201
|
Yang G, Pan H, Wei Y, Yang J, Zhang Z, Chen S, Wan W. Directional Mushroom-Derived Scaffold for Microenvironment Regulation in Infected Bone Defects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407730. [PMID: 39961061 DOI: 10.1002/adma.202407730] [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: 05/30/2024] [Revised: 12/11/2024] [Indexed: 03/27/2025]
Abstract
Infected bone defects are a common clinical condition, but conventional treatments often fail to achieve the desired outcomes, including addressing antibiotic resistance and preventing nonunion complications. In the presented study, a functionalized decellularized mushroom stem scaffold is developed composed of its naturally aligned channels, Zn2+/curcumin MOFs, hydroxyapatite minerals, and icariin. In vitro, It is found that functionalized acellular mushroom stem scaffold can control bacterial infections through Zn2+/curcumin MOFs. The naturally aligned channels guide bone mesenchymal stem cells (BMSCs) migration, and the components adsorbed on the acellular substrate further promote the migration of BMSCs. Moreover, these functional components further accelerated the polarization of M2 macrophage and osteogenic differentiation of BMSCs. In vivo, the functionalized decellularized mushroom stem scaffold cleared infected bacteria within 3 days, induced extracellular matrix secretion and alignment, and promoted new bone formation to cover defects within 8 weeks. The functionalized decellularized mushroom stem scaffold provides a promising strategy for treating infectious bone defects.
Collapse
Affiliation(s)
- Ganghua Yang
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Hao Pan
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Yuxuan Wei
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Jianqiu Yang
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zihan Zhang
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Wenbing Wan
- Department of Orthopaedic Surgery, Institute of Orthopedics of Jiangxi Province and Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
202
|
Xiao J, You K, Lu D, Guan S, Wu H, Gao J, Tang Y, Yu S, Gao B. Cell-Derived Basal Membrane-Like Extracellular Matrix Promotes Endothelial Cell Expansion and Functionalization. J Biomed Mater Res A 2025; 113:e37893. [PMID: 40059713 DOI: 10.1002/jbm.a.37893] [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: 10/23/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
Engineering cellular microenvironments with biomaterials is an effective strategy for endothelial cell expansion and functionality in vascular tissue engineering. The basement membrane (BM) is a natural vascular endothelium microenvironment that plays an important role in promoting rapid expansion and function of endothelial cells. However, mimicking the crucial function of BM with an ideal biomaterial remains challenging. In this study, we developed a cell-derived decellularized extracellular matrix (c-dECM) paper to mimic the role of BM in endothelial cell expansion and function. The results showed that c-dECM paper was a stable, biocompatible, and biodegradable scaffold that significantly promoted endothelial cell expansion by modulating cell migration, adhesion, and proliferation both in vivo and in vitro. Moreover, the biomimetic c-dECM paper can profoundly promote endothelial cell function by increasing the synthesis and release of nitric oxide (NO) and prostaglandin I2 (PGI2) and upregulating the expression of anticoagulant and vascularized genes, including thrombomodulin (THBD), tissue factor pathway inhibitor (TFPI), endothelial growth factor (VEGF) and endoglin (CD105). These data indicate that the c-dECM is a potential biomaterial for constructing vascular tissue engineering scaffolds or developing in vitro models to study the functional mechanisms of endothelial cells.
Collapse
Affiliation(s)
- Jiangwei Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Kai You
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Daohuan Lu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Shuwen Guan
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Hengpeng Wu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Jing Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Yadong Tang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Shan Yu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Chemical Engineering Department, Ningbo Key Laboratory of High Performance Petroleum Resin Preparation Engineering and Technology, Ningbo, China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- National Engineering Research Center for Healthcare Devices, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
203
|
Zhao J, Sarkar N, Ren Y, Pathak AP, Grayson WL. Engineering next-generation oxygen-generating scaffolds to enhance bone regeneration. Trends Biotechnol 2025; 43:540-554. [PMID: 39343620 PMCID: PMC11867879 DOI: 10.1016/j.tibtech.2024.09.006] [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: 03/14/2024] [Revised: 08/08/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
In bone, an adequate oxygen (O2) supply is crucial during development, homeostasis, and healing. Oxygen-generating scaffolds (OGS) have demonstrated significant potential to enhance bone regeneration. However, the complexity of O2 delivery and signaling in vivo makes it challenging to tailor the design of OGS to precisely meet this biological requirement. We review recent advances in OGS and analyze persisting engineering and translational hurdles. We also discuss the potential of computational and machine learning (ML) models to facilitate the integration of novel imaging data with biological readouts and advanced biomanufacturing technologies. By elucidating how to tackle current challenges using cutting-edge technologies, we provide insights for transitioning from traditional to next-generation OGS to improve bone regeneration in patients.
Collapse
Affiliation(s)
- Jingtong Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Naboneeta Sarkar
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Yunke Ren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Arvind P Pathak
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA; Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
204
|
Zhao X, Yu Y, Xu X, Zhang Z, Chen Z, Gao Y, Zhong L, Chen J, Huang J, Qin J, Zhang Q, Tang X, Yang D, Zhu Z. Machine Learning-Assisted High-Throughput Screening of Nanozymes for Ulcerative Colitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417536. [PMID: 39801185 DOI: 10.1002/adma.202417536] [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: 11/12/2024] [Revised: 12/13/2024] [Indexed: 03/06/2025]
Abstract
Ulcerative colitis (UC) is a chronic gastrointestinal inflammatory disorder with rising prevalence. Due to the recurrent and difficult-to-treat nature of UC symptoms, current pharmacological treatments fail to meet patients' expectations. This study presents a machine learning-assisted high-throughput screening strategy to expedite the discovery of efficient nanozymes for UC treatment. Therapeutic requirements, including antioxidant property, acid stability, and zeta potential, are quantified and predicted by using a machine learning model. Non-quantifiable attributes, including intestinal barrier repair efficacy and biosafety, are assessed via high-throughput screening. Feature significance analysis, sure independence screening, and sparsifying operator symbolic regression reveal the high-dimensional structure-activity relationships between material features and therapeutic needs. SrDy2O4 with high stability, low toxicity, targeting ability, and reactive oxygen species (ROS) scavenging capability is identified, which reduces ROS production, lowers cytochrome C levels in cytoplasm, and inhibits apoptosis in intestinal epithelial cells by stabilizing the mitochondrial membrane potential. Mice treated with SrDy2O4 show improvements in colon length and body weight compared with dextran sodium sulfate salt-treated model group. Transcriptomic and 16S rRNA sequencing analyses show that SrDy2O4 boosts beneficial gut bacteria, and decreases pathogenic bacteria, thereby effectively restoring gut microbiota balance. Moreover, SrDy2O4 offers the advantage of X-ray imaging without side effects.
Collapse
Affiliation(s)
- Xianguang Zhao
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Yixin Yu
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| | - Xudong Xu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Ziqi Zhang
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| | - Zhen Chen
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| | - Yubo Gao
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| | - Liang Zhong
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jiajie Chen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jiaxin Huang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jie Qin
- Department of Radiology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Qingyun Zhang
- Central Laboratory Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Xuemei Tang
- Central Laboratory Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Dongqin Yang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Central Laboratory Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Zhiling Zhu
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| |
Collapse
|
205
|
Li W, Zhao Y, Cheng Z, Niu F, Ding J, Bai Y, Li Z, Midgley AC, Zhu M. Fine-tuning of porous microchannelled silk fibroin scaffolds for optimal tissue ingrowth. MATERIALS & DESIGN 2025; 251:113711. [DOI: 10.1016/j.matdes.2025.113711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
|
206
|
Xia Y, Li X, Huang F, Wu Y, Liu J, Liu J. Design and advances in antioxidant hydrogels for ROS-induced oxidative disease. Acta Biomater 2025; 194:80-97. [PMID: 39900274 DOI: 10.1016/j.actbio.2025.01.057] [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: 09/30/2024] [Revised: 01/14/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
Reactive oxygen species (ROS) play a crucial role in human physiological processes, but oxidative stress caused by excessive ROS may lead to a variety of acute and chronic diseases. Despite the development of various strategies and biomaterials, an efficiently and broadly applied method for treatment of ROS-induced oxidative disease remains a bottleneck. Aiming to improve the local oxidative stress environment, numerous bioactive hydrogels with antioxidant properties have emerged and are proven to quickly and continuously eliminate excessive ROS. To deeply understand the design principles and applications of antioxidant hydrogels is highly beneficial for designing antioxidant hydrogels for treatment of oxidative disease. This review provides a detailed summary of recent advances in design and applications of antioxidant hydrogels for various ROS-induced oxidative diseases. In this review, the kinds of antioxidant components in antioxidant hydrogels are outlined in detail. Additionally, the crosslinking methods and the biomedical applications of antioxidant hydrogels are widely summarized and discussed, especially focusing on their usage in different types of diseases and the attention given to the treatment of diseases such as skin wounds, myocardial infarction, and osteoarthritis. Finally, the future development direction of antioxidant hydrogel is further proposed. STATEMENT OF SIGNIFICANCE: Oxidative stress is a pivotal biochemical process that plays a critical role in cellular homeostasis. Excessive cellular oxidative stress triggers an inflammatory response, which is implicated in a spectrum of associated diseases. Given the critical need for managing oxidative stress, antioxidant therapies have become a vital focus in medical research. Hydrogels have garnered substantial interest among biomaterial scientists due to their hydrophilic nature and biocompatibility. The review delves into the realm of antioxidant hydrogels, encompassing the classification of antioxidant components, the synthesis and fabrication of hydrogels, and a comprehensive overview of the biological applications and challenges of these antioxidant hydrogels. Aiming to provide new perspectives for researchers in developing cutting-edge therapeutic approaches that leverage antioxidant hydrogels.
Collapse
Affiliation(s)
- Yi Xia
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Xinyi Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Fan Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yuanhao Wu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jinjian Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
207
|
Liang J, Zou Y, Ju H, Lv Y. Rapid angiogenic hydrogel nanofiber scaffold promotes random flap regeneration and modulates inflammation. Wound Repair Regen 2025; 33:e70028. [PMID: 40231635 DOI: 10.1111/wrr.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 02/04/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025]
Abstract
Random skin flaps are commonly used to cover thick skin wounds. However, necrosis frequently occurs when the flap's aspect ratio exceeds 2:1. Promoting angiogenesis and regulating inflammation are essential for treating ischemia-reperfusion injury in random flaps. In this study, calcium-doped silica nanoparticles loaded with deferoxamine (CD) were created through physical adsorption by rapidly mixing biodegradable calcium-doped silica nanoparticles (CS) with deferoxamine (DFO). A gelatin methacryloyl (GM) hydrogel nanofiber scaffold containing 1% (w/v) CD (GM/CD-1) was subsequently produced using electrospinning technology. The GM/CD-1 scaffold showed excellent biocompatibility and significantly promoted flap regeneration in mice, achieving a 96.17 ± 3.17% flap survival rate at 14 days. Additionally, it effectively stimulated hair follicle growth and exhibited an inflammatory-modulating effect. These features suggest that the GM/CD-1 scaffold could be valuable for clinical applications in flap regeneration and other tissue engineering fields.
Collapse
Affiliation(s)
- Junyan Liang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, China
| | - Yang Zou
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, China
- School of Environmental Engineering, Wuhan Textile University, Wuhan, China
| | - Haiyan Ju
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, China
- College of Chemistry and Chemical Engineering, Wuhan Textile University, Wuhan, China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, China
| |
Collapse
|
208
|
Su X, Yang J, Xu Z, Wei L, Yang S, Li F, Sun M, Hu Y, He W, Zhao C, Chen L, Yuan Y, Qin L, Hu N. Fibrous scaffolds loaded with BMSC-derived apoptotic vesicles promote wound healing by inducing macrophage polarization. Genes Dis 2025; 12:101388. [PMID: 39759117 PMCID: PMC11697094 DOI: 10.1016/j.gendis.2024.101388] [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: 03/11/2024] [Accepted: 06/21/2024] [Indexed: 01/07/2025] Open
Abstract
Macrophages play a key role in wound healing. Dysfunction of their M0 polarization to M2 leads to disorders of the wound immune microenvironment and chronic inflammation, which affects wound healing. Regulating the polarization of M0 macrophages to M2 macrophages is an effective strategy for treating wound healing. Mesenchymal stem cells (MSCs) deliver endogenous regulatory factors via paracrine extracellular vesicles, which may play a key role in wound healing, and previous studies have shown that apoptotic bodies (ABs) are closely associated with inflammation regression and macrophage polarization. However, the specific regulatory mechanisms involved in ABs remain unknown. In the present study, we designed an MSC-AB (MSC-derived AB)-loaded polycaprolactone (PCL) scaffold, evaluated the macrophage phenotype and skin wound inflammation in vivo and in vitro, and explored the ability of MSC-AB-loaded PCL scaffolds to promote wound healing. Our data suggest that the PCL scaffold regulates the expression of the CCL-1 gene by targeting the delivery of mmu-miR-21a-5p by local sustained-release MSC-ABs, and drives M0 macrophages to program M2 macrophages to regulate inflammation and angiogenesis, thereby synergistically promoting wound healing. This study provides a promising therapeutic strategy and experimental basis for treating various diseases associated with imbalances in proinflammatory and anti-inflammatory immune responses.
Collapse
Affiliation(s)
- Xudong Su
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Jianye Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Zhenghao Xu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Li Wei
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Shuhao Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Feilong Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Min Sun
- Department of Knee Joint Sports Injury, Sichuan Provincial Orthopedic Hospital, Chengdu, Sichuan 610042, China
| | - Yingkun Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Wenge He
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Chen Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Li Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Yonghua Yuan
- Research Center for Pharmacodynamic Evaluation Engineering Technology of Chongqing, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Leilei Qin
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| | - Ning Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Orthopedics, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
209
|
Zhang B, Fan K. Design and application of ferritin-based nanomedicine for targeted cancer therapy. Nanomedicine (Lond) 2025; 20:481-500. [PMID: 39895329 PMCID: PMC11875477 DOI: 10.1080/17435889.2025.2459056] [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: 10/15/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
Owing to its unique structure and favorable biocompatibility, ferritin has been widely studied as a promising drug carrier over the past two decades. Since the identification of its inherent tumor-targeting property due to unique recognition ablity of the transferrin receptor 1 (TfR1), ferritin-based nanomedicine has attracted widespread attention and triggered a research surge in the field of targeted cancer therapy. Along with progress in structure studies and modification technology, diverse strategies have been carried out to equip ferritin with on-demand functions, further improving the antitumor efficacy and in vivo safety of ferritin-based cancer therapy. In this review, we highlight the structure-based rational design of ferritin and summarize the design strategies in detail from two main perspectives: multifunctional modification and drug loading. In particular, the critical issues that need attention in the design are discussed in depth. Furthermore, we provide an overview of the latest advances in the application of ferritin-based nanomedicines in chemotherapy, phototherapy and immunotherapy, with particular emphasis on emerging therapeutic approaches among these therapies.
Collapse
Affiliation(s)
- Baoli Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| |
Collapse
|
210
|
Tu T, Hsu Y, Yang C, Shyong Y, Kuo C, Liu Y, Shih S, Lin C. Variations in ECM Topography, Fiber Alignment, Mechanical Stiffness, and Cellular Composition Between Ventral and Dorsal Ligamentum Flavum Layers: Insights Into Hypertrophy Pathogenesis. JOR Spine 2025; 8:e70033. [PMID: 39886656 PMCID: PMC11780719 DOI: 10.1002/jsp2.70033] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Background Previous studies have suggested that changes in the composition of the extracellular matrix (ECM) play a significant role in the development of ligamentum flavum hypertrophy (LFH) and the histological differences between the ventral and dorsal layers of the hypertrophied ligamentum flavum. Although LFH is associated with increased fibrosis in the dorsal layer, comprehensive research exploring the characteristics of the ECM and its mechanical properties in both regions is limited. Furthermore, the distribution of fibrosis-associated myofibroblasts within LFH remains poorly understood. This study aimed to bridge the existing knowledge gap concerning the intricate relationships between ECM characteristics, mechanical properties, and myofibroblast expression in LFH. Methods Histological staining, scanning electron microscopy, and atomic force microscopy were used to analyze the components, alignment, and mechanical properties of the ECM. Immunostaining and western blot analyses were performed to assess the distribution of myofibroblasts in LF tissues. Results There were notable differences between the dorsal and ventral layers of the hypertrophic ligamentum flavum. Specifically, the dorsal layer exhibited higher collagen content and disorganized fibrous alignment, resulting in reduced stiffness. Immunohistochemistry analysis revealed a significantly greater presence of α-smooth muscle actin (αSMA)-stained cells, a marker for myofibroblasts, in the dorsal layer. Conclusions This study offers comprehensive insights into LFH by elucidating the distinctive ECM characteristics, mechanical properties, and cellular composition disparities between the ventral and dorsal layers. These findings significantly enhance our understanding of the pathogenesis of LFH and may inform future research and therapeutic strategies.
Collapse
Affiliation(s)
- Ting‐Yuan Tu
- Department of Biomedical Engineering, College of EngineeringNational Cheng Kung UniversityTainanTaiwan
- Medical Device Innovation CenterNational Cheng Kung UniversityTainanTaiwan
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan
| | - Yu‐Chia Hsu
- Department of Orthopedic Surgery, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Chia‐En Yang
- Department of Biomedical Engineering, College of EngineeringNational Cheng Kung UniversityTainanTaiwan
| | - Yan‐Jye Shyong
- Department of Clinical Pharmacy and Pharmaceutical SciencesNational Cheng Kung UniversityTainanTaiwan
| | - Cheng‐Hsiang Kuo
- International Center for Wound Repair and RegenerationNational Cheng Kung UniversityTainanTaiwan
- Department of Biochemistry and Molecular Biology, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Yuan‐Fu Liu
- Department of Orthopedic Surgery, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Shu‐Shien Shih
- Medical Device Innovation CenterNational Cheng Kung UniversityTainanTaiwan
| | - Cheng‐Li Lin
- Medical Device Innovation CenterNational Cheng Kung UniversityTainanTaiwan
- Department of Orthopedic Surgery, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Musculoskeletal Research Center, Innovation HeadquartersNational Cheng Kung UniversityTainanTaiwan
- Skeleton Materials and Bio‐Compatibility Core Lab, Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
211
|
Zhang Y, Huang Q, Lei F, Qian W, Zhang C, Wang Q, Liu C, Ji H, Wang F. Exploring New Bioorthogonal Catalysts: Scaffold Diversity in Catalysis for Chemical Biology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404431. [PMID: 39921286 PMCID: PMC11884534 DOI: 10.1002/advs.202404431] [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: 04/25/2024] [Revised: 01/11/2025] [Indexed: 02/10/2025]
Abstract
Bioorthogonal catalysis has revolutionized the field of chemical biology by enabling selective and controlled chemical transformations within living systems. Research has converged on the development of innovative catalyst scaffolds, seeking to broaden the scope of bioorthogonal reactions, boost their efficiency, and surpass the limitations of conventional catalysts. This review provides a comprehensive overview of the latest advancements in bioorthogonal catalyst research based on different scaffold materials. Through an in-depth analysis of fabrication strategies and applications of bioorthogonal catalysts, this review discusses the design principles, mechanisms of action, and applications of these novel catalysts in chemical biology. Current challenges and future directions in exploring the scaffold diversity are also highlighted. The integration of diverse catalyst scaffolds offers exciting prospects for precise manipulation of biomolecules and the development of innovative therapeutic strategies in chemical biology. In addition, the review fills in the gaps in previous reviews, such as in fully summarizing the presented scaffold materials applied in bioorthogonal catalysts, emphasizing the potential impact on advancing bioorthogonal chemistry, and offering prospects for future development in this field.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Qizhen Huang
- School of Public HealthNantong UniversityNantong226019China
| | - Fang Lei
- School of Public HealthNantong UniversityNantong226019China
| | - Wanlong Qian
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Chengfeng Zhang
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Qi Wang
- School of Public HealthNantong UniversityNantong226019China
| | - Chaoqun Liu
- School of PharmacyHenan UniversityKaifeng475004China
| | - Haiwei Ji
- School of Public HealthNantong UniversityNantong226019China
| | - Faming Wang
- School of Public HealthNantong UniversityNantong226019China
| |
Collapse
|
212
|
Wan X, Yao H, Wei Z, Gao D, Zheng D, Xu B, Xie M. Heterogeneous porous hypoxia-mimicking scaffolds propel urethral reconstruction by promoting angiogenesis and regulating inflammation. Biomaterials 2025; 314:122833. [PMID: 39277947 DOI: 10.1016/j.biomaterials.2024.122833] [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: 04/21/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
The nasty urine microenvironment (UME) impedes neourethral regeneration by inhibiting angiogenesis and inducing an excessive inflammatory response. Cellular adaptation to hypoxia improves regeneration in numerous tissues. In this study, heterogeneous porous hypoxia-mimicking scaffolds were fabricated for urethral reconstruction via promoting angiogenesis and modulating the inflammatory response based on sustained release of dimethyloxalylglycine (DMOG) to promote HIF-1α stabilization. Such scaffolds exhibit a two-layered structure: a dense layer composed of electrospun poly (l-lactic acid) (PLLA) nanofibrous mats and a loose layer composed of a porous gelatin matrix incorporated with DMOG-loaded mesoporous silica nanoparticles (DMSNs) and coated with poly(glycerol sebacate) (PGS). The modification of PGS could significantly increase rupture elongation, making the composite scaffolds more suitable for urethral tissue regeneration. Additionally, sustained release of DMOG from the scaffold facilitates proliferation, migration, tube formation, and angiogenetic gene expression in human umbilical vein endothelial cells (HUVECs), as well as stimulates M2 macrophage polarization and its regulation of HUVECs migration and smooth muscle cell (SMCs) contractile phenotype. These effects were downstream of the stabilization of HIF-1α in HUVECs and macrophages under hypoxia-mimicking conditions. Furthermore, the scaffold achieved better urethral reconstruction in a rabbit urethral stricture model, including an unobstructed urethra with a larger urethral diameter, increased regeneration of urothelial cells, SMCs, and neovascularization. Our results indicate that heterogeneous porous hypoxia-mimicking scaffolds could promote urethral reconstruction via facilitating angiogenesis and modulating inflammatory response.
Collapse
Affiliation(s)
- Xiang Wan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Haijun Yao
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ziwei Wei
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Dajun Gao
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Dachao Zheng
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Minkai Xie
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
213
|
Lin H, Gao Y, Zhu L, Guo Y, Zhang L, Xie J, Yang D, Liu J, Dong Q, Zhu Z. Rational Design of Single‐Atom Nanozymes for Combination Cancer Immunotherapy. ADVANCED FUNCTIONAL MATERIALS 2025; 35. [DOI: 10.1002/adfm.202416563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Indexed: 02/03/2025]
Abstract
AbstractRemodeling of the tumor immune microenvironment and enhancement of antitumor immune responses are necessary to overcome immunotherapy resistance in tumors. However, tumor heterogeneity and complexity of immune evasion mechanisms pose significant therapeutic challenges. Nanozymes exhibit enzyme‐like characteristics and unique nanomaterial properties, showing potential for tumor therapy. However, design of effective nanozymes remains complex, inefficient, and functionally limited. Therefore, in this study, a novel strategy combining rationally designed single‐atom nanozymes (SAzymes) with immune checkpoint blockade (ICB) therapy is established. Molybdenum SAzymes supported on graphitic carbon nitride (Mo SAs) are constructed using 25 transition metal candidates from the 4th to 6th periods based on high‐throughput calculations and optimal piezoelectric‐enhanced multienzyme‐like activities. Upon activation by ultrasound, Mo SAs exerted potent therapeutic effects against ICB‐resistant tumors and remodeled the tumor immune microenvironment by inducing tumor immunogenic cell death, alleviating tumor hypoxia, and modulating chemokine expression in tumors. Combination of Mo SAs with anti‐programmed death protein‐1 antibodies further enhanced their antitumor efficacy, highlighting their potential to treat ICB‐resistant tumors.
Collapse
Affiliation(s)
- Hanchao Lin
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Yonghui Gao
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Le Zhu
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
| | - Yu Guo
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Lumin Zhang
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
| | - Jiali Xie
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Dongqin Yang
- Department of Laboratory Medicine Huashan Hospital Fudan University 12 Middle Urumqi Road Shanghai 200040 China
| | - Jing Liu
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Qiongzhu Dong
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Zhiling Zhu
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
- Key Laboratory of Optic‐electric Sensing and Analytical Chemistry for Life Science MOE; Shandong Key Laboratory of Biochemical Analysis Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| |
Collapse
|
214
|
Zhang J, Jiao D, Qi X, Zhang Y, Liu X, Pan T, Gao H, Liu Z, Ding D, Feng G. An Albumin-Photosensitizer Supramolecular Assembly with Type I ROS-Induced Multifaceted Tumor Cell Deaths for Photodynamic Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410405. [PMID: 39804949 PMCID: PMC11884554 DOI: 10.1002/advs.202410405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/19/2024] [Indexed: 01/16/2025]
Abstract
Photodynamic therapy holds great potentials in cancer treatment, yet its effectiveness in hypoxic solid tumor is limited by the oxygen-dependence and insufficient oxidative potential of conventional type II reactive oxygen species (ROS). Herein, the study reports a supramolecular photosensitizer, BSA@TPE-BT-SCT NPs, through encapsulating aggregation-enhanced emission photosensitizer by bovine serum albumin (BSA) to significantly enhance ROS, particularly less oxygen-dependent type I ROS for photodynamic immunotherapy. The abundant type I ROS generated by BSA@TPE-BT-SCT NPs induce multiple forms of programmed cell death, including apoptosis, pyroptosis, and ferroptosis. These multifaceted cell deaths synergistically facilitate the release of damage-associated molecular patterns and antitumor cytokines, thereby provoking robust antitumor immunity. Both in vitro and in vivo experiments confirmed that BSA@TPE-BT-SCT NPs elicited the immunogenic cell death, enhance dendritic cell maturation, activate T cell, and reduce myeloid-derived suppressor cells, leading to the inhibition of both primary and distant tumors. Additionally, BSA@TPE-BT-SCP NPs also exhibited excellent antitumor performance in a humanized mice model, evidenced by a reduction in senescent T cells among these activated T cells. The findings advance the development of robust type I photosensitizers and unveil the important role of type I ROS in enhancing multifaceted tumor cell deaths and antitumor immunogenicity.
Collapse
Affiliation(s)
- Jingtian Zhang
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Di Jiao
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Xinwen Qi
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Yufan Zhang
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Xiaoang Liu
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Tengwu Pan
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Heqi Gao
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Zhaoyun Liu
- Department of HematologyTianjin Medical University General HospitalTianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone ControlTianjin Institute of HematologyTianjin300052China
| | - Dan Ding
- Frontiers Science Center for Cell ResponsesState Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of Educationand College of Life SciencesNankai UniversityTianjin300071China
| | - Guangxue Feng
- Guangdong Provincial Key Laboratory of Luminescence from Molecular AggregatesState Key Laboratory of Luminescent Materials and DevicesSchool of Materials Science and EngineeringAIE InstituteSouth China University of TechnologyGuangzhou510640China
| |
Collapse
|
215
|
Chen L, Yin Y, Li J, Li Q, Zhu Z, Li J. LINC00525 promotes cell proliferation and collagen expression through feedforward regulation of TGF-β signaling in hypertrophic scar fibroblasts. Burns 2025; 51:107353. [PMID: 39740483 DOI: 10.1016/j.burns.2024.107353] [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: 09/02/2024] [Revised: 11/24/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
The etiology of hypertrophic scar formation continues to elude researchers, despite advancements in the understanding of skin scarring. Several long non-coding RNAs (lncRNAs) have been implicated in the pathogenesis of hypertrophic scars, yet the role and molecular mechanisms of LINC00525 in this process remain unclear. This study demonstrates that LINC00525 enhances cell proliferation and collagen expression through knockdown and overexpression techniques. Further analysis, including nuclear and cytoplasmic localization studies, RNA pull-down assays, bioinformatics predictions, and PCR validation, reveals that LINC00525 interacts with miR-29a-5p. The downregulation of LINC00525 enhances the expression of miR-29a-5p and suppresses the TGF-β/Smad signaling pathway. Additionally, TGF-β1 induces the upregulation of LINC00525. Collectively, these findings indicate that LINC00525 operates through a feedforward mechanism to regulate TGF-β signaling in hypertrophic scar fibroblasts. This research offers novel insights for the prevention and treatment of scars.
Collapse
Affiliation(s)
- Ling Chen
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China
| | - Yiliang Yin
- Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China
| | - Jingyun Li
- Nanjing Women and Children's Healthcare Institute, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China
| | - Qian Li
- Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Jun Li
- Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), Nanjing 210004, China.
| |
Collapse
|
216
|
Luo Q, Li Z, Sun W, Wang G, Yao H, Wang G, Liu B, Ding J. Myocardia-Injected Synergistically Anti-Apoptotic and Anti-Inflammatory Poly(amino acid) Hydrogel Relieves Ischemia-Reperfusion Injury. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2420171. [PMID: 39906023 DOI: 10.1002/adma.202420171] [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: 12/21/2024] [Indexed: 02/06/2025]
Abstract
Reperfusion therapy is the most effective treatment for acute myocardial infarction, but its efficacy is frequently limited by ischemia-reperfusion injury (IRI). While antioxidant and anti-inflammatory therapies have shown significant potential in alleviating IRI, these strategies have not yielded satisfactory clinical outcomes. For that, a thermo-sensitive myocardial-injectable poly(amino acid) hydrogel of methoxy poly(ethylene glycol)45-poly(L-methionine20-co-L-alanine10) (mPEG45-P(Met20-co-Ala10), PMA) loaded with FTY720 (PMA/FTY720) is developed to address IRI through synergistic anti-apoptotic and anti-inflammatory effects. Upon injection into the ischemic myocardium, the PMA aqueous solution undergoes a sol-to-gel phase transition and gradually degrades in response to reactive oxygen species (ROS), releasing FTY720 on demand. PMA acts synergistically with FTY720 to inhibit cardiomyocyte apoptosis and modulate pro-inflammatory M1 macrophage polarization toward anti-inflammatory M2 macrophages by clearing ROS, thereby mitigating the inflammatory response and promoting vascular regeneration. In a rat IRI model, PMA/FTY720 reduces the apoptotic cell ratio by 81.8%, increases vascular density by 34.0%, and enhances left ventricular ejection fraction (LVEF) by 12.8%. In a rabbit IRI model, the gel-based sustained release of FTY720 enhanced LVEF by an additional 7.2% compared to individual treatment. In summary, the engineered PMA hydrogel effectively alleviates IRI through synergistic anti-apoptosis and anti-inflammation actions, offering valuable clinical potential for treating myocardial IRI.
Collapse
Affiliation(s)
- Qiang Luo
- Department of Cardiology, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun, 130041, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun, 130041, P. R. China
| | - Wei Sun
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun, 130041, P. R. China
| | - Guoliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Haochen Yao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Basic Medicine, Jilin University, 126 Xinmin Street, Changchun, 130061, P. R. China
| | - Guoqing Wang
- Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Basic Medicine, Jilin University, 126 Xinmin Street, Changchun, 130061, P. R. China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun, 130041, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, P. R. China
| |
Collapse
|
217
|
Sun Y, Li Y, Ding X, Xu P, Jing X, Cong H, Hu H, Yu B, Xu FJ. An NIR-responsive hydrogel loaded with polydeoxyribonucleotide nano-vectors for enhanced chronic wound healing. Biomaterials 2025; 314:122789. [PMID: 39260030 DOI: 10.1016/j.biomaterials.2024.122789] [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: 06/07/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
Chronic diabetic wounds are difficult to treat due to imbalanced inflammatory responses, high blood glucose levels, and bacterial infections. Novel therapeutic approaches based on nucleic acid analogues have been proposed, with unique advantages in improving angiogenesis, increasing collagen synthesis, and exerting anti-inflammatory effects. However, the inherent electronegativity of nucleic acids makes them less susceptible to cellular uptake. In this paper, a kind of near infrared (NIR)-responsive nanocomposite hydrogel loaded with nucleic acid vectors was proposed for promoting wound healing. The redox system composed of molybdenum disulphide nanosheets (MoS2 NSs) initiated the copolymerization of quaternized chitosan containing double bonds and N-isopropylacrylamide (NIPAAm) to form the matrix. In addition, MoS2 NSs with photothermal conversion performance endow the nanocomposite hydrogel to have NIR-response property and act as physical crosslinking points in the matrix. Polydeoxyribonucleotides (PDRN), which have the effect of promoting wound healing, were made into nucleic acid vectors, and loaded into the NIR-responsive hydrogel. MoS2 NSs can convert NIR irradiation into heat, causing phase transitions of temperature-sensitive segments that trigger volume contraction of the hydrogel to extrude the nucleic acid vector. Promoting angiogenesis, slowing inflammation, and guiding tissue regeneration were demonstrated in the diabetic wound model treated with the NIR-responsive nanocomposite hydrogel.
Collapse
Affiliation(s)
- Yanzhen Sun
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Yao Li
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Xiaokang Ding
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Pan Xu
- Department of Respiration, Binzhou Medical University Hospital, Binzhou, 256500, China
| | - Xiaodong Jing
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China.
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China.
| | - Fu-Jian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China.
| |
Collapse
|
218
|
Zhao Y, Cheng J, Li Z, Wang J, Chen X. Nanozymes in Biomedical Applications: Innovations Originated From Metal-Organic Frameworks. Adv Healthc Mater 2025; 14:e2402066. [PMID: 39319491 DOI: 10.1002/adhm.202402066] [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: 06/05/2024] [Revised: 08/18/2024] [Indexed: 09/26/2024]
Abstract
Nanozymes exhibit significant potential in medical theranostics, environmental protection, energy development, and biopharmaceuticals due to their exceptional catalytic performance. Compared with natural enzymes, nanozymes have the advantages of simple preparation and purification, convenient production and low cost. Therefore, it is very important to prepare nanozymes quickly and efficiently, which not only helps to expand their application scope, but also can further exert their great potential in various fields. Metal-organic frameworks (MOF) materials serve as versatile substrates for constructing nanozymes, offering unique advantages like adjustable structure, high specific surface area, and porous channels. MOF coordination nodes constructed from metal ions or metal clusters have unique properties that can be leveraged to tailor nanozyme characteristics for different applications. This review describes and analyzes recent methods for constructing nanozymes using MOF materials, and explores their application prospects in biomedicine. By expounding the preparation techniques and biomedical applications of nanozymes, this review aims to inspire researchers to develop innovative nanozyme materials and explore new application directions.
Collapse
Affiliation(s)
- Yuewu Zhao
- College of Pharmacy, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou, 253023, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Junjie Cheng
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Zhen Li
- College of Pharmacy, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou, 253023, China
| | - Jine Wang
- College of Pharmacy, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou, 253023, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
| |
Collapse
|
219
|
Jiang C, Miao T, Xing X, Schilling KJ, Lenhard N, Wang L, McDowell S, Nilsson BL, Wang H, Zhang X. Masquelet Inspired in Vivo Engineered Extracellular Matrix as Functional Periosteum for Bone Defect Repair and Reconstruction. Adv Healthc Mater 2025; 14:e2404975. [PMID: 39840608 PMCID: PMC11913577 DOI: 10.1002/adhm.202404975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/09/2025] [Indexed: 01/23/2025]
Abstract
The Masquelet technique that combines a foreign body reaction (FBR)-induced vascularized tissue membrane with staged bone grafting for reconstruction of segmental bone defect has gained wide attention in Orthopedic surgery. The success of Masquelet hinges on its ability to promote formation of a "periosteum-like" FBR-induced membrane at the bone defect site. Inspired by Masquelet's technique, here a novel approach is devised to create periosteum mimetics from decellularized extracellular matrix (dECM), engineered in vivo through FBR, for reconstruction of segmental bone defects. The approach involved 3D printing of polylactic acid (PLA) template with desired pattern/architecture, followed by subcutaneous implantation of the template to form tissue, and depolymerization and decellularization to generate dECM with interconnected channels. The dECM matrices produces from the same mice (autologous) or from different mice (allogenic) are used as a functional periosteum for repair of structural bone allograft in a murine segmental bone defect model. This study shows that autologous dECM performed better than allogenic dECM, further permitting local delivery of low dose BMP-2 to enhance allograft incorporation. The success of this current approach can establish a new line of versatile, patient-specific, and periosteum-like autologous dECM for bone regeneration, offering personalized therapeutics to patients with impaired healing.
Collapse
Affiliation(s)
- Chen Jiang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Tianfeng Miao
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Xiaojie Xing
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Kevin J Schilling
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Nicholas Lenhard
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Lichen Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Susan McDowell
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, NY, 14627, USA
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| |
Collapse
|
220
|
Roointan A, Xu R, Corrie S, Hagemeyer CE, Alt K. Nanotherapeutics in Kidney Disease: Innovations, Challenges, and Future Directions. J Am Soc Nephrol 2025; 36:500-518. [PMID: 39705082 PMCID: PMC11888965 DOI: 10.1681/asn.0000000608] [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: 08/28/2024] [Accepted: 12/17/2024] [Indexed: 12/22/2024] Open
Abstract
The treatment and management of kidney diseases present a significant global challenge, affecting over 800 million individuals and necessitating innovative therapeutic strategies that transcend symptomatic relief. The application of nanotechnology to therapies for kidney diseases, while still in its early stages, holds transformative potential for improving treatment outcomes. Recent advancements in nanoparticle-based drug delivery leverage the unique physicochemical properties of nanoparticles for targeted and controlled therapeutic delivery to the kidneys. Current research is focused on understanding the functional and phenotypic changes in kidney cells during both acute and chronic conditions, allowing for the identification of optimal target cells. In addition, the development of tailored nanomedicines enhances their retention and binding to key renal membranes and cell populations, ultimately improving localization, tolerability, and efficacy. However, significant barriers remain, including inconsistent nanoparticle synthesis and the complexity of kidney-specific targeting. To overcome these challenges, the field requires advanced synthesis techniques, refined targeting strategies, and the establishment of animal models that accurately reflect human kidney diseases. These efforts are critical for the clinical application of nanotherapeutics, which promise novel solutions for kidney disease management. This review evaluates a substantial body of in vivo research, highlighting the prospects, challenges, and opportunities presented by nanotechnology-mediated therapies and their potential to transform kidney disease treatment.
Collapse
Affiliation(s)
- Amir Roointan
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- NanoTheranostics Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Rong Xu
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Simon Corrie
- Department of Chemical and Biological Engineering, Monash University, Melbourne, Victoria, Australia
| | - Christoph E. Hagemeyer
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Karen Alt
- NanoTheranostics Laboratory, Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
221
|
Bi X, Wang Z, He J. Recent advances in biomimetic nanodelivery systems for the treatment of myocardial ischemia reperfusion injury. Colloids Surf B Biointerfaces 2025; 247:114414. [PMID: 39626610 DOI: 10.1016/j.colsurfb.2024.114414] [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: 11/09/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025]
Abstract
Myocardial ischemia/reperfusion injury (MIRI) is a significant challenge in the treatment of myocardial infarction, a leading cause of global mortality due to irreversible cardiac damage. Biomimetic nanodelivery systems offer promising therapeutic strategies to address MIRI. In this review, we comprehensively investigate the underlying pathophysiological mechanisms of MIRI and discuss recent advances in biomimetic nanodelivery systems including cell membrane-coated nanoparticles, exosomes, and nanoenzymes as innovative approaches for MIRI treatment. We emphasize the advantages and potential of biomimetic strategies in enhancing therapeutic efficacy, assess the preclinical effectiveness of these nanodelivery systems, and discuss the challenges associated with translating these approaches into clinical practice. This paper aims to provide new perspectives on biomimetic strategies for MIRI treatment, contributing to the development of effective drug delivery systems.
Collapse
Affiliation(s)
- Xiaojun Bi
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Ze Wang
- Dalian Medical University, Liaoning 116044, China
| | - Jingteng He
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
222
|
Mai Y, Wu S, Zhang P, Chen N, Wu J, Wei F. The anti-oxidation related bioactive materials for intervertebral disc degeneration regeneration and repair. Bioact Mater 2025; 45:19-40. [PMID: 39588482 PMCID: PMC11585838 DOI: 10.1016/j.bioactmat.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 11/27/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent chronic spinal condition characterized by the deterioration of the intervertebral discs (IVD), leading to structural damage and associated pain. This degenerative process is closely linked to oxidative stress injury, which plays a pivotal role in its onset and progression. Oxidative stress in IVDD results from the excessive production of reactive oxygen species (ROS) and impaired ROS clearance mechanisms, disrupting the redox balance within the intervertebral disc. Consequently, oxidative stress contributes to the degradation of the extracellular matrix (ECM), promotes cell apoptosis, and exacerbates disc tissue damage. Current treatment options for IVDD face significant challenges in effectively alleviating the oxidative stress-induced damage and facilitating disc tissue repair. However, recent advancements in biomaterials have opened new avenues of hope for IVDD treatment by addressing oxidative stress. In this review, we first provide an overview of the pathophysiological process of IVDD and explore the mechanisms and pathways associated with oxidative stress injury. Then, we delve into the current research on antioxidant biomaterials employed in the treatment of IVDD, and outline the advantages and limitations of hydrogel, nanomaterials, polyphenol and inorganic materials. Finally, we propose the future research direction of antioxidant biomaterials in IVDD treatment. The main idea of this review is shown in Scheme 1.
Collapse
Affiliation(s)
- Yingjie Mai
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Siying Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science & Technology (Guangzhou), Nansha, Guangzhou, Guangdong Province, 511400, China
| | - Penghui Zhang
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Ningning Chen
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science & Technology (Guangzhou), Nansha, Guangzhou, Guangdong Province, 511400, China
- Division of Life Science, The Hong Kong University of Science & Technology, Hong Kong SAR, 999077, China
| | - Fuxin Wei
- Department of Orthopaedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, 518107, China
| |
Collapse
|
223
|
Auguet-Lara M, Skrivergaard S, Therkildsen M, Rasmussen MK, Young JF. Development of a biomarker panel for cell characterization intended for cultivated meat. Exp Cell Res 2025; 446:114467. [PMID: 39978714 DOI: 10.1016/j.yexcr.2025.114467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Cultivated meat has in recent years been suggested as a sustainable alternative to produce meat at large-scale. Several aspects of cultivated meat production have demonstrated significant progress. However, there are still many questions regarding the cell culture, media composition, and the production itself to be answered and optimized. Finding good starter cell populations is a challenge to address and requires robust tools to characterize the cell populations. Detailed analysis is required to identify each type of cell within the skeletal muscle niche leads to optimized cultivated meat production at large-scale. In this study, we developed a set of biomarkers, using digital droplet PCR (ddPCR) and Immunofluorescence (IF) staining, to identify specific cell types within a heterogeneous cell population isolated from skeletal muscle tissue. We showed that combining Neural Cell Adhesion Molecule (NCAM), Calponin 1 (CNN1), and Fibronectin (FN), can be a powerful approach to predict the growth of skeletal myotubes, smooth muscle mesenchymal cells (SMMCs), and myofibroblasts, respectively. Moreover, early cell-cell interactions of fibroblastic cells were observed to be triggered through thin actin filaments containing CNN1 protein, to form, subsequently, myofibroblast networks. Besides, Myogenic Differentiation 1 (MyoD) is the key marker to detect skeletal muscle growth, whereas Myogenic Factor 5 (MyF5) can be expressed in myogenic and non-myogenic cells. MyF5 was detected at differentiation stages within the myotube nuclei, suggesting an unknown role during myotube formation.
Collapse
|
224
|
Zong X, Xu X, Pang DW, Huang X, Liu AA. Fine-Tuning Electron Transfer for Nanozyme Design. Adv Healthc Mater 2025; 14:e2401836. [PMID: 39015050 DOI: 10.1002/adhm.202401836] [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: 05/17/2024] [Revised: 06/22/2024] [Indexed: 07/18/2024]
Abstract
Nanozymes, with their versatile composition and structural adaptability, present distinct advantages over natural enzymes including heightened stability, customizable catalytic activity, cost-effectiveness, and simplified synthesis process, making them as promising alternatives in various applications. Recent advancements in nanozyme research have shifted focus from serendipitous discovery toward a more systematic approach, leveraging machine learning, theoretical calculations, and mechanistic explorations to engineer nanomaterial structures with tailored catalytic functions. Despite its pivotal role, electron transfer, a fundamental process in catalysis, has often been overlooked in previous reviews. This review comprehensively summarizes recent strategies for modulating electron transfer processes to fine-tune the catalytic activity and specificity of nanozymes, including electron-hole separation and carrier transfer. Furthermore, the bioapplications of these engineered nanozymes, including antimicrobial treatments, cancer therapy, and biosensing are also introduced. Ultimately, this review aims to offer invaluable insights for the design and synthesis of nanozymes with enhanced performance, thereby advancing the field of nanozyme research.
Collapse
Affiliation(s)
- Xia Zong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Xinran Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, and Frontiers Science Center for Cell Responses, Nankai University, Tianjin, 300071, P. R. China
| | - An-An Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
225
|
Xu DJ, Wang GT, Zhong Q. Extracellular matrix gene set and microRNA network in intestinal ischemia-reperfusion injury: Insights from RNA sequencing for diagnosis and therapy. World J Gastrointest Surg 2025; 17:100034. [DOI: 10.4240/wjgs.v17.i2.100034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
Intestinal ischemia-reperfusion injury (IIRI) is a complex and severe pathophysiological process characterized by oxidative stress, inflammation, and apoptosis. In recent years, the critical roles of extracellular matrix (ECM) genes and microRNAs (miRNAs) in IIRI have garnered widespread attention. This review aims to systematically summarize the diagnostic and therapeutic potential of ECM gene sets and miRNA regulatory networks in IIRI. First, we review the molecular mechanisms of IIRI, focusing on the dual role of the ECM in tissue injury and repair processes. The expression changes and functions of ECM components such as collagen, elastin, and matrix metalloproteinases during IIRI progression are deeply analyzed. Second, we systematically summarize the regulatory roles of miRNAs in IIRI, particularly the mechanisms and functions of miRNAs such as miR-125b and miR-200a in regulating inflammation, apoptosis, and ECM remodeling. Additionally, this review discusses potential diagnostic biomarkers and treatment strategies based on ECM genes and miRNAs. We extensively evaluate the prospects of miRNA-targeted therapy and ECM component modulation in preventing and treating IIRI, emphasizing the clinical translational potential of these emerging therapies. In conclusion, the diagnostic and therapeutic potential of ECM gene sets and miRNA regulatory networks in IIRI provides new directions for further research, necessitating additional clinical and basic studies to validate and expand these findings for improving clinical outcomes in IIRI patients.
Collapse
Affiliation(s)
- Dao-Jian Xu
- Department of Emergency Medicine, Taizhou Municipal Hospital, Taizhou 318000, Zhejiang Province, China
| | - Guo-Tao Wang
- Department of Emergency Medicine, Taizhou Municipal Hospital, Taizhou 318000, Zhejiang Province, China
| | - Qiang Zhong
- Department of Emergency Medicine, Taizhou Municipal Hospital, Taizhou 318000, Zhejiang Province, China
| |
Collapse
|
226
|
Cao T, Yuan W, Gao Y, Zou X, Tian B, Shi M, Feng W, Li F. Stealth Nanoparticles with a "Self-Consuming" Shell for Long-Term Blood Vessel Imaging. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11811-11819. [PMID: 39957207 DOI: 10.1021/acsami.4c20986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
The development of lanthanide-doped upconversion nanoparticle (UCNP)-based imaging with minimal autofluorescence and improved penetration depth is important in medical applications. Exogenous nanocarriers readily adsorb plasma proteins following intravenous administration (<0.5 min), resulting in the formation of a protein corona on the fixed surface. The protein corona facilitates UCNP interception by the immune system, preventing targeted delivery to disease sites. In this study, we report a novel surface-camouflaging strategy using lanthanide hydroxyl carbonate that is slowly dissolved by physiological phosphate in serum. The "self-consuming" inorganic-shell-modified UCNPs (denoted as UCSP-PEG) effectively reduce protein corona adhesion by more than 90% through a dissociation effect associated with the amphiphilic poly(ethylene glycol) (PEG)-modified UCNPs as determined in an ex vivo assay. The UCSP-PEG exhibits a prolonged blood circulation time (t1/2 = 73.9 ± 9.5 min), 185 times that of camouflage materials without the "stealth" feature, and can employ upconverted luminescence (UCL) imaging to monitor tumor-related blood vessels for at least 120 min. Based on the superior optical properties of UCSP-PEG, the application of a UCL dual-channel stereoscope magnification imaging system has enabled the observation of capillaries with high resolution, offering a powerful tool for monitoring biological activities at the fine tissue level. This work provides a novel "stealth" nanovehicle, resisting blood protein adhesion based on a "self-consuming" effect that can significantly advance tissue imaging and target-specific cancer diagnosis.
Collapse
Affiliation(s)
- Tianye Cao
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Wei Yuan
- Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, Institute of Optoelectronics, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Yilin Gao
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Xianmei Zou
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Bo Tian
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Mei Shi
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Wei Feng
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| | - Fuyou Li
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200438, P. R. China
| |
Collapse
|
227
|
Hyun J, Lee SY, An J, Lee YB, Bhang SH. Strengthening the cellular function of dermal fibroblasts and dermal papilla cells using nanovesicles extracted from stem cells using blue light-based photobiomodulation technology. Biomater Sci 2025; 13:1209-1221. [PMID: 39902823 DOI: 10.1039/d4bm01591f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Human dermal fibroblasts (hDFs) play a critical role in skin health by producing extracellular matrix (ECM) components essential for structural stability, while hair follicle dermal papilla cells (HFDPCs) are key to hair follicle growth and regeneration. However, factors such as UV radiation, oxidative stress, and aging impair the functions of hDFs and HFDPCs, leading to decrement in ECM production and skin maintenance and hair loss conditions like alopecia. Recent advances in nanovesicles (NVs) derived from human adipose-derived stem cells (hADSCs) have shown an innovative way in the regenerative medicine field, particularly with promise for enhancing the functionality of diverse cell types. NVs, filled with diverse bioactive molecules, are non-immunogenic, biologically stable, and capable of promoting cellular activities. To further enhance the therapeutic potential of NVs, photobiomodulation (PBM) using blue light has emerged as a promising application. Optimized blue light irradiation can induce moderate levels of reactive oxygen species production in hADSCs, activating signaling pathways that upregulate angiogenic and regenerative markers in hADSCs. In this study, blue light-irradiated NVs demonstrated superior efficacy in promoting hDF proliferation, ECM synthesis, and the functionality of HFDPCs, resulting in enhanced skin maintenance and hair follicle regeneration. This approach presents a safer and more efficient way for treating skin and hair disorders, highlighting the potential use of blue light-irradiated NVs as an innovative therapeutic strategy.
Collapse
Affiliation(s)
- Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Sang Yoon Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jiseon An
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - You Bin Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
228
|
Chen X, Zhang J, Lin T, Zhou F, Li F, Xue T, Zhong Q, Lee W, Chen G, Wang H, Ju E, Li M, Tao Y. Bioactive Decellularized Extracellular Matrix Platform Integrating Multifunctional Nanozymes and Cell-Laden Microgels for Acute Liver Failure Treatment. ACS NANO 2025; 19:6890-6910. [PMID: 39950852 DOI: 10.1021/acsnano.4c13709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Mesenchymal stem cell (MSC) therapy has emerged as a promising alternative approach for treating acute liver failure (ALF) while confronting the shortage of low efficiency and poor engraftment within a hostile liver milieu. In this study, we establish a bioactive decellularized extracellular matrix (dECM) platform that incorporates dihydrolipoic acid (DHLA)-protected Pt nanoclusters doped with Cu (PtCu-DHLA) nanozymes and cell-laden microgels. The PtCu-DHLA nanozymes, selected for their versatility, function as antioxidant, anti-inflammatory, pro-proliferative, and pro-angiogenic agents, enhancing ALF alleviation and providing an optimal microenvironment for MSC transplantation. Additionally, a methacrylic anhydride (MA)-modified porcine liver-derived decellularized extracellular matrix (PLdECM) hydrogel (PLdECMMA) has been developed for the construction of microgels via microfluidic devices. Interferon γ (IFNγ) preconditioned MSCs encapsulated in PLdECMMA microgels exhibit enhanced immunomodulating activity and prolonged survival. PtCu-DHLA nanozymes and cell-laden microgels are codelivered by leveraging the PLdECM hydrogel for orthotopic transplantation. The transplanted dECM platform enables an efficient and successful rescue of CCl4-induced ALF by counteracting oxidative stress, suppressing inflammatory storms, and promoting cellular regeneration. Overall, this study highlights a synergistic and reinforced strategy that combines biomimetic nanozymes with MSC therapy, offering significant potential for ALF treatment and broader applications in regenerative medicine.
Collapse
Affiliation(s)
- Xiaodie Chen
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jiabin Zhang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Tong Lin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Feng Zhou
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Fenfang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Tiantian Xue
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qingguo Zhong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Weijen Lee
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Guipan Chen
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Enguo Ju
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-Sen University, Guangzhou 510275, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
229
|
Wu Y, Meng L, Zhan S, Li M, Huang J, Chen X, Chen L, Gao X, Chen H, Chen H, Zhong Y, Xu L, Xu Y. ITIH5-mediated fibroblast/macrophage crosstalk exacerbates cardiac remodelling after myocardial infarction. J Transl Med 2025; 23:224. [PMID: 39994656 PMCID: PMC11852866 DOI: 10.1186/s12967-025-06244-5] [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: 06/26/2024] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Myocardial infarction (MI) and subsequent ischaemic cardiomyopathy (ICM) are the primary causes of heart failure. Inter-α trypsin inhibitor heavy chain 5 (ITIH5) is an extracellular matrix (ECM) protein and has been identified as a myocardial marker of ICM. However, its diagnostic value in patients with ICM and its function and molecular mechanism in regulating cardiac repair and remodelling after MI remain unknown. METHODS Three microarray datasets including 117 ICM and 152 non-failing (NF) myocardial tissue samples were merged and analysed. Peripheral blood and clinical information were collected from 53 patients with ICM and 40 NF controls. The effects of ITIH5 on cellular interactions and cardiac remodelling was studied using ITIH5 RNAi adeno-associated virus and mouse MI model in vivo and in fibroblast-macrophage co-culture model in vitro. RESULTS ITIH5 was upregulated in the myocardial tissue and peripheral blood of patients with ICM and could be an independent risk factor for ICM. Experiments in mice suggested that ITIH5 promotes cardiac fibrotic remodelling at all phases after MI. Downregulation of ITIH5 increased the risk of death within 7 d after MI but inhibited ventricular remodelling and improved cardiac function on the long-term. ITIH5 promotes the primary cardiac fibroblasts (CFs) proliferation, migration, and improves survival rather than activiation. Morover, ITIH5 directly promotes macrophage tissue infiltration, maturation, and profibrotic phenotype transformation, thereby promoting fibrotic remodelling. By using fibroblast-macrophage co-culture model, we demonstrated ITIH5 enhanced the fibroblast/macrophage crosstalk manifest as macrophage profibrotic phenotype transformation and CFs activation, mainly by enhancing the hyaluronan stability, the ability of ITIH5 to bind macrophage CD44 receptors and the downstream activation of the signal transduction and activator of transcription 3 pathway in macrophages. CONCLUSIONS ITIH5 could be used as a diagnostic marker for ICM. Moreover, ITIH5 expression was upregulated after MI, which accelerated ECM-fibroblast-macrophage interaction, thereby promoting macrophage profibrotic phenotype transformation, CFs activation, and cardiac fibrotic remodelling.
Collapse
Affiliation(s)
- Yirong Wu
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
| | - Li Meng
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Siyao Zhan
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
| | - Miaofu Li
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
| | - Jiamin Huang
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xuechun Chen
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
| | - Liuying Chen
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
| | - Xiaofei Gao
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
| | - Hao Chen
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Huimin Chen
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yigang Zhong
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China.
| | - Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China.
- Translational Medicine Research Center, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China.
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People'S Hospital, Westlake University School of Medicine, Zhejiang, 310006, China.
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
230
|
Zhang Y, Yang Y, Yin Z, Huang L, Wang J. Nanozyme-based wearable biosensors for application in healthcare. iScience 2025; 28:111763. [PMID: 39906563 PMCID: PMC11791255 DOI: 10.1016/j.isci.2025.111763] [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] [Indexed: 02/06/2025] Open
Abstract
Recent years have witnessed tremendous advances in wearable sensors, which play an essential role in personalized healthcare for their ability for real-time sensing and detection of human health information. Nanozymes, capable of mimicking the functions of natural enzymes and addressing their limitations, possess unique advantages such as structural stability, low cost, and ease of mass production, making them particularly beneficial for constructing recognition units in wearable biosensors. In this review, we aim to delineate the latest advancements in nanozymes for the development of wearable biosensors, focusing on key developments in nanozyme immobilization strategies, detection technologies, and biomedical applications. The review also highlights the current challenges and future perspectives. Ultimately, it aims to provide insights for future research endeavors in this rapidly evolving area.
Collapse
Affiliation(s)
- Yingcong Zhang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yiran Yang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhixin Yin
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lin Huang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
231
|
Fu L, Ren H, Wang C, Zhao Y, Zou B, Zhang X. Formation of PEG-PLGA Microspheres for Controlled Release of Simvastatin and Carvacrol: Enhanced Lipid-Lowering Efficacy and Improved Patient Compliance in Hyperlipidemia Therapy. Polymers (Basel) 2025; 17:574. [PMID: 40076067 PMCID: PMC11902393 DOI: 10.3390/polym17050574] [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: 01/16/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Polymer-based drug-controlled release systems offer greater efficacy and potency than conventional therapies. However, prominent drug side effects, lower circulation, and low drug loading capabilities limit their application range. In this work, the combination of Simvastatin (SIV) and Carvacrol (CAV) into PEG-PLGA microspheres (SIV-CAV-PP-MS) was achieved via an emulsification-solvent evaporation technique, resulting in microspheres characterized by high encapsulation efficiency and reduced particle size. In vitro studies demonstrated that the cumulative drug release increased with higher SIV and CAV levels in the release medium, reaching 88.91% and 89.35% at 25 days. Pharmacokinetic analysis revealed that the concentrations of SIV and CAV reached their maximum levels at approximately seven days in the SIV-CAV-PP-MS group, which indicates that using PEG-PLGA as a carrier significantly delays drug release. In vivo, evaluation demonstrated that the SIV-CAV-PP-MS high-dose group and positive drug control group showed reductions in low-density lipoprotein cholesterol levels by 0.39-fold and 0.36-fold compared to the Hyperlipidemia model group, and the addition of CAV significantly enhanced the lipid-lowering effects of SIV. Histological examinations indicated that the SIV-CAV-PP-MS medium-dose group displayed histological features more closely resembling those of normal mice compared to the Simvastatin control group, with a well-organized hepatocyte structure, a significant reduction in lipids, and improved liver health. The prepared polymeric microsphere utilizing SIV and SAV will be a promising dosage form for hyperlipidemia disease patients, with superior lipid-lowering efficacy and improved patient compliance.
Collapse
Affiliation(s)
- Lin Fu
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (L.F.); (H.R.); (C.W.); (B.Z.)
| | - Hengxin Ren
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (L.F.); (H.R.); (C.W.); (B.Z.)
| | - Chaoxing Wang
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (L.F.); (H.R.); (C.W.); (B.Z.)
| | - Yaxin Zhao
- College of Heilongjiang, University of Chinese Medicine, Jiamusi 154007, China;
| | - Bohang Zou
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (L.F.); (H.R.); (C.W.); (B.Z.)
| | - Xiangyu Zhang
- College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (L.F.); (H.R.); (C.W.); (B.Z.)
| |
Collapse
|
232
|
Cerveró-Varona A, Prencipe G, Peserico A, Canciello A, House AH, Santos HA, Perugini M, Sulcanese L, Takano C, Miki T, Iannetta A, Russo V, Mattioli M, Barboni B. Amniotic epithelial Cell microvesicles uptake inhibits PBMCs and Jurkat cells activation by inducing mitochondria-dependent apoptosis. iScience 2025; 28:111830. [PMID: 39967871 PMCID: PMC11834128 DOI: 10.1016/j.isci.2025.111830] [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: 08/01/2024] [Revised: 10/15/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Amniotic epithelial cells (AECs) exhibit significant immunomodulatory and pro-regenerative properties, largely due to their intrinsic paracrine functions that are currently harnessed through the collection of their secretomes. While there is increasing evidence of the role of bioactive components freely secreted or carried by exosomes, the bioactive cargo of AEC microvesicles (MVs) and their crosstalk with the immune cells remains to be fully explored. We showed that under intrinsic conditions or in response to LPS, AEC-derived MV carries components such as lipid-mediated signaling molecules, ER, and mitochondria. They foster the intra/interspecific mitochondrial transfer into immune cells (PBMCs and Jurkat cells) in vitro and in vivo on the zebrafish larvae model of injury. The internalization of MV cargoes through macropinocytosis induces hyperpolarization of PBMC mitochondrial membranes and triggers MV-mediated apoptosis. This powerful immune suppressive mechanism triggered by AEC-MV cargo delivery paves the way for controlled and targeted cell-free therapeutic approaches.
Collapse
Affiliation(s)
- Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Andrew H. House
- Helsinki University Lipidomics Unit, Helsinki Institute for Life Science (HiLIFE), Biocenter 3, Viikinkaari 1, 00790 Helsinki, Finland
| | - Hélder A. Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Monia Perugini
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Chika Takano
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Toshio Miki
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Annamaria Iannetta
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Mauro Mattioli
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
233
|
Yang D, Zhou Y, Wang M, Yuan Y, Feng Y, Fang Y, Li G, Liu D, Yao X. Protein Precoating with Concentration-Dependent Manner Breaks through the Biomacromolecular Barrier of Transferrin-Functionalized Nanoparticle in Intestinal Mucosa. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4231-4239. [PMID: 39918287 DOI: 10.1021/acs.jafc.4c08452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Biomacromolecules in physiological environments would adsorb onto the nanoparticles (NPs) to form corona layers, in which protein coronas (PCs) are the major constituent. PCs always play diverse influences on the fate of NPs in vitro and in vivo, especially for active-targeting NPs (e.g., transferrin-modified nanoparticles, Tf-NP). In order to eliminate the inhibition of PCs on the efficiency of Tf-NP, the precoated Tf-NP with bovine serum albumin (BSA, B@Tf-NP) was designed to fabricate an "active PCs" (PCs formed by artificial modification) against the "passive PCs" (PCs formed in the biological environments), which was inspired by the formation pattern of PCs. The results indicated that B@Tf-NP had similar particle size, dispersion, and physical stability with Tf-NP. Surprisingly, B@Tf-NP enhanced the cellular uptake in enterocytes and permeability in intestinal tract of mice. Notably, the concentration ratio of BSA to Tf that could ensure Tf revealed timely during the interacted process was considered to be appropriate. These findings provide an easy while efficient design platform for active-targeting NPs to overcome the biomacromolecular barrier in oral administration.
Collapse
Affiliation(s)
- Dan Yang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
- Xi'an Key Laboratory of Antiviral and Antimicrobial Resistant Bacteria Therapeutics Research, Xi'an 710021, China
| | - Yao Zhou
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
- Xi'an Key Laboratory of Antiviral and Antimicrobial Resistant Bacteria Therapeutics Research, Xi'an 710021, China
| | - Mengqi Wang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
- Xi'an Key Laboratory of Antiviral and Antimicrobial Resistant Bacteria Therapeutics Research, Xi'an 710021, China
| | - Ying Yuan
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Yuqi Feng
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Yapeng Fang
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guoliang Li
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Dechun Liu
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi 710072, China
| | - Xiaolin Yao
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| |
Collapse
|
234
|
Cheng N, Luo Q, Yang Y, Shao N, Nie T, Deng X, Chen J, Zhang S, Huang Y, Hu K, Luo L, Xiao Z. Injectable pH Responsive Conductive Hydrogel for Intelligent Delivery of Metformin and Exosomes to Enhance Cardiac Repair after Myocardial Ischemia-Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2410590. [PMID: 39965141 DOI: 10.1002/advs.202410590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/23/2024] [Indexed: 02/20/2025]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a leading cause of complications and high mortality associated with acute myocardial infarction. Injectable hydrogel emerges as a promising biomaterial for myocardial repair due to their ability to mimic the mechanical and electrophysiological properties of heart tissue. In this study, an injectable conductive hydrogel is developed that responds to the weakly acidic microenvironment of ischemic injury, enabling the intelligent release of metformin and exosomes to enhance cardiac repair following MIRI. This multifunctional hydrogel demonstrates self-healing properties, shear-thinning injectability, electrical conductivity, and an elastic modulus comparable to natural myocardium, alongside excellent biocompatibility. At the cellular level, the hydrogel system exhibits significant antioxidant, anti-apoptotic, improvement of electrophysiological characteristics, mitochondrial protection and angiogenic effects, with transcriptome sequencing revealing the effective activation of the PI3K/AKT, VEGF, and AMPK signaling pathways. In vivo studies further confirm that the hydrogel treatment reduces infarct size, cardiac fibrosis and incidence of arrhythmia, while improving ventricular ejection fraction and facilitating the restoration of cardiac function after MIRI. In conclusion, an injectable pH-responsive conductive hydrogel is presented that enables the intelligent delivery of metformin and exosomes, offering a promising and novel therapeutic approach for enhancing cardiac repair and treating MIRI.
Collapse
Affiliation(s)
- Nianlan Cheng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Qiao Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yongqing Yang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Ni Shao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Tianqi Nie
- Central laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Xiujiao Deng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Liangping Luo
- Department of Radiology and Nuclear Medicine, The Fifth Affiliated Hospital of Jinan University (Shenhe People's Hospital), Heyuan, 517000, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Department of Radiology and Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| |
Collapse
|
235
|
Chen X, Dai CM, Zhang B, Zhang WX, Huang ZH, Jiang JY, Hu SQ, Ma JH, Feng JF. RGD hydrogel-loaded ADSC extracellular vesicles mitigate uranium-induced renal injury via TLR4/NF-κB pathway inhibition. J Nanobiotechnology 2025; 23:114. [PMID: 39962465 PMCID: PMC11834392 DOI: 10.1186/s12951-025-03176-6] [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: 08/08/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Uranium-induced kidney damage represents a major health concern due to its toxic effects, including mitochondrial dysfunction and inflammation. Mitochondrial DNA (mtDNA)-mediated pyroptosis is a critical pathway in the pathogenesis of renal injury. The toll-like receptor 4 / nuclear factor-kappa B (TLR4/NF-κB) signaling pathway plays a pivotal role in this process. Recent studies have shown that extracellular vesicles derived from adipose-derived stem cells (ADSCs-EVs) possess therapeutic potential due to their anti-inflammatory and regenerative properties. Incorporating ADSCs-EVs into arginine-glycine-aspartate (RGD), hydrogels may enhance their stability and therapeutic efficacy in vivo. This study aims explore the molecular mechanism by which RGD hydrogel-loaded ADSCs-EVs modulate mtDNA-mediated pyroptosis by suppressing the TLR4/NF-κB signaling pathway to alleviate uranium-induced kidney injury. RESULTS Repairing mitochondrial dysfunction was found to mitigate mtDNA leakage, thereby inhibiting renal pyroptosis. ADSCs-EVs alleviated uranium-induced renal cell damage by suppressing the TLR4/NF-κB signaling pathway. In vivo animal experiments confirmed that RGD hydrogel-loaded ADSCs-EVs enhanced their stability in the body and improved their therapeutic efficacy against kidney injury. CONCLUSION Our findings reveal that RGD hydrogel-loaded ADSCs-EVs effectively inhibit the TLR4/NF-κB signaling pathway, preventing mtDNA-mediated pyroptosis and alleviating uranium-induced kidney damage. This elucidation provides a novel strategy for utilizing RGD hydrogel-loaded ADSCs-EVs in treating kidney injury.
Collapse
Affiliation(s)
- Xi Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, 621010, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Affiliated School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, Sichuan, 621000, China
| | - Chun-Mei Dai
- NHC Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Affiliated School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, Sichuan, 621000, China
- Department of Medical Laboratory, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Bin Zhang
- NHC Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Affiliated School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, Sichuan, 621000, China
- Department of Medical Laboratory, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Wan-Xin Zhang
- NHC Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Affiliated School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, Sichuan, 621000, China
- Department of Medical Laboratory, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Zheng-Hong Huang
- NHC Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Affiliated School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, Sichuan, 621000, China
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Jiu-Yi Jiang
- NHC Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Affiliated School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, Sichuan, 621000, China
- Department of Medical Laboratory, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shi-Qi Hu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, 621010, China
| | - Jia-Hua Ma
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, 621010, China.
| | - Jia-Fu Feng
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, 621010, China.
- NHC Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Affiliated School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, Sichuan, 621000, China.
| |
Collapse
|
236
|
Hu P, Zheng J, Wang H, Li Y, Ye T, Li Q, Lan X, Liu C, Liu C. Supramolecular Nanozymes Based on Self-Assembly of Biomolecule for Cancer Therapy. Int J Nanomedicine 2025; 20:2043-2057. [PMID: 39990286 PMCID: PMC11842878 DOI: 10.2147/ijn.s496831] [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: 09/27/2024] [Accepted: 01/10/2025] [Indexed: 02/25/2025] Open
Abstract
Natural enzyme systems possess extraordinary functions and characteristics, making them highly appealing for use in eco-friendly technologies and innovative cancer treatments. However, their inherent instability and structural complexity often limit their practical applications, leading to the exploration of biomolecular nanozyme alternatives. Supramolecular nanozymes, constructed using self-assembly techniques and various non-covalent interactions, have emerged as a promising solution. Amino acids, peptides, and protein motifs offer flexible building blocks for constructing these nanozymes. Importantly, the well-defined structural regulation mechanisms of biomolecular nanozymes, along with their unique properties as fundamental biological modules in living systems-such as selectivity, permeability, retention, and biocompatibility-present new opportunities for cancer therapy. This review highlights recent advances in supramolecular self-assembled nanozymes, including peroxidases, oxidases, catalases, superoxide dismutases, and other nanozyme systems, as building blocks for tumor therapy. Additionally, it discusses precise functional modulation through supramolecular non-covalent interactions and their therapeutic applications in targeting the tumor microenvironment. These studies provide valuable insights that may inspire the design of novel supramolecular nanozymes with enhanced catalytic selectivity, biocompatibility, and tumor-killing efficacy.
Collapse
Affiliation(s)
- Pengcheng Hu
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Jilu Zheng
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Hongjuan Wang
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Yongxin Li
- College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Tao Ye
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
- School of Clinical Medicine, Shandong second Medical University, Weifang, Shandong, 261053, People’s Republic of China
| | - Quanjun Li
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Xiaopeng Lan
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Chunzhao Liu
- College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Chunlei Liu
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| |
Collapse
|
237
|
Teng Y, Wang X, Song L, Yang J, Hou S, Lv Q, Jiang L, Guan Y, Shi J. 3D printed polycaprolactone/poly (L-lactide-co- ϵ-caprolactone) composite ureteral stent with biodegradable and antibacterial properties. Biomed Mater 2025; 20:025026. [PMID: 39908677 DOI: 10.1088/1748-605x/adb2ce] [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: 10/16/2024] [Accepted: 02/05/2025] [Indexed: 02/07/2025]
Abstract
The clinical application of biodegradable ureteral stents holds significant potential. There is an urgent need to develop new materials for ureteral stents to address the limitations related to performance degradation and antibacterial properties observed in current designs. Here, we developed a Polycaprolactone (PCL)/Poly (L-lactide-co-ϵ-caprolactone) (PLCL) composite ureteral stent by three-dimensional (3D) printing, which exhibits biodegradable and antibacterial properties. Silver nanoparticles (AgNPs) were bonded to the surface of the stent through the polymerization of dopamine (PDA) and coating with type I collagen (Col I). The ureteral stent (PP-PDA-Ag-Col) had a densely spiraled structure and higher hydrophilicity. The release behavior of silver ions from the stent was found to be slow and continuous when coated with AgNPs, which can enable long-term antibacterial effects after being implantedin vivo. Additionally,in vitrodegradation experiments demonstrated that the different ratios of ureteral stents degraded slowly in artificial urine over 6 weeks without compromising functionality. The stent exhibits excellent hemocompatibility and cell compatibility. The subcutaneous implantation experiment in Sprague-Dawley rats showed that the PP-PDA-Ag-Col stent degraded slowlyin vivoand had good biocompatibility. The stent PCL5/PLCL5 was the most promising ureteral stent regarding antibacterial, mechanical properties, and degradation. The novel 3D-printed PP-PDA-Ag-Col stent exhibits biocompatibility for safein vivotransplantation and antibacterial properties that reduce reliance on antibiotics. Additionally, its biodegradability eliminates the need for secondary surgical removal, making it a promising option for the clinical application of ureteral stents.
Collapse
Affiliation(s)
- Yanjiao Teng
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, People's Republic of China
- Key Laboratory for Disaster Medicine Technology, Tianjin 300072, People's Republic of China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, People's Republic of China
| | - Xinyan Wang
- Tianjin Children's Hospital/Tianjin University Children's Hospital, Tianjin 300074, People's Republic of China
- Department of Urology, Dalian Women and Children's Medical Group, Dalian 116037, People's Republic of China
| | - Lin Song
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210000, People's Republic of China
| | - Jianing Yang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, People's Republic of China
- Key Laboratory for Disaster Medicine Technology, Tianjin 300072, People's Republic of China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, People's Republic of China
| | - Shike Hou
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, People's Republic of China
- Key Laboratory for Disaster Medicine Technology, Tianjin 300072, People's Republic of China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, People's Republic of China
| | - Qi Lv
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, People's Republic of China
- Key Laboratory for Disaster Medicine Technology, Tianjin 300072, People's Republic of China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, People's Republic of China
| | - Li Jiang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Tianjin 300021, People's Republic of China
| | - Yong Guan
- Tianjin Children's Hospital/Tianjin University Children's Hospital, Tianjin 300074, People's Republic of China
| | - Jie Shi
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, People's Republic of China
- Key Laboratory for Disaster Medicine Technology, Tianjin 300072, People's Republic of China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, People's Republic of China
| |
Collapse
|
238
|
Habiburrohman MR, Jamilludin MA, Cahyati N, Herdianto N, Yusuf Y. Fabrication and in vitro cytocompatibility evaluation of porous bone scaffold based on cuttlefish bone-derived nano-carbonated hydroxyapatite reinforced with polyethylene oxide/chitosan fibrous structure. RSC Adv 2025; 15:5135-5150. [PMID: 39963456 PMCID: PMC11831101 DOI: 10.1039/d4ra08457h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
A novel porous bone scaffold based on nano-carbonated hydroxyapatite reinforced with fibrous-like structured polyethylene oxide/chitosan network (nCHA/PEO/CS) was introduced and fabricated via freeze-drying. Prior to this, the nCHA was synthesized through a hydrothermal reaction based on cuttlefish bone (CFB, Sepia officinalis). The raw cuttlefish bone (raw-CFB) was first decomposed to obtain cuttlefish bone-derived calcium oxide (CaO-CFB) by calcination at 1000 °C, which was used for synthesizing nCHA. The chemical composition analysis showed that the nCHA formed AB-type CHA with a high carbonate content of 7.38 wt%, which is in the range of carbonate content in native bone (2-9 wt%). The Ca/P molar ratio of nCHA was 1.712, very close to the Ca/P of biological apatite of 1.71. Morphological analysis revealed that nCHA consists of nanosized particles, potentially offering a large surface area to volume to promote ion exchange and cell interaction. The excellent physicochemical and morphological properties of nCHA proposed suitability as a bone scaffold precursor combined with PEO and CS. The nCHA/PEO/CS scaffolds were freeze-dried with varying PEO/CS concentrations. Physicochemical analysis indicated that increasing the PEO/CS concentration decreased the crystallinity of the scaffold, causing it to be lower than the nCHA crystallinity, which may be beneficial for cell growth. Morphological analysis revealed that the scaffold structure comprised nCHA cross-linked within a fibrous-like structured PEO/CS network, which appropriately mimics the fibrous structure of extracellular matrix (ECM) in natural bone. However, the nCHA/PEO/CS-11 scaffold formed more appropriate pores with suitable porosity for cell development, blood vessel formation, and nutrient perfusion. The nCHA/PEO/CS-11 scaffold also demonstrated sufficient compressive strength and good swelling behavior, which may favor bone regeneration. The nCHA/PEO/CS-11 scaffold demonstrated high cytocompatibility and facilitated the adherence of MC3T3E1 cells on the scaffold surface. The nCHA/PEO/CS-11 scaffold also promoted cell osteogenic differentiation. Owing to its desirable and suitable characteristics, the nCHA/PEO/CS-11 scaffold is promising in bone tissue engineering.
Collapse
Affiliation(s)
- Musyafa Riziq Habiburrohman
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada Yogyakarta 55281 Indonesia
| | - Muhammad Amir Jamilludin
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada Yogyakarta 55281 Indonesia
| | - Nilam Cahyati
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada Yogyakarta 55281 Indonesia
| | - Nendar Herdianto
- Research Centre for Advanced Material, National Research and Innovation Agency (BRIN) South Tangerang 15314 Indonesia
| | - Yusril Yusuf
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada Yogyakarta 55281 Indonesia
| |
Collapse
|
239
|
Gao C, Chen Y, Wen X, Han R, Qin Y, Li S, Tang R, Zhou W, Zhao J, Sun J, Li Z, Tan Z, Wang D, Zhou C. Plant-derived exosome-like nanoparticles in tissue repair and regeneration. J Mater Chem B 2025; 13:2254-2271. [PMID: 39817682 DOI: 10.1039/d4tb02394c] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
This article reviews plant-derived exosome-like nanoparticles (ELNs), and highlights their potential in regenerative medicine. Various extraction techniques, including ultracentrifugation and ultrafiltration, and their impact on ELN purity and yield were discussed. Characterization methods such as microscopy and particle analysis are found to play crucial roles in defining ELN properties. This review is focused on exploring the therapeutic potential of ELNs in tissue repair, immune regulation, and antioxidant activities. Further research and optimization methods for extraction of ELNs to realize clinical potential applications are necessary.
Collapse
Affiliation(s)
- Canyu Gao
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Yang Chen
- Center of Medical Product Technical Inspection, Chengdu, 610015, China
| | - Xingyue Wen
- 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, 610041, China
| | - Ruiying Han
- 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, 610041, China
| | - Yuxiang Qin
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Sijie Li
- Department of Burn and Plastic Reconstructive Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong Tang
- 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, 610041, China
| | - Weikai Zhou
- 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, 610041, China
| | - Junyu Zhao
- 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, 610041, China
| | - Jianxun Sun
- 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, 610041, China
| | - Zhengyong Li
- Department of Burn and Plastic Reconstructive Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhen Tan
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Deli Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Changchun Zhou
- College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
240
|
Ge Y, Wu L, Mei S, Wu J. Nanomaterials: Promising Tools for the Diagnosis and Treatment of Myocardial Infarction. Int J Nanomedicine 2025; 20:1747-1768. [PMID: 39958320 PMCID: PMC11829642 DOI: 10.2147/ijn.s500146] [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: 10/10/2024] [Accepted: 01/24/2025] [Indexed: 02/18/2025] Open
Abstract
Myocardial infarction (MI) is the leading cause of mortality from cardiovascular diseases. Rapid diagnosis and effective treatment are critical for improving patient prognosis. Although current diagnostic and therapeutic approaches have made significant progress, they still face challenges such as ischemia-reperfusion injury, microcirculatory disorders, adverse cardiac remodeling, and inflammatory responses. These issues highlight the urgent need for innovative solutions. Nanomaterials, with their diverse types, excellent physicochemical properties, biocompatibility, and targeting capabilities, offer promising potential in addressing these challenges. Advances in nanotechnology have increasingly drawn attention to the application of nanomaterials in both diagnosing and treating myocardial infarction. We summarize the pathophysiological mechanisms and staging of myocardial infarction. We systematically review the applications of nanomaterials in MI diagnosis, including the detection of biomarkers and imaging techniques, as well as in MI treatment, encompassing anti-inflammatory effects, antioxidant stress, inhibition of fibrosis, promotion of angiogenesis, and cardiac conduction repair. We analyze the existing challenges and provide insights into future research directions and potential solutions. Specifically, we discuss the need for rigorous safety assessments, long-term efficacy studies, and the development of robust strategies for translating laboratory findings into clinical practice. In conclusion, nanotechnology holds significant promise as a new strategy for diagnosing and treating myocardial infarction. Its potential to enhance clinical outcomes and revolutionize patient care makes it an exciting area of research with practical applications in real-world clinical settings.
Collapse
Affiliation(s)
- Yanmin Ge
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130000, People’s Republic of China
| | - Lincong Wu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130000, People’s Republic of China
| | - Shuyang Mei
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130000, People’s Republic of China
| | - Junduo Wu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130000, People’s Republic of China
| |
Collapse
|
241
|
Castro C, Patin J, Jajkiewicz C, Chizelle F, Cerpa CO, Tessier A, Le Pogam E, Fellah I, Baró I, Charpentier F, Derangeon M. Long QT syndrome type 3 gain-of-function of Na v1.5 increases ventricular fibroblasts proliferation and pro-fibrotic factors. Commun Biol 2025; 8:216. [PMID: 39934335 PMCID: PMC11814334 DOI: 10.1038/s42003-025-07636-5] [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: 07/05/2023] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
The long QT syndrome type 3 (LQT3) is a cardiac channelopathy caused by gain-of-function mutations in the SCN5A gene, encoding the sodium channel Nav1.5. As Nav1.5 is expressed in cardiomyocytes but also in cardiac fibroblasts, we investigated whether the LQT3-causing p.ΔQKP1507-1509 (ΔQKP) SCN5A mutation alters cardiac fibroblast phenotype. Primary cultured ventricular fibroblasts from Scn5a+/ΔQKP knock-in mice showed increased proliferation, survival, expression of transforming growth factor-β (TGF-β) and activation of its canonical pathway, and reduced α-smooth muscle actin expression. Ventricular tissue from Scn5a+/ΔQKP mice exhibited augmented fibroblast populations and fibrosis. Inhibiting TGF-β receptor, sodium current or Scn5a expression decreased Scn5a+/ΔQKP fibroblast proliferation, while veratridine increased proliferation of control fibroblasts, mimicking Nav1.5 gain-of-function. Lastly, abnormal calcium signaling underlied the increased proliferation of Scn5a+/ΔQKP fibroblasts. Our study shows that cardiac fibroblasts carrying the ΔQKP-SCN5A mutation exhibit an abnormal, proliferative phenotype, paving the way for better understanding the role of cardiac fibroblasts in LQT3.
Collapse
Affiliation(s)
- Claire Castro
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Justine Patin
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Cyrielle Jajkiewicz
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Franck Chizelle
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Cynthia Ore Cerpa
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Agnès Tessier
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Eva Le Pogam
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Imen Fellah
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Isabelle Baró
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Flavien Charpentier
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - Mickaël Derangeon
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France.
| |
Collapse
|
242
|
Zhang S, Li R, Song M, Han J, Fan X. Exploration of M2 macrophage membrane as a biotherapeutic agent and strong synergistic therapeutic effects in ischemic stroke. J Control Release 2025; 378:476-489. [PMID: 39561947 DOI: 10.1016/j.jconrel.2024.11.033] [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: 07/18/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
The macrophage-derived membrane is widely applied in the targeting nanocarrier for its inflammatory tendency and long circulation ability due to the preservation of membrane protein. Few studies reported the application of macrophage membranes as biotherapeutic agents. To verify the ability of macrophage membrane as a biotherapeutic agent, ischemic stroke was selected as the model disease. Inspired by the features of macrophages infiltrating the ischemic core and the talent of M2 macrophages in modulating the inflammatory microenvironment, an M2 macrophage membrane (M2M)-disguised poly lactic-co-glycolic acid nanoparticles loaded with baicalin (BA) (M2M@BANPs) is developed. The results in vivo and in vitro indicate that M2M@BANPs could efficiently and actively target ischemic brain tissue and accumulate in microglia and neurons due to the coating of M2M. Furthermore, M2M and M2M@BANPs exhibit significant therapeutic effects in salvaging brain tissue damage and neurological functional recovery by reprogramming microglia from M1 to M2, reducing neutrophil infiltration and inhibiting neuronal apoptosis. Together, our fabrication provides a new insight and an applicative perspective for M2M in the therapy of ischemic stroke.
Collapse
Affiliation(s)
- Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Meiying Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jin Han
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
243
|
Hsu P, Kamijyo Y, Koike E, Ichikawa S, Zheng Y, Ohno T, Katayama S. Exosome-like nanovesicles derived from kale juice enhance collagen production by downregulating Smad7 in human skin fibroblasts. Front Nutr 2025; 12:1486572. [PMID: 39996007 PMCID: PMC11847687 DOI: 10.3389/fnut.2025.1486572] [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: 08/26/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Plant-derived exosome-like nanovesicles (ELNs) are critical mediators of cross-kingdom communication, modulating gene expression in animal cells despite their plant origin. In this study, we investigated the effects of glucoraphanin-enriched kale (GEK)-derived ELNs (GELNs) on collagen production in normal human dermal fibroblasts NB1RGB. The ELNs isolated from GEK juice powder had particle sizes similar to those of typical exosomes. GELNs increased type I collagen expression in NB1RGB cells significantly. Microarray analysis demonstrated that GELN-derived total RNA upregulated the expression of genes related to extracellular matrix formation, including those involved in collagen synthesis. Further investigation revealed that microRNA-enriched fraction of GELNs promoted collagen production by inhibiting the expression of Smad7. These findings suggest that GELNs and their microRNA content enhance collagen production through the downregulation of Smad7.
Collapse
Affiliation(s)
- Peihan Hsu
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Yuriko Kamijyo
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Emiri Koike
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Saki Ichikawa
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Yifeng Zheng
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | | | - Shigeru Katayama
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| |
Collapse
|
244
|
Einen C, Snipstad S, Wesche HF, Nordlund V, Devold EJ, Amini N, Hansen R, Sulheim E, Davies CDL. Impact of the tumor microenvironment on delivery of nanomedicine in tumors treated with ultrasound and microbubbles. J Control Release 2025; 378:656-670. [PMID: 39701458 DOI: 10.1016/j.jconrel.2024.12.037] [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: 09/04/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
The delivery of nanoparticles to tumors has been shown preclinically to be improved by microbubble-mediated ultrasound. However, the mechanisms and biological effects are not fully understood. In this study, we explored the influence of the tumor microenvironment on nanoparticle uptake and microdistribution both with and without ultrasound and microbubble treatment. Three murine tumor models, KPC (pancreatic ductal adenocarcinoma), 4T1 (triple negative mammary carcinoma) and CT26 (colon carcinoma), were characterized with respect to extracellular matrix composition, tumor stiffness and perfusion. KPC and 4T1 tumors presented higher levels of collagen and hyaluronic acid and were stiffer compared to CT26, whereas all three tumors had similar levels of sulfated glycosaminoglycans. Furthermore, the 4T1 tumors appeared poorly vascularized with a lower cell density compared to KPC and CT26. All three tumors presented similar nanoparticle uptake, but extravasated nanoparticles traveled significantly shorter in KPC tumors compared to 4T1 and CT26. The effect of ultrasound and microbubble treatment on the tumor uptake and penetration of polymer nanoparticles into the extracellular matrix were evaluated using a treatment protocol previously shown to increase nanoparticle delivery to tumors. Interestingly, we found a significant increase in nanoparticle uptake in the soft CT26 tumor, but no effect of the ultrasound treatment in the stiff KPC and 4T1 tumors, suggesting that tumor stiffness is an important parameter for treatment with ultrasound and microbubbles. Ultrasound treatment resulted in a modest but not statistically significant improvement in nanoparticle penetration through the extracellular matrix. In tumors demonstrating increased uptake of nanoparticles following ultrasound treatment, the uptake correlated positively with blood volume. These findings emphasize the importance of taking the tumor microenvironment into consideration when optimizing ultrasound parameters for delivery of nanomedicine.
Collapse
Affiliation(s)
- Caroline Einen
- Porelab and Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Håkon F Wesche
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Veronica Nordlund
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ella J Devold
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Institute for Research in Biomedicine, Bellinzona, Switzerland
| | | |
Collapse
|
245
|
Gansevoort M, Wentholt S, Li Vecchi G, de Vries M, Versteeg EMM, Boekema BKHL, Choppin A, Barritault D, Chiappini F, van Kuppevelt TH, Daamen WF. Next-Generation Biomaterials for Wound Healing: Development and Evaluation of Collagen Scaffolds Functionalized with a Heparan Sulfate Mimic and Fibroblast Growth Factor 2. J Funct Biomater 2025; 16:51. [PMID: 39997585 PMCID: PMC11856099 DOI: 10.3390/jfb16020051] [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: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Fibrosis after full-thickness wound healing-especially after severe burn wounds-remains a clinically relevant problem. Biomaterials that mimic the lost dermal extracellular matrix have shown promise but cannot completely prevent scar formation. We present a novel approach where porous type I collagen scaffolds were covalently functionalized with ReGeneRating Agent (RGTA®) OTR4120. RGTA® is a glycanase-resistant heparan sulfate mimetic that promotes regeneration when applied topically to chronic wounds. OTR4120 is able to capture fibroblast growth factor 2 (FGF-2), a heparan/heparin-binding growth factor that inhibits the activity of fibrosis-driving myofibroblasts. Scaffolds with various concentrations and distributions of OTR4120 were produced. When loaded with FGF-2, collagen-OTR4120 scaffolds demonstrated sustained release of FGF-2 compared to collagen-heparin scaffolds. Their anti-fibrotic potential was investigated in vitro by seeding primary human dermal fibroblasts on the scaffolds followed by stimulation with transforming growth factor β1 (TGF-β1) to induce myofibroblast differentiation. Collagen-OTR4120(-FGF-2) scaffolds diminished the gene expression levels of several myofibroblast markers. In absence of FGF-2 the collagen-OTR4120 scaffolds displayed an inherent anti-fibrotic effect, as the expression of two fibrotic markers (TGF-β1 and type I collagen) was diminished. This work highlights the potential of collagen-OTR4120 scaffolds as biomaterials to improve skin wound healing.
Collapse
Affiliation(s)
- Merel Gansevoort
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Sabine Wentholt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Gaia Li Vecchi
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Marjolein de Vries
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Elly M. M. Versteeg
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Bouke K. H. L. Boekema
- Burn Research Lab, Alliance of Dutch Burn Care, 1941 AJ Beverwijk, The Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | | | | | | | - Toin H. van Kuppevelt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Willeke F. Daamen
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
246
|
Liu B, Zhang H, Qin X. Amyloid Fibrils and Their Applications: Current Status and Latest Developments. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:255. [PMID: 39997818 PMCID: PMC11858031 DOI: 10.3390/nano15040255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025]
Abstract
Amyloid fibrils are one of the important forms of protein aggregates, first discovered in the pathological brain tissues of patients with various neurodegenerative diseases. They are considered the core pathological markers of different neurodegenerative diseases. In recent years, research has found that multiple proteins or peptides dynamically assemble to form functional amyloid-like nanofibrils under physiological conditions, exhibiting excellent mechanical properties, high environmental stability, and self-healing ability. Therefore, they have become a class of functional biological nanomaterials with important development potential. This article systematically reviews the latest progress in the preparation, functionalization, and application of amyloid-like nanofibrils in engineering and provides an outlook on possible future development directions.
Collapse
Affiliation(s)
| | - Hongnan Zhang
- Key Laboratory of Textile Science & Technology, College of Textiles, Donghua University, Ministry of Education, Shanghai 200051, China; (B.L.); (X.Q.)
| | | |
Collapse
|
247
|
Tang Q, Xie J, Wang Y, Dong C, Sun Q. Exosomes secreted by ATF3/Nrf2-mediated ferroptotic renal tubular epithelial cells promote M1/M2 ratio imbalance inducing renal interstitial fibrosis following ischemia and reperfusion injury. Front Immunol 2025; 16:1510500. [PMID: 39975560 PMCID: PMC11835872 DOI: 10.3389/fimmu.2025.1510500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
Background Severe renal ischemia and reperfusion injury (IRI) progresses to renal interstitial fibrosis (RIF) with limited therapeutic strategies. Although ferrptosis and macrophage polarization both play important roles in this model, their specific pathogenesis and interactions have not been elucidated. Therefore, we aimed to explore the mechanisms by which ferrotosis occurs in renal tubular epithelial cells (RTECs) and ferroptotic cell-derived exosomes induce macrophage polarization in IRI-related RIF model. Methods In vivo, C57BL/6J mice were randomly divided into four groups: sham group, ischemia and reperfusion (IR) group, IR + Ferrostatin-1 (Fer-1) group, and IR +ATF3 knockdown (ATFKD) group. In vitro, RTECs were divided into control (CON) group, hypoxia/reoxygenation (HR) group, HR +Fer-1 group, HR + siRNA-ATF3 (siATF3) group. Result Compared with the sham group, the IR group showed more severe kidney injury in HE staining, more collagen fibers in Masson staining, and higher α-SMA expression levels in immunohistochemistry. Total iron and MDA content increased while GSH content decreased. The IR group had more significant mitochondrial damage and higher PTGS2 and TFRC mRNA levels than those in the sham group. Compared with the IR group, the above indexes were all alleviated in the IR+Fer-1 or IR+ATF3KD groups. In addition, the protein expressions of ATF3, Nrf2 and HO-1 in the IR group were increased than those in sham group. Compared with the IR group, ATF3 expressions in the IR+Fer-1 or IR+ATF3KD groups were decreased, and the protein contents of Nrf2 and HO-1 were further increased. Moreover, there were higher levels of M2 markers (Arg1, TGF-β and IL-10 mRNA) in the IR group than those in the sham group, and lower levels in the IR+Fer-1 group or in the IR+ATF3KD group compared with the IR group. The results of in vitro experiment are consistent with those of in vivo experiment. Mechanistically, the release of exosomes carrying miR-1306-5p by the HR group promoted more M2 macrophage. Conclusion ATF3 might accelerate the ferroptosis by inhibiting Nrf2/ARE pathway, and exosomes from ferroptotic cells reduced the M1/M2 macrophage ratio, promoting fibrosis.
Collapse
Affiliation(s)
- Qiao Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiatao Xie
- The First Clinical College of Wuhan University, Wuhan, China
| | - Yifei Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chong Dong
- Organ Transplantation Center, Tianjin First Central Hospital, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| | - Qian Sun
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
248
|
Xiao Y, Zhang S, Zhuo H, Zhang X, Zhu K, Chen W, You G, Chen H, Luo Q, Zhou H, Chen G. Dietary Natural Melanin Nanozymes Delay Aging and Ameliorate Neurodegeneration via Improving Gut Microbiota and Redox Homeostasis. ACS OMEGA 2025; 10:3610-3621. [PMID: 39926539 PMCID: PMC11800042 DOI: 10.1021/acsomega.4c08419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
Aging is an inevitable multifactor process that causes a decline in organ function and increases the risk of age-related diseases and death. Thus, the development of highly effective and safe therapeutic strategies to delay aging and age-related diseases is urgently required. In this study, we isolated natural melanin nanozymes (NMNs) from the ink sacs of live octopuses. The NMNs exhibited excellent superoxide-dismutase-mimicking and radical scavenging activities. In SAMP8 mice, treatment with NMNs improved their cognition and memory functions while restoring their aging-impaired liver function and lipid metabolism, thereby prolonging their lifespan. Moreover, the NMNs reversed metabolic changes in their aged brains and reconstructed their gut microbiota composition by enhancing microbial community diversity. Our findings indicate that NMNs treatment could be a promising approach for delaying aging and preventing age-associated physiological decline in humans.
Collapse
Affiliation(s)
- Yao Xiao
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Shikun Zhang
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Hailong Zhuo
- Department
of Transfusion, The Fifth Medical Center
of Chinese PLA General Hospital, Beijing 100071, China
| | - Xiaoyong Zhang
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Kai Zhu
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Wanyi Chen
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Guoxing You
- Academy
of Military Medical Sciences, Beijing 100850, China
| | | | - Qun Luo
- Department
of Transfusion, The Fifth Medical Center
of Chinese PLA General Hospital, Beijing 100071, China
| | - Hong Zhou
- Academy
of Military Medical Sciences, Beijing 100850, China
| | - Gan Chen
- Academy
of Military Medical Sciences, Beijing 100850, China
| |
Collapse
|
249
|
Bian X, Zhou L, Luo Z, Liu G, Hang Z, Li H, Li F, Wen Y. Emerging Delivery Systems for Enabling Precision Nucleic Acid Therapeutics. ACS NANO 2025; 19:4039-4083. [PMID: 39834294 DOI: 10.1021/acsnano.4c11858] [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: 01/22/2025]
Abstract
Nucleic acid therapeutics represent a highly promising treatment approach in modern medicine, treating diseases at the genetic level. However, these therapeutics face numerous challenges in practical applications, particularly regarding their stability, effectiveness, cellular uptake efficiency, and limitations in delivering them specifically to target tissues. To overcome these obstacles, researchers have developed various innovative delivery systems, including viral vectors, lipid nanoparticles, polymer nanoparticles, inorganic nanoparticles, protein carriers, exosomes, antibody oligonucleotide conjugates, and DNA nanostructure-based delivery systems. These systems enhance the therapeutic efficacy of nucleic acid drugs by improving their stability, targeting specificity, and half-life in vivo. In this review, we systematically discuss different types of nucleic acid drugs, analyze the major barriers encountered in their delivery, and summarize the current research progress in emerging delivery systems. We also highlight the latest advancements in the application of these systems for treating genetic diseases, infectious diseases, cancer, brain diseases, and wound healing. This review aims to provide a comprehensive overview of nucleic acid drug delivery systems' current status and future directions by integrating the latest advancements in nanotechnology, biomaterials science, and gene editing technologies, emphasizing their transformative potential in precision medicine.
Collapse
Affiliation(s)
- Xiaochun Bian
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhiwei Luo
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Guotao Liu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhongci Hang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Haohao Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Fengyong Li
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
250
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [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: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|