Furthermore, the highly heterogeneous nature of MSK conditions calls for a personalized approach to studying disease mechanisms and developing regenerative treatments. Organoids have emerged as an advanced approach to generating functional tissue/organ mimics in vitro, which hold promise in MSK regeneration, disease modeling, and therapeutic development. Herein, we review the preparation, characterization, and application of various MSK organoids. We highlight the potential of patient-specific organoids in the development of personalized medicine and discuss the challenges and opportunities in the future development of MSK organoids. STATEMENT OF SIGNIFICANCE: Despite decades of research, translation of MSK research into clinical applications remains limited, partially attributed to our inadequate understanding of disease mechanisms. To advance therapeutic development, there are critical needs for MSK disease models with higher clinical relevance and predictive power. Additionally, engineered constructs that closely mimic the structural and functional features of native MSK tissues are highly desirable. MSK organoids have emerged as a promising approach to meet the above requirements. To unleash the full potential of MSK organoids necessitates a comprehensive understanding of their categories, construction, development, functions, applications, and challenges. This review aims to fulfill this crucial need, aiming to accelerate the clinical translation of MSK organoid platforms to benefit millions of patients afflicted with MSK conditions.
Collapse
Affiliation(s)
- Yuwei Liu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China; Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, PR China
| | - Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Gang Lu
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong Special Administrative Region of China
| | - Xin Zhang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191 PR China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; Department of Pharmacology, Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen 518000, PR China.
| | - Rocky S Tuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong Special Administrative Region of China.
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong Special Administrative Region of China; Peter Hung Pain Research Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China.
| |
Collapse
|
4
|
Liao H, Fu B, Yang P, Chen K, Wei Y, Zeng C. Recent Advances in the Clinical Treatment of Osteochondral Lesions of the Talus (2021-2023): A Narrative Review. Orthop Surg 2025. [PMID: 40371488 DOI: 10.1111/os.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/16/2025] Open
Abstract
The management of osteochondral lesions of the talus (OLT) remains a challenging clinical issue. With advances in theory and technology, treatment options for OLT have expanded significantly. This review examines recent progress in the clinical treatment of OLT, focusing on studies published from 2021 to 2023. We searched PubMed, Embase, and Cochrane databases to identify relevant clinical treatments, including both nonsurgical and surgical approaches. Surgical advancements have primarily centered on microfracture, bone transplantation, cartilage transplantation, combined cell therapy, and biomaterials. Among these, some innovative methods, such as autologous costal cartilage transplantation and biological scaffolds, have yielded promising clinical outcomes. However, high-quality clinical studies are still lacking, particularly those exploring nonsurgical treatments and physical therapy. Future research may increasingly focus on integrating biomaterials with physical therapy, which holds potential for improving patient outcomes.
Collapse
Affiliation(s)
- Hanbin Liao
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Bowen Fu
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Panpan Yang
- Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Kai Chen
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Yuxuan Wei
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Canjun Zeng
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
5
|
Lin Y, Zhao L, Jin H, Gu Q, Lei L, Fang C, Pan X. Multifunctional applications of silk fibroin in biomedical engineering: A comprehensive review on innovations and impact. Int J Biol Macromol 2025; 309:143067. [PMID: 40222531 DOI: 10.1016/j.ijbiomac.2025.143067] [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: 02/20/2025] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Silk fibroin (SF) is a biomaterial naturally produced by certain insects (notably silkworms), animals such as spiders, or through recombinant methods in genetically modified organisms. Its exceptional mechanical properties, biocompatibility, degradability, and bioactivity have inspired extensive research. In biomedicine, SF has been utilized in various forms, including gels, membranes, microspheres, and more. It also demonstrates versatility for applications across medical devices, regenerative medicine, tissue engineering, and related fields. This review explores the current research status, advantages, limitations, and potential application pathways of SF in biomedical engineering. The objective is to stimulate innovative ideas and perspectives for research and applications involving silk.
Collapse
Affiliation(s)
- Yinglan Lin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.; Ningxia Medical University, Yinchuan 750004, China
| | - Lifen Zhao
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Hairong Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.; Ningxia Medical University, Yinchuan 750004, China
| | - Qiancheng Gu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China..
| | - Chunyan Fang
- Institute of Quality Standard and Testing Technology for Agro-Products, Sichuan Academy of Agricultural Sciences, Chengdu 610066, China.
| | - Xiaoyi Pan
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China..
| |
Collapse
|
6
|
Zhu Z, Yang Y, Jiang Y, Gu T, Siow L, Gao Y, Zheng Y, Xing K, Zhou S, Zhang C, Gong J, Liu Y, Yu M. DNA Hydrogels in Tissue Engineering: From Molecular Design to Next-Generation Biomedical Applications. Adv Healthc Mater 2025; 14:e2500192. [PMID: 40211647 DOI: 10.1002/adhm.202500192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/12/2025] [Indexed: 05/17/2025]
Abstract
DNA hydrogels have emerged as promising materials in tissue engineering due to their biocompatibility, programmability, and responsiveness to stimuli. Synthesized through physical and chemical crosslinking, these hydrogels can be categorized into functionalized types, such as those based on aptamers, and stimuli-responsive types that react to pH, temperature, and light. This review highlights their applications in tissue engineering, including drug delivery, cell culture, biosensing, and gene editing. DNA hydrogels can encapsulate therapeutic agents, support cell growth, and respond dynamically to environmental changes, making them ideal for tissue engineering. A comprehensive bibliometric analysis is included, identifying key research trends and emerging areas of interest in DNA hydrogel design, synthesis, and biomedical applications. The analysis provides a deeper understanding of the field's development and future research directions. Challenges such as mechanical strength, stability, and biosafety persist, but the integration of AI in hydrogel design shows promise for advancing their functionality in clinical applications.
Collapse
Affiliation(s)
- Ziyu Zhu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yemu Yang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Yun Jiang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Tianyi Gu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Lixuen Siow
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Yunxia Gao
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Yuxin Zheng
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), Republic of Singapore
| | - Kuoran Xing
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Siyi Zhou
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Chuhan Zhang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Jiaxing Gong
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Yu Liu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
7
|
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
|
8
|
Xie D, Chen B, Wang W, Guo W, Sun Z, Wang L, Shi B, Song Y, Su M. Nanocomposite Hydrogels and Micro/Nanostructures for Printing Organoids. ACS NANO 2025; 19:12458-12466. [PMID: 40162603 DOI: 10.1021/acsnano.4c17587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Organoids are 3D artificial miniature organs composed of a cluster of self-renewing and self-organizing cells in vitro, which mimic the functions of real organs. Nanotechnologies, including the preparation of nanomaterials and the fabrication of micro/nanostructures, have been proven to promote cell proliferation, guide cell differentiation, and regulate cell self-organization, showing great promise in engineering organoids. In this Perspective, different types of nanocomposite hydrogels for organoid culture are summarized, the effects of micro/nanostructures on organoid growth and development are discussed, and 3D bioprinting technologies for constructing organoid models are introduced.
Collapse
Affiliation(s)
- Daixi Xie
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Bingda Chen
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
| | - Wenqing Wang
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Wenjing Guo
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Zhiyuan Sun
- Macao Institute of Materials Science and Engineering, Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Long Wang
- Department of Orthopedics, the Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Bin Shi
- Department of Organ Transplantation, the Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Yanlin Song
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Meng Su
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| |
Collapse
|
9
|
Chen Z, Zhang H, Huang J, Weng W, Geng Z, Li M, Su J. DNA-encoded dynamic hydrogels for 3D bioprinted cartilage organoids. Mater Today Bio 2025; 31:101509. [PMID: 39925718 PMCID: PMC11803226 DOI: 10.1016/j.mtbio.2025.101509] [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: 11/21/2024] [Revised: 01/06/2025] [Accepted: 01/19/2025] [Indexed: 02/11/2025] Open
Abstract
Articular cartilage, composed of chondrocytes within a dynamic viscoelastic matrix, has limited self-repair capacity, posing a significant challenge for regeneration. Constructing high-fidelity cartilage organoids through three-dimensional (3D) bioprinting to replicate the structure and physiological functions of cartilage is crucial for regenerative medicine, drug screening, and disease modeling. However, commonly used matrix bioinks lack reversible cross-linking and precise controllability, hindering dynamic cellular regulation. Thus, encoding bioinks adaptive for cultivating cartilage organoids is an attractive idea. DNA, with its ability to be intricately encoded and reversibly cross-linked into hydrogels, offers precise manipulation at both molecular and spatial structural levels. This endows the hydrogels with viscoelasticity, printability, cell recognition, and stimuli responsiveness. This paper elaborates on strategies to encode bioink via DNA, emphasizing the regulation of predictable dynamic properties and the resulting interactions with cell behavior. The significance of these interactions for the construction of cartilage organoids is highlighted. Finally, we discuss the challenges and future prospects of using DNA-encoded hydrogels for 3D bioprinted cartilage organoids, underscoring their potential impact on advancing biomedical applications.
Collapse
Affiliation(s)
- Ziyu Chen
- 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
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jingtao Huang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201900, China
| | - Weizong Weng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Mengmeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- Sanming Institute of Translational Medicine, Fujian, 365004, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
10
|
Chen W, Liu D, Lu K, Xu M, Li D, Yan W, Chen S, Li B. Organoids of Musculoskeletal System for Disease Modeling, Drug Screening, and Regeneration. Adv Healthc Mater 2025; 14:e2402444. [PMID: 39610173 DOI: 10.1002/adhm.202402444] [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: 07/04/2024] [Revised: 10/30/2024] [Indexed: 11/30/2024]
Abstract
Musculoskeletal diseases have emerged as the leading cause of disability worldwide, with their prevalence increasing annually. In light of this escalating health challenge, organoids, an emerging technology in tissue engineering, offer promising solutions for disease modeling, drug screening, regeneration, and repair processes. The successful development of musculoskeletal organoids represents a significant breakthrough, providing a novel platform for studying musculoskeletal diseases and facilitating the discovery of new treatments. Moreover, organoids serve as valuable complements to traditional 2D culture methods and animal models, offering rich insights into musculoskeletal biology. This review provides an overview of organoid technology, outlining the construction processes of various musculoskeletal organoids and highlighting their similarities and differences. Furthermore, the challenges associated with organoid technology in musculoskeletal systems are discussed and insights into future perspectives are offered.
Collapse
Affiliation(s)
- Weicheng Chen
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Dachuan Liu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Kai Lu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Mengping Xu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Di Li
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Wei Yan
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Song Chen
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| | - Bin Li
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215000, P. R. China
| |
Collapse
|
11
|
Wu X, Hu Y, Sheng S, Yang H, Li Z, Han Q, Zhang Q, Su J. DNA-based hydrogels for bone regeneration: A promising tool for bone organoids. Mater Today Bio 2025; 31:101502. [PMID: 39911372 PMCID: PMC11795821 DOI: 10.1016/j.mtbio.2025.101502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
DNA-based hydrogels stand out for bone regeneration due to their exceptional biocompatibility and programmability. These hydrogels facilitate the formation of spatial bone structures through bulk hydrogel fabricating, microsphere formatting, and 3D printing. Furthermore, the bone microenvironment can be finely tuned by leveraging the degradation products, nanostructure, targeting, and delivery capabilities inherent to DNA-based materials. In this review, we underscore the advantages of DNA-based hydrogels, detailing their composition, gelation techniques, and structure optimization. We then delineate three critical elements in the promotion of bone regeneration using DNA-based hydrogels: (i) osteogenesis driven by phosphate ions, plasmids, and oligodeoxynucleotides (ODNs) that enhance mineralization and promote gene and protein expression; (ii) vascularization facilitated by tetrahedral DNA nanostructures (TDNs) and aptamers, which boosts gene expression and targeted release; (iii) immunomodulation achieved through loaded factors, TDNs, and bound ions that stimulate macrophage polarization and exhibit antibacterial properties. With these advantages and properties, these DNA-based hydrogels can be used to construct bone organoids, providing an innovative tool for disease modeling and therapeutic applications in bone tissue engineering. Finally, we discuss the current challenges and future prospects, emphasizing the potential impacts and applications in regenerative medicine.
Collapse
Affiliation(s)
- Xiang Wu
- 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
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Materials Science and Engineering, Shanghai University, Shanghai, 200444, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Huijian Yang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Clinical Laboratory, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Zuhao Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Qinglin Han
- Department of Orthopedics, The Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Qin Zhang
- 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
- Sanming Institute of Translational Medicine, Sanming, 365004, 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 Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
12
|
Shanto PC, Park S, Fahad MAA, Park M, Lee BT. 3D bio-printed proteinaceous bioactive scaffold loaded with dual growth factor enhanced chondrogenesis and in situ cartilage regeneration. Bioact Mater 2025; 46:365-385. [PMID: 39845130 PMCID: PMC11751550 DOI: 10.1016/j.bioactmat.2024.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Articular cartilage has a limited self-healing capacity, leading to joint degeneration and osteoarthritis over time. Therefore, bioactive scaffolds are gaining attention as a promising approach to regenerating and repairing damaged articular cartilage through tissue engineering. In this study, we reported on a novel 3D bio-printed proteinaceous bioactive scaffolds combined with natural porcine cancellous bone dECM, tempo-oxidized cellulose nanofiber (TOCN), and alginate carriers for TGF-β1, FGF-18, and ADSCs to repair cartilage defects. The characterization results demonstrate that the 3D scaffolds are physically stable and facilitate a controlled dual release of TGF-β1 and FGF-18. Moreover, the key biological proteins within the bioactive scaffold actively interact with the biological systems to create a favorable microenvironment for cartilage regeneration. Importantly, the in vitro, in vivo, and in silico simulation showed that the scaffolds promote stem cell recruitment, migration, proliferation, and ECM deposition, and synergistic effects of TGF-β1/FGF-18 with the bioactive scaffolds significantly regulate stem cell chondrogenesis by activating the PI3K/AKT and TGFβ1/Smad4 signaling pathways. After implantation, the proteinaceous bioactive scaffold led to the regeneration of mechanically robust, full-thickness cartilage tissue that closely resembles native cartilage. Thus, these findings may provide a promising approach for regulating stem cell chondrogenesis and treating in situ cartilage regeneration.
Collapse
Affiliation(s)
- Prayas Chakma Shanto
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Seongsu Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Md Abdullah Al Fahad
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Myeongki Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
- Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
13
|
Gao L, Li Y, Liu G, Lin X, Tan Y, Liu J, Li R, Zhang C. Mechanical properties and biocompatibility characterization of 3D printed collagen type II/silk fibroin/hyaluronic acid scaffold. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:564-586. [PMID: 39388283 DOI: 10.1080/09205063.2024.2411797] [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: 07/19/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
Damage to articular cartilage is irreversible and its ability to heal is minimal. The development of articular cartilage in tissue engineering requires suitable biomaterials as scaffolds that provide a 3D natural microenvironment for the development and growth of articular cartilage. This study aims to investigate the applicability of a 3D printed CSH (collagen type II/silk fibroin/hyaluronic acid) scaffold for constructing cartilage tissue engineering. The results showed that the composite scaffold had a three-dimensional porous network structure with uniform pore sizes and good connectivity. The hydrophilicity of the composite scaffold was 1071.7 ± 131.6%, the porosity was 85.12 ± 1.6%, and the compressive elastic modulus was 36.54 ± 2.28 kPa. The creep and stress relaxation constitutive models were also established, which could well describe the visco-elastic mechanical behavior of the scaffold. The biocompatibility experiments showed that the CSH scaffold was very suitable for the adhesion and proliferation of chondrocytes. Under dynamic compressive loading conditions, it was able to promote cell adhesion and proliferation on the scaffold surface. The 3D printed CSH scaffold is expected to be ideal for promoting articular cartilage regeneration.
Collapse
Affiliation(s)
- Lilan Gao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
- Tianjin Key Laboratory of Bone Implant Interface Functionalization and Personality Research, Just Medical Equipment (Tianjin) Co., Ltd, Tianjin, China
| | - Yali Li
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
| | - Gang Liu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
| | - Xianglong Lin
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
| | - Yansong Tan
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
| | - Jie Liu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
| | - Ruixin Li
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, China
| | - Chunqiu Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, China
- Tianjin Key Laboratory of Bone Implant Interface Functionalization and Personality Research, Just Medical Equipment (Tianjin) Co., Ltd, Tianjin, China
| |
Collapse
|
14
|
Huang H, Xiao L, Fang L, Lei M, Liu Z, Gao S, Lei Q, Lei J, Wei R, Lei Y, Xue L, Geng Z, Cai L, Yan F. Static Topographical Cue Combined with Dynamic Fluid Stimulation Enhances the Macrophage Extracellular Vesicle Yield and Therapeutic Potential for Bone Defects. ACS NANO 2025; 19:8667-8691. [PMID: 39998493 DOI: 10.1021/acsnano.4c15201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Extracellular vesicles (EVs) hold promise for tissue regeneration, but their low yield and limited therapeutic efficacy hinder clinical translation. Bioreactors provide a larger culture surface area and stable environment for large-scale EV production, yet their ability to enhance EV therapeutic efficacy is limited. Physical stimulation, by inducing cell differentiation and modulating EV cargo composition, offers a more efficient, cost-effective, and reproducible approach compared to the cargo loading of EVs and biochemical priming of parental cells. Herein, the effects of a 3D-printed perfusion bioreactor with a topographical cue on the macrophage EV yield and bioactivity were assessed. The results indicate that the bioreactor increased the EV yield 12.5-fold and enhanced bioactivity in promoting osteogenic differentiation and angiogenesis via upregulated miR-210-3p. Mechanistically, fluid shear stress activates Piezo1, triggering Ca2+ influx and Yes-associated protein (YAP) nuclear translocation, promoting EV secretion and enhancing macrophage M2 polarization in conjunction with morphological changes guided by aligned topography. Moreover, a porous electrospun membrane-hydrogel composite scaffold loaded with bioreactor-derived EVs exhibited outstanding efficacy in promoting osteogenic differentiation and angiogenesis in a rat cranial defect model. This study presents a scalable, cost-effective, and stable platform for the production of therapeutic EVs, potentially overcoming key challenges in translating EV-based therapies to the clinic.
Collapse
Affiliation(s)
- Huayi Huang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| | - Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| | - Lucheng Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| | - Ming Lei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| | - Zhibo Liu
- Plastic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009 Zhejiang, China
| | - Shijie Gao
- Orthopedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, 899 Pinghai Road, Suzhou 215006, Jiangsu, China
| | - Qingjian Lei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| | - Jun Lei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| | - Renxiong Wei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| | - Yifeng Lei
- The Institute of Technological Science, School of Power and Mechanical Engineering, Wuhan University, 299 Bayi Road, Wuchang District, Wuhan 430072, Hubei, China
| | - Longjian Xue
- The Institute of Technological Science, School of Power and Mechanical Engineering, Wuhan University, 299 Bayi Road, Wuchang District, Wuhan 430072, Hubei, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, 99 Shangda Road, Baoshan District, Shanghai 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, 99 Shangda Road, Baoshan District, Shanghai 200444, China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| | - Feifei Yan
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, 169 Donghu Street, Wuchang District, Wuhan 430071, Hubei, China
| |
Collapse
|
15
|
Lai W, Geliang H, Bin X, Wang W. Effects of hydrogel stiffness and viscoelasticity on organoid culture: a comprehensive review. Mol Med 2025; 31:83. [PMID: 40033190 PMCID: PMC11877758 DOI: 10.1186/s10020-025-01131-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
As an emerging technology, organoids are promising new tools for basic and translational research in disease. Currently, the culture of organoids relies mainly on a type of unknown composition scaffold, namely Matrigel, which may pose problems in studying the effect of mechanical properties on organoids. Hydrogels, a new material with adjustable mechanical properties, can adapt to current studies. In this review, we summarized the synthesis of recent advance in developing definite hydrogel scaffolds for organoid culture and identified the critical parameters for regulating mechanical properties. In addition, classified by different mechanical properties like stiffness and viscoelasticity, we concluded the effect of mechanical properties on the development of organoids and tumor organoids. We hope this review enhances the understanding of the development of organoids by hydrogels and provides more practical approaches to investigating them.
Collapse
Affiliation(s)
- Wei Lai
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hu Geliang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xu Bin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
16
|
He S, Wang B, Hou H, Zhang Y, Zhang Z, Zhao F, Su M. Silk fibroin protein-templated gold nanoclusters for in vivo fluorescence imaging. Photochem Photobiol Sci 2025; 24:467-477. [PMID: 40113738 DOI: 10.1007/s43630-025-00699-7] [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: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Protein-templated synthesis has been proved to be an effective approach for building high-performance fluorescent bioimaging agents. Nevertheless, the high cost of proteins has restricted its wide application. Silk fibroin is a low cost natural protein with superior properties, which holds significant application prospects in many biomedical fields. However, its application potential in the biomedical imaging field remains to be explored. Herein, we report a one-pot green synthesis of high performance gold nanoclusters (AuNCs) using silk fibroin as stabilizer. The SF-AuNCs (~ 1.8 nm) shows a red emission around 600 nm with a large Stokes shift of 200 nm and a quantum yield of 5.42% which is comparable with that of fluorescence proteins. Meanwhile, the lifetime is as high as 3.47 μs which is about three magnitude orders higher than that of most molecular dyes and fluorescence proteins. Moreover, the stability is also greatly enhanced than that of the classical GSH-AuNCs. The SF-AuNCs is also well performed in cell labeling and in vivo imaging in zebrafish. This work not only provides a promising high performance protein fluorescence nano agent for bioimaging, but also expands the potential of the silk fibroin application in the biomedical imaging field.
Collapse
Affiliation(s)
- Shanshan He
- College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Baozhu Wang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Huixin Hou
- Key Laboratory of Surface and Interface Science of Polymer Materials of Zhejiang Province, School of Chemistry and Chemical Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yueyue Zhang
- Key Laboratory of Surface and Interface Science of Polymer Materials of Zhejiang Province, School of Chemistry and Chemical Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zhijun Zhang
- Key Laboratory of Surface and Interface Science of Polymer Materials of Zhejiang Province, School of Chemistry and Chemical Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- Shengzhou Innovation Research Institute of Zhejiang Sci-Tech University, Shengzhou, 312400, China.
| | - Feng Zhao
- College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Miao Su
- College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- Shengzhou Innovation Research Institute of Zhejiang Sci-Tech University, Shengzhou, 312400, China.
| |
Collapse
|
17
|
Li R, Wang J, Lin Q, Yin Z, Zhou F, Chen X, Tan H, Su J. Mechano-Responsive Biomaterials for Bone Organoid Construction. Adv Healthc Mater 2025; 14:e2404345. [PMID: 39740101 DOI: 10.1002/adhm.202404345] [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: 11/05/2024] [Revised: 12/08/2024] [Indexed: 01/02/2025]
Abstract
Mechanical force is essential for bone development, bone homeostasis, and bone fracture healing. In the past few decades, various biomaterials have been developed to provide mechanical signals that mimic the natural bone microenvironment, thereby promoting bone regeneration. Bone organoids, emerging as a novel research approach, are 3D micro-bone tissues that possess the ability to self-renew and self-organize, exhibiting biomimetic spatial characteristics. Incorporating mechano-responsive biomaterials in the construction of bone organoids presents a promising avenue for simulating the mechanical bone microenvironment. Therefore, this review commences by elucidating the impact of mechanical force on bone health, encompassing both cellular interactions and alterations in bone structure. Furthermore, the most recent applications of mechano-responsive biomaterials within the realm of bone tissue engineering are highlighted. Three different types of mechano-responsive biomaterials are introduced with a focus on their responsive mechanisms, construction strategies, and efficacy in facilitating bone regeneration. Based on a comprehensive overview, the prospective utilization and future challenges of mechano-responsive biomaterials in the construction of bone organoids are discussed. As bone organoid technology advances, these biomaterials are poised to become powerful tools in bone regeneration.
Collapse
Affiliation(s)
- Ruiyang Li
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Jian Wang
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Qiushui Lin
- Department of Spine Surgery, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, P. R. China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, P. R. China
| | - Xiao Chen
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Hongbo Tan
- Department of Orthopedics, The 920th Hospital of Joint Logistics Support Force, Yunnan, 650020, P. R. China
| | - Jiacan Su
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
18
|
Chen J, Li Q, Li H, Lv C, Yu H, Feng Q, Dong H. Injectable acellular matrix microgel assembly with stem cell recruitment and chondrogenic differentiation functions promotes microfracture-based articular cartilage regeneration. Bioact Mater 2025; 44:220-235. [PMID: 39497706 PMCID: PMC11533518 DOI: 10.1016/j.bioactmat.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/29/2024] [Accepted: 10/14/2024] [Indexed: 11/07/2024] Open
Abstract
Articular cartilage repair and regeneration is still a significant challenge despite years of research. Although microfracture techniques are commonly used in clinical practice, the newborn cartilage is usually fibrocartilage rather than hyaline cartilage, which is mainly attributed to the inadequate microenvironment for effectively recruiting, anchoring, and inducing bone marrow mesenchymal stem cells (BMSCs) to differentiate into hyaline cartilage. This paper introduces a novel cartilage acellular matrix (CACM) microgel assembly with excellent microporosity, injectability, tissue adhesion, BMSCs recruitment and chondrogenic differentiation capabilities to improve the microfracture-based articular cartilage regeneration. Specifically, the sustained release of simvastatin (SIM) from the SIM@CACM microgel assembly efficiently recruits BMSCs in the early stage of cartilage regeneration, while the abundant interconnected micropores and high specific area assure the quick adhesion, proliferation and infiltration of BMSCs. Additionally, the active factors within the CACM matrix, appropriate mechanical properties of the microgel assembly, and excellent tissue adhesion provide a conductive environment for the continuous chondrogenic differentiation of BMSCs into hyaline cartilage. Owing to the synergistic effect of the above-mentioned factors, good articular cartilage repair and regeneration is achieved.
Collapse
Affiliation(s)
- Junlin Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510641, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Qingtao Li
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510641, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510641, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Haofei Li
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510641, China
| | - Chuhan Lv
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Hongbo Yu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510641, China
| | - Qi Feng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510641, China
| | - Hua Dong
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510641, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510641, China
| |
Collapse
|
19
|
Wang Z, Zhao F, Lang H, Ren H, Zhang Q, Huang X, He C, Xu C, Tan C, Ma J, Duan S, Wang Z. Organoids in skin wound healing. BURNS & TRAUMA 2025; 13:tkae077. [PMID: 39759541 PMCID: PMC11697111 DOI: 10.1093/burnst/tkae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025]
Abstract
Stem cells (SCs) can self-replicate and differentiate into multiple lineages. Organoids, 3D cultures derived from SCs, can replicate the spatial structure and physiological characteristics of organs in vitro. Skin organoids can effectively simulate the physiological structure and function of skin tissue, reliably restoring the natural skin ecology in various in vitro environments. Skin organoids have been employed extensively in skin development and pathology research, offering valuable insights for drug screening. Moreover, they play crucial roles in skin regeneration and tissue repair. This in-depth review explores the construction and applications of skin organoids in wound healing, with a focus on their construction process, including skin appendage integration, and significant advancements in wound-healing research.
Collapse
Affiliation(s)
- Zitong Wang
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang, Liaoning 110013, China
| | - Hongxin Lang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang, Liaoning 110013, China
| | - Haiyue Ren
- Department of Pathology, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No. 1 Hospital), No. 215 Zhongshan Street, Wuhan, Hubei 430022, China
| | - Qiqi Zhang
- Department of Pathology, Chengdu Third People's Hospital, No. 82 Qinglong Street, Chengdu, Sichuan 610031, China
| | - Xing Huang
- Department of Anaesthesiology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yantaxi Road, Xi'an, Shanxi 710061, China
| | - Cai He
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Chengcheng Xu
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Chiyu Tan
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Jiajie Ma
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Shu Duan
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| |
Collapse
|
20
|
Hashemi-Afzal F, Fallahi H, Bagheri F, Collins MN, Eslaminejad MB, Seitz H. Advancements in hydrogel design for articular cartilage regeneration: A comprehensive review. Bioact Mater 2025; 43:1-31. [PMID: 39318636 PMCID: PMC11418067 DOI: 10.1016/j.bioactmat.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
This review paper explores the cutting-edge advancements in hydrogel design for articular cartilage regeneration (CR). Articular cartilage (AC) defects are a common occurrence worldwide that can lead to joint breakdown at a later stage of the disease, necessitating immediate intervention to prevent progressive degeneration of cartilage. Decades of research into the biomedical applications of hydrogels have revealed their tremendous potential, particularly in soft tissue engineering, including CR. Hydrogels are highly tunable and can be designed to meet the key criteria needed for a template in CR. This paper aims to identify those criteria, including the hydrogel components, mechanical properties, biodegradability, structural design, and integration capability with the adjacent native tissue and delves into the benefits that CR can obtain through appropriate design. Stratified-structural hydrogels that emulate the native cartilage structure, as well as the impact of environmental stimuli on the regeneration outcome, have also been discussed. By examining recent advances and emerging techniques, this paper offers valuable insights into developing effective hydrogel-based therapies for AC repair.
Collapse
Affiliation(s)
- Fariba Hashemi-Afzal
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Hooman Fallahi
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, 14115-111, Iran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104 USA
| | - Fatemeh Bagheri
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Maurice N. Collins
- School of Engineering, Bernal Institute and Health Research Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 16635-148, Iran
| | - Hermann Seitz
- Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Justus-von-Liebig-Weg 6, 18059 Rostock, Germany
- Department Life, Light & Matter, University of Rostock, Albert-Einstein-Straße 25, 18059 Rostock, Germany
| |
Collapse
|
21
|
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
|
22
|
Shen C, Zhou Z, Li R, Yang S, Zhou D, Zhou F, Geng Z, Su J. Silk fibroin-based hydrogels for cartilage organoids in osteoarthritis treatment. Theranostics 2025; 15:560-584. [PMID: 39744693 PMCID: PMC11671376 DOI: 10.7150/thno.103491] [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: 09/10/2024] [Accepted: 11/06/2024] [Indexed: 01/11/2025] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by cartilage degeneration. It can cause severe pain, deformity and even amputation risk. However, existing clinical treatment methods for cartilage repair present certain deficiencies. Meanwhile, the repair effect of cartilage tissue engineering is also unsatisfactory. Cartilage organoids are multicellular aggregates with cartilage-like three-dimensional structure and function. On the one hand, cartilage organoids can be used to explore the pathogenesis of OA by constructing disease models. On the other hand, it can be used as filler for rapid cartilage repair. Extracellular matrix (ECM)-like three-dimensional environment is the key to construct cartilage organoids. Silk fibroin (SF)-based hydrogels not only have ECM-like structure, but also have unique mechanical properties and remarkable biocompatibility. Therefore, SF-based hydrogels are considered as ideal biomaterials for constructing cartilage organoids. In this review, we reviewed the studies of cartilage organoids and SF-based hydrogels. The advantages of SF-based hydrogels in constructing cartilage organoids and the iterative optimization of cartilage organoids through designing hydrogels by using artificial intelligence (AI) calculation are also discussed. This review aims to provide a theoretical basis for the treatment of OA using SF-based biomaterials and cartilage organoids.
Collapse
Affiliation(s)
- Congyi Shen
- 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
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ziyang Zhou
- 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
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ruiyang Li
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shike Yang
- 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
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Anesthesiology, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Dongyang Zhou
- 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
| | - 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
|
23
|
Lu J, Gao Z, He W, Lu Y. Harnessing the potential of hyaluronic acid methacrylate (HAMA) hydrogel for clinical applications in orthopaedic diseases. J Orthop Translat 2025; 50:111-128. [PMID: 39886531 PMCID: PMC11779684 DOI: 10.1016/j.jot.2024.11.004] [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] [Received: 07/11/2024] [Revised: 09/29/2024] [Accepted: 11/12/2024] [Indexed: 02/01/2025] Open
Abstract
The treatment of orthopaedic diseases, such as fractures and osteoarthritis, remains a significant challenge due to the complex requirements for mechanical strength and tissue repair. Hydrogels based on hyaluronic acid methacrylate (HAMA) show promise as tissue engineering materials for these conditions. Hyaluronic acid (HA) is a natural component of the extracellular matrix, known for its good compatibility. The mechanical strength of HAMA-based hydrogels can be adjusted through crosslinking and by combining them with other materials. This review provides an overview of recent research on HAMA-based hydrogels for tissue engineering applications in orthopaedic diseases. First, we summarize the techniques for the preparation and characterization of HAMA hydrogels. Next, we offer a detailed review of the use of HAMA-based hydrogels in treating conditions such as cartilage injuries, bone defects, and meniscus injuries. Additionally, we discuss the applications of HAMA-based hydrogels in other diseases related to orthopaedics. Finally, we point out the challenges and propose future directions for the clinical translation of HAMA-based hydrogels. Translational potential statement HAMA-based hydrogels show strong translational potential in orthopaedics due to their biocompatibility, adjustable mechanical properties, and regenerative capabilities. With ongoing research, these hydrogels are well-positioned for clinical applications, particularly in cartilage repair, meniscus injuries, and osteoarthritis treatment.
Collapse
Affiliation(s)
- Junliang Lu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, 16 West Huangjiahu Road, Wuhan, Hubei, 430061, China
| | - Zhifei Gao
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou, Guangdong, 51282, China
| | - Wei He
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, 16 West Huangjiahu Road, Wuhan, Hubei, 430061, China
- Department of Orthopedics, Hubei Provincial Hospital of Traditional Chinese Medicine, 856 Luoyu Road, Wuhan, Hubei, 430061, China
- Hubei Shizhen Laboratory, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, 856 Luoyu Road, Wuhan, Hubei, 430061, China
| | - Yao Lu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou, Guangdong, 51282, China
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou, Guangdong, 51282, China
| |
Collapse
|
24
|
Shi Y, Han X, Zou S, Liu G. Nanomaterials in Organoids: From Interactions to Personalized Medicine. ACS NANO 2024; 18:33276-33292. [PMID: 39609736 DOI: 10.1021/acsnano.4c13330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Organoids are three-dimensional models of microscopic organisms created through the self-organization of various types of stem cells. They are widely unitized in personalized medicine due to their capacity to replicate the structure and functionality of native organs. Meanwhile, nanotechnology has been integrated into diagnostic and therapeutic tools to manage an array of medical conditions, given its unique characteristics of nanoscale. Nanomaterials have demonstrated potential in developing innovative and effective organoids. With a focus on studying the interaction of nanomaterials and organoid technology in personalized medicine, this Review examines the role of nanomaterials in regulating the fate of stem cells to construct different types of organoids. It also explores the potential of nanotechnology to create 3D microenvironments for organoids. Finally, perspectives and challenges of applying nanotechnology for organoids development toward the translation of personalized medicine are discussed.
Collapse
Affiliation(s)
- Ying Shi
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Xin Han
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Siyi Zou
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Guozhen Liu
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
25
|
Zhang S, Zhong R, Younis MR, He H, Xu H, Li G, Yang R, Lui S, Wang Y, Wu M. Hydrogel Applications in the Diagnosis and Treatment of Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:65754-65778. [PMID: 39366948 DOI: 10.1021/acsami.4c11855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Glioblastoma multiforme (GBM), a common malignant neurological tumor, has boundaries indistinguishable from those of normal tissue, making complete surgical removal ineffective. The blood-brain barrier (BBB) further impedes the efficacy of radiotherapy and chemotherapy, leading to suboptimal treatment outcomes and a heightened probability of recurrence. Hydrogels offer multiple advantages for GBM diagnosis and treatment, including overcoming the BBB for improved drug delivery, controlled drug release for long-term efficacy, and enhanced relaxation properties of magnetic resonance imaging (MRI) contrast agents. Hydrogels, with their excellent biocompatibility and customizability, can mimic the in vivo microenvironment, support tumor cell culture, enable drug screening, and facilitate the study of tumor invasion and metastasis. This paper reviews the classification of hydrogels and recent research for the diagnosis and treatment of GBM, including their applications as cell culture platforms and drugs including imaging contrast agents carriers. The mechanisms of drug release from hydrogels and methods to monitor the activity of hydrogel-loaded drugs are also discussed. This review is intended to facilitate a more comprehensive understanding of the current state of GBM research. It offers insights into the design of integrated hydrogel-based GBM diagnosis and treatment with the objective of achieving the desired therapeutic effect and improving the prognosis of GBM.
Collapse
Affiliation(s)
- Shuaimei Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Renming Zhong
- Radiotherapy Physics & Technology Center, Cancer Center, West China Hospital, Chengdu, Sichuan 610041, P. R. China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Hualong He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Hong Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Ruiyan Yang
- Department of Biology, Macalester College, Saint Paul, Minnesota 55105, United States
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
26
|
A S S, G MK. In vitro chondrogenic potential of marine biocomposite hydrogel construct for cartilage tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:2845-2866. [PMID: 39431438 DOI: 10.1080/09205063.2024.2391223] [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: 07/01/2024] [Accepted: 08/07/2024] [Indexed: 10/22/2024]
Abstract
Cartilage tissue engineering (CTE) is a field of regenerative medicine focused on constructing ideal substitutes for injured cartilage by effectively combining cells, scaffolds, and stimulatory factors. In vitro CTE employing chondrocytes and biopolymer-based hydrogels has the potential to repair damaged cartilage. In this research, primary chondrocytes were extracted from the rib cartilage of rats and seeded on a hydrogel construct named HACF, which is made from hydroxyapatite, alginate, chitosan, and fucoidan. We then evaluated in vitro chondrogenesis on HACF cartilage construct. The results revealed that the primary chondrocytes were successfully isolated from rat rib cartilage by collagenase D digestion and HACF cartilage construct was effectively synthesized. Chondrocyte viability and its differentiation inside the scaffold HACF were determined by MTT assay, NRU assay, live/dead assay, DAPI nuclear staining, flow cytometry analysis (FCA), mRNA expression studies, and quantification of extracellular matrix components in the HACF scaffold. The findings indicated excellent chondrocyte viability within the HACF scaffold, with no noticeable changes in morphology. Apoptosis was not detected in the chondrocytes cultured on these hydrogels, as confirmed by DAPI staining, live/dead assay, and FCA. This demonstrates that the cells were capable of proliferating, dividing, multiplying, and maintaining their integrity on HACF scaffold. The results also showed more collagen deposition and glycosaminoglycan synthesis showing the good health of chondrocytes on the HACF construct. It indicates that HACF is an ideal scaffold supporting stable cartilage matrix production, highlighting its suitability for cartilage tissue engineering.
Collapse
Affiliation(s)
- Sumayya A S
- Assistant Professor, Department of Biochemistry, T.K.M. College of Arts and Science, kollam-5, kerala, India
| | | |
Collapse
|
27
|
Ge JY, Wang Y, Li QL, Liu FK, Lei QK, Zheng YW. Trends and challenges in organoid modeling and expansion with pluripotent stem cells and somatic tissue. PeerJ 2024; 12:e18422. [PMID: 39619184 PMCID: PMC11608026 DOI: 10.7717/peerj.18422] [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: 04/28/2024] [Accepted: 10/08/2024] [Indexed: 03/10/2025] Open
Abstract
The increasing demand for disease modeling, preclinical drug testing, and long waiting lists for alternative organ substitutes has posed significant challenges to current limitations in organoid technology. Consequently, organoid technology has emerged as a cutting-edge tool capable of accurately recapitulating the complexity of actual organs in physiology and functionality. To bridge the gaps between basic research and pharmaceutical as well as clinical applications, efforts have been made to develop organoids from tissue-derived stem cells or pluripotent stem cells. These developments include optimizing starting cells, refining culture systems, and introducing genetic modifications. With the rapid development of organoid technology, organoid composition has evolved from single-cell to multi-cell types, enhancing their level of biomimicry. Tissue structure has become more refined, and core challenges like vascularization are being addressed actively. These improvements are expected to pave the way for the construction of organoid atlases, automated large-scale cultivation, and universally compatible organoid biobanks. However, major obstacles remain to be overcome before urgently proof-of-concept organoids can be readily converted to practical applications. These obstacles include achieving structural and functional summarily to native tissue, remodeling the microenvironment, and scaling up production. This review aims to summarize the status of organoid development and applications, highlight recent progress, acknowledge existing limitations and challenges, and provide insights into future advancements. It is expected that this will contribute to the establishment of a reliable, scalable, and practical platform for organoid production and translation, further promoting their use in the pharmaceutical industry and regenerative medicine.
Collapse
Affiliation(s)
- Jian-Yun Ge
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
- Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
- Innovation and Transformation Center, University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yun Wang
- Institute of Regenerative Medicine, and Department of Dermatology, Affilated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Dermatology, The First People’s Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Qi-Lin Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Fan-Kai Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Quan-Kai Lei
- Institute of Regenerative Medicine, and Department of Dermatology, Affilated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun-Wen Zheng
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
- Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
- Institute of Regenerative Medicine, and Department of Dermatology, Affilated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
Zou Y, Wang Z, Shi H, Hu J, Hu W. Soybean Isoflavones Alleviate Osteoarthritis Through Modulation of the TSC1/mTORC1 Signaling Pathway to Reduce Intrachondral Angiogenesis. Immunol Invest 2024; 53:1439-1455. [PMID: 39360672 DOI: 10.1080/08820139.2024.2410737] [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: 10/04/2024]
Abstract
BACKGROUND The incidence of osteoarthritis (OA) is increasing, yet its pathogenesis remains largely unknown. Recent studies suggest that abnormal subchondral bone remodeling plays a crucial role in OA development, highlighting a gap in clinical treatments targeting this aspect. Soybean Isoflavone (SI) has shown potential in treating OA, although its mechanisms are not fully understood. METHODS This research investigated the effects of SI on subchondral bone remodeling in an OA rat model, assessing joint damage, OARSI scores, and type H vessel formation (CD31hiEmcnhi expression). Additionally, the expression of ALP, OCN, BMP, and TSC1 was evaluated to determine involvement of the mTORC1 pathway. In vitro studies on IL-1β-induced osteoblasts further examined the impact of SI on TSC1/mTORC1 signaling and related markers. RESULTS SI treatment reduced joint damage and OARSI scores in the rat OA model, significantly decreasing CD31hiEmcnhi expression, indicating a reduction in type H vessel formation. SI also downregulated ALP, OCN, and BMP expression while upregulating TSC1, suggesting inhibition of the mTORC1 signaling pathway and VEGF release. In vitro, SI increased TSC1 expression and decreased mTORC1 signaling, VEGF, ALP, OCN, and BMP levels in IL-1β-induced osteoblasts. CONCLUSION SI targets the TSC1/mTORC1 signaling pathway to suppress osteoblast activation and VEGF release, inhibiting type H vessel formation and slowing abnormal subchondral bone remodeling. These findings provide a novel therapeutic approach for OA by focusing on subchondral bone remodeling mechanisms.
Collapse
Affiliation(s)
- Yang Zou
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhaoyang Wang
- Department of Orthopedics, Chengdu Bayi Orthopedic Hospital, Chengdu, China
| | - Hangchu Shi
- Department of Orthopedics, The Third People's Hospital of Yuhang District, Hangzhou, China
| | - Jiong Hu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Hu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
32
|
Shuai Y, Zheng M, Kundu SC, Mao C, Yang M. Bioengineered Silk Protein-Based 3D In Vitro Models for Tissue Engineering and Drug Development: From Silk Matrix Properties to Biomedical Applications. Adv Healthc Mater 2024; 13:e2401458. [PMID: 39009465 DOI: 10.1002/adhm.202401458] [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: 04/21/2024] [Revised: 06/22/2024] [Indexed: 07/17/2024]
Abstract
3D in vitro model has emerged as a valuable tool for studying tissue development, drug screening, and disease modeling. 3D systems can accurately replicate tissue microstructures and physiological features, mirroring the in vivo microenvironment departing from conventional 2D cell cultures. Various 3D in vitro models utilizing biomacromolecules like collagen and synthetic polymers have been developed to meet diverse research needs and address the complex challenges of contemporary research. Silk proteins, bearing structural and functional similarities to collagen, have been increasingly employed to construct advanced 3D in vitro systems, surpassing the limitations of 2D cultures. This review examines silk proteins' composition, structure, properties, and functions, elucidating their role in 3D in vitro models. Furthermore, recent advances in biomedical applications involving silk-based organoid models are discussed. In particular, the unique physiological attributes of silk matrix constituents in in vitro tissue constructs are highlighted, providing a meticulous evaluation of their importance. Additionally, it outlines the current research hurdles and complexities while contemplating future avenues, thereby paving the way for developing complex and biomimetic silk protein-based microtissues.
Collapse
Affiliation(s)
- Yajun Shuai
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Meidan Zheng
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, P. R. China
| | - Mingying Yang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
33
|
Guo L, Li P, Rong X, Wei X. Key roles of the superficial zone in articular cartilage physiology, pathology, and regeneration. Chin Med J (Engl) 2024:00029330-990000000-01274. [PMID: 39439390 DOI: 10.1097/cm9.0000000000003319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT The superficial zone (SFZ) of articular cartilage is an important interface that isolates deeper zones from the microenvironment of the articular cavity and is directly exposed to various biological and mechanical stimuli. The SFZ is not only a crucial structure for maintaining the normal physiological function of articular cartilage but also the earliest site of osteoarthritis (OA) cartilage degeneration and a major site of cartilage progenitor cells, suggesting that the SFZ might represent a key target for the early diagnosis and treatment of OA. However, to date, SFZ research has not received sufficient attention, accounting for only about 0.58% of cartilage tissue research. The structure, biological composition, function, and related mechanisms of the SFZ in the physiological and pathological processes of articular cartilage remain unclear. This article reviews the key role of the SFZ in articular cartilage physiology and pathology and focuses on the characteristics of SFZ in articular cartilage degeneration and regeneration in OA, aiming to provide researchers with a systematic understanding of the current research status of the SFZ of articular cartilage, hoping that scholars will give more attention to the SFZ of articular cartilage in the future.
Collapse
Affiliation(s)
- Li Guo
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Pengcui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xueqin Rong
- Department of Pain Medicine Center, Central Hospital of Sanya, Sanya, Hainan 572000, China
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| |
Collapse
|
34
|
Liu X, Ouyang Q, Yao X, Zhang Y. A facile nanopattern modification of silk fibroin electrospun scaffold and the corresponding impact on cell proliferation and osteogenesis. Regen Biomater 2024; 11:rbae117. [PMID: 39575301 PMCID: PMC11580685 DOI: 10.1093/rb/rbae117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/12/2024] [Accepted: 09/07/2024] [Indexed: 11/24/2024] Open
Abstract
As a well-known natural protein biomaterial, silk fibroin (SF) has shown broad application prospects in typical biomedical fields. However, the mostly used SF from Bombyx mori silkworm lacks specific cell adhesion sites and other bioactive peptide sequences, and there is still significant room for further improvement of their biological functions. Therefore, it is crucial to develop a facile and effective modification strategy for this widely researched biomaterial. In this study, the SF electrospun scaffold has been chosen as a typical SF biomaterial, and air plasma etching has been adopted as a facile nanopattern modification strategy to promote its biological functions. Results demonstrated that the plasma etching could feasibly and effectively create nano-island-like patterns on the complex surface of SF scaffolds, and the detailed nanopattern features could be easily regulated by adjusting the etching time. In addition, the mesenchymal stem cell responses have illustrated that the nanopattern modification could significantly regulate corresponding cell behaviors. Compared with the non-etched scaffold, the 10 min-etched scaffolds (10E scaffold) significantly promoted stem cell proliferation and osteogenic differentiation. Moreover, 10E scaffold has also been confirmed to effectively accelerate vascularization and ectopic osteogenesis in vivo using a rat subcutaneous implantation model. However, the mentioned promoting effects would be weakened or even counteracted with the increase of etching time. In conclusion, this facile modification strategy demonstrated great application potential for promoting cell proliferation and differentiation. Thus, it provided useful guidance to develop excellent SF-based scaffolds suitable for bone and other tissue engineering.
Collapse
Affiliation(s)
- Xiaojiao Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Qinjun Ouyang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Xiang Yao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Yaopeng Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| |
Collapse
|
35
|
Yang S, Wu H, Peng C, He J, Pu Z, Lin Z, Wang J, Hu Y, Su Q, Zhou B, Yong X, Lan H, Hu N, Hu X. From the microspheres to scaffolds: advances in polymer microsphere scaffolds for bone regeneration applications. BIOMATERIALS TRANSLATIONAL 2024; 5:274-299. [PMID: 39734699 PMCID: PMC11681185 DOI: 10.12336/biomatertransl.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 12/31/2024]
Abstract
The treatment and repair of bone tissue damage and loss due to infection, tumours, and trauma are major challenges in clinical practice. Artificial bone scaffolds offer a safer, simpler, and more feasible alternative to bone transplantation, serving to fill bone defects and promote bone tissue regeneration. Ideally, these scaffolds should possess osteoconductive, osteoinductive, and osseointegrative properties. However, the current first-generation implants, represented by titanium alloys, have shown poor bone-implant integration performance and cannot meet the requirements for bone tissue repair. This has led to increased research on second and third generation artificial bone scaffolds, which focus on loading bioactive molecules and cells. Polymer microspheres, known for their high specific surface areas at the micro- and nanoscale, exhibit excellent cell and drug delivery behaviours. Additionally, with their unique rigid structure, microsphere scaffolds can be constructed using methods such as thermal sintering, injection, and microsphere encapsulation. These scaffolds not only ensure the excellent cell drug loading performance of microspheres but also exhibit spatial modulation behaviour, aiding in bone repair within a three-dimensional network structure. This article provides a summary and discussion of the use of polymer microsphere scaffolds for bone repair, focusing on the mechanisms of bone tissue repair and the current status of clinical bone grafts, aimed at advancing research in bone repair.
Collapse
Affiliation(s)
- Shuhao Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Haoming Wu
- School of Preclinical Medicine of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Chao Peng
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Jian He
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Zhengguang Pu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Zhidong Lin
- The Second Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jun Wang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yingkun Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Qiao Su
- West China School of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Bingnan Zhou
- School of Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan Province, China
| | - Hai Lan
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan Province, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
36
|
Zhang P, Qin Q, Cao X, Xiang H, Feng D, Wusiman D, Li Y. Hydrogel microspheres for bone regeneration through regulation of the regenerative microenvironment. BIOMATERIALS TRANSLATIONAL 2024; 5:205-235. [PMID: 39734698 PMCID: PMC11681181 DOI: 10.12336/biomatertransl.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/12/2024] [Accepted: 09/13/2024] [Indexed: 12/31/2024]
Abstract
Bone defects are a prevalent category of skeletal tissue disorders in clinical practice, with a range of pathogenic factors and frequently suboptimal clinical treatment effects. In bone regeneration of bone defects, the bone regeneration microenvironment-composed of physiological, chemical, and physical components-is the core element that dynamically coordinates to promote bone regeneration. In recent years, medical biomaterials with bioactivity and functional tunability have been widely researched upon and applied in the fields of tissue replacement/regeneration, and remodelling of organ structure and function. The biomaterial treatment system based on the comprehensive regulation strategy of bone regeneration microenvironment is expected to solve the clinical problem of bone defect. Hydrogel microspheres (HMS) possess a highly specific surface area and porosity, an easily adjustable physical structure, and high encapsulation efficiency for drugs and stem cells. They can serve as highly efficient carriers for bioactive factors, gene agents, and stem cells, showing potential advantages in the comprehensive regulation of bone regeneration microenvironment to enhance bone regeneration. This review aims to clarify the components of the bone regeneration microenvironment, the application of HMS in bone regeneration, and the associated mechanisms. It also discusses various preparation materials and methods of HMS and their applications in bone tissue engineering. Furthermore, it elaborates on the relevant mechanisms by which HMS regulates the physiological, chemical, and physical microenvironment in bone regeneration to achieve bone regeneration. Finally, we discuss the future prospects of the HMS system application for comprehensive regulation of bone regeneration microenvironment, to provide novel perspectives for the research and application of HMS in the bone tissue engineering field.
Collapse
Affiliation(s)
- Pengrui Zhang
- Department of Orthopaedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Qiwei Qin
- Department of Orthopaedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Xinna Cao
- Department of Orthopaedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Honglin Xiang
- Department of Orthopaedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Dechao Feng
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Yuling Li
- Department of Orthopaedics, Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| |
Collapse
|
37
|
Bousso I, Genin G, Thomopoulos S. Achieving tendon enthesis regeneration across length scales. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2024; 31:100547. [PMID: 39219714 PMCID: PMC11364215 DOI: 10.1016/j.cobme.2024.100547] [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] [Indexed: 09/04/2024]
Abstract
Surgical reattachment of tendon to bone is a clinical challenge, with unacceptably high retear rates in the early period after repair. A primary reason for these repeated tears is that the multiscale toughening mechanisms found at the healthy tendon enthesis are not regenerated during tendon-to-bone healing. The need for technologies to improve these outcomes is pressing, and the tissue engineering community has responded with many advances that hold promise for eventually regenerating the multiscale tissue interface that transfers loads between the two dissimilar materials, tendon, and bone. This review provides an assessment of the state of these approaches, with the aim of identifying a critical agenda for future progress.
Collapse
Affiliation(s)
- Ismael Bousso
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Guy Genin
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO USA
| | - Stavros Thomopoulos
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- Department of Orthopaedic Surgery, Columbia University, New York, NY USA
| |
Collapse
|
38
|
Wu R, Li W, Yang P, Shen N, Yang A, Liu X, Ju Y, Lei L, Fang B. DNA hydrogels and their derivatives in biomedical engineering applications. J Nanobiotechnology 2024; 22:518. [PMID: 39210464 PMCID: PMC11360341 DOI: 10.1186/s12951-024-02791-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Deoxyribonucleotide (DNA) is uniquely programmable and biocompatible, and exhibits unique appeal as a biomaterial as it can be precisely designed and programmed to construct arbitrary shapes. DNA hydrogels are polymer networks comprising cross-linked DNA strands. As DNA hydrogels present programmability, biocompatibility, and stimulus responsiveness, they are extensively explored in the field of biomedicine. In this study, we provide an overview of recent advancements in DNA hydrogel technology. We outline the different design philosophies and methods of DNA hydrogel preparation, discuss its special physicochemical characteristics, and highlight the various uses of DNA hydrogels in biomedical domains, such as drug delivery, biosensing, tissue engineering, and cell culture. Finally, we discuss the current difficulties facing DNA hydrogels and their potential future development.
Collapse
Affiliation(s)
- Rui Wu
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Wenting Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences School of Basic Medicine, Peking Union Medical College, Beijing, 100000, China
| | - Pu Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Naisi Shen
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Anqi Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xiangjun Liu
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yikun Ju
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China.
| | - Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
39
|
Fan MH, Pi JK, Zou CY, Jiang YL, Li QJ, Zhang XZ, Xing F, Nie R, Han C, Xie HQ. Hydrogel-exosome system in tissue engineering: A promising therapeutic strategy. Bioact Mater 2024; 38:1-30. [PMID: 38699243 PMCID: PMC11061651 DOI: 10.1016/j.bioactmat.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/24/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Characterized by their pivotal roles in cell-to-cell communication, cell proliferation, and immune regulation during tissue repair, exosomes have emerged as a promising avenue for "cell-free therapy" in clinical applications. Hydrogels, possessing commendable biocompatibility, degradability, adjustability, and physical properties akin to biological tissues, have also found extensive utility in tissue engineering and regenerative repair. The synergistic combination of exosomes and hydrogels holds the potential not only to enhance the efficiency of exosomes but also to collaboratively advance the tissue repair process. This review has summarized the advancements made over the past decade in the research of hydrogel-exosome systems for regenerating various tissues including skin, bone, cartilage, nerves and tendons, with a focus on the methods for encapsulating and releasing exosomes within the hydrogels. It has also critically examined the gaps and limitations in current research, whilst proposed future directions and potential applications of this innovative approach.
Collapse
Affiliation(s)
- Ming-Hui Fan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Jin-Kui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Yan-Lin Jiang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Qian-Jin Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Xiu-Zhen Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Fei Xing
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Chen Han
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, 610212, PR China
| |
Collapse
|
40
|
Wang J, Wu Y, Li G, Zhou F, Wu X, Wang M, Liu X, Tang H, Bai L, Geng Z, Song P, Shi Z, Ren X, Su J. Engineering Large-Scale Self-Mineralizing Bone Organoids with Bone Matrix-Inspired Hydroxyapatite Hybrid Bioinks. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309875. [PMID: 38642033 DOI: 10.1002/adma.202309875] [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: 09/23/2023] [Revised: 04/02/2024] [Indexed: 04/22/2024]
Abstract
Addressing large bone defects remains a significant challenge owing to the inherent limitations in self-healing capabilities, resulting in prolonged recovery and suboptimal regeneration. Although current clinical solutions are available, they have notable shortcomings, necessitating more efficacious approaches to bone regeneration. Organoids derived from stem cells show great potential in this field; however, the development of bone organoids has been hindered by specific demands, including the need for robust mechanical support provided by scaffolds and hybrid extracellular matrices (ECM). In this context, bioprinting technologies have emerged as powerful means of replicating the complex architecture of bone tissue. The research focused on the fabrication of a highly intricate bone ECM analog using a novel bioink composed of gelatin methacrylate/alginate methacrylate/hydroxyapatite (GelMA/AlgMA/HAP). Bioprinted scaffolds facilitate the long-term cultivation and progressive maturation of extensive bioprinted bone organoids, foster multicellular differentiation, and offer valuable insights into the initial stages of bone formation. The intrinsic self-mineralizing quality of the bioink closely emulates the properties of natural bone, empowering organoids with enhanced bone repair for both in vitro and in vivo applications. This trailblazing investigation propels the field of bone tissue engineering and holds significant promise for its translation into practical applications.
Collapse
Affiliation(s)
- Jian Wang
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Yan Wu
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Guangfeng Li
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
- Department of Trauma Orthopedics, Zhongye Hospital, Shanghai, 200941, P. R. China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, P. R. China
| | - Xiang Wu
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Miaomiao Wang
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Xinru Liu
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Hua Tang
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Long Bai
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Zhen Geng
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Peiran Song
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Zhongmin Shi
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai, 200233, P. R. China
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Jiacan Su
- Institute of Translational Medicine, Musculoskeletal Organoid Research Center, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| |
Collapse
|
41
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [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: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
42
|
Dai W, Liang J, Guo R, Zhao Z, Na Z, Xu D, Li D. Bioengineering approaches for the endometrial research and application. Mater Today Bio 2024; 26:101045. [PMID: 38600921 PMCID: PMC11004221 DOI: 10.1016/j.mtbio.2024.101045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/07/2024] [Accepted: 03/29/2024] [Indexed: 04/12/2024] Open
Abstract
The endometrium undergoes a series of precise monthly changes under the regulation of dynamic levels of ovarian hormones that are characterized by repeated shedding and subsequent regeneration without scarring. This provides the potential for wound healing during endometrial injuries. Bioengineering materials highlight the faithful replication of constitutive cells and the extracellular matrix that simulates the physical and biomechanical properties of the endometrium to a larger extent. Significant progress has been made in this field, and functional endometrial tissue bioengineering allows an in-depth investigation of regulatory factors for endometrial and myometrial defects in vitro and provides highly therapeutic methods to alleviate obstetric and gynecological complications. However, much remains to be learned about the latest progress in the application of bioengineering technologies to the human endometrium. Here, we summarize the existing developments in biomaterials and bioengineering models for endometrial regeneration and improving the female reproductive potential.
Collapse
Affiliation(s)
- Wanlin Dai
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Junzhi Liang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Renhao Guo
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China
| | - Zhongyu Zhao
- Innovation Institute, China Medical University, Shenyang, China
| | - Zhijing Na
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China
| | - Dake Xu
- Shenyang National Laboratory for Materials Science, Northeastern University, Shenyang, China
| | - Da Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China
| |
Collapse
|