1
|
Xing X, Gong Z, Chen C, Lin Y, Liu P, Xiao T, Yu H, Li Y, Lin Y, Tan G, Ning C, Wu Z, Wang L, Zhou L. Injectable bioresponsive bone adhesive hydrogels inhibit NLRP3 inflammasome on demand to accelerate diabetic fracture healing. Biomaterials 2025; 317:123059. [PMID: 39731843 DOI: 10.1016/j.biomaterials.2024.123059] [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: 10/08/2024] [Revised: 12/16/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Diabetes is associated with excessive inflammation, which negatively impacts the fracture healing process and delays bone repair. Previously, growing evidence indicated that activation of the nod-like receptor (NLR) family, such as nod-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome induces a vicious cycle of chronic low-grade inflammatory responses in diabetic fracture. Here, we describe the synthesis of a bone adhesive hydrogel that can be locally injected into the fracture site and releases a natural inhibitor of NLRP3 (rutin) in response to pathological cue reactive oxygen species activity (ROS). The hydrogel (denoted as RPO) was facilely formed by the cross-linking of rutin-functionalized gelatin, poly(vinyl alcohol), and oxidized starch based on the dynamic schiff base and boronate ester bond. Specifically, rutin is conjugated in the RPO hydrogel via a ROS linker and is released as the linker is cleaved by active ROS. In vitro studies demonstrate that RPO hydrogel effectively mitigates oxidative stress, alleviates mitochondrial dysfunction, and limits the overactivation of NLRP3 inflammasome in bone marrow derived macrophages, thereby promoting osteogenic differentiation of bone marrow mesenchymal stem cells. In a diabetic rat fracture model, RPO hydrogel significantly accelerates bone repair by modulating the inflammatory microenvironment. Our results demonstrate that local, on-demand NLRP3 inhibition for the treatment of diabetic fracture is achievable by using an injectable bioresponsive adhesive RPO hydrogel.
Collapse
Affiliation(s)
- Xudan Xing
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Zunlei Gong
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Chuke Chen
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Yeyin Lin
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Peiyi Liu
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Tianhua Xiao
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China; School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Hui Yu
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Yuanxin Li
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Yucong Lin
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Guoxin Tan
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Chengyun Ning
- School of Materials Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510641, PR China
| | - Zenghui Wu
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China.
| | - Le Wang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China.
| | - Lei Zhou
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, PR China.
| |
Collapse
|
2
|
Son G, Song J, Park JC, Kim HN, Kim H. Fusogenic lipid nanoparticles for rapid delivery of large therapeutic molecules to exosomes. Nat Commun 2025; 16:4799. [PMID: 40410169 PMCID: PMC12102247 DOI: 10.1038/s41467-025-59489-5] [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: 09/03/2024] [Accepted: 04/25/2025] [Indexed: 05/25/2025] Open
Abstract
Exosomes, as cell-derived lipid nanoparticles, are promising drug carriers because they can traverse challenging physiological barriers such as the blood-brain barrier (BBB). However, a major obstacle in utilizing exosomes as drug carriers is loading large therapeutic molecules without compromising the structural integrity of embedded biomolecules. Here, we introduce a membrane fusion method utilizing fusogenic lipid nanoparticles, cubosomes, to load large molecules into exosomes in a non-destructive manner. When the drug-loaded cubosome and exosome solutions are simply mixed, membrane fusion is completed in just 10 min. Our method effectively loads doxorubicin and immunoglobulin G into exosomes. Moreover, even the most challenging molecule-mRNA-is loaded with nearly 100% efficiency, demonstrating the versatility of our approach. In terms of biological behavior, the resulting hybrid exosomes preserve the functional behavior of exosomes in BBB uptake and penetration. Surprisingly, controlling exosome-to-cubosome ratios allows precise control over BBB uptake and transport. Furthermore, these hybrid exosomes retain cell-specific delivery properties, preserving the targeted delivery functions dictated by their exosomal origin. This study demonstrates the feasibility of a mix-and-load method for rapid and efficient drug loading into exosomes, with significant potential for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Gamsong Son
- Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jiyoung Song
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jae Chul Park
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hong Nam Kim
- Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea.
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea.
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Hojun Kim
- Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea.
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
3
|
Zhang C, Zhu C, Chen X, Chen X, Zhang D, Zhao H, Zhang J, Zhang Y, Xu W, Zhao X, Hu Y, Wei W, Xu J, Li Y, Wu B. Natural regeneration-inspired sequential delivery of synergistic growth factors for structural and functional endometrial regeneration. Acta Biomater 2025:S1742-7061(25)00375-7. [PMID: 40412507 DOI: 10.1016/j.actbio.2025.05.050] [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: 01/24/2025] [Revised: 05/16/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Large-scale deep endometrial injury has a serious impact on the reproductive health of women, necessitating the development of novel therapeutic approaches. Treatment strategies using single factor may not perfectly match the intricate and dynamic process of endometrial regeneration. In light of the sequential progression of vascularization and endometrial remodeling observed during the regeneration of injured endometrium, a dual growth factor sequential delivery system is prepared by loading IGF-1 onto hydrogel microspheres and blending with an outer bulk hydrogel containing VEGF. The controlled degradation of hydrogel facilitates the sequential release of the two factors, thereby fostering the vascularization, migration and proliferation of endometrial cells in vitro. Animal experiments have proved that the hydrogel system can promote the regeneration of endometrial structure through vascular remodeling, glandular regeneration, and proliferation of endometrial cells, and simultaneously improve the rate of embryo implantation and live birth, which further indicates the functional reconstruction of the injured endometrium. Consequently, drawing inspiration from the sequential process of endometrial regeneration, this study provides innovative strategies for structural and functional restoration of the endometrium. STATEMENT OF SIGNIFICANCE: This research presents an innovative approach to the treatment of injured endometrium through a sequential dual growth factor delivery system. The system involves the incorporation of IGF-1 onto hydrogel microspheres, which are subsequently embedded within a GelMA hydrogel matrix containing VEGF. Unlike conventional hydrogel-based therapeutic strategies that involve the loading of growth factors, the developed delivery system is engineered in accordance with the vascularization and endometrial remodeling processes inherent to the regeneration of injured endometrial tissue. It facilitates the initial release of VEGF to stimulate the formation of blood vessels, followed by a gradual release of IGF-1 during the intermediate phase of endometrial regeneration to promote tissue remodeling. Pre-clinical animal studies have demonstrated that this innovative delivery strategy effectively restores the structure and function of the endometrium, suggesting significant potential for clinical application.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Chengcheng Zhu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Xiao Chen
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Xuzhi Chen
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Di Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Huafei Zhao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Junwen Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Yu Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Wanwan Xu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Xiaofeng Zhao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Yingying Hu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000
| | - Wei Wei
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000.
| | - Jian Xu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China, 310006.
| | - Yu Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000.
| | - Bingbing Wu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China, 322000.
| |
Collapse
|
4
|
Drohat P, Baron M, Kaplan LD, Best TM, Kouroupis D. Long-Acting Extracellular Vesicle-Based Biologics in Osteoarthritis Immunotherapy. Bioengineering (Basel) 2025; 12:525. [PMID: 40428144 PMCID: PMC12109516 DOI: 10.3390/bioengineering12050525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 05/01/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by low-grade inflammation, cartilage breakdown, and persistent pain. Despite its prevalence, current therapeutic strategies primarily focus on symptom management rather than modifying disease progression. Monoclonal antibodies and cytokine inhibitors targeting inflammatory pathways, including TNF-α and IL-1, have shown promise but remain limited by inconsistent efficacy and safety concerns. Long-acting biologic therapies-ranging from extended-release formulations, such as monoclonal antibodies and cytokine inhibitors, to gene therapy approaches-have emerged as promising strategies to enhance treatment durability and improve patient outcomes. Extracellular vesicles (EVs) have gained particular attention as a novel delivery platform due to their inherent stability, biocompatibility, and ability to transport therapeutic cargo, including biologics and immunomodulatory agents, directly to joint tissues. This review explores the evolving role of EVs in OA treatment, highlighting their ability to extend drug half-life, improve targeting, and modulate inflammatory responses. Additionally, strategies for EV engineering, including endogenous and exogenous cargo loading, genetic modifications, and biomaterial-based delivery systems, are discussed.
Collapse
Affiliation(s)
- Philip Drohat
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (P.D.); (M.B.); (L.D.K.); (T.M.B.)
| | - Max Baron
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (P.D.); (M.B.); (L.D.K.); (T.M.B.)
| | - Lee D. Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (P.D.); (M.B.); (L.D.K.); (T.M.B.)
| | - Thomas M. Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (P.D.); (M.B.); (L.D.K.); (T.M.B.)
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (P.D.); (M.B.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute, Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
5
|
Zheng Y, Fu L, Zhang Z, Wu J, Yuan X, Ding Z, Ning C, Sui X, Liu S, Guo Q. Three-Dimensional Bioprinting of Growth Differentiation Factor 5-Preconditioned Mesenchymal Stem Cell-Derived Exosomes Facilitates Articular Cartilage Endogenous Regeneration. ACS NANO 2025; 19:15281-15301. [PMID: 40245443 DOI: 10.1021/acsnano.4c13492] [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: 04/19/2025]
Abstract
The repair of articular cartilage defects remains a major regenerative and clinical challenge. Exosomes (Exos) derived from mesenchymal stem cells (MSCs) have good application potential in cartilage tissue engineering. Numerous studies have indicated that appropriate preconditioning methods can promote the therapeutic effect of Exos. Growth differentiation factor 5 (GDF-5) plays a critical role in chondrogenesis and regeneration. In this study, GDF-5 was used to precondition synovial mesenchymal stem cells (SMSCs) to increase the chondrogenic-promoting effect of Exos (G-Exos). In addition, we demonstrated that G-Exos rich in miR-383-3p increased the chondrogenic potential of SMSCs by activating the Kdm2a/SOX2 signaling pathway. On this basis, G-Exos were loaded into a glycyrrhizic acid/methacrylate-acylated hyaluronic acid (GA/HA/G-Exos) scaffold via digital light processing (DLP) bioprinting to maintain bioactivity and sustained release. The GA/HA/G-Exos scaffolds not only presented significant biological properties in vitro but also significantly promoted the remodeling of the joint cavity regenerative microenvironment and the regeneration of articular cartilage in Sprague-Dawley rats. This study provides a promising cell-free regenerative strategy for cartilage defect repair via the use of engineered exofunctionalized biological scaffolds.
Collapse
Affiliation(s)
- Yazhe Zheng
- Guizhou Medical University, Guiyang, Guizhou Province 550004, People's Republic of China
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Liwei Fu
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Zhichao Zhang
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Jiang Wu
- Guizhou Medical University, Guiyang, Guizhou Province 550004, People's Republic of China
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Xun Yuan
- Guizhou Medical University, Guiyang, Guizhou Province 550004, People's Republic of China
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Zhengang Ding
- Guizhou Medical University, Guiyang, Guizhou Province 550004, People's Republic of China
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Chao Ning
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Xiang Sui
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Shuyun Liu
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| | - Quanyi Guo
- Guizhou Medical University, Guiyang, Guizhou Province 550004, People's Republic of China
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Institute of Orthopedics, Beijing Key Laboratory ofRegenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Chinese PLA General Hospital, Beijing 100853, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, People's Republic of China
| |
Collapse
|
6
|
Liang W, Yang R, Qin L, Liang T, Chen W. Current Status and Perspectives of Research on Polymer Hydrogels in the Treatment and Protection of Osteoarthritis. Macromol Biosci 2025:e2500016. [PMID: 40271818 DOI: 10.1002/mabi.202500016] [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: 01/07/2025] [Revised: 04/04/2025] [Indexed: 04/25/2025]
Abstract
Arthritis is a degenerative disease characterized by chronic cartilage degeneration. It affects hundreds of millions of people worldwide and often has serious consequences such as joint pain and swelling, limited mobility, and joint deformity. However, conventional treatments still struggle to achieve satisfactory results. Finding more effective treatments for arthritis remains an important clinical challenge. As hydrogels have a unique 3D spatial mesh structure, significant material interaction ability, adjustable mechanical properties, and good biodegradability, they can provide a suitable cellular or tissue microenvironment, and their potential in scaffolding effect, lubrication, anti-inflammatory effect, or drug or cellular delivery is expected to be a potent therapeutic approach for the treatment of osteoarthritis. In this review, three aspects of hydrogel products for osteoarthritis treatment are comprehensively summarized and discussed, namely, material selection and gel design, exploration of cross-linking mechanisms, and mechanisms of hydrogel therapy for osteoarthritis, and focus on the advantages and limitations of their clinical applications, which point out the direction of the development strategy of innovative products in this field, applied research, and clinical transformation.
Collapse
Affiliation(s)
- Wanjun Liang
- School of Pharmaceutical Sciences, Institute of Materia Medica, Shandong Academy of Medical Sciences, State Key Laboratory of Advanced Drug Delivery and Release System, Shandong First Medical University, Jinan, 250117, China
| | - Rui Yang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Lijing Qin
- School of Pharmaceutical Sciences, Institute of Materia Medica, Shandong Academy of Medical Sciences, State Key Laboratory of Advanced Drug Delivery and Release System, Shandong First Medical University, Jinan, 250117, China
| | - Tongjuan Liang
- School of Pharmaceutical Sciences, Institute of Materia Medica, Shandong Academy of Medical Sciences, State Key Laboratory of Advanced Drug Delivery and Release System, Shandong First Medical University, Jinan, 250117, China
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
7
|
Yu L, Wang W, Lv C, Chen Q, Yang P, Qi Z, Yu H, Cao R, Li W, Qin Y, Ge G, Liu P, Zhu L, Sun H, Geng D, Zhang L. Dual functional hydrogel of osteoclastic-inhibition and osteogenic-stimulation for osteoporotic bone defect regeneration. Mater Today Bio 2025; 31:101550. [PMID: 40018058 PMCID: PMC11867540 DOI: 10.1016/j.mtbio.2025.101550] [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: 12/05/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 03/01/2025] Open
Abstract
Osteoporotic bone regeneration poses significant challenges due to the complexity of the condition. Osteoporosis, a degenerative disorder, results from an imbalance in bone homeostasis driven by dysregulation of osteoblast and osteoclast activity. This complicates the treatment of osteoporosis and its related bone injuries in clinical practice. Despite the development of various polymer scaffolds for bone defect repair, achieving effective regeneration in osteoporotic bones-especially when combined with osteoporosis medications-remains difficult. In this study, we designed a drug delivery system composed of mesoporous bioactive glass (MBG) and photo-crosslinked hyaluronic acid methacrylate (HAMA). This system, loaded with the osteogenesis-promoting peptide DWIVA (D5) and the osteoclastogenesis-inhibiting drug alendronate (ALN), is gelled using a light initiator and 405 nm wavelength light. The MBG@D5-Gel complex enables the controlled spatiotemporal release of these agents, markedly enhancing bone regeneration in osteoporotic conditions within ovariectomized rats by inhibiting osteoclastogenesis and bone resorption while promoting osteogenic differentiation and mineralization. This dual-action system synergistically regulates osteoblast and osteoclast activity, optimizing the pathological microenvironment of osteoporosis and facilitating the repair of osteoporotic bone defects. MBG@D5-Gel holds great potential as an effective organic-inorganic hybrid biomimetic implant material for the treatment of osteoporotic bone defects.
Collapse
Affiliation(s)
- Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
- Department of Orthopedics, Qilu Hospital of Shangdong University, Shandong University, Jinan, 250100, Shandong, China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Chang Lv
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Qian Chen
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Peng Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Zhenrong Qi
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Haomiao Yu
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Ruiqi Cao
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| | - Wenhao Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yi Qin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Peilai Liu
- Department of Orthopedics, Qilu Hospital of Shangdong University, Shandong University, Jinan, 250100, Shandong, China
| | - Lixin Zhu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Houyi Sun
- Department of Orthopedics, Qilu Hospital of Shangdong University, Shandong University, Jinan, 250100, Shandong, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Liang Zhang
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Xicheng District, Beijing, 100050, China
| |
Collapse
|
8
|
Su W, Nie Y, Zheng S, Yao Y. Recent Research on Chondrocyte Dedifferentiation and Insights for Regenerative Medicine. Biotechnol Bioeng 2025; 122:749-760. [PMID: 39716991 DOI: 10.1002/bit.28915] [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: 07/26/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Chondrocytes maintain the balance of the extracellular matrix by synthesizing glycoproteins, collagen, proteoglycans and hyaluronic acid. Chondrocyte dedifferentiation refers to a process in which chondrocytes lose their mature differentiated phenotype and transform into a fibroblast-like morphology with fewer differentiated stages and inferior function under external stimulation. The important mechanism of homeostasis loss in osteoarthritis (OA) is a change in the chondrocyte phenotype. The dedifferentiation markers of chondrocytes are upregulated in OA, and the pathogenic factors related to OA have also been shown to enhance chondrocyte dedifferentiation. In this review, we compile recent studies on chondrocyte dedifferentiation, with an emphasis on potential markers and the underlying mechanisms of dedifferentiation, as well as the current research progress in inhibiting dedifferentiation or achieving redifferentiation. A deep understanding of chondrocyte dedifferentiation would not only support the pathogenesis of OA theoretically but also provide insightful ideas for regenerative medicine to manipulate the functional phenotype of cells.
Collapse
Affiliation(s)
- Weixian Su
- Department of Joint Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou Medical University, Guangzhou, China
| | - Yupeng Nie
- Department of Joint Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou Medical University, Guangzhou, China
| | - Shicong Zheng
- Department of Joint Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Chen Z, Zheng X, Mu Z, Lu W, Zhang H, Yan J. Intelligent Nanomaterials Design for Osteoarthritis Managements. SMALL METHODS 2025:e2402263. [PMID: 40159773 DOI: 10.1002/smtd.202402263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/12/2025] [Indexed: 04/02/2025]
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disorder, characterized by progressive joint degradation, pain, and diminished mobility, all of which collectively impair patients' quality of life and escalate healthcare expenditures. Current treatment options are often inadequate due to limited efficacy, adverse side effects, and temporary symptom relief, underscoring the urgent need for more effective therapeutic strategies. Recent advancements in nanomaterials and nanomedicines offer promising solutions by improving drug bioavailability, reducing side effects and providing targeted therapeutic benefits. This review critically examines the pathogenesis of OA, highlights the limitations of existing treatments, and explores the latest innovations in intelligent nanomaterials design for OA therapy, with an emphasis on their engineered properties, therapeutic mechanisms, and translational potential in clinical application. By compiling recent findings, this work aims to inspire further exploration and innovation in nanomedicine, ultimately advancing the development of more effective and personalized OA therapies.
Collapse
Affiliation(s)
- Zhihao Chen
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xuan Zheng
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhengzhi Mu
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun, 130022, China
| | - Weijie Lu
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- Department of Orthopedics, Yanjiang Hospital, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Haiyuan Zhang
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiao Yan
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
10
|
Huang J, Chen H, Luo Z, Nie M, Wang J, Lu L, Zhao Y. Genetically Engineered Stromal Cell Exosomes from High-Throughput Herringbone Microfluidics. ACS NANO 2025; 19:10568-10577. [PMID: 40043164 DOI: 10.1021/acsnano.5c01773] [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: 03/19/2025]
Abstract
Stromal cell-derived exosomes have demonstrated their value in the field of biomedical engineering. However, the low production and specific requirements of different diseases limited the practical efficacy of these exosomes and restricted their wider applications. Here, we presented a method to culture genetically engineered mesenchymal stromal cells (MSC) that overexpressed the hepatocyte growth factor (HGF) in microfluidics and harvest mass HGF overexpressed exosomes for wound healing. The microfluidic chips were featured with herringbone grooves and micropillar arrays, where sufficient fluidic mechanical stimuli and efficient nutrient delivery were promoted by a turbulent vortex. It was demonstrated that the production of exosomes was much higher than by the traditional flask cell culture, along with higher HGF content. In addition, the MSCHGF-secreted exosomes were applied for wound healing in diabetic rat model, showing superior angiogenesis, cell migration, and immune modulation capabilities. These features indicated that the genetically engineered MSCHGF exosomes from high-throughput herringbone microfluidics possess great potential for wound healing and related biomedical applications.
Collapse
Affiliation(s)
- Junjie Huang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Hanxu Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhiqiang Luo
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Nie
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Jinglin Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ling Lu
- Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing 210096, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing 210096, China
| |
Collapse
|
11
|
Yang JJ, He SQ, Huang B, Wang PX, Xu F, Lin X, Liu J. A bibliometric and visualized analysis of extracellular vesicles in degenerative musculoskeletal diseases (from 2006 to 2024). Front Pharmacol 2025; 16:1550208. [PMID: 40183074 PMCID: PMC11966045 DOI: 10.3389/fphar.2025.1550208] [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: 12/23/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Background With the rapid development of extracellular vesicles (EVs) in regenerative medicine research, they have become a promising new direction in the mechanistic, diagnosis and treatment studies of degenerative musculoskeletal diseases (DMDs), and has attracted increasing attention. However, there is currently a lack of comprehensive and objective summary analysis to help researchers quickly and conveniently understand the development trajectory and future trends of this field. Method This study collected articles and reviews published from 2006 to 2024 on EVs in DMDs from the Web of Science database. Bibliometric and visual analysis was conducted using several tools, including Microsoft Excel Office, VOSviewer, CiteSpace, Pajek, and R packages. Results 1,182 publications were included in the analysis from 2006 to 2024. Notably, there was a rapid increase in the number of publications starting in 2016, suggesting that this field remains in a developmental stage. Co-authorship analysis revealed that China ranked first in terms of publications, whereas the United States led in citations. The journal with the highest number of publications was International Journal of Molecular Sciences (INT J MOL SCI). The most prolific authors were Ragni, E with 23 publications, while the most cited author was Toh, WS. Additionally, nine of the top 10 institutions were from China, with Shanghai Jiao Tong University leading in the number of publications. The most cited article was "MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity", authored by Zhang, S, and published in BIOMATERIALS in 2018. Conclusion This study, through bibliometric and visual analysis, clearly illustrates the collaborative relationships among countries, authors, institutions, and journals, providing valuable insights for researchers seeking academic collaboration opportunities. Moreover, the analysis of keywords and citations allows researchers to better understand key research hotspots and frontiers in this field, and points toward promising directions for future research. The growing interest in EV research in DMDs over recent years indicates increasing attention and a dynamic progression in this field.
Collapse
Affiliation(s)
- Jun-Jie Yang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, The Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Sha-Qi He
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Huang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Peng-Xin Wang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Medical Imaging in Hunan Province, Department of Radiology Quality Control Center in Hunan Province, Changsha, China
| |
Collapse
|
12
|
He CF, Qiao TH, Ren XC, Xie M, Gao Q, Xie CQ, Wang P, Sun Y, Yang H, He Y. Printability in Multi-material Projection-Based 3-Dimensional Bioprinting. RESEARCH (WASHINGTON, D.C.) 2025; 8:0613. [PMID: 40041038 PMCID: PMC11876545 DOI: 10.34133/research.0613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/13/2025] [Accepted: 01/24/2025] [Indexed: 03/06/2025]
Abstract
Accurately reconstructing the intricate structure of natural organisms is the long-standing goal of 3-dimensional (3D) bioprinting. Projection-based 3D printing boasts the highest resolution-to-manufacturing time ratio among all 3D-printing technologies, rendering it a highly promising technique in this field. However, achieving standardized, high-fidelity, and high-resolution printing of composite structures using bioinks with diverse mechanical properties remains a marked challenge. The root of this challenge lies in the long-standing neglect of multi-material printability research. Multi-material printing is far from a simple physical assembly of different materials; rather, effective control of material interfaces is a crucial factor that governs print quality. The current research gap in this area substantively hinders the widespread application and rapid development of multi-material projection-based 3D bioprinting. To bridge this critical gap, we developed a multi-material projection-based 3D bioprinter capable of simultaneous printing with 6 materials. Building upon this, we established a fundamental framework for multi-material printability research, encompassing its core logic and essential process specifications. Furthermore, we clarified several critical issues, including the cross-linking behavior of multicomponent bioinks, mechanical mismatch and interface strength in soft-hard composite structures, the penetration behavior of viscous bioinks within hydrogel polymer networks, liquid entrapment and adsorption phenomena in porous heterogeneous structures, and error source analysis along with resolution evaluation in multi-material printing. This study offers a solid theoretical foundation and guidance for the quantitative assessment of multi-material projection-based 3D bioprinting, holding promise to advance the field toward higher precision and the reconstruction of more intricate biological structures.
Collapse
Affiliation(s)
- Chao-fan He
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu Laboratory, School of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
| | - Tian-hong Qiao
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu Laboratory, School of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
| | - Xu-chao Ren
- School of Computer Science,
Xi’an Shiyou University, Xi’an 710065, China
| | - Mingjun Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu Laboratory, School of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
| | - Qing Gao
- EFL-Tech,
Suzhou Yongqinquan Intelligent Equipment Co., Ltd, Suzhou 215101, China
| | - Chao-qi Xie
- EFL-Tech,
Suzhou Yongqinquan Intelligent Equipment Co., Ltd, Suzhou 215101, China
| | - Peng Wang
- EFL-Tech,
Suzhou Yongqinquan Intelligent Equipment Co., Ltd, Suzhou 215101, China
| | - Yuan Sun
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu Laboratory, School of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu Laboratory, School of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering,
Zhejiang University, Hangzhou 310027, China
- The Second Affiliated Hospital of Zhejiang University,
Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
13
|
Chen X, Tian B, Wang Y, Zheng J, Kang X. Potential and challenges of utilizing exosomes in osteoarthritis therapy (Review). Int J Mol Med 2025; 55:43. [PMID: 39791222 PMCID: PMC11759586 DOI: 10.3892/ijmm.2025.5484] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
Exosomes are integral to the pathophysiology of osteoarthritis (OA) due to their roles in mediating intercellular communication and regulating inflammatory processes. Exosomes are integral to the transport of bioactive molecules, such as proteins, lipids and nucleic acids, which can influence chondrocyte behavior and joint homeostasis. Given their properties of regeneration and ability to target damaged tissues, exosomes represent a promising therapeutic avenue for OA treatment. Exosomes have potential in promoting cartilage repair, reducing inflammation and improving overall joint function. However, several challenges remain, including the need for standardized isolation and characterization methods, variability in exosomal content, and regulatory hurdles. The present review aims to provide a comprehensive overview of the current understanding of exosome mechanisms in OA and their therapeutic potential, while also addressing the ongoing challenges faced in translating these findings into clinical practice. By consolidating existing research, the present review aims to pave the way for future studies aimed at optimizing exosome‑based therapies for effective OA management.
Collapse
Affiliation(s)
| | | | | | - Jiang Zheng
- Department of Joint Surgery, Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| | - Xin Kang
- Department of Joint Surgery, Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| |
Collapse
|
14
|
Chen M, Liu Y, Cao Y, Zhao C, Liu Q, Li N, Liu Y, Cui X, Liu P, Liang J, Fan Y, Wang Q, Zhang X. Remodeling the Proinflammatory Microenvironment in Osteoarthritis through Interleukin-1 Beta Tailored Exosome Cargo for Inflammatory Regulation and Cartilage Regeneration. ACS NANO 2025; 19:4924-4941. [PMID: 39848926 DOI: 10.1021/acsnano.4c16785] [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: 01/25/2025]
Abstract
Osteoarthritis (OA) presents a significant therapeutic challenge, with few options for preserving joint cartilage and repairing associated tissue damage. Inflammation is a pivotal factor in OA-induced cartilage deterioration and synovial inflammation. Recently, exosomes derived from human umbilical cord mesenchymal stem cells (HucMSCs) have gained recognition as a promising noncellular therapeutic modality, but their use is hindered by the challenge of harvesting a sufficient number of exosomes with effective therapeutic efficacy. Given that HucMSCs are highly sensitive to microenvironmental signals, we hypothesized that priming HucMSCs within a proinflammatory environment would increase the number of exosomes secreted with enhanced anti-inflammatory properties. Subsequent miRNA profiling and pathway analysis confirmed that interleukin-1 beta (IL-1β)-induced exosomes (C-Exos) exert positive effects through miRNA regulation and signaling pathway modulation. In vitro experiments revealed that C-Exos enhance chondrocyte functionality and cartilage matrix production, as well as macrophage polarization, thereby enhancing cartilage repair. C-Exos were encapsulated in hyaluronic acid hydrogel microspheres (HMs) to ensure sustained release, leading to substantial improvements in the inflammatory microenvironment and cartilage regeneration in a rat OA model. This study outlines a strategy to tailor exosome cargo for anti-inflammatory and cartilage regenerative purposes, with the functionalized HMs demonstrating potential for OA treatment.
Collapse
Affiliation(s)
- Manyu Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Yuhan Liu
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Yi Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Chengkun Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Quanying Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Na Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29 Wangjiang Road, Chengdu 610000, China
| | - Yuan Liu
- Orthopedics Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Xiaolin Cui
- Cardiac and Osteochondral Tissue Engineering (COTE) Group, School of medicine, the Chinese University of Hong Kong, Shenzhen 518172, China
- Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch 8011, New Zealand
| | - Pengcheng Liu
- Chengdu Xiangyakanglin Biotechnology Co., Ltd, Chengdu 610213, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29 Wangjiang Road, Chengdu 610000, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| |
Collapse
|
15
|
Liu L, Liu W, Han Z, Shan Y, Xie Y, Wang J, Qi H, Xu Q. Extracellular Vesicles-in-Hydrogel (EViH) targeting pathophysiology for tissue repair. Bioact Mater 2025; 44:283-318. [PMID: 39507371 PMCID: PMC11539077 DOI: 10.1016/j.bioactmat.2024.10.017] [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: 04/06/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
Regenerative medicine endeavors to restore damaged tissues and organs utilizing biological approaches. Utilizing biomaterials to target and regulate the pathophysiological processes of injured tissues stands as a crucial method in propelling this field forward. The Extracellular Vesicles-in-Hydrogel (EViH) system amalgamates the advantages of extracellular vesicles (EVs) and hydrogels, rendering it a prominent biomaterial in regenerative medicine with substantial potential for clinical translation. This review elucidates the development and benefits of the EViH system in tissue regeneration, emphasizing the interaction and impact of EVs and hydrogels. Furthermore, it succinctly outlines the pathophysiological characteristics of various types of tissue injuries such as wounds, bone and cartilage injuries, cardiovascular diseases, nerve injuries, as well as liver and kidney injuries, underscoring how EViH systems target these processes to address related tissue damage. Lastly, it explores the challenges and prospects in further advancing EViH-based tissue regeneration, aiming to impart a comprehensive understanding of EViH. The objective is to furnish a thorough overview of EViH in enhancing regenerative medicine applications and to inspire researchers to devise innovative tissue engineering materials for regenerative medicine.
Collapse
Affiliation(s)
- Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266003, China
| | - Zeyu Han
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yansheng Shan
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Jialu Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Quanchen Xu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| |
Collapse
|
16
|
Kong K, Li B, Chang Y, Zhao C, Qiao H, Jin M, Wu X, Fan W, Wang L, Qi Y, Xu Y, Zhai Z, Ma P, Li H. Delivery of FGF18 using mRNA-LNP protects the cartilage against degeneration via alleviating chondrocyte senescence. J Nanobiotechnology 2025; 23:34. [PMID: 39844298 PMCID: PMC11753171 DOI: 10.1186/s12951-025-03103-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease with an immense unmet medical need. FGF18 protein is a potential regenerative factor for cartilage repair. However, traditional protein delivery methods have limited efficacy due to the short lifetime and shallow infiltration. RESULTS In this work, we discovered that lipid nanoparticle (LNP) can infiltrate and deliver FGF18 mRNA deeper in the cartilage than proteins. After mRNA UTR optimization and chemical modification, the expression of FGF18 can last up to 6 days in the cartilage. Furthermore, delivering FGF18 mRNA activates FOXO3a-autophagy pathway, which protects against chondrocyte degeneration and senescence. Local intra-articular injection of FGF18 mRNA-LNP significantly alleviates OA symptoms in DMM and senile OA models. Sustained expression and accessibility of FGF18-mRNA to deeper chondrocytes makes LNP-mRNA more effective than FGF18 recombinant protein. CONCLUSIONS In summary, this study presents a novel approach superior to recombinant protein alone and holds promise as a new therapeutic strategy for OA.
Collapse
Affiliation(s)
- Keyu Kong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Baixing Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Yongyun Chang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Chen Zhao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Hua Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Minghao Jin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Xinru Wu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Wenxuan Fan
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Liao Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China
| | - Yansong Qi
- Orthopedic Center (Sports Medicine Center), Inner Mongolia People's Hospital, Hohhot, 010017, P.R. China
| | - Yongsheng Xu
- Orthopedic Center (Sports Medicine Center), Inner Mongolia People's Hospital, Hohhot, 010017, P.R. China.
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China.
| | - Peixiang Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China.
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, P.R. China.
| |
Collapse
|
17
|
Zhou X, Chen S, Pich A, He C. Advanced Bioresponsive Drug Delivery Systems for Promoting Diabetic Vascularized Bone Regeneration. ACS Biomater Sci Eng 2025; 11:182-207. [PMID: 39666445 DOI: 10.1021/acsbiomaterials.4c02037] [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: 12/14/2024]
Abstract
The treatment of bone defects in diabetes mellitus (DM) patients remains a major challenge since the diabetic microenvironments significantly impede bone regeneration. Many abnormal factors including hyperglycemia, elevated oxidative stress, increased inflammation, imbalanced osteoimmune, and impaired vascular system in the diabetic microenvironment will result in a high rate of impaired, delayed, or even nonhealing events of bone tissue. Stimuli-responsive biomaterials that can respond to endogenous biochemical signals have emerged as effective therapeutic systems to treat diabetic bone defects via the combination of microenvironmental regulation and enhanced osteogenic capacity. Following the natural bone healing processes, coupling of angiogenesis and osteogenesis by advanced bioresponsive drug delivery systems has proved to be of significant approach for promoting bone repair in DM. In this Review, we have systematically summarized the mechanisms and therapeutic strategies of DM-induced impaired bone healing, outlined the bioresponsive design for drug delivery systems, and highlighted the vascularization strategies for promoting bone regeneration. Accordingly, we then overview the recent advances in developing bioresponsive drug delivery systems to facilitate diabetic vascularized bone regeneration by remodeling the microenvironment and modulating multiple regenerative cues. Furthermore, we discuss the development of adaptable drug delivery systems with unique features for guiding DM-associated bone regeneration in the future.
Collapse
Affiliation(s)
- Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- Institute for Technical and Macromolecular Chemistry, Functional and Interactive Polymers, RWTH Aachen University, Aachen 52074, Germany
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen 52074, Germany
| | - Shuo Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Andrij Pich
- Institute for Technical and Macromolecular Chemistry, Functional and Interactive Polymers, RWTH Aachen University, Aachen 52074, Germany
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University, Aachen 52074, Germany
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
18
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
19
|
Wang P, Zhao H, Chen W, Guo Y, Zhang S, Xing X, Yang S, Wang F, Wang J, Shao Z, Zhang Y. Cell-free osteoarthritis treatment with dual-engineered chondrocyte-targeted extracellular vesicles derived from mechanical loading primed mesenchymal stem cells. J Tissue Eng 2025; 16:20417314241312563. [PMID: 39926048 PMCID: PMC11806476 DOI: 10.1177/20417314241312563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/23/2024] [Indexed: 02/11/2025] Open
Abstract
Osteoarthritis (OA) is an age-related chronic inflammatory disease, predominantly characterized by chondrocyte senescence and extracellular matrix (ECM) degradation. Although mesenchymal stem cells (MSCs) derived extracellular vesicles (EVs) are promising for promoting cartilage regeneration, their clinical application is limited by inconsistent therapeutic effects and insufficient targeting capabilities. Mechanical loading shows potential to optimize MSC-EVs for OA treatment, while the underlying mechanism is not clear. In this study, EVs derived from mechanical loading-primed MSCs (ML-EVs) demonstrate prominent efficacy in maintaining ECM homeostasis and relieving chondrocyte senescence, thereby mitigating OA. Subsequent miRNA sequencing reveals that ML-EVs exert their effects by delivering miR-27b-3p, which targets ROR1 mRNA in chondrocytes and suppresses downstream NF-κB pathways. By modulating the ROR1/NF-κB axis, miR-27b-3p effectively restrains ECM degradation and chondrocyte senescence. To optimize therapeutic efficacy of EVs, miR-27b-3p is overexpressed within EVs (miROE-EVs), and a chondrocyte-targeted peptide (CTP) is conjugated to their surface, thereby constructing dual-engineered chondrocyte-targeted EVs (CTP/miROE-EVs). CTP/miROE-EVs exhibit excellent ability to specifically target cartilage and ameliorate OA pathology. In conclusion, this study underscores the critical role of mechanical loading in augmenting effectiveness of EVs in mitigating OA and introduces dual-engineered EVs that specifically target chondrocytes, providing a promising therapeutic strategy for OA.
Collapse
Affiliation(s)
- Peng Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haiyue Zhao
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- School of Medicine, Nankai University, Tianjin, China
| | - Wei Chen
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuhui Guo
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuo Zhang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xin Xing
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuai Yang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- School of Medicine, Nankai University, Tianjin, China
| | - Fengkun Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- School of Medicine, Nankai University, Tianjin, China
| | - Juan Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingze Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Zhang H, Yuan S, Zheng B, Wu P, He X, Zhao Y, Zhong Z, Zhang X, Guan J, Wang H, Yang L, Zheng X. Lubricating and Dual-Responsive Injectable Hydrogels Formulated From ZIF-8 Facilitate Osteoarthritis Treatment by Remodeling the Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407885. [PMID: 39604796 DOI: 10.1002/smll.202407885] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/29/2024] [Indexed: 11/29/2024]
Abstract
Osteoarthritis (OA) is a progressively developing condition primarily characterized by the deterioration of articular cartilage and the proliferation of bone, along with ongoing inflammation. Although the precise pathogenesis remains somewhat elusive, restoring the homeostatic balance of the intra-articular microenvironment is crucial for the management of OA. Intra-articular injection of medication is one of the most direct and effective treatment methods; however, most injectable drugs used for osteoarthritis treatment, due to their rapid breakdown, quick release, poor biological activity, and frequent injections, leading to increased risk of infection and suboptimal therapeutic outcomes. In this study, a lubricating and dual-responsive injectable hydrogel based on zeolitic imidazolate frameworks-8 (ZIF-8) impregnated with Quercetin (Que) is designed, which can facilitate OA treatment by remodeling the microenvironment. The prepared injectable nanocomposite hydrogel (MH/CCM@ZIF-8@Que) exhibits pH and reactive oxygen species (ROS) responsiveness, alongside a controllable release of bioactive substances to modulate the microenvironment of bone tissue, thereby mitigating synovitis and the degeneration of cartilage matrix, while simultaneously facilitating cartilage repair. This developed thermosensitive injectable hydrogel, which effectively balances lubrication with the controlled release of bioactive substances, represents a highly promising therapeutic approach for osteoarthritis.
Collapse
Affiliation(s)
- Hongtao Zhang
- Department of Orthopedics, Zhongshan Torch Development Zone People's Hospital, Zhongshan, 528437, P. R. China
| | - Shiguo Yuan
- Department of Orthopaedic, Hainan Hospital, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, 570203, P. R. China
- Department of Orthopaedic, Hainan Traditional Chinese Medicine Hospital, Hainan Medical University, Haikou, 570203, P. R. China
| | - Boyuan Zheng
- Department of Sports Medicine, The First Affiliated Hospital, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Guangdong Provincial Key Laboratory of Speed Capability, Jinan University, Guangzhou, 510630, P. R. China
| | - Peng Wu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Xiuming He
- Department of Orthopedics, Zhongshan Torch Development Zone People's Hospital, Zhongshan, 528437, P. R. China
| | - Yi Zhao
- Department of Orthopedics, the Third Hospital of Shijiazhuang, Shijiazhuang, 050011, P. R. China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, P. R. China
| | - Xiaofang Zhang
- Department of Pharmacy, the First Affiliated Hospital, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Science and Technology Planning Project of Guangzhou, Jinan University, Guangzhou, 510630, P. R. China
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, P. R. China
| | - Jian Guan
- Department of Orthopedics, the Third Hospital of Shijiazhuang, Shijiazhuang, 050011, P. R. China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Guangdong Provincial Key Laboratory of Speed Capability, Jinan University, Guangzhou, 510630, P. R. China
| | - Lei Yang
- Department of Orthopedics, Key Laboratory of Hepatosplenic Surgery of Ministry of Education, NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 163711, P. R. China
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Guangdong Provincial Key Laboratory of Speed Capability, Jinan University, Guangzhou, 510630, P. R. China
| |
Collapse
|
21
|
Edirisinghe O, Ternier G, Alraawi Z, Suresh Kumar TK. Decoding FGF/FGFR Signaling: Insights into Biological Functions and Disease Relevance. Biomolecules 2024; 14:1622. [PMID: 39766329 PMCID: PMC11726770 DOI: 10.3390/biom14121622] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Fibroblast Growth Factors (FGFs) and their cognate receptors, FGFRs, play pivotal roles in a plethora of biological processes, including cell proliferation, differentiation, tissue repair, and metabolic homeostasis. This review provides a comprehensive overview of FGF-FGFR signaling pathways while highlighting their complex regulatory mechanisms and interconnections with other signaling networks. Further, we briefly discuss the FGFs involvement in developmental, metabolic, and housekeeping functions. By complementing current knowledge and emerging research, this review aims to enhance the understanding of FGF-FGFR-mediated signaling and its implications for health and disease, which will be crucial for therapeutic development against FGF-related pathological conditions.
Collapse
Affiliation(s)
- Oshadi Edirisinghe
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Gaëtane Ternier
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| | - Zeina Alraawi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| | - Thallapuranam Krishnaswamy Suresh Kumar
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA;
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| |
Collapse
|
22
|
Bader J, Brigger F, Leroux JC. Extracellular vesicles versus lipid nanoparticles for the delivery of nucleic acids. Adv Drug Deliv Rev 2024; 215:115461. [PMID: 39490384 DOI: 10.1016/j.addr.2024.115461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Extracellular vesicles (EVs) are increasingly investigated for delivering nucleic acid (NA) therapeutics, leveraging their natural role in transporting NA and protein-based cargo in cell-to-cell signaling. Their synthetic counterparts, lipid nanoparticles (LNPs), have been developed over the past decades as NA carriers, culminating in the approval of several marketed formulations such as patisiran/Onpattro® and the mRNA-1273/BNT162 COVID-19 vaccines. The success of LNPs has sparked efforts to develop innovative technologies to target extrahepatic organs, and to deliver novel therapeutic modalities, such as tools for in vivo gene editing. Fueled by the recent advancements in both fields, this review aims to provide a comprehensive overview of the basic characteristics of EV and LNP-based NA delivery systems, from EV biogenesis to structural properties of LNPs. It addresses the primary challenges encountered in utilizing these nanocarriers from a drug formulation and delivery perspective. Additionally, biodistribution profiles, in vitro and in vivo transfection outcomes, as well as their status in clinical trials are compared. Overall, this review provides insights into promising research avenues and potential dead ends for EV and LNP-based NA delivery systems.
Collapse
Affiliation(s)
- Johannes Bader
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Finn Brigger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
23
|
Hwang HS, Lee CS. Exosome-Integrated Hydrogels for Bone Tissue Engineering. Gels 2024; 10:762. [PMID: 39727520 DOI: 10.3390/gels10120762] [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: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Exosome-integrated hydrogels represent a promising frontier in bone tissue engineering, leveraging the unique biological properties of exosomes to enhance the regenerative capabilities of hydrogels. Exosomes, as naturally occurring extracellular vesicles, carry a diverse array of bioactive molecules that play critical roles in intercellular communication and tissue regeneration. When combined with hydrogels, these exosomes can be spatiotemporally delivered to target sites, offering a controlled and sustained release of therapeutic agents. This review aims to provide a comprehensive overview of the recent advancements in the development, engineering, and application of exosome-integrated hydrogels for bone tissue engineering, highlighting their potential to overcome current challenges in tissue regeneration. Furthermore, the review explores the mechanistic pathways by which exosomes embedded within hydrogels facilitate bone repair, encompassing the regulation of inflammatory pathways, enhancement of angiogenic processes, and induction of osteogenic differentiation. Finally, the review addresses the existing challenges, such as scalability, reproducibility, and regulatory considerations, while also suggesting future directions for research in this rapidly evolving field. Thus, we hope this review contributes to advancing the development of next-generation biomaterials that synergistically integrate exosome and hydrogel technologies, thereby enhancing the efficacy of bone tissue regeneration.
Collapse
Affiliation(s)
- Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
24
|
Sun M, Ma B, Pan Z, Zhao Y, Tian L, Fan Y, Kong W, Wang J, Xu B, Ao Y, Guo Q, Wang X, Peng X, Li X, Cheng J, Miao L, Wang K, Hu X. Targeted Therapy of Osteoarthritis via Intra-Articular Delivery of Lipid-Nanoparticle-Encapsulated Recombinant Human FGF18 mRNA. Adv Healthc Mater 2024; 13:e2400804. [PMID: 39363784 PMCID: PMC11582510 DOI: 10.1002/adhm.202400804] [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: 03/01/2024] [Revised: 07/08/2024] [Indexed: 10/05/2024]
Abstract
Fibroblast growth factor 18 (FGF18) emerges as a promising therapeutic target for osteoarthritis (OA). In this study, a novel articular cavity-localized lipid nanoparticle (LNP) named WG-PL14 is developed. This optimized formulation has a nearly 30-fold increase in mRNA expression as well as better articular cavity enrichment compared to commercial lipids MC3 when performing intra-articular injection. Then, a mRNA sequence encoding recombinant human FGF18 (rhFGF18) for potential mRNA therapy in OA is optimized. In vitro assays confirm the translation of rhFGF18 mRNA into functional proteins within rat and human chondrocytes, promoting cell proliferation and extracellular matrix (ECM) synthesis. Subsequently, the therapeutic efficacy of the LNP-rhFGF18 mRNA complex is systematically assessed in a mouse OA model. The administration exhibits several positive outcomes, including an improved pain response, upregulation of ECM-related genes (e.g., AGRN and HAS2), and remodeling of subchondral bone homeostasis compared to a control group. Taken together, these findings underscore the potential of localized LNP-rhFGF18 mRNA therapy in promoting the regeneration of cartilage tissue and mitigating the progression of OA.
Collapse
Affiliation(s)
- Mengze Sun
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Bin Ma
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Zihang Pan
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Yun Zhao
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Liangliang Tian
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Yifei Fan
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Weijing Kong
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Junyan Wang
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Boyang Xu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Yingfang Ao
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Quanyi Guo
- Institute of OrthopedicsThe Fourth Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLANo. 28 Fuxing Road, Haidian DistrictBeijing100853China
| | - Xi Wang
- State Key Laboratory of Female Fertility PromotionClinical Stem Cell Research CenterPeking University Third HospitalBeijing100191China
| | - Xiaohong Peng
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Xiaoxia Li
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Jin Cheng
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Lei Miao
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Kai Wang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Xiaoqing Hu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| |
Collapse
|
25
|
Selvadoss A, Baby HM, Zhang H, Bajpayee AG. Harnessing exosomes for advanced osteoarthritis therapy. NANOSCALE 2024; 16:19174-19191. [PMID: 39323205 PMCID: PMC11799831 DOI: 10.1039/d4nr02792b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
Exosomes are nanosized, lipid membrane vesicles secreted by cells, facilitating intercellular communication by transferring cargo from parent to recipient cells. This capability enables biological crosstalk across multiple tissues and cells. Extensive research has been conducted on their role in the pathogenesis of degenerative musculoskeletal diseases such as osteoarthritis (OA), a chronic and painful joint disease that particularly affects cartilage. Currently, no effective treatment exists for OA. Given that exosomes naturally modulate synovial joint inflammation and facilitate cartilage matrix synthesis, they are promising candidates as next generation nanocarriers for OA therapy. Recent advancements have focused on engineering exosomes through endogenous and exogenous approaches to enhance their joint retention, cartilage and chondrocyte targeting properties, and therapeutic content enrichment, further increasing their potential for OA drug delivery. Notably, charge-reversed exosomes that utilize electrostatic binding interactions with cartilage anionic aggrecan glycosaminoglycans have demonstrated the ability to penetrate the full thickness of early-stage arthritic cartilage tissue following intra-articular administration, maximizing their therapeutic potential. These exosomes offer a non-viral, naturally derived, cell-free carrier for OA drug and gene delivery applications. Efforts to standardize exosome harvest, engineering, and property characterization methods, along with scaling up production, will facilitate more efficient and rapid clinical translation. This article reviews the current state-of-the-art, explores opportunities for exosomes as OA therapeutics, and identifies potential challenges in their clinical translation.
Collapse
Affiliation(s)
- Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Ambika G Bajpayee
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
26
|
Zhao H, Wang T, Fang X, Xu T, Li J, Jing S, Chen G, Liu Y, Sheng G. 2D MXene Nanosheets with ROS Scavenging Ability Effectively Delay Osteoarthritis Progression. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1572. [PMID: 39404298 PMCID: PMC11478061 DOI: 10.3390/nano14191572] [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: 07/15/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
MXenes nanosheets with high conductivity, hydrophilicity, and excellent reactive oxygen species (ROS) scavenging ability have shown promise in treating various degenerative diseases correlated with abnormal ROS accumulation. Herein, the therapeutic potential of Ti3C2Tx nanosheets, which is the most widely investigated MXene material, in delaying osteoarthritis (OA) progression is demonstrated. In vitro experiments indicate the strong ROS scavenging capacity of Ti3C2Tx nanosheets and their acceptable biocompatibility. Ti3C2Tx nanosheets effectively protect chondrocytes from cell death induced by oxidative stress. In addition, Ti3C2Tx nanosheets demonstrate a prominent anti-inflammatory effect and the ability to restore homeostasis between anabolic activities and catabolic activities in chondrocytes. Furthermore, RNA sequencing reveals the potential mechanism underlying the Ti3C2Tx nanosheet-mediated therapeutic effect. Finally, the in vivo curative effect of Ti3C2Tx nanosheets is verified using a rat OA model. Histological staining and immunohistochemical analyses indicate that Ti3C2Tx nanosheets effectively ameliorate OA progression. Conclusively, the in vitro and in vivo experiments suggest that Ti3C2Tx nanosheets could be a promising and effective option for OA treatment.
Collapse
Affiliation(s)
- Hongqi Zhao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianqi Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xuan Fang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Shaoze Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Guangzi Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
27
|
Chu Y, Zhao Z, Schreiber S, Zeng H, Chen L. Size-Controllable and pH-Sensitive Whey Protein Microgels as High-Performance Aqueous Biolubricants. ACS APPLIED MATERIALS & INTERFACES 2024; 16:46909-46922. [PMID: 39172030 DOI: 10.1021/acsami.4c09771] [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: 08/23/2024]
Abstract
Developing efficient aqueous biolubricants has become a significant focus of research due to their prevalence in biotribological contacts and enormous potential in soft matter applications. In this study, size-controllable, pH-sensitive whey protein microgels were prepared using a water-in-water emulsion template method from protein-polysaccharide phase separation. The granular hydrogel from the protein microgels exhibited superior lubricity, obtaining 2.7-fold and 1.7-fold reductions in coefficient of friction (μ) compared to native protein and human saliva (μ = 0.30 compared to 0.81 and 0.52, respectively). The microgels also exhibited outstanding load-bearing capabilities, sustaining lubrication under normal forces up to 5 N. Microgels with a smaller size (1 μm) demonstrated better lubricating performance than 6 and 20 μm microgels. The exceptional lubricity was from a synergistic effect of the ball-bearing mechanism and the hydration state of the microgels. Particularly at pH 7.4, the hydration layer surrounding highly negative charges contributed to the electrostatic repulsion among the swollen microgels, leading to an improved buffer ability to separate contact surfaces and effective rolling behavior. Such pH-dependent repulsion was evidenced using a surface forces apparatus that the adhesion between the whey protein-coated surfaces and protein-mica surfaces decreased from 4.49 to 0.97 mN/m and from 7.89 to 0.36 mN/m, respectively, with pH increasing from the isoelectronic point to 7.4. Our findings fundamentally elucidated the tribo-rheological properties and lubrication mechanisms of the whey protein microgels with excellent biocompatibility and environmental responsiveness, offering novel insights for their food and biomedical applications requiring aqueous biolubrication.
Collapse
Affiliation(s)
- Yifu Chu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada
| | - Ziqian Zhao
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Canada
| | - Sabina Schreiber
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada
| | - Hongbo Zeng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton T6G 1H9, Canada
| | - Lingyun Chen
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada
| |
Collapse
|
28
|
Lai C, Cheng X, Yuan T, Fang P, Qian H, Jiang H, Meng J, Zhao J, Bao N, Zhang L. A novel mechanism behind irreversible development of cartilage degradation driven articular cartilage defects revealed by rat model: The chain reaction initiated by extracellular vesicles delivered LOC102546541. Int Immunopharmacol 2024; 137:112467. [PMID: 38875997 DOI: 10.1016/j.intimp.2024.112467] [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: 04/04/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Articular cartilage defects (ACD) are injuries with a diameter greater than 3 mm, resulting from wear and tear on joints. When the diameter of the defect exceeds 6 mm, it can further damage the surrounding joint cartilage, causing osteoarthritis (OA). Try to explain why OA is an irreversible disease, we hypothesize that damaged articular chondrocytes (DAC) may have reduced capacities to repair cartilage because its extracellular vesicle (EVs) that might directly contribute to OA formation. METHODS In this study, DAC-EVs and AC-EVs were isolated using ultracentrifugation. Next-generation sequencing was employed to screen for a pathogenic long non-coding RNA (lncRNA). After verifying its function in vitro, the corresponding small interfering RNA (siRNA) was constructed and loaded into extracellular vesicles, which were then injected into the knee joint cavities of rats. RESULTS The results revealed that DAC-EVs packaged lncRNA LOC102546541 acts as a competitive endogenous RNA (ceRNA) of MMP13, down-regulating miR-632. Consequently, the function of MMP13 in degrading the extracellular matrix is enhanced, promoting the development of osteoarthritis. CONCLUSIONS This study uncovered a novel mode of OA pathogenesis using rat models, which DAC deliver pathogenic LOC102546541 packaged EVs to normal articular chondrocytes, amplifying the degradation of the extracellular matrix. Nonetheless, the functions of highly homologous human gene of LOC102546541 need to be verified in the future.
Collapse
Affiliation(s)
- Chengteng Lai
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xi Cheng
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Yuan
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Peng Fang
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hong Qian
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hui Jiang
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jia Meng
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jianning Zhao
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Nirong Bao
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Lei Zhang
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
29
|
Hou J, Lin Y, Zhu C, Chen Y, Lin R, Lin H, Liu D, Guan D, Yu B, Wang J, Wu H, Cui Z. Zwitterion-Lubricated Hydrogel Microspheres Encapsulated with Metformin Ameliorate Age-Associated Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402477. [PMID: 38874373 PMCID: PMC11321630 DOI: 10.1002/advs.202402477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/17/2024] [Indexed: 06/15/2024]
Abstract
Chondrocyte senescence and reduced lubrication play pivotal roles in the pathogenesis of age-related osteoarthritis (OA). In the present study, highly lubricated and drug-loaded hydrogel microspheres are designed and fabricated through the radical polymerization of sulfobetaine (SB)-modified hyaluronic acid methacrylate using microfluidic technology. The copolymer contains a large number of SB and carboxyl groups that can provide a high degree of lubrication through hydration and form electrostatic loading interactions with metformin (Met@SBHA), producing a high drug load for anti-chondrocyte senescence. Mechanical, tribological, and drug release analyses demonstrated enhanced lubricative properties and prolonged drug dissemination of the Met@SBHA microspheres. RNA sequencing (RNA-seq) analysis, network pharmacology, and in vitro assays revealed the extraordinary capacity of Met@SBHA to combat chondrocyte senescence. Additionally, inducible nitric oxide synthase (iNOS) has been identified as a promising protein modulated by Met in senescent chondrocytes, thereby exerting a significant influence on the iNOS/ONOO-/P53 pathway. Notably, the intra-articular administration of Met@SBHA in aged mice ameliorated cartilage senescence and OA pathogenesis. Based on the findings of this study, Met@SBHA emerges as an innovative and promising strategy in tackling age-related OA serving the dual function of enhancing joint lubrication and mitigating cartilage senescence.
Collapse
Affiliation(s)
- Jiahui Hou
- Devision of Orthopaedics and TraumatologyDepartment of OrthopaedicsNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration MedicineNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yanpeng Lin
- Department of RadiologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Chencheng Zhu
- Devision of Orthopaedics and TraumatologyDepartment of OrthopaedicsNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration MedicineNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yupeng Chen
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Rongmin Lin
- Devision of Orthopaedics and TraumatologyDepartment of OrthopaedicsNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration MedicineNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Hancheng Lin
- Devision of Orthopaedics and TraumatologyDepartment of OrthopaedicsNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration MedicineNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Dahai Liu
- School of MedicineFoshan UniversityFoshanGuangdong528000China
| | - Daogang Guan
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Bin Yu
- Devision of Orthopaedics and TraumatologyDepartment of OrthopaedicsNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration MedicineNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jun Wang
- School of MedicineFoshan UniversityFoshanGuangdong528000China
| | - Hangtian Wu
- Devision of Orthopaedics and TraumatologyDepartment of OrthopaedicsNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration MedicineNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhuang Cui
- Devision of Orthopaedics and TraumatologyDepartment of OrthopaedicsNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration MedicineNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| |
Collapse
|
30
|
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
|
31
|
Lin J, Jia S, Cao F, Huang J, Chen J, Wang J, Liu P, Zeng H, Zhang X, Cui W. Research Progress on Injectable Microspheres as New Strategies for the Treatment of Osteoarthritis Through Promotion of Cartilage Repair. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202400585] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Indexed: 07/07/2024]
Abstract
AbstractOsteoarthritis (OA) is a degenerative disease caused by a variety of factors with joint pain as the main symptom, including fibrosis, chapping, ulcers, and loss of cartilage. Traditional treatment can only delay the progression of OA, and classical delivery system have many side effects. In recent years, microspheres have shown great application prospects in the field of OA treatment. Microspheres can support cells, reproduce the natural tissue microenvironment in vitro and in vivo, and are an efficient delivery system for the release of drugs or biological agents, which can promote cell proliferation, migration, and differentiation. Thus, they have been widely used in cartilage repair and regeneration. In this review, preparation processes, basic materials, and functional characteristics of various microspheres commonly used in OA treatment are systematically reviewed. Then it is introduced surface modification strategies that can improve the biological properties of microspheres and discussed a series of applications of microsphere functionalized scaffolds in OA treatment. Finally, based on bibliometrics research, the research development, future potential, and possible research hotspots of microspheres in the field of OA therapy is systematically and dynamically evaluated. The comprehensive and systematic review will bring new understanding to the field of microsphere treatment of OA.
Collapse
Affiliation(s)
- Jianjing Lin
- Department of Sports Medicine and Rehabilitation Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
| | - Shicheng Jia
- Department of Sports Medicine and Rehabilitation Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
- Shantou University Medical College Shantou Guangdong 515041 P. R. China
| | - Fuyang Cao
- Department of Orthopedics Second Hospital of Shanxi Medical University Taiyuan Shanxi 030001 P. R. China
| | - Jingtao Huang
- Shantou University Medical College Shantou Guangdong 515041 P. R. China
| | - Jiayou Chen
- Department of Sports Medicine and Rehabilitation Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
- Shantou University Medical College Shantou Guangdong 515041 P. R. China
| | - Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200025 P. R. China
| | - Peng Liu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
| | - Hui Zeng
- Shenzhen Second People's Hospital (First Affiliated Hospital of Shenzhen University) Shenzhen Guangdong 518035 China
| | - Xintao Zhang
- Department of Sports Medicine and Rehabilitation Peking University Shenzhen Hospital Shenzhen Guangdong 518036 P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200025 P. R. China
| |
Collapse
|
32
|
Su Z, Yang T, Wu X, Liu P, Nuermaimaiti Y, Ran Y, Wang P, Cao P. Comparative Analysis and Regeneration Strategies for Three Types of Cartilage. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 38970440 DOI: 10.1089/ten.teb.2024.0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Cartilage tissue, encompassing hyaline cartilage, fibrocartilage, and elastic cartilage, plays a pivotal role in the human body because of its unique composition, structure, and biomechanical properties. However, the inherent avascularity and limited regenerative capacity of cartilage present significant challenges to its healing following injury. This review provides a comprehensive analysis of the current state of cartilage tissue engineering, focusing on the critical components of cell sources, scaffolds, and growth factors tailored to the regeneration of each cartilage type. We explore the similarities and differences in the composition, structure, and biomechanical properties of the three cartilage types and their implications for tissue engineering. A significant emphasis is placed on innovative strategies for cartilage regeneration, including the potential for in situ transformation of cartilage types through microenvironmental manipulation, which may offer novel avenues for repair and rehabilitation. The review underscores the necessity of a nuanced approach to cartilage tissue engineering, recognizing the distinct requirements of each cartilage type while exploring the potential of transforming one cartilage type into another as a flexible and adaptive repair strategy. Through this detailed examination, we aim to broaden the understanding of cartilage tissue engineering and inspire further research and development in this promising field.
Collapse
Affiliation(s)
- Zhan Su
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinze Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiran Liu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yisimayili Nuermaimaiti
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuxuan Ran
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Pinyin Cao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Yang L, Li W, Zhao Y, Shang L. Magnetic Polysaccharide Mesenchymal Stem Cells Exosomes Delivery Microcarriers for Synergistic Therapy of Osteoarthritis. ACS NANO 2024; 18:20101-20110. [PMID: 39039744 DOI: 10.1021/acsnano.4c01406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease that afflicts more than 250 million people worldwide, impairing their mobility and quality of life. However, conventional drug therapy is palliative. Exosomes (Exo), although with the potential to fundamentally repair cartilage, face challenges in their efficient enrichment and delivery. In this study, we developed magnetic polysaccharide hydrogel particles as microcarriers for synergistic therapy of OA. The microcarriers were composed of modified natural polysaccharides, hyaluronic acid (HAMA), and chondroitin sulfate (CSMA), and were generated from microfluidic electrospray in combination with a cryogelation process. Magnetic nanoparticles with spiny structures capable of capturing stem cell Exo were encapsulated within the microcarriers together with an anti-inflammatory drug diclofenac sodium (DS). The released DS and Exo from the microcarriers had a synergistic effect in alleviating the OA symptoms and promoting cartilage repair. The in vitro and in vivo results demonstrated the excellent performance of the microcarrier for OA treatment. We believe this work has potential for Exo therapy of OA and other related diseases.
Collapse
Affiliation(s)
- Lei Yang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Wenzhao Li
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Luoran Shang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
34
|
Ma T, Xu G, Gao T, Zhao G, Huang G, Shi J, Chen J, Song J, Xia J, Ma X. Engineered Exosomes with ATF5-Modified mRNA Loaded in Injectable Thermogels Alleviate Osteoarthritis by Targeting the Mitochondrial Unfolded Protein Response. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21383-21399. [PMID: 38626424 DOI: 10.1021/acsami.3c17209] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Osteoarthritis (OA) progression is highly associated with chondrocyte mitochondrial dysfunction and disorders of catabolism and anabolism of the extracellular matrix (ECM) in the articular cartilage. The mitochondrial unfolded protein response (UPRmt), which is an integral component of the mitochondrial quality control (MQC) system, is essential for maintaining chondrocyte homeostasis. We successfully validated the pivotal role of activating transcription factor 5 (ATF5) in upregulating the UPRmt, mitigating IL-1β-induced inflammation and mitochondrial dysfunction, and promoting balanced metabolism in articular cartilage ECM, proving its potential as a promising therapeutic target for OA. Modified mRNAs (modRNAs) have emerged as novel and efficient gene delivery vectors for nucleic acid therapeutic approaches. In this study, we combined Atf5-modRNA (modAtf5) with engineered exosomes derived from bone mesenchymal stem cells (ExmodAtf5) to exert cytoprotective effects on chondrocytes in articular cartilage via Atf5. However, the rapid localized metabolization of ExmodAtf5 limits its application. PLGA-PEG-PLGA (Gel), an injectable thermosensitive hydrogel, was used as a carrier of ExmodAtf5 (Gel@ExmodAtf5) to achieve a sustained release of ExmodAtf5. In vitro and in vivo, the use of Gel@ExmodAtf5 was shown to be a highly effective strategy for OA treatment. The in vivo therapeutic effect of Gel@ExmodAtf5 was evidenced by the preservation of the intact cartilage surface, low OARSI scores, fewer osteophytes, and mild subchondral bone sclerosis and cystic degeneration. Consequently, the combination of ExmodAtf5 and PLGA-PEG-PLGA could significantly enhance the therapeutic efficacy and prolong the exosome release. In addition, the mitochondrial protease ClpP enhanced chondrocyte autophagy by modulating the mTOR/Ulk1 pathway. As a result of our research, Gel@ExmodAtf5 can be considered to be effective at alleviating the progression of OA.
Collapse
Affiliation(s)
- Tiancong Ma
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Guangyu Xu
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Tian Gao
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Guanglei Zhao
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Gangyong Huang
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jingsheng Shi
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jie Chen
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jian Song
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jun Xia
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Xiaosheng Ma
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| |
Collapse
|
35
|
Chai W, Zhang M, He Y, Chai W. Characteristics of immune cells and causal relationship with chondromalacia: A two-sample, bidirectional mendelian randomization study. Mol Pain 2024; 20:17448069241289962. [PMID: 39313492 PMCID: PMC11528737 DOI: 10.1177/17448069241289962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/19/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024] Open
Abstract
Chondromalacia, characterized by the softening of cartilage, is a prevalent condition affecting joint health with complex etiology. The immune system's role in its pathogenesis has been implicated but remains to be fully elucidated. To address a critical knowledge gap, we conducted a two-sample Mendelian randomization analysis of 731 immune cell phenotypes, assessing parameters like fluorescence, cell count, and morphology. After sensitivity and pleiotropy checks, and applying a false discovery rate correction, our study linked 17 phenotypes to chondromalacia (p < .05). Among them, seven immune cell phenotypes were found to have a protective effect against chondromalacia (IVW: p < .05, OR <1), while 10 were considered risk factors (IVW:p < .05, OR >1). Despite the constraints of sample size and possible genetic differences among populations, our research has identified a notable genetic correlation between specific immune cell indicators and chondromalacia. This breakthrough sheds light on the pathophysiological mechanisms of the condition. The identification of protective and risk-associated immune cell phenotypes provides a foundation for further exploration of immunological mechanisms in chondromalacia and may pave the way for targeted interventions. Future research is warranted to validate these findings and explore their clinical implications.
Collapse
Affiliation(s)
- Weiwei Chai
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province(Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Mengwei Zhang
- Department of Emergency, Luoyang Orthopedic-Traumatological Hospital of Henan Province(Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Yan He
- Department of Radiology, The Third Afiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Weihao Chai
- Department of Graduate School, Xinjiang Medical University, Urumqi, China
| |
Collapse
|