1
|
Chen J, Chen J, Chen J, Lu R, Liu Z, Zhang Y, Zhang C. Pretreated exosomes by electrical stimulation accelerate bone regeneration. Bioact Mater 2025; 51:383-398. [PMID: 40491687 PMCID: PMC12148642 DOI: 10.1016/j.bioactmat.2025.04.019] [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: 01/13/2025] [Revised: 03/27/2025] [Accepted: 04/16/2025] [Indexed: 06/11/2025] Open
Abstract
Bone tissue engineering has attracted significant attention from both the research and clinical communities. Inspired by the inherent bioelectric properties of bone tissue, electrical stimulation is widely recognized as an external intervention that can induce osteogenesis, mineralization, and accelerate bone regeneration. However, the clinical application of electrical stimulation is limited by the complexity of the procedures and the use of cumbersome, invasive equipment. Exosomes, as an alternative to seed cells, can overcome many of the limitations associated with stem cell transplantation. Researchers aim to enhance exosomes' therapeutic potential for bone regeneration. While various pretreatments have been studied, there is currently no research investigating the role of exosomes pretreated with electrical stimulation in bone tissue regeneration. In this study, we pretreated bone marrow mesenchymal stem cells (BMSCs) with electrical stimulation and isolated the resulting exosomes (Elec-exo). A series of in vitro experiments determined that 150 μA is the optimal condition for electrical stimulation. Mechanistically, proteomic analysis revealed an enrichment of proteins involved in "Oxidative Phosphorylation" regulation within Elec-exo, and transcriptomic analysis indicated the activation of Pl3k-Akt and MAPK bone formation-related signaling pathways in the effector cells. Hydrogels, as a sustained-release scaffold, were used to deliver Elec-exo in vivo. In a rat femur defect model, Elec-exo loaded into chondroitin sulfate methacrylate (CSMA) hydrogel accelerated early bone tissue regeneration. In summary, our study explores the mechanisms by which electrical stimulation pretreatment enhances bone tissue regeneration and broadens the therapeutic application of exosomes in accelerating bone regeneration.
Collapse
Affiliation(s)
- Jialu Chen
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200236, China
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Jiahao Chen
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200236, China
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Renjie Lu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200236, China
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziyuan Liu
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yang Zhang
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Pharmacy, Shanghai Eighth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chi Zhang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China
| |
Collapse
|
2
|
Li Z, Ren K, Chen J, Zhuang Y, Dong S, Wang J, Liu H, Ding J. Bioactive hydrogel formulations for regeneration of pathological bone defects. J Control Release 2025; 380:686-714. [PMID: 39880040 DOI: 10.1016/j.jconrel.2025.01.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Bone defects caused by osteoporosis, infection, diabetes, post-tumor resection, and nonunion often cause severe pain and markedly increase morbidity and mortality, which remain a significant challenge for orthopedic surgeons. The precise local treatments for these pathological complications are essential to avoid poor or failed bone repair. Hydrogel formulations serve as injectable innovative platforms that overcome microenvironmental obstacles and as delivery systems for controlled release of various bioactive substances to bone defects in a targeted manner. Additionally, hydrogel formulations can be tailored for specific mechanical strengths and degradation profiles by adjusting their physical and chemical properties, which are crucial for prolonged drug retention and effective bone repair. This review summarizes recent advances in bioactive hydrogel formulations as three-dimensional scaffolds that support cell proliferation and differentiation. It also highlights their role as smart drug-delivery systems with capable of continuously releasing antibacterial agents, anti-inflammatory drugs, chemotherapeutic agents, and osteogenesis-related factors to enhance bone regeneration in pathological areas. Furthermore, the limitations of hydrogel formulations in pathological bone repair are discussed, and future development directions are proposed, which is expected to pave the way for the repair of pathological bone defects.
Collapse
Affiliation(s)
- Zuhao Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China
| | - Kaixuan Ren
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China; Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Jiajia Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China; The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 6822 Jinhu Road, Changchun 130021, China
| | - Yaling Zhuang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 6822 Jinhu Road, Changchun 130021, China
| | - Jincheng Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China
| | - He Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| |
Collapse
|
3
|
Guo J, Zhang T, Li M, Wang Q, Ding X. Synergistic interactions between physical exercise intervention, innovative materials, and neurovascular coupling in bone repair and injury recovery: a comprehensive review. Biomed Mater 2025; 20:032002. [PMID: 40138797 DOI: 10.1088/1748-605x/adc5c0] [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: 06/23/2024] [Accepted: 03/26/2025] [Indexed: 03/29/2025]
Abstract
Bone injury presents a prevalent challenge in clinical settings, with traditional treatment modalities exhibiting inherent limitations. Recent advancements have highlighted the potential of combining physical exercise intervention and innovative materials to enhance bone repair and recovery. This review explores the synergistic effects of physical exercise and novel materials in promoting bone regeneration, with a particular focus on the role of neurovascular coupling (NVC) mechanisms. Physical exercise not only stimulates bone cell function and blood circulation but also enhances the bioactivity of novel materials, such as nanofiber membranes and smart materials, which provide supportive scaffolds for bone cell attachment, proliferation, and differentiation. NVC, involving the interaction between neural activity and blood flow, is integral to the bone repair process, ensuring the supply of nutrients and oxygen to the injured site. Studies demonstrate that the combination of physical exercise and novel materials can accelerate bone tissue regeneration, with exercise potentially enhancing the bioactivity of materials and materials improving the effectiveness of exercise. However, challenges remain in clinical applications, including patient variability, material biocompatibility, and long-term stability. Optimizing the integration of physical exercise and novel materials for optimal therapeutic outcomes is a key focus for future research. This review examines the collaborative mechanisms between physical exercise, novel materials, and NVC, emphasizing their potential and the ongoing challenges in clinical settings. Further exploration is needed to refine their application and improve bone repair strategies.
Collapse
Affiliation(s)
- Jiejie Guo
- Central laboratory, Department of Clinical Laboratory, The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou 317500, People's Republic of China
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo 315000, People's Republic of China
| | - Ting Zhang
- Central laboratory, Department of Clinical Laboratory, The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou 317500, People's Republic of China
| | - Mengjia Li
- Central laboratory, Department of Clinical Laboratory, The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou 317500, People's Republic of China
| | - Qinwen Wang
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo 315000, People's Republic of China
| | - Xianting Ding
- Central laboratory, Department of Clinical Laboratory, The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou 317500, People's Republic of China
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| |
Collapse
|
4
|
Feng J, Wang F, Shao Y, Jin A, Lei L. Engineered protein-based materials for tissue repair: A review. Int J Biol Macromol 2025; 303:140674. [PMID: 39909268 DOI: 10.1016/j.ijbiomac.2025.140674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/19/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
The human body may suffer multiple injuries and losses due to various external factors, such as tumors, diseases, traffic accidents, and war conflicts. Under such circumstances, engineered protein-based materials, as an innovative adjunctive material, can not only effectively promote the natural repair process of tissues, but also greatly circumvent the negative effects and complications that may be associated with conventional surgery. In this review, we first trace the definition and development of engineered protein-based materials and explain in detail their mechanism of action in promoting tissue repair. Subsequently, the advantages and disadvantages of various engineered protein-based materials in tissue repair are analyzed by comparison. In addition, the present review reveals in depth how material properties can be optimized by scientific means to meet different tissue repair needs. In addition, we present in detail specific application cases of engineered protein-based materials in the field of tissue repair. Finally, we summarize current challenges in engineered protein-based materials and provide an outlook for the future. This review not only provides theoretical support for the further exploration and development of engineered protein-based materials in the field of tissue repair, but also provides valuable references and inspiration for research in related fields.
Collapse
Affiliation(s)
- Jiayin Feng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Fangyan Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.
| |
Collapse
|
5
|
Fu Z, Qiu H, Xu Y, Tan C, Wang H. Biological effects, properties and tissue engineering applications of polyhydroxyalkanoates: A review. Int J Biol Macromol 2025; 293:139281. [PMID: 39736299 DOI: 10.1016/j.ijbiomac.2024.139281] [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: 06/09/2024] [Revised: 12/10/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025]
Abstract
Polyhydroxyalkanoates (PHAs) are a group of polymers with a variety of monomers, which are extracted from microorganisms and plants. Due to its good biocompatibility, biodegradability, tunable mechanical property and piezoelectricity, PHAs have been widely used in biomedical fields, such as bone, cartilage, nerve, vascular and skin tissue engineering. This review focuses on the in vivo synthesis, metabolism and biological functions of PHA, and the applications of PHAs in the field of tissue engineering and commercial were also summarized and discussed. Moreover, this review hints the future perspective and research direction of PHA-based materials in the challenging field of tissue engineering. We hope that this review will catalyze the continued advancement and broadening of PHAs' applications in biomedicine.
Collapse
Affiliation(s)
- Zeyu Fu
- Beijing Joyinera biomaterial Technology Co., Ltd., Beijing, China
| | - He Qiu
- Beijing Joyinera biomaterial Technology Co., Ltd., Beijing, China; Department of Cosmetic and Plastic Surgery, West China School of Public Health and West China Fourth Hospital, Sichuan University, Sichuan, Chengdu, China
| | - Yuan Xu
- Beijing Joyinera biomaterial Technology Co., Ltd., Beijing, China
| | - Chang Tan
- Beijing Joyinera biomaterial Technology Co., Ltd., Beijing, China.
| | - Hang Wang
- Department of Cosmetic and Plastic Surgery, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Huang J, Li H, Mei Y, Yi P, Ren Y, Wang Y, Han L, Tang Q, Liu D, Chen W, An Y, Hu C. An Injectable Hydrogel Bioimplant Loaded with Engineered Exosomes and Triple Anti-Tuberculosis Drugs with Potential for Treating Bone and Joint Tuberculosis. Int J Nanomedicine 2025; 20:1285-1302. [PMID: 39911262 PMCID: PMC11794387 DOI: 10.2147/ijn.s480288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/12/2024] [Indexed: 02/07/2025] Open
Abstract
Purpose Treatment for bone and joint tuberculosis (BJTB) is challenging due to its refractory and recurrent nature. This study aimed to develop a bioimplantable scaffold with osteoinductive and antituberculosis characteristics to treat BJTB. Methods This scaffold is built on oxidized hyaluronic acid and carboxymethyl chitosan hydrogel mixed with hydroxyapatite as a bone tissue engineered material. In order to make the scaffold have the biological activity of promoting tissue repair, the engineered exosomes (Exoeng) were added innovatively. In addition, drug-loaded liposomes equipped with an aldehyde group on the surface are cross-linked with the amine group of the hydrogel skeleton to participate in the Schiff base reaction. Results The designed scaffold has characteristics of self-healing and injectability exhibit excellent anti-tuberculosis and promoting bone repair activities. Exoeng strongly stimulates cellular angiogenesis and osteogenic differentiation. The liposomes coated in hydrogel can release three kinds of anti-tuberculosis drugs smoothly and slowly, achieving a long term anti-tuberculosis. Conclusion The composite bio-scaffold shows good tissue repair and long-term anti-tuberculosis abilities, which expected to provide a viable treatment plan for bone-related BJTB.
Collapse
Affiliation(s)
- Jiayan Huang
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Han Li
- Department of Pharmacy, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, People’s Republic of China
| | - Yuting Mei
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Pengcheng Yi
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yunyao Ren
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yunjuan Wang
- Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Limei Han
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Qiusha Tang
- Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Dongfang Liu
- Medical School of Southeast University, Nanjing, People’s Republic of China
| | - Wei Chen
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yanli An
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Medical School, Zhong da Hospital, Southeast University, Nanjing, JiangsuPeople’s Republic of China
| | - Chunmei Hu
- Department of Tuberculosis, the Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
7
|
Xiang M, Guo Q, Liu Y, Zhang G, Liao C, Xiao L, Xiang M, Long S, Long Q, Guan X, Liu J. Low-intensity pulsed ultrasound enhances the osteogenic potential of PDLSCs-derived extracellular vesicles through COMP/PI3K/AKT. FASEB J 2025; 39:e70299. [PMID: 39792132 DOI: 10.1096/fj.202402463r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/23/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
The therapeutic potential of extracellular vesicles (EVs) in bone regeneration is noteworthy; however, their clinical application is impeded by low yield and limited efficacy. This study investigated the effect of low-intensity pulsed ultrasound (LIPUS) on the therapeutic efficacy of EVs derived from periodontal ligament stem cells (PDLSCs) and preliminarily explored its mechanism. PDLSCs were cultured with osteogenic media and stimulated with or without LIPUS, and then EVs and LIPUS-stimulated EVs (L-EVs) were isolated separately. We investigated the biological characteristics and effects of these two EVs on cell proliferation, migration, osteogenic differentiation, and bone regeneration in vivo and in vitro, and explored the potential mechanism by analyzing protein profiles. LIPUS significantly stimulated the secretion of PDLSCs-EVs, and L-EVs exhibited stronger efficacy in promoting cell proliferation, migration, and osteogenic differentiation, thereby enhancing new bone formation. LIPUS stimulation affected the protein profile of PDLSCs-EVs, and 42 proteins were upregulated and 4 proteins downregulated in L-EVs when compared with EVs. LIPUS significantly upregulated the level of cartilage oligomeric matrix protein (COMP) in EVs, which enhanced EVs' osteogenic ability via the PI3K/AKT pathway. This study proposes that LIPUS has potential as an optimization method for enhancing the therapeutic effects of EVs in tissue regeneration.
Collapse
Affiliation(s)
- Mingli Xiang
- GuiZhou University Medical College, Guiyang, China
| | - Qiushuang Guo
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Yulin Liu
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Gengchao Zhang
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Chengcheng Liao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Linlin Xiao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Meiling Xiang
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Sicen Long
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Qian Long
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaoyan Guan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Jianguo Liu
- GuiZhou University Medical College, Guiyang, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
8
|
Chen J, Dong S. Polymer-based antimicrobial strategies for periodontitis. Front Pharmacol 2025; 15:1533964. [PMID: 39834832 PMCID: PMC11743519 DOI: 10.3389/fphar.2024.1533964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Periodontitis is a chronic inflammatory condition driven by plaque-associated microorganisms, where uncontrolled bacterial invasion and proliferation impair host immune responses, leading to localized periodontal tissue inflammation and bone destruction. Conventional periodontal therapies face challenges, including incomplete microbial clearance and the rise of antibiotic resistance, limiting their precision and effectiveness in managing periodontitis. Recently, nanotherapies based on polymeric materials have introduced advanced approaches to periodontal antimicrobial therapy through diverse antimicrobial mechanisms. This review explored specific mechanisms, emphasizing the design of polymer-based agents that employ individual or synergistic antimicrobial actions, and evaluated the innovations and limitations of current strategies while forecasting future trends in antimicrobial development for periodontitis.
Collapse
Affiliation(s)
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
9
|
Wan X, Zhang W, Dai L, Chen L. The Role of Extracellular Vesicles in Bone Regeneration and Associated Bone Diseases. Curr Issues Mol Biol 2024; 46:9269-9285. [PMID: 39329900 PMCID: PMC11430372 DOI: 10.3390/cimb46090548] [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: 08/03/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoscale particles with a lipid bilayer membrane structure secreted by various cell types. Nearly all human cells secrete EVs, primarily mediating intercellular communication. In recent years, scientists have discovered that EVs can carry multiple biological cargos, such as DNA, non-coding RNAs (ncRNAs), proteins, cytokines, and lipids, and mediate intercellular signal transduction. Bone is a connective tissue with a nerve supply and high vascularization. The repair process after injury is highly complex, involving interactions among multiple cell types and biological signaling pathways. Bone regeneration consists of a series of coordinated osteoconductive and osteoinductive biological processes. As mediators of intercellular communication, EVs can promote bone regeneration by regulating osteoblast-mediated bone formation, osteoclast-mediated bone resorption, and other pathways. This review summarizes the biogenesis of EVs and the mechanisms by which EV-mediated intercellular communication promotes bone regeneration. Additionally, we focus on the research progress of EVs in various diseases related to bone regeneration. Finally, based on the above research, we explore the clinical applications of engineered EVs in the diagnosis and treatment of bone regeneration-related diseases.
Collapse
Affiliation(s)
- Xinyue Wan
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Lingyan Dai
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
| | - Liang Chen
- School of Medicine, Chongqing University, Chongqing 400030, China; (X.W.); (W.Z.); (L.D.)
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing University School of Medicine, Chongqing 400030, China
| |
Collapse
|
10
|
Li X, Pang Y, Guan L, Li L, Zhu Y, Whittaker AK, Yang B, Zhu S, Lin Q. Mussel-inspired antimicrobial hydrogel with cellulose nanocrystals/tannic acid modified silver nanoparticles for enhanced calvarial bone regeneration. Int J Biol Macromol 2024; 270:132419. [PMID: 38759859 DOI: 10.1016/j.ijbiomac.2024.132419] [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: 01/22/2024] [Revised: 03/20/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Bacterial infection is a serious challenge in the treatment of open bone defects, and reliance on antibiotic therapy may contribute to the emergence of drug-resistant bacteria. To solve this problem, this study developed a mineralized hydrogel (PVA-Ag-PHA) with excellent antibacterial properties and osteogenic capabilities. Silver nanoparticles (CNC/TA@AgNPs) were greenly synthesized using natural macromolecular cellulose nanocrystals (CNC) and plant polyphenolic tannins (TA) as stabilizers and reducing agents respectively, and then introduced into polyvinyl alcohol (PVA) and polydopamine-modified hydroxyapatite (PDA@HAP) hydrogel. The experimental results indicate that the PVA-Ag-PHA hydrogel, benefiting from the excellent antibacterial properties of CNC/TA@AgNPs, can not only eliminate Staphylococcus aureus and Escherichia coli, but also maintain a sustained sterile environment. At the same time, the HAP modified by PDA is uniformly dispersed within the hydrogel, thus releasing and maintaining stable concentrations of Ca2+ and PO43- ions in the local environment. The porous structure of the hydrogel with excellent biocompatibility creates a suitable bioactive environment that facilitates cell adhesion and bone regeneration. The experimental results in the rat critical-sized calvarial defect model indicate that the PVA-Ag-PHA hydrogel can effectively accelerate the bone healing process. Thus, this mussel-inspired hydrogel with antibacterial properties provides a feasible solution for the repair of open bone defects, demonstrating the considerable potential for diverse applications in bone repair.
Collapse
Affiliation(s)
- Xingchen Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Yuxuan Pang
- Department of Prosthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Lin Guan
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Lei Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Yanlin Zhu
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology. The University of Queensland Brisbane, QLD 4072, Australia.
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Song Zhu
- Department of Prosthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China.
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China.
| |
Collapse
|
11
|
Wang B, Huang Y, Cai Q, Du Z, Li X. Biomaterials for diabetic bone repair: Influencing mechanisms, multi-aspect progress and future prospects. COMPOSITES PART B: ENGINEERING 2024; 274:111282. [DOI: 10.1016/j.compositesb.2024.111282] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
|
12
|
Zhan W, Deng M, Huang X, Xie D, Gao X, Chen J, Shi Z, Lu J, Lin H, Li P. Pueraria lobata-derived exosome-like nanovesicles alleviate osteoporosis by enhacning autophagy. J Control Release 2023; 364:644-653. [PMID: 37967723 DOI: 10.1016/j.jconrel.2023.11.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023]
Abstract
Osteoporosis (OP) is the most common bone disorder worldwide, especially in postmenopausal women. However, many OP drugs are not suitable for long term use due to major adverse effects. Therefore, there is an urgent need to identify more effective and safe therapeutic drugs. Pueraria lobata has been reported to promote osteoblast growth in bone regeneration, but the exact mechanisms still need further exploration. The current study found that Pueraria lobata-derived exosome-like nanovesicles (PELNs) promoting primary human bone mesenchymal stem cells (hBMSCs) differentiation and mineralization both in vitro and in ovariectomized (OVX)-induced osteoporotic rats. Interestingly, the relative abundance of harmful strains significantly decreased in the intestine of the osteoporosis SD rat model administrated PELNs via the regulation of trimethylamine-N-oxide (TMAO), a metabolite of gut microbiota. Moreover, RNA sequencing revealed that the osteogenic activity of PELNs is revealed to autophagy signaling. In vitro and in vivo experiments also showed that the treatment with PELNs promoted the differentiation and function of hBMSCs by elevating autophagy via the degradation of TMAO. Collectively, PELNs demonstrate promise as a therapeutic approach for OP, with TMAO emerging as a potential target of OP treatment.
Collapse
Affiliation(s)
- Weiqiang Zhan
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China; Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China
| | - Mingzhu Deng
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China; Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China
| | - Xinqia Huang
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China; Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China
| | - Dong Xie
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China
| | - Jiaxian Chen
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China
| | - Zhen Shi
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China
| | - Jiaxu Lu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China; Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China
| | - Hao Lin
- Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China.
| | - Peng Li
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, PR China.
| |
Collapse
|
13
|
Liu X, Xiao C, Xiao K. Engineered extracellular vesicles-like biomimetic nanoparticles as an emerging platform for targeted cancer therapy. J Nanobiotechnology 2023; 21:287. [PMID: 37608298 PMCID: PMC10463632 DOI: 10.1186/s12951-023-02064-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Abstract
Nanotechnology offers the possibility of revolutionizing cancer theranostics in the new era of precision oncology. Extracellular vesicles (EVs)-like biomimetic nanoparticles (EBPs) have recently emerged as a promising platform for targeted cancer drug delivery. Compared with conventional synthetic vehicles, EBPs have several advantages, such as lower immunogenicity, longer circulation time, and better targeting capability. Studies on EBPs as cancer therapeutics are rapidly progressing from in vitro experiments to in vivo animal models and early-stage clinical trials. Here, we describe engineering strategies to further improve EBPs as effective anticancer drug carriers, including genetic manipulation of original cells, fusion with synthetic nanomaterials, and direct modification of EVs. These engineering approaches can improve the anticancer performance of EBPs, especially in terms of tumor targeting effectiveness, stealth property, drug loading capacity, and integration with other therapeutic modalities. Finally, the current obstacles and future perspectives of engineered EBPs as the next-generation delivery platform for anticancer drugs are discussed.
Collapse
Affiliation(s)
- Xinyi Liu
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunxiu Xiao
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Xiao
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Tianfu Jingcheng Laboratory (Frontier Medical Center), Chengdu, 610041, China.
| |
Collapse
|
14
|
Lin SW, Tsai JC, Shyong YJ. Drug delivery of extracellular vesicles: Preparation, delivery strategies and applications. Int J Pharm 2023; 642:123185. [PMID: 37391106 DOI: 10.1016/j.ijpharm.2023.123185] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Extracellular vesicles (EV) are cell-originated vesicles exhibited with characteristics similar to the parent cells. Several studies have suggested the therapeutic potential of EV since they played as an intercellular communicator and modulate disease microenvironment, and thus EV has been widely studied in cancer management and tissue regeneration. However, merely application of EV revealed limited therapeutic outcome in different disease scenario and co-administration of drugs may be necessary to exert proper therapeutic effect. The method of drug loading into EV and efficient delivery of the formulation is therefore important. In this review, the advantages of using EV as drug delivery system compared to traditional synthetic nanoparticles will be emphasized, followed by the method of preparing EV and drug loading. The pharmacokinetic characteristics of EV was discussed, together with the review of reported delivery strategies and related application of EV in different disease management.
Collapse
Affiliation(s)
- Shang-Wen Lin
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan
| | - Jui-Chen Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan
| | - Yan-Jye Shyong
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan.
| |
Collapse
|
15
|
Huang X, Li Y, Liao H, Luo X, Zhao Y, Huang Y, Zhou Z, Xiang Q. Research Advances on Stem Cell-Derived Extracellular Vesicles Promoting the Reconstruction of Alveolar Bone through RANKL/RANK/OPG Pathway. J Funct Biomater 2023; 14:jfb14040193. [PMID: 37103283 PMCID: PMC10145790 DOI: 10.3390/jfb14040193] [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: 02/02/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023] Open
Abstract
Periodontal bone tissue defects and bone shortages are the most familiar and troublesome clinical problems in the oral cavity. Stem cell-derived extracellular vesicles (SC-EVs) have biological properties similar to their sources, and they could be a promising acellular therapy to assist with periodontal osteogenesis. In the course of alveolar bone remodeling, the RANKL/RANK/OPG signaling pathway is an important pathway involved in bone metabolism. This article summarizes the experimental studies of SC-EVs applied for the therapy of periodontal osteogenesis recently and explores the role of the RANKL/RANK/OPG pathway in their mechanism of action. Their unique patterns will open a new field of vision for people, and they will help to advance a possible future clinical treatment.
Collapse
Affiliation(s)
- Xia Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
- Department of Orthodontics, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Yuxiao Li
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Hui Liao
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Xin Luo
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Yueping Zhao
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Yadong Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Zhiying Zhou
- School of Stomatology, Jinan University, Guangzhou 510632, China
- Department of Orthodontics, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|