1
|
Ahmed W, Li S, Liang M, Peng P, Muhammad W, Wang Q, Gao C. Selenium-containing polyurethane nanofibers with MnO 2 nanoparticles and gelsevirine promote diabetic wound healing by modulation of ROS and inflammation. BIOMATERIALS ADVANCES 2025; 173:214289. [PMID: 40158270 DOI: 10.1016/j.bioadv.2025.214289] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Reactive oxygen species (ROS) and subsequent inflammatory cascades hinder the healing of diabetic wounds, which should be tackled simultaneously when designing wound dressings. In this study, ROS-responsive di‑selenium-containing polyurethane nanofibers (PUF) loaded with manganese dioxide nanoparticles (MnO2 NPs) and gelsevirine (GSV) with an average diameter of 0.6 ± 10 μm, were prepared to specifically target ROS and inflammation control, thereby enhancing healing in diabetic wounds. The resulting nanofibers exhibited a porous architecture and enhanced drug delivery capability. The MnO2/GSV/PUF dressing showed negligible cytotoxicity with cell viability over 80 %, effective ROS-scavenging ability of 87 %, and inhibition of pro-inflammatory cytokine expression. This dual mechanism of ROS-scavenging and inflammation modulation accelerated the healing of diabetic wounds up to 92 % in 14 d in a full-thickness diabetic wound model, as evidenced by reduced cytokine levels, enhanced epithelialization, and uniform collagen deposition with the highest percentage of 60 %.
Collapse
Affiliation(s)
- Wajiha Ahmed
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shifen Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Min Liang
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312099, China
| | - Pai Peng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Wali Muhammad
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Qiaoxuan Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312099, China.
| |
Collapse
|
2
|
Wu L, Wang Y, Zhao X, Zhao T, Li J, Kuang Y, He Y, Yang S, Gu Z, Mao H. A self-adhesive hierarchical nanofiber patch for dynamic and multistage management of full-thickness cutaneous wounds. J Nanobiotechnology 2025; 23:448. [PMID: 40524144 DOI: 10.1186/s12951-025-03513-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/28/2025] [Indexed: 06/19/2025] Open
Abstract
Full-thickness cutaneous wounds pose a significant threat to global health due to their complex healing demands. Standard clinical wound dressings often fall short in providing the adaptability and functionality required for the entire healing process. While hierarchically engineered nanofiber dressings have shown advancement in wound management, challenges such as material compatibility and interfacial bonding during their design have limited both manufacturing and therapeutic outcomes. This study introduces a self-adhesive hierarchical nanofiber (SAHN) patch designed to provide a comprehensive and dynamic approach to wound care. The SAHN patch strategically integrates synthetic biodegradable poly(ester carbonate) with natural bioactive components, forming a seamless dual-layer system that offers both immediate protection and sustained bioactivity to support tissue regeneration. In vitro and in vivo studies demonstrate the patch's superior interlayer adhesion, soft tissue adhesion, controlled degradation, and robust antibacterial capabilities. These features collectively safeguard the wound microenvironment, facilitate hemostasis, manage inflammation, and accelerate wound closure. Our findings highlight the transformative potential of the SAHN patch in improving traditional wound care, overcoming the manufacturing challenges associated with hierarchical nanofiber dressings, and offering a promising solution for dynamic and multistage management of full-thickness cutaneous wounds that aligns with the natural progression of tissue repair.
Collapse
Affiliation(s)
- Lihuang Wu
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Yuqi Wang
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Xinyue Zhao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Tingting Zhao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Junhua Li
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Yi Kuang
- College of Chemical and Materials Engineering, Zhejiang A&F University, Lin'an, 311300, China
| | - Yiyan He
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China.
| | - Shengxiang Yang
- College of Chemical and Materials Engineering, Zhejiang A&F University, Lin'an, 311300, China.
| | - Zhongwei Gu
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China.
| |
Collapse
|
3
|
Xu L, Zhang J, Luo J, Cui Y, Chen J, Zeng B, Deng Z, Shao L. "Double-sided protector" Janus hydrogels for skin and mucosal wound repair: applications, mechanisms, and prospects. J Nanobiotechnology 2025; 23:387. [PMID: 40426120 PMCID: PMC12117736 DOI: 10.1186/s12951-025-03438-3] [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: 02/27/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Skin and mucous membranes serve as crucial barrier tissues within the human body. Defective wound healing not only inflicts pain but also heightens the risk of infection and impairs immune function. Janus hydrogels possess two-sided distinct asymmetric structures that endow them with diverse properties such as high water absorbency, flexibility, anti-adhesion ability etc. These hydrogels also exhibit great potential in biofluid transport, drug delivery and promoting tissue repair. Currently, research efforts predominantly concentrate on the preparation techniques, properties, and biomedical applications. This review summarized its structural characteristics and different forms of designations, and focused on the possible mechanisms, the existing problems and improvement strategies for the skin and mucous tissues wound, aiming to provide new design ideas for repairing complex skin and mucous membrane tissue defects.
Collapse
Affiliation(s)
- Laijun Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
- School of Stomatology, Changsha Medical University, Changsha, 410219, China
| | - Junyi Zhang
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Junsi Luo
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Yiteng Cui
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China
| | - Jinhong Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Bin Zeng
- School of Stomatology, Changsha Medical University, Changsha, 410219, China
| | - Zhiyuan Deng
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, 410000, China.
- School of Stomatology, Changsha Medical University, Changsha, 410219, China.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China.
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
4
|
Nazar LA, Al-Salman SS, Torki SH, Al-Musawi MH, Najafinezhad A, Noory P, Rajab ES, Khosravi N, Talebi S, Azamian F, Valizadeh H, Sharifianjazi F, Tavamaishvili K, Mohabbatkhah M, Shahriari-Khalaji M, Nasiri-Harchegani S, Mehrjoo M, Tavakoli M, Mirhaj M. 3D printed PHB-dextran-whitlockite porous construct coated with sildenafil-loaded nanofibers: a hybrid scaffold for craniofacial reconstruction. Int J Biol Macromol 2025; 314:144352. [PMID: 40389012 DOI: 10.1016/j.ijbiomac.2025.144352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 05/02/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
In this study, a novel hybrid scaffold comprising 3D-printed porous polyhydroxybutyrate (PHB), dextran (Dex), and magnesium-doped whitlockite (WL) nanoparticles was developed, which were further enhanced with an electrospun nanofibrous coating composed of Dex and Pluronic F127 (F127) loaded with Sildenafil (Sil) for use in craniofacial regeneration. This design was intended to improve the solubility of sildenafil and enable controlled release. Scanning electron microscopy (SEM) revealed a well-integrated structure between the 3D-printed strands and electrospun nanofibers. The scaffold exhibited sustained release of Sil over 28 days, with mechanical testing showing a compressive strength of 3.70 ± 0.33 MPa and an elastic modulus of 49.04 ± 4.62 MPa. Non-toxicity was confirmed via MTT assay on the MG63 cell line, and qRT-PCR results indicated significantly higher expression levels of collagen I, RUNX2, osteocalcin, VEGF, and CD31 markers associated with osteogenesis and angiogenesis. Following implantation in a rat calvarial defect model, the scaffold demonstrated robust osteogenic activity and new bone tissue formation over an eight-week period. This innovative scaffold design offers a promising solution for overcoming the challenges in craniofacial defect repair by integrating bioactive materials with advanced drug delivery systems, leading to more effective tissue regeneration strategies.
Collapse
Affiliation(s)
- Lara Ali Nazar
- Department of Chemistry, College of Sciences, Mustansiriyah University, Baghdad, Iraq.
| | - Sarah Sameer Al-Salman
- Department of Clinical Laboratory Sciences, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq.
| | - Sumyah Hasan Torki
- Department of Plant Biotechnology College of Biotechnology, Al-Nahrain University, Baghdad, Iraq.
| | - Mastafa H Al-Musawi
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq.
| | - Aliakbar Najafinezhad
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Parastoo Noory
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Eslah Shakir Rajab
- Department of Microbiology, College of Science, Al_Karkh University for Science, Baghdad, Iraq.
| | - Negin Khosravi
- School of Science and Health, The University of Georgia, Tbilisi, Georgia.
| | - Sina Talebi
- Department of Orthopaedics, Isfahan University of Medical Science, Isfahan, Iran
| | - Fariba Azamian
- Department of Materials Science and Nanotechnology, Sharif University of Technology, International Campus-Kish, 794117-76655 Kish, Iran.
| | - Hamideh Valizadeh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fariborz Sharifianjazi
- Center for Advanced Materials and Structures, School of Science and Technology, The University of Georgia, Tbilisi 0171, Georgia; Department of Civil Engineering, School of Science and Technology, The University of Georgia, 0171, Tbilisi, Georgia.
| | - Ketevan Tavamaishvili
- Georgian American University, School of Medicine, 10 Merab Aleksidze Str., Tbilisi 0160, Georgia.
| | - Mehdi Mohabbatkhah
- Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | | | - Sepideh Nasiri-Harchegani
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Morteza Mehrjoo
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Mohamadreza Tavakoli
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Marjan Mirhaj
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| |
Collapse
|
5
|
Wang S, Wu J, Ren K, Zhang Y, Gao F, Chen Y, Chen C, Lu J. Platelet-Rich Plasma-Derived Exosome-Encapsulated Hydrogels Accelerate Diabetic Wound Healing by Inhibiting Fibroblast Ferroptosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27923-27936. [PMID: 40315047 DOI: 10.1021/acsami.5c02705] [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: 05/04/2025]
Abstract
Platelet-rich plasma-derived exosomes (PRP-Exos) have recently been considered an optimized strategy for diabetic wound treatment, yet the potential role of PRP-Exos in diabetic wound healing is still unclear. This study aims to investigate the potential mechanisms of PRP-Exos in diabetic wound healing and to utilize the hydrogel Pluronic F127 as a carrier to maintain the sustained release of encapsulated PRP-Exos. PRP-Exos were isolated from the blood of healthy individuals and characterized, followed by co-culturing with isolated diabetic human skin fibroblasts (Diabetes HSF). RNA sequencing (RNA-seq) was used to analyze the effect of PRP-Exos on the transcriptome of normal and Diabetes HSF, screening and validating the crucial mechanism and target gene. Then, a hydrogel composed of Pluronic F127 and PRP-Exos (PRP-Exos/Gel) was constructed and applied in the diabetic mouse models to evaluate the effect and mechanism. RNA-seq analysis revealed that PRP-Exos significantly upregulated the expression of FosB in Diabetes HSF. Further intervention in the expression of FosB in Diabetes HSF showed that knocking down FosB induced ferroptosis in Diabetes HSF, characterized by decreased cell viability, increased oxidative stress, and increased iron ion levels, along with downregulation of GPX4 and SLC7A11 expression, while ACSL4 expression was increased; conversely, overexpression of FosB had the opposite effect. Subsequently, adding PRP-Exos to FosB-knocked down Diabetes HSF significantly weakened the inhibitory effect of PRP-Exos on ferroptosis in diabetic fibroblasts. The synthesized PRP-Exos/Gel exhibited significant thermosensitivity and sustained release of exosomes. In animal experiments, the PRP-Exos/Gel showed significant anti-inflammatory effects, evidenced by an increased proportion of M2 macrophages and a decreased proportion of central granule cells in wound tissue, and inhibited fibroblast ferroptosis, thereby accelerating wound healing. Overall, the constructed PRP-Exos/Gel displays a continuous release of exosomes and promotes diabetic wound healing by suppressing inflammatory responses and fibroblast ferroptosis, which provides new insights and methods for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Shanzheng Wang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Jianyue Wu
- Department of Orthopaedics, Xishan People's Hospital of Wuxi, Wuxi, Jiangsu 214105, PR China
| | - Ke Ren
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Yuehou Zhang
- Department of Burn and Plastic Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| | - Feifei Gao
- Department of Orthopaedics, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214400, PR China
| | - Yaofei Chen
- Department of Orthopaedics, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214400, PR China
| | - Changhong Chen
- Department of Orthopaedics, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214400, PR China
| | - Jun Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, PR China
| |
Collapse
|
6
|
Liu D, Yu T, Ma S, Su L, Zhong S, Wang W, Liu Y, Yu JA, Gao M, Chen Y, Xu H, Liu Y. Insulin/PHMB-grafted sodium alginate hydrogels improve infected wound healing by antibacterial-prompted macrophage inflammatory regulation. J Nanobiotechnology 2025; 23:328. [PMID: 40319298 PMCID: PMC12048987 DOI: 10.1186/s12951-025-03398-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Non-healing chronic wounds with high susceptibility to infection represent a critical challenge in modern healthcare. While growth factors play a pivotal role in regulating chronic wound repair, their therapeutic efficacy is compromised in infected microenvironments. Current wound dressings inadequately address the dual demands of sustained bioactive molecule delivery and robust antimicrobial activity. RESULTS In this study, we developed a sodium alginate hydrogel (termed P-SA/Ins), which incorporated polyhexamethylene biguanide (PHMB) grafting and long-acting glargine insulin loading. P-SA/Ins exhibited the favorable physicochemical performance, biocompatibility and antibacterial efficacy against both Gram-negative and Gram-positive pathogens through inhibition of bacterial proliferation and biofilm formation. Glargine insulin was applied to prolonged insulin delivery. P-SA/Ins treatment attenuated S. aureus induced pro-inflammatory cytokine cascades in macrophages. The evaluation in vivo using a rat model with S. aureus infected wound demonstrated that P-SA/Ins significantly enhanced wound healing and optimized skin barrier through antimicrobial-mediated modulation of macrophage polarization and subsequent inflammatory cytokine profiling. CONCLUSIONS Our findings demonstrate that P-SA/Ins promotes wound healing and restores epidermal barrier integrity, indicating its potential as a therapeutic dressing for chronic wound healing, particularly in cases with infection risk.
Collapse
Affiliation(s)
- Dan Liu
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Tianyi Yu
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Shan Ma
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Lefeng Su
- College of Chemistry & Materials Science, Shanghai Normal University, No. 100 Guilin Road, Shanghai, 200234, China
| | - Shan Zhong
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Wenao Wang
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yang Liu
- Department of Pediatric Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jia-Ao Yu
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Min Gao
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yunsheng Chen
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - He Xu
- College of Chemistry & Materials Science, Shanghai Normal University, No. 100 Guilin Road, Shanghai, 200234, China.
| | - Yan Liu
- Department of Burn, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
7
|
Kazemi N, Hassanzadeh-Tabrizi SA, Koupaei N, Ghomi H, Masaeli E. Incorporation of forsterite nanoparticles in a 3D printed polylactic acid/polyvinylpyrrolidone scaffold for bone tissue regeneration applications. Int J Biol Macromol 2025; 305:141046. [PMID: 39954877 DOI: 10.1016/j.ijbiomac.2025.141046] [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: 05/24/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/17/2025]
Abstract
Three-dimensional (3D) printing has facilitated the fabrication of customized scaffolds for the repair of complex bone defects. In this study, 3D-printed scaffolds composed of a mixture of polylactic acid-polyvinylpyrrolidone (PLA-PVP) incorporating different amounts of forsterite (F; Mg2SiO4) nanoparticles were fabricated using fused deposition modeling (FDM) technique. The incorporation of PVP and F nanoparticles into the PLA scaffold significantly decreased the water drop contact angle. The mechanical properties of the PLA-PVP scaffold were enhanced with the addition of 10 % F nanoparticles, as the compressive yield strength increased from 10.8 to 16.0 MPa and the elastic modulus from 83.52 to 108.41 MPa. However, the addition of F nanoparticles increased the degradation rate of the PLA-PVP scaffold over 8 weeks. Importantly, the addition of 10 % F nanoparticles into the PLA-PVP scaffold improved bioactivity and formation of apatite deposits on the scaffold after 4 weeks of immersion in simulated body fluid. Moreover, the PLA-PVP/10F scaffold showed strong MG63 cell adhesion and proliferation, as well as promoting osteogenic differentiation of rat bone marrow mesenchymal stem cells. At last, these findings suggest the PLA-PVP/10F scaffold is a promising candidate for application in bone defect repair.
Collapse
Affiliation(s)
- Nafise Kazemi
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - S A Hassanzadeh-Tabrizi
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran.
| | - Narjes Koupaei
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Hamed Ghomi
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Elahe Masaeli
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
8
|
Yang H, Chen Y, Rong Y, Zhou Y, Li S, Li X, Wu H, Lv D, Cao X, Wang P, Zhu J, Tang B, Hu Z. Multifunctional hydrogel targeting senescence to accelerate diabetic wound healing through promoting angiogenesis. J Nanobiotechnology 2025; 23:177. [PMID: 40050885 PMCID: PMC11884173 DOI: 10.1186/s12951-025-03274-5] [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: 01/25/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025] Open
Abstract
Diabetic wound healing remains a significant clinical challenge because of hyperglycaemia-induced cellular senescence, impaired angiogenesis, and chronic inflammation. To address these issues, we developed a multifunctional hydrogel (GelMA/PNS/Alg@IGF-1) that integrates gelatine methacryloyl (GelMA), Panax notoginseng saponins (PNS), and sodium alginate microspheres encapsulating insulin-like growth factor-1 (IGF-1). This hydrogel was engineered to achieve gradient and sustained release of bioactive agents to target senescence and promote vascular repair. In vitro studies demonstrated that the hydrogel significantly reduced oxidative stress, suppressed senescence markers and senescence-associated secretory phenotypes, and restored endothelial cell function under high-glucose conditions by inhibiting NF-κB pathway activation. Transcriptomic analysis revealed the modulation of pathways linked to inflammation, apoptosis, and angiogenesis. This hydrogel accelerated diabetic wound closure in a rat model in vivo and enhanced collagen deposition, granulation tissue formation, and neovascularization. Furthermore, the hydrogel mitigated oxidative stress and cellular senescence and promoted tissue remodelling. The synergistic effects of PNS and IGF-1 within the hydrogel established a pro-regenerative microenvironment to address both pathological ageing and vascular dysfunction. These findings highlight GelMA/PNS/Alg@IGF-1 as a promising therapeutic platform for diabetic wound management, as this material offers dual anti-senescence and proangiogenic efficacy to overcome the complexities of chronic wound healing.
Collapse
Affiliation(s)
- Hao Yang
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yongfei Chen
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yanchao Rong
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuxi Zhou
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuting Li
- Department of Plastic Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaohui Li
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Honglin Wu
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Dongming Lv
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoling Cao
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Peng Wang
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiayuan Zhu
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| | - Bing Tang
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| | - Zhicheng Hu
- Department of Burn and Wound Repair, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|