1
|
Wu J, Qian Y, Yang K, Zhang S, Zeng E, Luo D. Innate immune cells in vascular lesions: mechanism and significance of diversified immune regulation. Ann Med 2025; 57:2453826. [PMID: 39847394 PMCID: PMC11758805 DOI: 10.1080/07853890.2025.2453826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/18/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Angiogenesis is a complex physiological process. In recent years, the immune regulation of angiogenesis has received increasing attention, and innate immune cells, which are centred on macrophages, are thought to play important roles in vascular neogenesis and development. Various innate immune cells can act on the vasculature through a variety of mechanisms, with commonalities as well as differences and synergistic effects, which are crucial for the progression of vascular lesions. In recent years, monotherapy with antiangiogenic drugs has encountered therapeutic bottlenecks because of the short-term effect of 'vascular normalization'. The combination treatment of antiangiogenic therapy and immunotherapy breaks the traditional treatment pattern. While it has a remarkable curative effect and survival benefits, it also faces many challenges. This review focuses on innate immune cells and mainly introduces the regulatory mechanisms of monocytes, macrophages, natural killer (NK) cells, dendritic cells (DCs) and neutrophils in vascular lesions. The purpose of this paper was to elucidate the underlying mechanisms of angiogenesis and development and the current research status of innate immune cells in regulating vascular lesions in different states. This review provides a theoretical basis for addressing aberrant angiogenesis in disease processes or finding new antiangiogenic immune targets in inflammation and tumor.
Collapse
Affiliation(s)
- Jinjing Wu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yulu Qian
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Kuang Yang
- Queen Mary University of London, Nanchang University, Nanchang, China
| | - Shuhua Zhang
- Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Cardiovascular Research Institute, Nanchang, Jiangxi, China
| | - Erming Zeng
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Daya Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Lan X, Johnston E, Ning T, Chen G, Haglund L, Li J. Immunomodulatory bioadhesive technologies. Biomaterials 2025; 321:123274. [PMID: 40156979 DOI: 10.1016/j.biomaterials.2025.123274] [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: 11/12/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Bioadhesives have found significant use in medicine and engineering, particularly for wound care, tissue engineering, and surgical applications. Compared to traditional wound closure methods such as sutures and staples, bioadhesives offer advantages, including reduced tissue damage, enhanced healing, and ease of implementation. Recent progress highlights the synergy of bioadhesives and immunoengineering strategies, leading to immunomodulatory bioadhesives capable of modulating immune responses at local sites where bioadhesives are applied. They foster favorable therapeutic outcomes such as reduced inflammation in wounds and implants or enhanced local immune responses to improve cancer therapy efficacy. The dual functionalities of bioadhesion and immunomodulation benefit wound management, tissue regeneration, implantable medical devices, and post-surgical cancer management. This review delves into the interplay between bioadhesion and immunomodulation, highlighting the mechanobiological coupling involved. Key areas of focus include the modulation of immune responses through chemical and physical strategies, as well as the application of these bioadhesives in wound healing and cancer treatment. Discussed are remaining challenges such as achieving long-term stability and effectiveness, necessitating further research to fully harness the clinical potential of immunomodulatory bioadhesives.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Evan Johnston
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Tianqin Ning
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Pine Ave W, Montreal, Quebec, H3A 1A3, Canada
| | - Lisbet Haglund
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Shriners Hospital for Children, 1003 Decarie Blvd, Montreal, Quebec, H4A 0A9, Canada.
| | - Jianyu Li
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada.
| |
Collapse
|
3
|
Zhou X, Wang P, Xie L, Chan YK, Jiao Z, Shu R, Bai D, Lai S, Deng Y. Molybdoenzymes-emulating bio-heterojunction hydrogel with rapid disinfection and macrophage reprogramming for wound regeneration. Biomaterials 2025; 320:123284. [PMID: 40121831 DOI: 10.1016/j.biomaterials.2025.123284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Developing hydrogel dressings with the capabilities to accommodate irregular wounds and provide a cascade disinfective-regenerative microenvironment for wound repair is of great importance to combating pathogenic bacteria-infected wounds but remains an ongoing challenge. To address the conundrum, we devise a molybdoenzymes-emulating bio-heterojunction (M-bioHJ) doped double network (DN) hydrogel dressing for bacterial-infected wound healing. The near-infrared (NIR) photothermal effect of the M-bioHJ facilitates the exchange of multiple dynamic crosslinking sites in the hydrogel, endowing the hydrogel with photo-remote reprocessing capabilities to completely accommodate the encountered irregular wounds and ultimately accomplish the admirable therapeutic effect. Meanwhile, the introduced M-bioHJ shows NIR light-enhanced photodynamic activity to induce a massive engendering of reactive oxygen species (ROS), allowing rapid sterilization without reliance on exogenous hydrogen peroxide. Furthermore, the Mo ions released from the M-bioHJ-encapsulated hydrogel can play a crucial role in reprogramming the macrophage phenotype and determining tissue regeneration. Both in vitro and in vivo evidences authenticate the accelerated healing potential of infected wounds through the synergistic effects of photo-reprocessing, disinfection, and macrophage-reprogramming facilitated by the hydrogel. These findings highlight the promising application prospects of such neoteric M-bioHJ-encapsulated hydrogel dressings for wound disinfection and tissue regeneration.
Collapse
Affiliation(s)
- Xiong Zhou
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Peiqi Wang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lu Xie
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, 999077, Hong Kong, China
| | - Zheng Jiao
- Swanson School of Engineering, University of Pittsburgh, Pittsburgh, 15261, USA
| | - Rui Shu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shuangquan Lai
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, Guangdong 519000, China
| | - Yi Deng
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
4
|
Wu P, Jin L, Jiang W, Zhou Y, Lin L, Lin H, Chen H. Smart bandages for wound monitoring and treatment. Biosens Bioelectron 2025; 283:117522. [PMID: 40334449 DOI: 10.1016/j.bios.2025.117522] [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: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/09/2025]
Abstract
Wound management plays a crucial role in nursing care as it facilitates effective wound healing and prevents infections. To overcome limitations associated with traditional treatment methods, various smart bandages have been developed. The monitoring of wound parameters and the implementation of targeted treatments are crucial aspects of smart bandage development. Smart bandages, as cutting-edge flexible wearable medical devices, integrate various sensing technologies, providing new insights for dynamic monitoring and personalized treatment of chronic wounds. This paper systematically summarizes the applications and developments of smart bandages in monitoring wound environmental parameters, focusing on two major detection methods: colorimetric sensing and electrochemical sensing. Colorimetric sensors typically rely on color changes induced by physiological parameters, which can not only be identified by the naked eye but also combined with image recognition algorithms for physiological parameter detection. Electrochemical sensors, on the other hand, modify electrodes with specific enzymes and detect physiological parameters through the electrical signals generated by redox reactions. In addition to sensing, this paper further explores the integrated application of three smart therapeutic strategies in smart bandages, including promoting cell proliferation and angiogenesis through electrical stimulation, achieving controlled drug release via responsive materials, and utilizing photothermal materials for efficient antibacterial treatment of wounds. Finally, the paper delves into the challenges these bandages face in system integration and clinical translation, and discusses their potential in personalized wound care.
Collapse
Affiliation(s)
- Ping Wu
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Long Jin
- Department of Pathology, Fujian Provincial hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China
| | - Wanying Jiang
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Yingzhang Zhou
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Lisheng Lin
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Hongxin Lin
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China.
| | - Hu Chen
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
5
|
Ju R, Li Y, Sui D, Xu FJ. Polyaminoglycoside nanosystem expressing antimicrobial peptides for multistage chronic wound management. J Control Release 2025; 382:113657. [PMID: 40122239 DOI: 10.1016/j.jconrel.2025.113657] [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: 11/24/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Chronic wounds are difficult to heal due to pathogenic microbial colonization and dysregulation of healing cascades, necessitating novel therapeutic strategies. This study developed a multifunctional nanosystem by integrating the antimicrobial peptide LL37 with cationic polyaminoglycoside (SS-HPT), constructing a self-sustaining "AMP factory" to achieve multi-stage modulation of the wound healing. Validation through cell-level experiments and in vivo dual models (mechanical injury and bacterial infection) in immunocompromised rats demonstrated the system's unique dual intracellular-extracellular pathogen-killing capability, significantly accelerating the wound healing process. Transcriptomic analysis revealed that its mechanism involves the dual effects of suppressing pro-inflammatory factor expression and activating tissue repair pathways. Histological evidence confirmed that the system promotes angiogenesis, enhances re-epithelialization rates, and guides orderly collagen fiber deposition. This nanosystem, combining efficient AMP delivery and integrated therapeutic strategies, achieves three-dimensional synergy in microbial clearance, immune microenvironment regulation, and tissue matrix remodeling, providing theoretical and technical foundations for a paradigm shift in chronic wound treatment.
Collapse
Affiliation(s)
- Rui Ju
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education) and Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yang Li
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education) and Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Dandan Sui
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education) and Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education) and Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
6
|
He YT, Geng XY, Chang MY, Li FF, Du XL, Chen BZ, Guo XD. Harnessing innovation in microneedle technology for Women's healthcare. J Control Release 2025; 382:113706. [PMID: 40220870 DOI: 10.1016/j.jconrel.2025.113706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/17/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
Women's health management plays a crucial role in modern healthcare, encompassing the prevention, detection, and treatment of female diseases. However, existing technologies often face challenges, such as the invasiveness and discomfort associated with serological testing and injection-based therapies. Microneedles, as an emerging technology in biomedical engineering, demonstrate significant advantages. These micron-sized transdermal devices are applicable in a range of applications, from drug delivery to interstitial fluid sampling, and their painless, minimally invasive nature significantly enhances medication compliance. In recent years, microneedles have been widely utilized in women's health management, showing promising results in early disease prevention and subsequent treatment. Although there are reviews about microneedles applied in disease treatment management, few of them focus on the application of microneedles in the prevention and early detection of women's disease. Herein, we present a comprehensive overview of the current application status of microneedles in women's health management, with a special emphasis on their design and mechanism for disease prevention, and treatment in women. Finally, we discuss the advantages and limitations of microneedles in women's health management, and propose suggestions for future research direction.
Collapse
Affiliation(s)
- Yu Ting He
- State Key Laboratory of Organic-Inorganic Composites (Beijing University of Chemical Technology), Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xin Yao Geng
- State Key Laboratory of Organic-Inorganic Composites (Beijing University of Chemical Technology), Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ming Yu Chang
- State Key Laboratory of Organic-Inorganic Composites (Beijing University of Chemical Technology), Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Fei Fei Li
- State Key Laboratory of Organic-Inorganic Composites (Beijing University of Chemical Technology), Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xin Ling Du
- State Key Laboratory of Organic-Inorganic Composites (Beijing University of Chemical Technology), Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Bo Zhi Chen
- State Key Laboratory of Organic-Inorganic Composites (Beijing University of Chemical Technology), Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xin Dong Guo
- State Key Laboratory of Organic-Inorganic Composites (Beijing University of Chemical Technology), Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
7
|
Zhou X, Chen S, Savitz B, Yu N, Perdikis G, Duckworth K, Dean Y, Long X, Lineaweaver W. Comparative efficacy of different functional hydrogel dressings in healing diabetic foot ulcer: A systematic review and network meta-analysis. Diabetes Obes Metab 2025; 27:3431-3441. [PMID: 40197692 DOI: 10.1111/dom.16367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 04/10/2025]
Abstract
AIMS Functional hydrogel dressings offer a promising therapeutic approach, and optimizing their formulations is crucial for improving diabetic foot ulcers (DFUs) outcomes. This study explores the comparative efficacy of different functional hydrogel dressings in DFUs treatment. MATERIALS AND METHODS We conducted a systematic review and Bayesian network meta-analysis of randomized controlled trials evaluating functional hydrogel dressings for DFUs treatment. A comprehensive search was performed across PubMed, Embase, CENTRAL, CNKI and Web of Science from inception to June 2024. Bayesian network meta-analysis was employed to synthesize and compare the relative efficacy of hydrogel interventions, defined as the number of patients with complete wound closure. RESULTS In total, 23 studies involving 1671 patients with DFUs were included. The analysis revealed that immuno-regulating hydrogels (IRHs) had the highest effect estimate (2.2, 95% CI: 1.6, 3.2), compared with anti-bacterial hydrogels (ABHs) ranked last (1.3, 95% CI: 0.78, 2.3). Multi-functional hydrogels (MFHs) and proliferation-promoting hydrogels (PPHs) displayed intermediate effects (1.7, 95% CI: 1.2, 2.4). The relative efficacy ranking was IRH > MFH/PPH > ABH > placebo. The risk of adverse events was lower in functional hydrogel groups relative to placebo (0.75, 95% CI: 0.56, 0.96). Node-splitting analysis confirmed the consistency between direct and indirect evidence for IRH versus ABH. A funnel plot analysis indicated no significant publication bias, affirming the robustness of our findings. CONCLUSION This study provides a comprehensive evaluation of functional hydrogel dressings for DFUs treatment, highlighting the potential of IRH as the most effective option. These insights will guide future research and clinical applications to improve DFUs management.
Collapse
Affiliation(s)
- Xiaozhen Zhou
- Peking Union Medical College Hospital, Beijing, China
| | - Shida Chen
- Peking Union Medical College Hospital, Beijing, China
| | - Benjamin Savitz
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nanze Yu
- Peking Union Medical College Hospital, Beijing, China
| | - Galen Perdikis
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kylie Duckworth
- Medical University of South Carolina College of Medicine, Charleston, South Carolina, USA
| | - Yomna Dean
- Alexandria University, Alexandria, Egypt
| | - Xiao Long
- Peking Union Medical College Hospital, Beijing, China
| | | |
Collapse
|
8
|
Zheng Y, Li Q, Jin X, Zhu M, Liang Q, Wu Y, Pan F, Qiu H, Wang X, Lu D, Huang H. W-GA nanodots with multienzyme activities alleviate the inflammatory microenvironment in the treatment of acute wounds. Mater Today Bio 2025; 32:101662. [PMID: 40166380 PMCID: PMC11957797 DOI: 10.1016/j.mtbio.2025.101662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
Acute wounds present a significant clinical challenge due to delayed healing, which is often exacerbated by elevated levels of reactive oxygen species (ROS). These high ROS concentrations hinder the natural healing process, leading to prolonged recovery and increased risk of complications. W-GA nanodots, synthesized via a simple coordination method, have emerged as promising solutions, demonstrating multifunctional enzymatic activity that effectively scavenges ROS. To explore the underlying mechanisms of ROS-induced oxidative stress, we conducted RNA sequencing on macrophages exposed to H2O2. The results revealed significant regulation of key stress response pathways, including substantial upregulation of the "p53 signaling pathway" and the "HIF-1 signaling pathway," both of which are essential for cellular adaptation to oxidative stress. By alleviating oxidative stress, W-GA nanodots not only accelerate wound repair but also improve overall healing outcomes. Notably, RNA sequencing of animal tissue samples revealed that W-GA nanodots activate the "Wnt signaling pathway," further promoting wound healing. These findings underscore the potential of W-GA nanodots as a novel therapeutic strategy for enhancing wound healing and treating oxidative stress-related conditions, positioning them as promising candidates for future clinical applications in wound care and inflammatory diseases.
Collapse
Affiliation(s)
- Yang Zheng
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Qingrong Li
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, PR China
| | - Xu Jin
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Mengmei Zhu
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, PR China
| | - Qian Liang
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Yingjie Wu
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Fuqiang Pan
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Houhuang Qiu
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, PR China
| | - Decheng Lu
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| | - Huiqiao Huang
- Research Center of Nanomedicine Technology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, PR China
| |
Collapse
|
9
|
Liang X, Chen H, Zhang R, Xu Z, Zhang G, Xu C, Li Y, Zhang L, Xu FJ. Herbal micelles-loaded ROS-responsive hydrogel with immunomodulation and microenvironment reconstruction for diabetic wound healing. Biomaterials 2025; 317:123076. [PMID: 39805188 DOI: 10.1016/j.biomaterials.2024.123076] [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: 11/12/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Persistent inflammation is a major cause of diabetic wounds that are difficult to heal. This is manifested in diabetic wounds with excessive reactive oxygen clusters (ROS), advanced glycation end products (AGE) and other inflammatory factors, and difficulty in polarizing macrophages toward inhibiting inflammation. Berberine is a natural plant molecule that inhibits inflammation; however, its low solubility limits its biological function through cytosis. In this study, we designed F127 micelles to encapsulate berberine with the aim of improving its solubility and bioavailability. Meanwhile, in order to achieve effective drug delivery at the wound site, we designed an injectable ferrocene-cyclodextrin self-assembled oxidation-reactive supramolecular hydrogel drug delivery system. Cellular experiments have shown that the hydrogel can reduce intracellular ROS and AGE production, attenuate cellular damage, promote macrophage polarization toward inhibition of inflammation, and reduce the secretion of inflammatory factors. In an animal model of diabetic mice, this hydrogel dressing reduces the level of inflammation in diabetic wounds, optimizes collagen deposition in diabetic wounds, and ultimately achieves high-quality diabetic wound healing. The work offers a straightforward and effective solution to the challenge of administering hydrophobic anti-inflammatory agents in the context of diabetic wound therapy.
Collapse
Affiliation(s)
- Xiaoyang Liang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Honggui Chen
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Rui Zhang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zhixuan Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Guo Zhang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Chen Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yang Li
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Lei Zhang
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
10
|
Feng L, Peng Q, Miao L, Cai C, Tay FR, Zhou S, Zhang Y, Liu Z, Wang X, Jiao Y, Guo R. "Monitor-and-treat" that integrates bacterio-therapeutics and bio-optics for infected wound management. Bioact Mater 2025; 48:118-134. [PMID: 40034807 PMCID: PMC11872670 DOI: 10.1016/j.bioactmat.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 03/05/2025] Open
Abstract
Wound infections are one of the major threats to human health, accounting for millions of deaths annually. Real-time monitoring, accurate diagnosis, and on-demand therapy are crucial to minimizing complications and saving lives. Herein, we propose a "monitor-and-treat" strategy for infected wound management by integrating the emerging development of bacterio-therapeutics and bio-optics. The upper layer consists of gelatin methacryloyl (GelMA)-collagen III methacryloyl (Col3MA) (GC), Reuterin (Reu) isolated from the probiotic Lactobacillus reuteri (L. reuteri) and microfluidic safflower polysaccharide (SPS)@GelMA microspheres using 3D printing technology. The lower layer is made of acryloylated glycine (ACG) hydrogel with tissue adhesion capability, which enables the hydrogel to adapt to the movement and stretching of the skin. By integrating temperature-sensitive polydimethylsiloxane (PDMS) optical fibers, the ACG-GC/Reu/SPS-PDMS hydrogel could accurately and steadily sense and send wound temperature information to intelligent devices for real-time monitoring of the healing status ("monitor"). The double-layered hydrogel not only inhibited bacterial survival and colonization (97.4 % against E. coli and 99 % against S. aureus), but also exhibited remarkable hemostatic properties. Furthermore, it was conducive to L929 cell proliferation and pro-angiogenesis, and promoted the polarization of pro-inflammatory M1 macrophages to the anti-inflammatory M2-phenotype, therefore creating a favorable immune microenvironment at the wound site. Animal experiments using SD rats and Bama minipigs demonstrated that this hydrogel promoted wound closure, directed polarization to M2 macrophages, alleviated inflammation, enhanced neovascularization, therefore accelerating infected wound healing ("treat"). In addition, RNA-Seq analysis revealed the mechanism of action of ACG-GC/Reu/SPS-PDMS hydrogel in modulating key signaling pathways, including down-regulation of AMPK, IL-17, and NF-κB signaling pathways, activation of NLRP3 inflammatory vesicles, and enrichment of MAPK, TGF-β, PI3K-Akt, TNF, and VEGF signaling pathways. The modulation of these signaling pathways suggests that hydrogels play an important role in the molecular mechanisms that promote wound healing and tissue regeneration. Therefore, the design of this study provides an innovative and multifunctional bandage strategy that can significantly improve pathologic diagnosis and wound treatment.
Collapse
Affiliation(s)
- Longbao Feng
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrie Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, PR China
| | - Qing Peng
- Central Laboratory of the Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, PR China
| | - Li Miao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, PR China
| | - Chenghao Cai
- Department of Burns & Wound Care Center, The 2nd Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310009, PR China
| | - Franklin R. Tay
- The Graduate School, Augusta University, Augusta, GA, 30912, USA
| | - Shuqin Zhou
- Department of Anesthesiology of the Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, PR China
| | - Ying Zhang
- Central Laboratory of the Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, PR China
| | - Zonghua Liu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrie Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, PR China
| | - Xingang Wang
- Department of Burns & Wound Care Center, The 2nd Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310009, PR China
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, PR China
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Centre for Drug Carrie Development, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, PR China
| |
Collapse
|
11
|
Feng G, Zhou X, Fang X, He Y, Lin T, Mu L, Yang H, Wu J. A non-bactericidal glycine-rich peptide enhances cutaneous wound healing in mice via the activation of the TLR4/MAPK/NF-κB pathway. Biochem Pharmacol 2025; 236:116912. [PMID: 40164342 DOI: 10.1016/j.bcp.2025.116912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Although the antibacterial properties of glycine-rich peptides from prokaryotes to eukaryotes have been well characterized, their role in skin wound healing remains poorly understood, especially non-bactericidal glycine-rich peptides. Herein, a novel glycine-rich (46.5%) peptide (Smaragin, SRGSRGGRGGRGGGGRGGRGRSGSGSSIAGGGSRGSRGGSQYA) was identified from the skin of the tree frog Zhangixalus smaragdinus. Unlike other glycine-rich peptides, Smaragin showed no antimicrobial activity in vitro but significantly enhance wound healing in full-thickness dermal wounds in mice. In comparison with other wound healing-promoting peptides, Smaragin did not directly affect the proliferation and migration of keratinocytes, vascular endothelial cells, and fibroblasts. However, it notably increased phagocytes infiltration at the wound site by 0.5-day post-injury. Smaragin was not a direct chemoattractant for phagocytes, but it stimulated macrophages to secrete chemokines CXCL1 and CXCL2, which indirectly enhanced the migration of phagocytes, keratinocytes and vascular endothelial cells. Moreover, Smaragin promoted the polarization of macrophages from a pro-inflammatory M1-type to an anti-inflammatory M2 phenotype at the wound, which is associated with angiogenic activity. As expected, CD31, the most common analyzed marker of angiogenesis, showed a significant increase in vascular network area. Subsequent studies revealed that Smaragin promoted the chemokine level and polarization of macrophages via the TLR4/MAPK/NF-κB pathway, which enhanced the number of phagocytes and the regeneration of the epidermis and blood vessels at the wound, thereby accelerating skin wound healing in mice. These findings highlight the skin healing properties of non-bactericidal glycine-rich peptides and display the potential of Smaragin as a promising candidate for developing effective wound healing therapies.
Collapse
Affiliation(s)
- Guizhu Feng
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Xiaoyan Zhou
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Xiaojie Fang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Yanmei He
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Ting Lin
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Lixian Mu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China.
| | - Hailong Yang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China.
| | - Jing Wu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
12
|
Yang M, Xu Y, Cheng Q, He Y, Xu Z, Mu C, Ge L, Li D. Injectable Polysaccharide-Based Hydrogel with Glucose Responsiveness as an Immunoregulatory Platform for Enhanced Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40402095 DOI: 10.1021/acsami.5c06112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Persistent excessive inflammatory response in diabetic wounds caused by the imbalance of the immune microenvironment leads to delayed or nonhealing of the wounds. Timely attenuation of inflammation through immunoregulation is a crucial strategy to accelerate diabetic wound closure. Here, the protocatechuic acid (PCA)- and deferoxamine (DFO)-loaded polysaccharide-based immunoregulatory hydrogel (POCP@D) was developed by the dual-cross-linking strategy of borate ester bonds and imine bonds to promote advanced healing of full-thickness diabetic wounds. The POCP@D hydrogel showed good tissue adhesiveness property, flexibility, mechanical strength, injectability, self-healing, and glucose-responsive drug release properties, besides strong broad-spectrum antibacterial and antioxidant activities. The POCP@D hydrogel acted as an immunoregulatory platform to remold the in vivo immune microenvironment of diabetic wounds by regulating the M2-type macrophage polarization and timely relieving wound inflammation, thus promoting collagen deposition, angiogenesis, and the development of diabetic wounds from the inflammatory stage to the proliferative stage and ultimately achieving high-quality skin tissue regeneration. Overall, our developed immunoregulatory hydrogel held great potential for refractory diabetic wound therapy in clinical settings.
Collapse
Affiliation(s)
- Minxuan Yang
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yongbin Xu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, P. R. China
| | - Qingsu Cheng
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Yiruo He
- West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Zhilang Xu
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Changdao Mu
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Liming Ge
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Defu Li
- Department of Pharmaceutics and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
13
|
Lu J, Guo Y, Yang Z, Xie M, Zhang S, Li K, Yang J, Xue S, Xu D, Yan K, Liu Y. Calycosin-7-glucoside-Loaded Hydrogel Promotes Wound Healing in Gestational Diabetes Mellitus. ACS APPLIED BIO MATERIALS 2025; 8:4186-4199. [PMID: 40300146 DOI: 10.1021/acsabm.5c00290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
The prevalence of gestational diabetes mellitus (GDM) is currently on the rise globally, which heightens the risk of adverse pregnancy outcomes and subsequently increases the likelihood of cesarean delivery. GDM can induce hyperglycemic conditions in cesarean wounds, leading to delayed wound healing and complications such as itching, pain, and scarring. These complications significantly impact the quality of life and mental health of mothers. Furthermore, there is a lack of effective clinical prevention strategies. Consequently, the need to improve wound healing after cesarean sections in women with GDM is a pressing concern that warrants our attention. To maximize the therapeutic impact and extend the bioavailability of calycosin-7-glucoside (CG), it was integrated into a hybridized hydrogel (GOHA) as a drug carrier to create the GOHACG hydrogel. Bases on our tests, the GOHACG hydrogel demonstrated a strong capacity for water absorption, appropriate pore size, and good biocompatibility to adjust to the in situ surroundings of the wound. GOHACG also promoted epidermal regeneration, collagen deposition, angiogenesis, and the conversion of macrophages from the M1 to M2 phenotype. Indicating a reduction in the inflammatory response, accelerated wound repair, and minimized skin scarring in a postcesarean delivery model involving gestational diabetic mellitus mice. In brief, the GOHACG possesses significant properties that enhance wound healing in GDM model, suggesting its potential effects in treating wound healing of GDM.
Collapse
Affiliation(s)
- Jicong Lu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yingying Guo
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Zhaoyuan Yang
- The 988 Hospital of the Joint Service Support Force of the Chinese People's Liberation Army, Zhengzhou 450000, China
| | - Mengxia Xie
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shuangyu Zhang
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Keji Li
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jingjing Yang
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shanhui Xue
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Da Xu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Kanglu Yan
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yuehua Liu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
14
|
Abdollahzadeh N, Vatanchian M, Oroojalian F, Enderami SE, Amani A, Salarinia R. Fibromodulin-overexpressing fibroblast cells increase wound contraction, improve scar quality and enhance angiogenesis: an in-vivo study. BMC Biotechnol 2025; 25:40. [PMID: 40389960 PMCID: PMC12090437 DOI: 10.1186/s12896-025-00975-5] [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: 08/05/2024] [Accepted: 05/05/2025] [Indexed: 05/21/2025] Open
Abstract
INTRODUCTION Fibromodulin, a small leucine rich proteoglycan has been suggested to have prominent role in wound healing. On the other hand, fibroblast cells, due to their ability to secrete growth factors and control inflammation in the wound area, have been proposed as effective approaches in cell therapy for wounds. In the current study we attempted to improve treatment results using a combination of fibroblast and fibromodulin features. METHOD Fibroblast cells were isolated from the skin and transfected with a vector carrying the fibromodulin gene. Following the assessment of fibromodulin protein production, the effect of transfected fibroblast cells was studied in an animal wound model. RESULTS Flow cytometry analysis showed high expression of the CD90 marker (97.2%) and very low expression of the CD34 marker (0.47%). Additionally, enzyme-linked immunosorbent assay (ELISA) findings confirmed high expression of the fibromodulin gene in the transfected fibroblast cells. In vivo studies demonstrated that the animals treated with fibroblast cells transfected with fibromodulin (V + G+) exhibited significantly improved wound contraction on day 7 (i.e., contraction percentage: 21.79 ± 9.96%, compared with 7.23 ± 2.30% in the PBS-treated group). Histopathological studies also indicated improvements in angiogenesis score and collagen density score in the animals treated with the V + G + group. CONCLUSION The results of this study showed that fibroblast cells expressing the fibromodulin gene improve wound contraction and some histological parameters in the deep wound model of the rat.
Collapse
Affiliation(s)
- Negar Abdollahzadeh
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehran Vatanchian
- Department of Anatomical Sciences and Pathology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Ehsan Enderami
- Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Medical Biotechnology, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amir Amani
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Reza Salarinia
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| |
Collapse
|
15
|
Khajornpipat P, Reamtong O, Aunpad R. Rational engineering unlocks the therapeutic potential of WHP1: A revolutionary peptide poised to advance wound healing. PLoS One 2025; 20:e0323363. [PMID: 40367225 PMCID: PMC12077786 DOI: 10.1371/journal.pone.0323363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Treatment of chronic or non-healing wounds has faced a considerable clinical challenge and impose several detrimental effects on individuals, society, the healthcare system, and the economy. Bioactive peptides have been employed to accelerate wound healing in active wound treatment efficiently and effectively. In the current study, a novel wound-healing peptide, WHP1, was designed from 23 existing wound-healing peptides by a rational template-assisted approach. It demonstrated the ability to enhance migration and proliferation of human keratinocyte cell lines (HaCaT) without exhibiting cytotoxic effects on human red blood cells and HaCaT cells. By quantitative proteomic analysis, WHP1 exerted a multifaceted role on diverse cellular processes in human keratinocyte. Notably, it increased the expression of intracellular proteins of HaCaT cells involved in cell cycle regulation and focal adhesion, including centromeric histone H3 variant CENPA, ubiquitin-conjugating enzyme E2 C, thyroid receptor-interacting protein 6, and ribosomal components essential for cell adhesion and migration. WHP1 upregulated the key enzyme glyceraldehyde-3-phosphate dehydrogenase, orchestrating metabolic biosynthesis particularly glycolysis, cell cycle regulation, and cytoskeletal processes. An intriguing observation was the antioxidant activity of WHP1, protecting cells from reactive oxygen species-induced senescence. This is consistent with the upregulation of GAPDH expression and reduction of histone H2A.J levels. WHP1 also stimulated macrophages to secrete transforming growth factor-β (TGF-β), a crucial growth factor necessary for the remodeling phase of wound healing. This investigation highlighted the feasibility of rational design to create novel wound-healing peptides. Such advancements hold promise for improving patients' quality of life and elevating the standard of care in contemporary healthcare.
Collapse
Affiliation(s)
- Patcharin Khajornpipat
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Ratchaneewan Aunpad
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| |
Collapse
|
16
|
Mao H, Jiang X, Liang J, Zhang L, Yang Z, Chen Z, Qiao J, An X, Li X, Xie G, Liu HW, Xiao L. FOSL1 promotes keratinocyte migration and wound repair by modulating the IL17 signaling pathway. Sci Rep 2025; 15:16457. [PMID: 40355666 PMCID: PMC12069625 DOI: 10.1038/s41598-025-99128-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Abstract
Keratinocytes, the most important cell type constituting the epidermis, migrate to restore the epithelial barrier during wound healing and are a crucial step in wound healing. This study utilized bioinformatics analysis of comprehensive expression datasets of aberrantly expressed genes in wound healing to identify the abnormal expression of the critical transcription factor Fos-like antigen-1 (FOSL1), which is involved in various diseases. Currently, there is limited research on the role of FOSL1 in wound healing, and its molecular mechanisms remain unclear. This study explores the role and regulatory mechanisms of FOSL1 in the wound-healing process. A comprehensive expression dataset of abnormal genes in wound repair was constructed by bioinformatics analysis. Mouse trauma models and mouse wound splint models were constructed to verify the role of FOSL1 in vivo. Real-time quantitative polymerase chain reaction (qRT-PCR), immunoblot, immunofluorescence staining, and HE staining were used to confirm the analysis, and FOSL1 was used as the target in the wound healing process. At the cellular level, using 5'-ethynyl-2'-deoxyuridine (EdU) assay, Transwell assay, Migration assay, western blotting and immunofluorescence, FOSL1 promoted the molecular mechanism of wound repair by regulating the proliferation and migration of keratinocytes through IL-17 signaling pathway. Bioinformatics analysis revealed differential expression of FOSL1 during wound healing. In the mouse back wound model, qRT-PCR, western blotting (WB), and immunofluorescence staining showed significant upregulation of FOSL1 and IL-17 expression during wound tissue healing, indicating a close association between FOSL1 and mouse wound healing. In the mouse wound splinting model, subcutaneous injection of recombinant FOSL1 protein contributed to wound surface healing. Overexpression of FOSL1 in HaCaT cells promoted their proliferation and migration abilities. When IL-17 inhibitor was added to HaCaT cells, both FOSL1 overexpression and knockdown inhibited the proliferation and migration abilities of HaCaT cells. Thus, this study confirms that FOSL1 promotes keratinocyte proliferation and migration through the IL-17 signaling pathway, facilitating wound healing in epidermal wound repair. The results of this study indicate that FOSL1 plays a key role in epidermal wound healing, and elucidate a new molecular mechanism by which FOSL1 promotes keratinocyte proliferation and migration through IL-17 signaling pathway in epidermal wound repair, thereby promoting wound healing.
Collapse
Affiliation(s)
- Haoran Mao
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
- Key Laboratory of Regenerative Medicine, Innovative Technology Research Institute of Tissue Repair and Regeneration, Ministry of Education, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Xiao Jiang
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
- Key Laboratory of Regenerative Medicine, Innovative Technology Research Institute of Tissue Repair and Regeneration, Ministry of Education, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Jiaji Liang
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
- Key Laboratory of Regenerative Medicine, Innovative Technology Research Institute of Tissue Repair and Regeneration, Ministry of Education, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Lei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Bengbu Medical University, Bengbu, 233080, Anhui, People's Republic of China
| | - Zixian Yang
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
- Key Laboratory of Regenerative Medicine, Innovative Technology Research Institute of Tissue Repair and Regeneration, Ministry of Education, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Zhijing Chen
- Medical Cosmetic Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, People's Republic of China
| | - Jinlong Qiao
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
- Key Laboratory of Regenerative Medicine, Innovative Technology Research Institute of Tissue Repair and Regeneration, Ministry of Education, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Xifeng An
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
- Key Laboratory of Regenerative Medicine, Innovative Technology Research Institute of Tissue Repair and Regeneration, Ministry of Education, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Xuangu Li
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
- Key Laboratory of Regenerative Medicine, Innovative Technology Research Institute of Tissue Repair and Regeneration, Ministry of Education, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Guanghui Xie
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Hong-Wei Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China.
- Key Laboratory of Regenerative Medicine, Innovative Technology Research Institute of Tissue Repair and Regeneration, Ministry of Education, Guangzhou, 510630, Guangdong, People's Republic of China.
| | - Liling Xiao
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China.
| |
Collapse
|
17
|
Cheng M, Hou Y, Chen Q, Ge S, Chen C, Zheng X, Zhang C. Exploring the wound-healing mechanism of Cayratia japonica extract: A combined experimental and network pharmacology study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119810. [PMID: 40239878 DOI: 10.1016/j.jep.2025.119810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wound healing is a complex biological process and remains a significant challenge due to the lack of effective therapeutic drugs. Cayratia japonica (CJG), a traditional folk medicine, has been widely used for its anti-inflammatory and efficacy in treating traumatic injuries. AIM OF THE STUDY This study aimed to investigate the wound-healing effects of CJG and elucidate its underlying mechanism. METHODS First, the phytochemical composition of CJG was identified using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS), and its potential wound-healing mechanisms were predicated via network pharmacology. Next, in vivo experiments were conducted by dividing subjects into control, CJG (1.5-6 mg/cm2), and bFGF (150 IU/cm2) groups to assess its therapeutic efficacy. Finally, the mechanism of CJG and its key bioactive component, luteolin-7-O-glucoside (LUT-7G), were explained through polymerase chain reaction (PCR), Western blotting, histopathology, immunofluorescence, plasmid transfection, colony formation unit assays, and cellular thermal shift assay (CETSA). RESULTS LC-MS/MS identified 15 major constituents of CJG and 102 potential wound healing-related targets. Network pharmacology analysis revealed key enriched pathways, including AMPK, TNF, and metabolic pathways. In vivo, CJG significantly accelerated wound-healing by inhibiting inflammatory responses, promoting angiogenesis, and modulating collagen deposition. In vitro, LUT-7G treatment markedly enhanced the proliferation and migration of HaCaT and HSF cells. Mechanistically, LUT-7G exerted its wound-healing effects by activating the AMPK/CTHRC1/TGF-β1 signaling pathway in HaCaT cells. In conclusion, CJG significantly promotes wound healing by regulating AMPK signaling pathways, indicating its promising clinical application prospects.
Collapse
Affiliation(s)
- Mengqin Cheng
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Yi Hou
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 21000, China
| | - Qi Chen
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Shanchun Ge
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Ce Chen
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Xueping Zheng
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 21000, China.
| | - Chaofeng Zhang
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
18
|
Gong T, Fan X, Wu M, Chen Z, Xia Z. Post-transcriptional regulation of diabetic wound healing by junctional adhesion molecule A/miR-106b axis. Burns 2025; 51:107527. [PMID: 40359641 DOI: 10.1016/j.burns.2025.107527] [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: 11/24/2024] [Revised: 04/25/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Despite advancements in molecular science and biomaterial technology, the mechanisms underlying the impaired healing of diabetic wounds remain unclear. In this study, we investigated the post-transcriptional regulation of diabetic wound healing using JAM-A. METHODS Mouse wound models, hematoxylin and eosin staining analysis, and scratch wound assays were used to investigate the effects of JAM-A 3'-UTR on the re-epithelialization of diabetic wounds, whereas RNA pulldown, microRNA-seq, and bioinformatics analyses were performed to identify key miRNA players and predict their target genes. In situ hybridization, immunohistochemistry, western blotting, and polymerase chain reaction (PCR) were used to confirm the alternative splicing of JAM-A 3'-UTR in diabetic conditions. CCK-8 proliferation assays, scratch wound assays, PCR, western blotting, and dual-luciferase assays were performed to study the changes in cell proliferation and migration induced by miR-106b-5p modification and confirm the target gene. RESULTS JAM-A 3'-UTR accelerated re-epithelialization in diabetic mouse wounds. Shortened splicing was found in the 3'-UTR of JAM-A under diabetic conditions, leading to the excessive release of miR-106b-5p while the promoter of miR-106b was activated. Furthermore, upregulated miR-106b-5p over-activated cell proliferation and inhibited cell migration in diabetic wound keratinocytes by suppressing the target gene PTEN/TIAM1 and regulating the AKT and RAC1 pathways, thereby impairing wound re-epithelialization. CONCLUSIONS We identified alternative splicing of JAM-A 3'-UTR in diabetic conditions, which caused the excessive release of miR-106b. Upregulation of miR-106b reduced the expression of its target genes, PTEN and TIAM1, which led to hyperactive proliferation and impaired migration of keratinocytes, thereby dysregulating wound re-epithelialization.
Collapse
Affiliation(s)
- Teng Gong
- Burn & Wound Repair Department, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Burn Institute, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Burn Medical Center, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China
| | - Xiaoming Fan
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, No. 168 Changhai Road, Shanghai 200082, China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200082, China
| | - Minjuan Wu
- Department of Histology and Embryology, Naval Medical University, No. 800 Xiangyin Road, Shanghai 200082, China.
| | - Zhaohong Chen
- Burn & Wound Repair Department, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Burn Institute, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Burn Medical Center, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China.
| | - Zhaofan Xia
- Burn & Wound Repair Department, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Burn Institute, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Burn Medical Center, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, Fujian 350001, China; Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, No. 168 Changhai Road, Shanghai 200082, China; Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200082, China.
| |
Collapse
|
19
|
Petit N, Gomes A, Chang YYJ, Da Silva J, Leal EC, Carvalho E, Gomes P, Browne S. Development of a bioactive hyaluronic acid hydrogel functionalised with antimicrobial peptides for the treatment of chronic wounds. Biomater Sci 2025. [PMID: 40331923 DOI: 10.1039/d5bm00567a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Chronic wounds present significant clinical challenges due to delayed healing and high infection risk. This study presents the development and characterisation of acrylated hyaluronic acid (AcHyA) hydrogels functionalised with gelatin (G) and the antimicrobial peptide (AMP) PP4-3.1 to enhance cellular responses while providing antimicrobial activity. AcHyA-G and AcHyA-AMP hydrogels were formed via thiol-acrylate crosslinking, enabling in situ AcHyA hydrogel formation with stable mechanical properties across varying gelatin concentrations. Biophysical characterisation of AcHyA-G hydrogels showed rapid gelation, elastic behaviour, uniform mesh size, and consistent molecular diffusion across all formulations. Moreover, the presence of gelatin enhanced stability without affecting the hydrogel's degradation kinetics. AcHyA-G hydrogels supported the adhesion and spreading of key cell types involved in wound repair (dermal fibroblasts and endothelial cells), with 0.5% gelatin identified as the optimal effective concentration. Furthermore, the conjugation of the AMP conferred bactericidal activity against Staphylococcus aureus and Escherichia coli, two of the most prevalent bacterial species found in chronically infected wounds. These results highlight the dual function of AcHyA-AMP hydrogels in promoting cellular responses and antimicrobial activity, offering a promising strategy for chronic wound treatment. Further in vivo studies are needed to evaluate their efficacy, including in diabetic foot ulcers.
Collapse
Affiliation(s)
- Noémie Petit
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland.
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Ana Gomes
- LAQV-REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Portugal
| | - Yu-Yin Joanne Chang
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland.
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Jessica Da Silva
- University of Coimbra, Institute of Interdisciplinary Research, Doctoral Program in Experimental Biology and Biomedicine (PDBEB), 3004-504 Coimbra, Portugal
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ermelindo C Leal
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Eugénia Carvalho
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paula Gomes
- LAQV-REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Portugal
| | - Shane Browne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Ireland.
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
20
|
Lyu Z, Wu Y, Hu F, Zheng X, Ma D, Xu Z, Ding Y, Liu X, Huo S. Controlled release of ionic carrier hydrogels for sequential immunomodulation to facilitate stage-specific treatment of infectious wound. Biomaterials 2025; 322:123376. [PMID: 40349534 DOI: 10.1016/j.biomaterials.2025.123376] [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: 03/28/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025]
Abstract
Infected wounds present a significant clinical challenge, exacerbated by antibiotic resistance, which complicates effective treatment. This study introduces a hydrogel (CC/AP@CM) embedded with core-shell bioactive glass nanoparticles designed for the controlled, sequential release of copper (Cu2+) and magnesium (Mg2+) ions. The hydrogel is crosslinked via a Schiff base reaction, endowing it with injectable, self-healing, and adhesive properties. Notably, the bilayer structure of the bioactive glass within the hydrogel allows an initial release of Cu2+ ions to trigger an early-stage pro-inflammatory and antimicrobial response, followed by Mg2+ ions that support tissue repair and an anti-inflammatory environment. This design aligns with natural wound healing stages, promoting a shift in macrophage polarization from the M1 to M2 phenotype, effectively balancing antibacterial defense with tissue regeneration. The hydrogel demonstrated robust antibacterial efficacy against MRSA, increased angiogenesis, and enhanced fibroblast proliferation and migration in vitro. In a murine wound model, it significantly accelerated wound closure and immune activation, including responses from dendritic cells and T cells. These findings suggest that this hydrogel, through its stage-specific immunomodulatory properties and temporally controlled ion release, offers a promising strategy for treating complex wound infections, supporting both immune defense and tissue healing.
Collapse
Affiliation(s)
- Zhuocheng Lyu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yuezhou Wu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fei Hu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xu Zheng
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dajun Ma
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhenjiang Xu
- Department of Orthopedic Surgery Spine Center Changzheng Hospital Navy Medical University, Shanghai, China
| | - Yurun Ding
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Xuesong Liu
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiaotong University, China.
| | - Shicheng Huo
- Department of Orthopedic Surgery Spine Center Changzheng Hospital Navy Medical University, Shanghai, China.
| |
Collapse
|
21
|
Zhan Y, Sun H, Zhang Z, Chen X, Xu Z, He Y, Tao S, Fan L, Tan J. Chitosan and polyvinyl alcohol-based bilayer electrospun nanofibrous membrane incorporated with astaxanthin promotes diabetic wound healing by addressing multiple factors. Int J Biol Macromol 2025; 311:143921. [PMID: 40324499 DOI: 10.1016/j.ijbiomac.2025.143921] [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: 12/09/2024] [Revised: 04/27/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Delayed diabetic wound regeneration can be attributed to multiple underlying factors, including bacterial infection, endogenous reactive oxygen species (ROS), impaired angiogenesis and exaggerated inflammatory response. Here, a bilayer electrospun nanofibrous membrane (ENM) was fabricated through sequential electrospinning to accelerate diabetic wound healing by addressing aforementioned challenges. For the purpose, nano Zinc Oxide was mixed into chitosan as the bottom layer of ENM (CS/ZnO NPs), while astaxanthin (AST) was encapsulated in a composite nanofibrous membrane of polyvinyl alcohol, chitosan and Ti3C2TX MXene (PVA/CS/MXene) as the upper layer, thus preparing the bilayer CZ/PCM@AST ENM, which reflected the therapeutic properties of spatial structure distribution and time series on diabetic wounds. The bilayer CZ/PCM@AST ENM was verified to possess sufficient biocompatibility and effective antibacterial properties on E. coli and S. aureus. Furthermore, the ENM facilitated sustained AST release at inflammatory sites, effectively scavenging excessive ROS and inhibiting inflammatory responses, ultimately accelerating diabetic wound healing, as demonstrated through both in vitro and in vivo evaluations. In summary, the multi-effect combination strategy improved complicated pathological microenvironment of wound sites, thereby presenting a promising method in diabetic wound treatment.
Collapse
Affiliation(s)
- Yuhang Zhan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Huixuan Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 430070, China
| | - Zhihan Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Xi Chen
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Zhengping Xu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 430070, China
| | - Yifeng He
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Shengxiang Tao
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China.
| | - Lihong Fan
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 430070, China.
| | - Jinhai Tan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
22
|
Raju V, Reddy R, Javan AC, Hajihossainlou B, Weissleder R, Guiseppi-Elie A, Kurabayashi K, Jones SA, Faghih RT. Tracking inflammation status for improving patient prognosis: A review of current methods, unmet clinical needs and opportunities. Biotechnol Adv 2025; 82:108592. [PMID: 40324661 DOI: 10.1016/j.biotechadv.2025.108592] [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: 12/20/2024] [Revised: 04/07/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Inflammation is the body's response to infection, trauma or injury and is activated in a coordinated fashion to ensure the restoration of tissue homeostasis and healthy physiology. This process requires communication between stromal cells resident to the tissue compartment and infiltrating immune cells which is dysregulated in disease. Clinical innovations in patient diagnosis and stratification include measures of inflammatory activation that support the assessment of patient prognosis and response to therapy. We propose that (i) the recent advances in fast, dynamic monitoring of inflammatory markers (e.g., cytokines) and (ii) data-dependent theoretical and computational modeling of inflammatory marker dynamics will enable the quantification of the inflammatory response, identification of optimal, disease-specific biomarkers and the design of personalized interventions to improve patient outcomes - multidisciplinary efforts in which biomedical engineers may potentially contribute. To illustrate these ideas, we describe the actions of cytokines, acute phase proteins and hormones in the inflammatory response and discuss their role in local wounds, COVID-19, cancer, autoimmune diseases, neurodegenerative diseases and aging, with a central focus on cardiac surgery. We also discuss the challenges and opportunities involved in tracking and modulating inflammation in clinical settings.
Collapse
Affiliation(s)
- Vidya Raju
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA
| | - Revanth Reddy
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA
| | | | - Behnam Hajihossainlou
- Department of Infectious Diseases, Harlem Medical Center, and Columbia University, New York, 10032, NY, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Department of Systems Biology, Harvard Medical School, and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Anthony Guiseppi-Elie
- Department of Biomedical Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B), and Department of Electrical and Computer Engineering, Texas A & M University, College Station, 77843, TX, USA; Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, Houston, 77030, TX, USA; ABTECH Scientific, Inc., Biotechnology Research Park, Richmond, 23219, Virginia, USA
| | - Katsuo Kurabayashi
- Department of Mechanical and Aerospace Engineering, New York University, New York 11201, NY, USA
| | - Simon A Jones
- Division of Infection and Immunity, and School of Medicine, Cardiff University, UK; Systems Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Rose T Faghih
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA.
| |
Collapse
|
23
|
Khaity A, Albakri K, Al-dardery NM, Yousef YA, Foppiani JA, Lin SJ. Adipose-Derived Stem Cell Therapy in Hypertrophic and Keloid Scars: A Systematic Review of Experimental Studies. Plast Surg (Oakv) 2025; 33:318-328. [PMID: 40351807 PMCID: PMC12059430 DOI: 10.1177/22925503231195017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 06/23/2023] [Accepted: 07/09/2023] [Indexed: 05/14/2025] Open
Abstract
Introduction: Hypertrophic and keloid scars are abnormal tissue growth that can be disfiguring, for which the available treatment has not yielded consistent results. Therefore, this study aimed to evaluate the capability of Adipose tissue-derived stem cell (ADSC) therapy in treating these scars. Methods: A literature search was conducted on PubMed, Scopus, Cochrane Library, and Web of Science from inception until July 2022. We included experimental studies that evaluated ADSCs as a therapy for hypertrophic and keloid scars in both in-vivo and in-vitro models. Results: Our findings extracted from 12 included studies demonstrated that ADSCs have a promising potential in reducing collagen deposition, proliferation, and migration rates of fibroblast, decreasing gene/protein expression of scar-related molecules including levels of TGF-β1 and lowering intracellular signal pathway-related molecules of hypertrophic and keloid scars in both models. However, no significant difference (P > .05) was found in the hypertrophic scar in-vitro models in terms of DCN gene expression. Conclusion: Ultimately, the current studies included in this systematic review support the use of ADSCs to alleviate hypertrophic and keloid scars.
Collapse
Affiliation(s)
| | - Khaled Albakri
- Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | | | - Yaman A.S. Yousef
- Faculty of Medicine, Elrazi University, Khartoum, Sudan
- Faculty of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Jose A. Foppiani
- 1st Faculty of Medicine, Charles University, Prague, Czech Republic
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Samuel J. Lin
- Division of Plastic and Reconstructive Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
24
|
Kim YG, Lee Y, Oh HJ, Chu J, Min GH, Kim K, Lee J, Baik S, Kim D, Kim CK, Soh M, Namgoong S, Hyeon T. Ceria-Nanoparticle-Entangled Reticulation for Angiogenic and Therapeutic Embrocation for Multifactorial Approach to Treat Diabetic Wound. Adv Healthc Mater 2025; 14:e2404667. [PMID: 40143658 DOI: 10.1002/adhm.202404667] [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: 11/21/2024] [Revised: 03/06/2025] [Indexed: 03/28/2025]
Abstract
The therapeutic efficacy of a nanomedicine or a natural biomaterial can vary in different disorders due to their complex pathophysiology. A nanomedicine that is capable of not only targeting specific pathological cues through functional ligands but also optimizing the therapeutic efficacy of its components throughout the intricate pathways involved in complex disorders is highly desired. Here, ceria-nanoparticle-entangled reticulation for angiogenic and therapeutic embrocation (CERATE), composed of hyaluronic acid, levofloxacin, and the as-synthesized ceria nanoparticles is developed. CERATE is formulated in situ as a rigid nanoparticle-based network that integrates its components intimately using highly diluted concentrations, thereby augmenting the therapeutic efficiency of its individual components. The physical states of CERATE can be altered freely while retaining its integrity, by adjusting the water proportion to accommodate diverse clinical needs. This physically robust CERATE can withstand enzymatical degradation, display antibacterial activity, scavenge reactive oxygen species, and improve the migration and proliferation of fibroblasts by activating the proangiogenic factors. CERATE accelerates the repair of diabetic wounds by promoting both the angiogenesis and the synthesis of collagen. The results demonstrate the effectiveness of a multifactorial approach involving the recruitment of minimally modified biofunctional ligands and nanomaterials altogether with synergistic efficacy in treating complex disorders.
Collapse
Affiliation(s)
- Young Geon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yunjung Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyun Jyung Oh
- Department of Plastic Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
- Department of Biomedical Science, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Jinyoung Chu
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Gyeong Ho Min
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kang Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaeseon Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seungmin Baik
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dokyoon Kim
- Department of Bionano Engineering, Hanyang University, Ansan, 15588, Republic of Korea
| | - Chi Kyung Kim
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Min Soh
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Center for Advanced Pharmaceutical Technology, HyeonTechNBio Inc., Seoul, 08826, Republic of Korea
| | - Sik Namgoong
- Department of Plastic Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
- Institute of Advanced Regeneration and Reconstruction, Korea University College of Medicine, Seoul, 08308, Republic of Korea
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 770 Welch Road, Suite 400, Palo Alto, CA, 94304, USA
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
25
|
Hu H, Sheng Q, Yang F, Wu X, Zhang Y, Wu S, Liu Y, Hu N, Fu C, Leong J, Deng R, Jiang Z, Chen J, Wang Z, Chen C, Chen F, Luo Y, Zeng Y, Yu Y, Xie H, Wang G, Zou L. Enhanced Skin Wound Healing Through Chemically Modified Messenger RNA Encoding Epidermal Growth Factor (EGF). Int Wound J 2025; 22:e70143. [PMID: 40320617 PMCID: PMC12050261 DOI: 10.1111/iwj.70143] [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: 08/12/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 05/08/2025] Open
Abstract
Efficient wound healing remains a formidable medical challenge in clinical practice, due to the prevalence of skin defects arising from diverse etiological factors. It is indisputable that epidermal growth factor (EGF) plays a pivotal role in wound repair. However, its clinical application through recombinant proteins encounters challenges, including a short half-life in vivo and high production costs. Addressing these limitations, recent advancements in chemically modified mRNA (cmRNA) technologies offer a promising alternative. This study explores the utilisation of cmRNA in a biocompatible citrate-saline formulation to encode EGF for therapeutic purposes, capitalising on the advantages of cmRNA's inherent stability and the formulation's compatibility with biological systems. CmRNA demonstrated high transfection efficiency in human immortalised keratinocyte (HaCaT) and normal human dermal fibroblasts (NHDF) cells (93.97% ± 1.25% and 90.37% ± 0.97%, respectively), resulting in efficient production of biologically active EGF protein. In vitro, EGF cmRNA significantly promoted HaCaT and NHDF cell cycle, proliferation and migration. In vivo, in vivo imaging system (IVIS) imaging of murine skin confirmed localised and sustained expression of Luciferase cmRNA, with signals detectable up to 11 days post-injection. Immunohistochemistry revealed protein expression in both epidermal and dermal layers as early as 1 h post-injection, peaking at 48 h, further corroborated by enzyme-linked immunosorbent assay (ELISA). In a full-thickness skin defect mouse model, EGF cmRNA significantly accelerated wound healing, with superior re-epithelialisation observed compared to controls by Day 6. Mitogen-activated protein kinase (MEK)/Extracellular signal-regulated kinase (ERK) and Ki67 mRNA expression levels were markedly increased, both in vitro and in vivo. By Day 14, histological and immunohistochemical analyses revealed that EGF cmRNA outperformed recombinant human EGF (rhEGF), as indicated by enhanced formation of hair follicles and cutaneous glands, better-organised collagen fibres, and a reduced collagen Type I/III ratio. No adverse effects were observed in major organs, confirming cmRNA's biosafety. These results highlight the therapeutic potential of EGF-encoding cmRNA as an effective and safe alternative for enhancing wound healing.
Collapse
Affiliation(s)
- Haiyang Hu
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Qianglong Sheng
- Department of PharmacologyInnovative Institute of Basic Medical Sciences of Zhejiang UniversityZhejiangHangzhouChina
- Geneheal Pharmacy LtdZhejiangHangzhouChina
| | - Fan Yang
- Department of PharmacologyInnovative Institute of Basic Medical Sciences of Zhejiang UniversityZhejiangHangzhouChina
- Geneheal Pharmacy LtdZhejiangHangzhouChina
| | - Xinyi Wu
- Nanchang University Queen Mary SchoolJiangxiChina
| | - Youlai Zhang
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Shuling Wu
- Department of PharmacologyInnovative Institute of Basic Medical Sciences of Zhejiang UniversityZhejiangHangzhouChina
- Geneheal Pharmacy LtdZhejiangHangzhouChina
| | - Yihu Liu
- Department of PharmacologyInnovative Institute of Basic Medical Sciences of Zhejiang UniversityZhejiangHangzhouChina
- Geneheal Pharmacy LtdZhejiangHangzhouChina
| | - Ningyan Hu
- Department of PharmacologyInnovative Institute of Basic Medical Sciences of Zhejiang UniversityZhejiangHangzhouChina
- Geneheal Pharmacy LtdZhejiangHangzhouChina
| | - Chenhong Fu
- Department of PharmacologyInnovative Institute of Basic Medical Sciences of Zhejiang UniversityZhejiangHangzhouChina
- Geneheal Pharmacy LtdZhejiangHangzhouChina
| | - Jialin Leong
- Department of PharmacologyInnovative Institute of Basic Medical Sciences of Zhejiang UniversityZhejiangHangzhouChina
- Geneheal Pharmacy LtdZhejiangHangzhouChina
| | - Rufei Deng
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Zhenyu Jiang
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Jiaxin Chen
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Zhenxing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Hunan Key Laboratory of AngmedicineChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Chunyuan Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Hunan Key Laboratory of AngmedicineChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Fei Chen
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Yixuan Luo
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Yuanlin Zeng
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| | - Yin Yu
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Hunan Key Laboratory of AngmedicineChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Gang Wang
- Department of PharmacologyInnovative Institute of Basic Medical Sciences of Zhejiang UniversityZhejiangHangzhouChina
- Geneheal Pharmacy LtdZhejiangHangzhouChina
| | - Lijin Zou
- Jiangxi Provincial Key Laboratory of Trauma, Burn and Pain Medicine, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangJiangxiChina
| |
Collapse
|
26
|
Zhang Y, Zheng Z, Zhu S, Xu L, Zhang Q, Gao J, Ye M, Shen S, Xing J, Wu M, Xu RX. Electroactive Electrospun Nanofibrous Scaffolds: Innovative Approaches for Improved Skin Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416267. [PMID: 40190057 PMCID: PMC12079356 DOI: 10.1002/advs.202416267] [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] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/05/2025] [Indexed: 05/16/2025]
Abstract
The incidence and burden of skin wounds, especially chronic and complex wounds, have a profound impact on healthcare. Effective wound healing strategies require a multidisciplinary approach, and advances in materials science and bioengineering have paved the way for the development of novel wound healing dressing. In this context, electrospun nanofibers can mimic the architecture of the natural extracellular matrix and provide new opportunities for wound healing. Inspired by the bioelectric phenomena in the human body, electrospun nanofibrous scaffolds with electroactive characteristics are gaining widespread attention and gradually emerging. To this end, this review first summarizes the basic process of wound healing, the causes of chronic wounds, and the current status of clinical treatment, highlighting the urgency and importance of wound dressings. Then, the biological effects of electric fields, the preparation materials, and manufacturing techniques of electroactive electrospun nanofibrous (EEN) scaffolds are discussed. The latest progress of EEN scaffolds in enhancing skin wound healing is systematically reviewed, mainly including treatment and monitoring. Finally, the importance of EEN scaffold strategies to enhance wound healing is emphasized, and the challenges and prospects of EEN scaffolds are summarized.
Collapse
Affiliation(s)
- Yang Zhang
- Department of RehabilitationThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
| | - Zhiyuan Zheng
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
| | - Shilu Zhu
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Liang Xu
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Qingdong Zhang
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Jie Gao
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Min Ye
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Shuwei Shen
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| | - Jinyu Xing
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
| | - Ming Wu
- Department of RehabilitationThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
| | - Ronald X. Xu
- Department of Precision Machinery and InstrumentationSchool of Engineering ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- School of Biomedical EngineeringDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027P. R. China
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhou215000China
| |
Collapse
|
27
|
Torregrossa M, Davies L, Hans-Günther M, Simon JC, Franz S, Rinkevich Y. Effects of embryonic origin, tissue cues and pathological signals on fibroblast diversity in humans. Nat Cell Biol 2025; 27:720-735. [PMID: 40263573 DOI: 10.1038/s41556-025-01638-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/18/2025] [Indexed: 04/24/2025]
Abstract
Fibroblasts, once perceived as a uniform cell type, are now recognized as a mosaic of distinct populations with specialized roles in tissue homeostasis and pathology. Here we provide a global overview of the expanding compendium of fibroblast cell types and states, their diverse lineage origins and multifaceted functions across various human organs. By integrating insights from developmental biology, lineage tracing and single-cell technologies, we highlight the complex nature of fibroblasts. We delve into their origination from embryonic mesenchyme and tissue-resident populations, elucidating lineage-specific behaviours in response to physiological cues. Furthermore, we highlight the pivotal role of fibroblasts in orchestrating tissue repair, connective tissue remodelling and immune modulation across diverse pathologies. This knowledge is essential to develop novel fibroblast-targeted therapies to restore steady-state fibroblast function and advance regenerative medicine strategies across multiple diseases.
Collapse
Affiliation(s)
- Marta Torregrossa
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Lindsay Davies
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Machens Hans-Günther
- Department for Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Sandra Franz
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany.
| | - Yuval Rinkevich
- Chinese Institutes for Medical Research, Beijing, China.
- Capital Medical University, Beijing, China.
| |
Collapse
|
28
|
Thota S, Begum R, Mutyala D, Bidarimath N, Thakur M, Sarkar B, Morehouse J, Yang S, Deb PK, Dorsey W, Batra S. Unraveling the Hsp70-ROS-autophagy axis in pentachlorophenol-challenged lung and liver epithelial cells. Arch Toxicol 2025; 99:2039-2062. [PMID: 40189663 DOI: 10.1007/s00204-025-03983-8] [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: 06/08/2024] [Accepted: 02/05/2025] [Indexed: 05/18/2025]
Abstract
Pentachlorophenol (PCP) was extensively utilized as an organochlorine pesticide and wood preservative in the United States from the 1930s until the Environmental Protection Agency (EPA) imposed restrictions due to concerns about its toxicity and potential carcinogenic properties. Although it is no longer widely used, PCP remains a concern due to its environmental persistence and potential for long-term health effects. Significant occupational and environmental exposures have likely occurred, with the health and economic costs of PCP exposure potentially being substantial given its known toxicity. Notably, PCP exhibits rapid absorption through both the skin and respiratory system and has been shown to cause hepatotoxicity, developmental toxicity, immunotoxicity, irritation, and carcinogenicity in laboratory animal studies. PCP exposure induces oxidative stress, a key mechanism underlying its inflammatory and toxic effects, which can activate cellular stress responses including upregulation of heat shock protein 70 (Hsp70). Previous studies in lung and liver epithelial cells have shown that Hsp70 and oxidative stress play pivotal roles in triggering autophagy. This study establishes the critical role of the Hsp70-reactive oxygen species (ROS)-autophagy axis in regulating cellular responses to PCP exposure in human alveolar (A549) and liver carcinoma (HepG2) epithelial cells. Our research elucidated the molecular mechanisms underlying PCP's cellular effects, demonstrating that its exposure resulted in increased expression of autophagy-related proteins (Beclin-1, LC3B, ATG12, and ATG16), subunits of NADPH oxidase (NCF-1, NCF-2, NOX2, and Rac), and antioxidant proteins (SOD and GPx) in both lung and liver cell types. Notably, PCP augmented the interaction between Hsp70 and the autophagy regulator Beclin-1. Pretreatment with the ROS inhibitor N-acetylcysteine or Hsp70 knockdown markedly reversed PCP-induced responses. Our in-silico protein-protein docking analysis and molecular dynamics simulation studies revealed enhanced interactions and/or stable confirmations maintained throughout the simulations for TLR4-Hsp70 and Hsp70-Beclin-1 complexes in the presence of PCP. These findings provide a strong foundation for future studies, employing in vivo experimental models and human populations to identify promising targets for PCP-induced toxicity and cellular injury. Furthermore, these findings may have far-reaching implications for public health and environmental policy, ultimately leading to the identification of biomarkers and the development of more effective interventions for environmentally induced toxicity and diseases.
Collapse
Affiliation(s)
- S Thota
- Department of Environmental Toxicology, Laboratory of Pulmonary Immunotoxicology, Southern University and A&M College, 129 Health Research Center, Baton Rouge, LA, 70813, USA
| | - R Begum
- Department of Environmental Toxicology, Laboratory of Pulmonary Immunotoxicology, Southern University and A&M College, 129 Health Research Center, Baton Rouge, LA, 70813, USA
| | - D Mutyala
- Department of Environmental Toxicology, Laboratory of Pulmonary Immunotoxicology, Southern University and A&M College, 129 Health Research Center, Baton Rouge, LA, 70813, USA
| | - N Bidarimath
- Department of Environmental Toxicology, Laboratory of Pulmonary Immunotoxicology, Southern University and A&M College, 129 Health Research Center, Baton Rouge, LA, 70813, USA
| | - M Thakur
- Department of Environmental Toxicology, Laboratory of Pulmonary Immunotoxicology, Southern University and A&M College, 129 Health Research Center, Baton Rouge, LA, 70813, USA
| | - B Sarkar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - J Morehouse
- Department of Computer Sciences, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - S Yang
- Department of Computer Sciences, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - P K Deb
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - W Dorsey
- Department of Biological Sciences, Grambling State University, Grambling, LA, USA
| | - S Batra
- Department of Environmental Toxicology, Laboratory of Pulmonary Immunotoxicology, Southern University and A&M College, 129 Health Research Center, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
29
|
Yang L, Shi F, Cao F, Wang L, She J, He B, Xu X, Kong L, Cai B. Neutrophils in Tissue Injury and Repair: Molecular Mechanisms and Therapeutic Targets. MedComm (Beijing) 2025; 6:e70184. [PMID: 40260014 PMCID: PMC12010766 DOI: 10.1002/mco2.70184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Tissue repair represents a highly intricate and ordered dynamic process, critically reliant on the orchestration of immune cells. Among these, neutrophils, the most abundant leukocytes in the body, emerge as the initial immune responders at injury sites. Traditionally recognized for their antimicrobial functions in innate immunity, neutrophils now garner attention for their indispensable roles in tissue repair. This review delves into their novel functions during the early stages of tissue injury. We elucidate the mechanisms underlying neutrophil recruitment and activation following tissue damage and explore their contributions to vascular network formation. Furthermore, we investigate the pivotal role of neutrophils during the initial phase of repair across different tissue types. Of particular interest is the investigation into how the fate of neutrophils influences overall tissue healing outcomes. By shedding light on these emerging aspects of neutrophil function in tissue repair, this review aims to pave the way for novel strategies and approaches in future organ defect repair, regeneration studies, and advancements in tissue engineering. The insights provided here have the potential to significantly impact the field of tissue repair and regeneration.
Collapse
Affiliation(s)
- Luying Yang
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Fan Shi
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Feng Cao
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Le Wang
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Jianzhen She
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Boling He
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiaoying Xu
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Liang Kong
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Bolei Cai
- Department of Oral and Maxillofacial SurgeryState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
30
|
Murali R, Balasubramanian RV, V S H, Kasoju N, N S R, Kartha RS, A P, A S, V AK, Nair RP, Bhatt A. Unravelling the wound healing efficiency of 3D bioprinted alginate-gelatin-diethylaminoethyl cellulose-fibrinogen based skin construct in a rat's full thickness wound model. Int J Biol Macromol 2025; 305:140816. [PMID: 39956240 DOI: 10.1016/j.ijbiomac.2025.140816] [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: 08/09/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
The skin, being the largest external organ, is highly vulnerable to injuries. To address the donor skin scarcity, tissue engineering and regenerative medicine have emerged as an alternative strategies to skin grafting over the past few decades. Recent advancements in 3D bioprinting technology however, have positioned it as a promising tool for constructing tissue that closely mimics in-vivo conditions, using a variety of biomaterials to meet the demands of tissue repair. An ideal 3D-printed tissue construct has the potential to provide an ideal tissue microenvironment, promoting wound healing while minimizing scar formation. In this study, we first optimized the bioink formulation by conducting toxicological evaluations, including skin irritation and skin sensitization tests, on two formulations of alginate-gelatin-DEAE cellulose, with and without fibrinogen. The addition of fibrinogen was found to reduce inflammation, making the fibrinogen-containing formulation the preferred choice for further studies. Further, we have analyzed the wound healing efficiency of 3D-printed dermal and epidermal-dermal constructs in rat full thickness wound model. Skin cells were isolated from rat tissue, and dermal, epidermal skin constructs were printed layer by layer using an alginate-gelatin-DEAE cellulose and fibrinogen-based bioink formulation, previously optimized in our laboratory. Full thickness excision wound was created and acellular, dermal and epidermal-dermal construct were implanted. Wound healing was analyzed by means of wound contraction, collagen synthesis, histopathological evaluation and gene expression analysis. Our results indicate that the epidermal-dermal construct promotes faster wound healing and enhanced angiogenesis compared to the dermal-alone and acellular constructs.
Collapse
Affiliation(s)
- Reshma Murali
- Division of Thrombosis Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India
| | - Rathina Vel Balasubramanian
- Division of Thrombosis Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India; 3D Printing and Biofabrication Group, Institute of Materials Science and Technology, TU Wien (Technische Universität Wien), Getreidemarkt 9/308, 1060 Vienna, Austria
| | - Harikrishnan V S
- Division of Laboratory Sciences, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram - 695012, Kerala, India
| | - Naresh Kasoju
- Division of Tissue Culture, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India
| | - Remya N S
- Division of Toxicology, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India
| | - Ranjith S Kartha
- Division of Thrombosis Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India
| | - Priyanka A
- Division of Thrombosis Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India
| | - Sabareeswaran A
- Division of Experimental Pathology, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India
| | - Anil Kumar V
- Division of Thrombosis Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India
| | - Renjith P Nair
- Division of Thrombosis Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India
| | - Anugya Bhatt
- Division of Thrombosis Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura, Thiruvananthapuram 695012, Kerala, India.
| |
Collapse
|
31
|
Song C, Yang J, Gu Z. Latest developments of microphysiological systems (MPS) in aging-related and geriatric diseases research: A review. Ageing Res Rev 2025; 107:102728. [PMID: 40058462 DOI: 10.1016/j.arr.2025.102728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Aging is a gradual and irreversible process accompanied by the decline in tissue function and a significantly increased risk of various aging-related and geriatric diseases. Especially in the paradoxical context of accelerated global aging and the widespread emergence of pandemics, aging-related and geriatric diseases have become leading causes of individual mortality and disability, drawing increasing attention from researchers and investors alike. Despite the utility of current in vitro systems and in vivo animal models for studying aging, these approaches are limited by insurmountable inherent constraints. In response, microphysiological systems (MPS), leveraging advances in tissue engineering and microfluidics, have emerged as highly promising platforms. MPS are capable of replicating key features of the tissue microenvironment within microfabricated devices, offering biomimetic tissue culture conditions that enhance the in vitro simulation of intact or precise human body structure and function. This capability improves the predictability of clinical trial outcomes while reducing time and cost. In this review, we focus on recent advancements in MPS used to study age-related and geriatric diseases, with particular emphasis on the application of organoids and organ-on-a-chip technologies in understanding cardiovascular diseases, cerebrovascular diseases, neurodegenerative diseases, fibrotic diseases, locomotor and sensory degenerative disorders, and rare diseases. And we aim to provide readers with critical guidelines and an overview of examples for modeling age-related and geriatric diseases using MPS, exploring mechanisms, treatments, drug screening, and other subsequent applications, from a physiopathological perspective, emphasizing the characteristic of age-related and geriatric diseases and their established correlations with the aging process. We also discuss the limitations of current models and propose future directions for MPS in aging research, highlighting the potential of interdisciplinary approaches to address unresolved challenges in the field.
Collapse
Affiliation(s)
- Chao Song
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Jiachen Yang
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
32
|
Zhang W, Liu Y, Zhang L, Shen X. Development of hyaluronic acid-based hydrogels for chronic diabetic wound healing: A review. Int J Biol Macromol 2025; 308:142273. [PMID: 40112998 DOI: 10.1016/j.ijbiomac.2025.142273] [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: 10/09/2024] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
This research delves into the advancements in chronic skin wound treatment, with a particular focus on diabetic foot ulcers, utilizing hyaluronic acid (HA)-based hydrogels. Hyaluronic acid, an integral component of the skin's extracellular matrix, plays a crucial role in process such as inflammation, angiogenesis, and tissue regeneration. Due to their three-dimensional network structure, biocompatibility, hydrophilicity, and gas exchange capabilities, HA-based hydrogels are considered highly suitable for promoting wound healing. Nonetheless, pure HA hydrogels exhibit limitations including insufficient mechanical strength and rapid release of encapsulated substances. To address these limitations, the incorporation of bioactive materials such as chitosan and collagen was investigated. This combination not only optimized mechanical strength and degradation rates but also enhanced antibacterial and anti-inflammatory properties. Furthermore, responsive hydrogel dressings were developed to adapt to the specific characteristics of the diabetic wound microenvironment, enabling on-demand drug release. These advancements present new perspectives for the treatment of diabetic foot ulcers.
Collapse
Affiliation(s)
- Wenhao Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Guangdong Engineering Technology Research Center of Offshore Environmental Pollution Control, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Yang Liu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Guangdong Engineering Technology Research Center of Offshore Environmental Pollution Control, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China.
| | - Ling Zhang
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Guangdong Engineering Technology Research Center of Offshore Environmental Pollution Control, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Xinni Shen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Guangdong Engineering Technology Research Center of Offshore Environmental Pollution Control, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| |
Collapse
|
33
|
Zhao L, Wang J, Pan Y, Tan F, Wang T, Ran H, Pang M, Zou X, Xu P, Chen A, Chen J, Li P, Zhou M, Guo X, Wang J, Yu X, Peng Y. Selenium-Albumin Nanoaccelerator Hydrogel Promotes Wound Healing by Antibacterial, Anti-Inflammatory and Antioxidant along with Inhibits Scar Formation via Downregulating CD36. Adv Healthc Mater 2025; 14:e2500699. [PMID: 40231588 DOI: 10.1002/adhm.202500699] [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] [Revised: 03/10/2025] [Indexed: 04/16/2025]
Abstract
Wounds repairing after skin damage or diabetes remain a vast medical challenge, which often faces infection, inflammation, oxidative stress, and skin scarring. Herein, a multifunctional selenium-albumin nanoaccelerator hydrogel (H-Se NPs-Gel) is constructed based on the self-assembly of human serum albumin (HSA) with selenium nanoparticles (Se NPs) using carbomer as the carrier, it has remarkable antibacterial, anti-inflammatory, antioxidant and inhibits scarring properties than Se NPs for wound healing. Compared with Se NPs, H-Se NPs exhibit smaller particle sizes, exceptional stability, better antibacterial activity against common bacteria and MRSA, and superior antioxidant and anti-inflammatory capabilities in vitro without remarkable toxicity on skin cells. Importantly, it exhibits superior efficacy to Se NPs-Gel in accelerating the healing of full-thickness skin defects and diabetic wounds in mice. Interestingly, in a hypertrophic scar (HTS) model, H-Se NPs-Gel is more effective than Se NPs-Gel in inhibiting collagen formation to suppress scarring, which is mediated by the inhibition of CD36. The antagonistic effect of H-Se NPs on CD36 is also proved with the CD36 overexpression model. Furthermore, H-Se NPs-Gel demonstrates excellent safety in mice without systemic toxicity. H-Se NPs-Gel is an effective and safe therapy strategy for promoting wound healing and reducing scar formation in clinic.
Collapse
Affiliation(s)
- Lulu Zhao
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Junyi Wang
- The Third Hospital of Mianyang, Sichuan Mental Health Center, The Affiliated Mianyang Hospital of Chongqing Medical University, Mianyang, 621000, China
| | - Yu Pan
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Fengjian Tan
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Tianlun Wang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Hongyan Ran
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Meiling Pang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xinrong Zou
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Ping Xu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Aijun Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jin Chen
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ping Li
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ming Zhou
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410083, China
| | - Xinhong Guo
- College of Biology, Hunan University, Changsha, 410082, China
| | - Jisheng Wang
- The Third Hospital of Mianyang, Sichuan Mental Health Center, The Affiliated Mianyang Hospital of Chongqing Medical University, Mianyang, 621000, China
| | - Xiufeng Yu
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yongbo Peng
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- The Third Hospital of Mianyang, Sichuan Mental Health Center, The Affiliated Mianyang Hospital of Chongqing Medical University, Mianyang, 621000, China
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
34
|
Toogood G, Evans R, Zhang L, Patel R, Meng S, Boda VK, Li W, Xu J. TRPC3 inhibition induces myofibroblast differentiation in diabetic dermal fibroblasts. Front Physiol 2025; 16:1577118. [PMID: 40370935 PMCID: PMC12075372 DOI: 10.3389/fphys.2025.1577118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025] Open
Abstract
Diabetic wounds present a significant healthcare challenge due to impaired healing mechanisms, with dermal fibroblasts playing a crucial role in tissue repair. This study investigates the role of transient receptor potential canonical-3 (TRPC3) in the dysfunction of diabetic fibroblasts and explores the therapeutic potential of TRPC3 inhibition. Findings reveal that TRPC3 expression is significantly elevated in diabetic dermal fibroblasts, which correlates with suppressed transforming growth factor-beta (TGF-β) signaling and impaired differentiation into myofibroblasts. Inhibiting TRPC3 effectively restores fibroblast functionality by upregulating TGF-β1 and its downstream effector, SMAD4. This restoration enhances the expression of key myofibroblast markers, such as α-smooth muscle actin (ACTA2) and type I collagen (COL1a1), which are essential for wound contraction and extracellular matrix remodeling. These results establish TRPC3 as a critical regulator of fibroblast activity and present TRPC3 inhibition as a promising therapeutic strategy for improving wound healing in diabetic patients.
Collapse
Affiliation(s)
- Gemma Toogood
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Robin Evans
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Liping Zhang
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Rima Patel
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Songmei Meng
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Vijay K. Boda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Junwang Xu
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
35
|
Qiu X, Xiang F, Liu H, Zhan F, Liu X, Bu P, Zhou B, Duan Q, Ji M, Feng Q. Electrical hydrogel: electrophysiological-based strategy for wound healing. Biomater Sci 2025; 13:2274-2296. [PMID: 40131331 DOI: 10.1039/d4bm01734j] [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: 03/26/2025]
Abstract
Wound healing remains a significant challenge in clinical practice, driving ongoing exploration of innovative therapeutic approaches. In recent years, electrophysiological-based wound healing strategies have gained considerable attention. Specifically, electrical hydrogels combine the synergistic effects of electrical stimulation and hydrogel properties, offering a range of functional benefits for wound healing, including antibacterial activity, real-time wound monitoring, controlled drug release, and electrical treatment. Despite significant progress made in electrical hydrogel research for wound healing, there is a lack of comprehensive, systematic reviews summarizing this field. In this review, we survey the latest advancements in electrical hydrogel technology. After analyzing the mechanisms of electrical stimulation in promoting wound healing, we establish a novel classification framework for electrical hydrogels based on their operational principles. The review further provides an in-depth evaluation of the therapeutic efficacy of these hydrogels in various types of wounds. Finally, we propose future directions and challenges for the development of electrical hydrogels for wound healing.
Collapse
Affiliation(s)
- Xingan Qiu
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing 404010, China.
- School of Medicine, Chongqing University, Chongqing 404010, China
| | - Feng Xiang
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Hong Liu
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing 404010, China.
- School of Medicine, Chongqing University, Chongqing 404010, China
| | - Fangbiao Zhan
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing 404010, China.
- School of Medicine, Chongqing University, Chongqing 404010, China
| | - Xuezhe Liu
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Pengzhen Bu
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Bikun Zhou
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Qiaojian Duan
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Ming Ji
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing 404010, China.
- School of Medicine, Chongqing University, Chongqing 404010, China
| | - Qian Feng
- Key laboratory of Biorheological Science and Technology, Ministry of Educations, Collage of Bioengineering, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
36
|
P LP, Ghosh T, Sri R, B V B. Advancements in polymer-based approaches in diabetic wound healing: a comprehensive review. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-15. [PMID: 40279098 DOI: 10.1080/09205063.2025.2492777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025]
Abstract
Diabetes, both Type 1 and Type 2, often leads to chronic wounds due to impaired healing processes, marked by prolonged inflammation, delayed blood vessel formation, and abnormal collagen production. These issues disrupt normal tissue regeneration, slowing healing. To address these challenges, polymer-based wound dressings are being explored as a promising solution. Natural polymers like alginate, chitosan, and hyaluronic acid, as well as synthetic ones like PCL, PLA, and PLGA, offer potential for more effective healing. These materials can be used in advanced delivery systems, such as nanofibrous scaffolds, nanoparticles, and hydrogels, which help deliver medications, maintain a moist healing environment, and stimulate cell growth. By improving the wound environment, polymer-based systems provide new hope for diabetic patients with slow-to-heal wounds, enhancing therapeutic outcomes and accelerating healing. These innovations could significantly improve the management of chronic wounds in diabetes.
Collapse
Affiliation(s)
| | - Tanmoy Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, MS Ramaiah University of Applied Sciences, Bengaluru, India
| | - Ramya Sri
- Department of Pharmaceutics, Faculty of Pharmacy, MS Ramaiah University of Applied Sciences, Bengaluru, India
| | - Basavaraj B V
- Department of Pharmaceutics, Faculty of Pharmacy, MS Ramaiah University of Applied Sciences, Bengaluru, India
| |
Collapse
|
37
|
Thiagarajan L, Sanchez-Alvarez R, Kambara C, Rajasekar P, Wang Y, Halloy F, Hall J, Stark HJ, Martin I, Boukamp P, Kurinna S. miRNA-29 regulates epidermal and mesenchymal functions in skin repair. FEBS Lett 2025. [PMID: 40285401 DOI: 10.1002/1873-3468.70051] [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: 11/19/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 04/29/2025]
Abstract
MicroRNAs (miRNAs) control organogenesis in mammals by inhibiting translation of mRNA. Skin is an excellent model to study the role of miRNAs in epidermis and the mesenchyme. Previous research demonstrated miRNA-29 family functions in skin; however, the mRNA targets and the downstream mechanisms of miRNA-29-mediated regulation are missing. We used the miRNA crosslinking and immunoprecipitation method to find direct targets of miRNA-29 in keratinocytes and fibroblasts from human skin. miRNA-29 inhibition using modified antisense oligonucleotides in 2D and 3D cultures of keratinocytes and fibroblasts enhanced cell-to-matrix adhesion through autocrine and paracrine mechanisms of miRNA-29-dependent tissue growth. We reveal a full transcriptome of human keratinocytes with enhanced adhesion to the matrix, which supports regeneration of the epidermis and is regulated by miRNA-29. Impact statement The functions of small, therapeutically targetable microRNA molecules identified in our study can provide a new approach to improve wound healing by restoring and enhancing the inner molecular mechanisms of a cell and its surrounding matrix. We also provide a plethora of new mRNA targets for follow-up studies of cell adhesion and extracellular matrix formation in humans.
Collapse
Affiliation(s)
- Lalitha Thiagarajan
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, UK
| | - Rosa Sanchez-Alvarez
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, UK
| | - Chiho Kambara
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, UK
| | | | - Yuluang Wang
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland
| | - François Halloy
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland
| | - Jonathan Hall
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland
| | | | - Iris Martin
- German Cancer Research Center, Heidelberg, Germany
| | | | - Svitlana Kurinna
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, UK
| |
Collapse
|
38
|
Gould LJ, Tomic-Canic M. A Call to Action: Adopting the New Wound Reporting in Animal and Human Preclinical Studies (WRAHPS) Guidelines. J Invest Dermatol 2025:S0022-202X(25)00363-X. [PMID: 40261226 DOI: 10.1016/j.jid.2025.02.152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 04/24/2025]
Affiliation(s)
- Lisa J Gould
- South Shore Hospital Center for Wound Healing, Weymouth, Massachusetts, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
39
|
Lei H, Cui H, Xia Y, Sun F, Zhang W. Illuminating Hope for Tumors: The Progress of Light-Activated Nanomaterials in Skin Cancer. Int J Nanomedicine 2025; 20:5081-5118. [PMID: 40264819 PMCID: PMC12013650 DOI: 10.2147/ijn.s506000] [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: 11/12/2024] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
Skin cancer is a common malignant tumor that poses significant global health and economic burdens. The main clinical types include malignant melanoma and non-melanoma. Complications such as post-surgical recurrence, wound formation, or disfigurement can severely impact the patient's mental well-being. Traditional treatments such as surgery, chemotherapy, radiation therapy, and immunotherapy often face limitations. These challenges not only reduce the effectiveness of treatments but also negatively impact patients' quality of life. Phototherapy, a widely used and long-standing method in dermatology, presents a promising alternative for skin cancer treatment. Light-triggered nanomaterials further enhance the potential of phototherapy by offering advantages such as improved therapeutic precision, controlled drug release, minimal invasiveness, and reduced damage to surrounding healthy tissues. This review summarizes the application of light-triggered nanomaterials in skin cancer treatment, focusing on the principles, advantages, and design strategies of photodynamic therapy (PDT), photothermal therapy (PTT), and photoacoustic therapy (PAT). In this manuscript we have an in-depth discussion on overcoming translational barriers, including strategies to enhance light penetration, mitigate toxicity, reduce production costs, and optimize delivery systems. Additionally, we discuss the challenges associated with their clinical translation, including limited light penetration in deep tissues, potential toxicity, high production costs, and the need for advanced delivery systems.
Collapse
Affiliation(s)
- Huaqing Lei
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, People’s Republic of China
| | - Hengqing Cui
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Institute of Aesthetic Plastic Surgery and Medicine, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yu Xia
- College of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Fujia Sun
- College of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Wenjun Zhang
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, People’s Republic of China
| |
Collapse
|
40
|
Chen J, Fu X, Ahmed AS, Hart DA, Zhou Z, Ackermann PW. Systematic Review of Relevant Biomarkers for Human Connective Tissue Repair and Healing Outcome: Implications for Understanding Healing Processes and Design of Healing Interventions. Adv Wound Care (New Rochelle) 2025. [PMID: 40248898 DOI: 10.1089/wound.2024.0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025] Open
Abstract
Objective: The healing process following connective tissue (CT) injuries is complex, resulting in variable and often suboptimal outcomes. Patients undergoing CT repair frequently experience healing failures, compromised function, and chronic degenerative diseases. The identification of biomarkers to guide improved clinical outcomes after CT injuries remains an emerging but promising field. [Figure: see text] [Figure: see text] Design: Systematic review. Data sources: Databases, including PubMed, MEDLINE Ovid, Web of Science, and Google Scholar, were searched up to August 2024. Eligibility criteria: To achieve the research objective, randomized control trials, cohort studies, and case-control studies on biomarkers associated with CT repair and healing outcomes were selected. The present analysis was confined to clinical and preclinical models, excluding imaging studies. The entire process of this systematic review adhered strictly to the guidelines outlined in the Preferred Reporting Items for Systematic Review and Meta-Analyses protocol checklist. Results: A total of 1,815 studies on biomarkers of CT repair were initially identified, with 75 studies meeting eligibility criteria and 55 passing quality assessments. For biomarkers associated with CT healing outcomes, 281 studies were considered, with 30 studies meeting eligibility criteria and 24 passing quality assessments. Twenty-one overlapping studies investigated the effects of biomarkers on both CT repair and healing outcomes. Specific biomarkers identified, and ranked from highest to lowest quality, include complement factor D, eukaryotic elongation factor-2, procollagen type I N-terminal propetide, procollagen type III N-terminal propetide, lactate, pyruvate, platelet-derived growth factor-BB, tissue inhibitor of metalloproteinase-3 (TIMP-3), cysteine-rich protein-1, plastin-3, periostin, protein S100-A11, vimentin, matrix metalloproteinases (MMP-2, MMP-7, and MMP-9), hepatocyte growth factor, interferon-γ, interleukins (IL-6, IL-8, and IL-10), MMP-1, MMP-3, tumor necrosis factor-α, fibroblast growth factor-2, IL-1α, chondroitin-6-sulfate, inter-alpha-trypsin inhibitor heavy chain-4, transforming growth factor-beta 1, vascular endothelial growth factor, C-C chemokine receptor 7, C-C chemokine ligand 19, IL-1β, IL-1Ra, IL-12p40, granulocyte-macrophage colony-stimulating factor (GM-CSF), and TIMP-1. Conclusions: All of the 37 identified potential biomarkers demonstrated regulatory effects on CT repair and mediated healing outcomes. Notably, the identified biomarkers from human studies can potentially play an essential role in the development of targeted treatment protocols to counteract compromised healing and can also serve as predictors for detecting CT healing processes and long-term outcomes.
Collapse
Affiliation(s)
- Junyu Chen
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, PR China
| | - Xiaoxue Fu
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, PR China
| | - Aisha S Ahmed
- Integrative Orthopedic Laboratory, Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David A Hart
- Department of Surgery, Faculty of Kinesiology, McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Canada
| | - Zongke Zhou
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, PR China
| | - Paul W Ackermann
- Integrative Orthopedic Laboratory, Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Trauma, Acute Surgery and Orthopedics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
41
|
He L, Xing S, Zhang W, Wang Y, Li Y, Chen J, Zhang J, Wang S, Zhao Q. Multifunctional dynamic chitosan-guar gum nanocomposite hydrogels in infection and diabetic wound healing. Carbohydr Polym 2025; 354:123316. [PMID: 39978920 DOI: 10.1016/j.carbpol.2025.123316] [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: 11/26/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
Traditional wound care methods are less effective for infectious and diabetic wounds, highlighting an urgent need for effective strategies. The study aimed to design a self-healing hydrogel with antibacterial, antioxidant, and photothermal capabilities to treat infectious and diabetic wounds. Silver nanoparticles (AgNPs) were loaded into mesoporous polydopamine (MPDA) nanoparticles to form Ag@MPDA nanoparticles. Ag@MPDA was incorporated into the cationic guar gum-chitosan-boric acid (CCB) hydrogel to obtain the PA-CCB hydrogel. PA-CCB hydrogel exhibited excellent self-healing and adhesive properties, adapting well to the dynamic wound environment. PA-CCB hydrogel combined with photothermal therapy (PTT) could effectively eradicated E. coli (99.9 %) and S. aureus (99.7 %). The PA-CCB hydrogel reduced excessive reactive oxygen species and promoted the migration of fibroblasts in vitro. In the infected mouse wound models, the PA-CCB hydrogel effectively inhibited bacteria. After combining with PTT, the antibacterial ability of the PA-CCB hydrogel was further enhanced. In the diabetic mouse wound models, the PA-CCB hydrogel reduced the inflammatory level of wound tissue. In both models, after combining with PTT, the PA-CCB hydrogel exhibited further improvements in angiogenesis, collagen deposition, and re-epithelialization. By integrating multifunctional hydrogel with PTT, the PA-CCB hydrogel exhibited broad application potential for infectious and diabetic wounds.
Collapse
Affiliation(s)
- Luning He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Shuyi Xing
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Weikang Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Ying Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Yian Li
- School of Libra Arts of Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Jinghao Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jinghai Zhang
- Department of Biomedical Engineering, School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
42
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation during Wound Closure. ACS Biomater Sci Eng 2025; 11:2249-2262. [PMID: 40029610 DOI: 10.1021/acsbiomaterials.4c02019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. Extra domain A (EDA) Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, EDA Fn organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional coactivator, yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate whether YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity, but on stiffer substrates, they decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest that there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
43
|
Zou Y, Yang Y, Pei J, Sun P, Wang Y. Ganoderma lucidum Polysaccharide/carboxymethyl Chitosan Hydrogels Modulate Macrophage Polarization for Wound Healing. Biomacromolecules 2025; 26:2675-2689. [PMID: 40153544 DOI: 10.1021/acs.biomac.5c00112] [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: 03/30/2025]
Abstract
Wound healing remains a global challenge for clinical and experimental research. Hydrogels prepared from natural polysaccharides show great potential in the wound healing process. In this study, novel hydrogels (G-GLP) were prepared using oxidized Ganoderma lucidum polysaccharides (OGLPs) and carboxymethyl chitosan via the Schiff base reaction, which did not require the addition of any chemical cross-linking agent. The hydrogels showed excellent mechanical properties and biocompatibility. Moreover, the hydrogels showed superior hemostatic performance in mouse liver trauma and tail amputation models. Importantly, G-GLP improved inflammation by promoting the polarization of the macrophage M2 subtype, inhibiting the M1 subtype and reducing intracellular levels of reactive oxygen species. In vivo experiments demonstrated that G-GLP accelerated healing in a total defect wound model by reducing inflammation and promoting blood vessel repair and collagen deposition. These results demonstrate that G-GLP has potential as an effective wound repair dressing.
Collapse
Affiliation(s)
- Yu Zou
- College of Food Science and Technology, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China
| | - Yuheng Yang
- College of Food Science and Technology, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China
| | - Jingying Pei
- College of Food Science and Technology, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China
| | - Peilong Sun
- College of Food Science and Technology, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou 310014, China
| | - Yan Wang
- College of Food Science and Technology, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou 310014, China
| |
Collapse
|
44
|
Chang W, Lu P, Li S, Xiang J, Liu J, Wang Y, Zhang L, Sun H. Injectable Functional Microspheres Capable of BMSC Recruitment and Osteogenic Induction for In Situ Bone Regeneration. ACS Biomater Sci Eng 2025; 11:2236-2248. [PMID: 40131171 DOI: 10.1021/acsbiomaterials.4c01720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Currently, bone defects remain a major challenge in clinical treatment. Recruiting target cells at the defect site and inducing them to differentiate into bone tissue are effective treatment methods. In previous studies, we used the CD271 antibody to construct bone marrow mesenchymal stem cell (BMSC) recruitment microspheres for the treatment of bone defects. However, the osteoconductivity of the microspheres themselves was poor, and the system lacked osteoinductivity, which affected the repair efficiency. In this study, we prepared submillimeter-sized porous chitosan (CS) microspheres through process optimization, and the BMSCs were able to directly adhere and proliferate on their surfaces. After the bioconjugation of the CD271 antibody, bone morphogenetic protein-2 (BMP-2) was further loaded onto the pore structure of microspheres to obtain the injectable microspheres with BMSC recruitment and osteogenic differentiation induction functions. Microspheres could efficiently recruit BMSCs through the combined action of the CD271 antibody and BMP-2 and further induce the recruited BMSCs, differentiating into osteoblasts through BMP-2, which ultimately exhibited promising bone regeneration ability in rats. We expect that the novel functional microspheres have great potential in biomedical applications for in situ treatment of bone defects.
Collapse
Affiliation(s)
- Wenliao Chang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, 123 Hexiang Road, Changzhou 213000, Jiangsu, China
| | - Peipei Lu
- Nursing Department, Changzhou Hygiene Vocational Technology College, Changzhou 213000, Jiangsu, China
| | - Shuxiang Li
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, 123 Hexiang Road, Changzhou 213000, Jiangsu, China
| | - Jinghua Xiang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, 123 Hexiang Road, Changzhou 213000, Jiangsu, China
| | - Jiachen Liu
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, 123 Hexiang Road, Changzhou 213000, Jiangsu, China
| | - Yimin Wang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, 123 Hexiang Road, Changzhou 213000, Jiangsu, China
| | - Lei Zhang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, Anhui, China
- China State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, Jiangsu, China
| | - Han Sun
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
- Changzhou Medical Center, Nanjing Medical University, 123 Hexiang Road, Changzhou 213000, Jiangsu, China
| |
Collapse
|
45
|
Kananivand M, Nouri F, Yousefi MH, Pajouhi A, Ghorbani H, Afkhami H, Razavi ZS. Mesenchymal stem cells and their exosomes: a novel approach to skin regeneration via signaling pathways activation. J Mol Histol 2025; 56:132. [PMID: 40208456 DOI: 10.1007/s10735-025-10394-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
Accelerating wound healing is a crucial objective in surgical and regenerative medicine. The wound healing process involves three key stages: inflammation, cell proliferation, and tissue repair. Mesenchymal stem cells (MSCs) have demonstrated significant therapeutic potential in promoting tissue regeneration, particularly by enhancing epidermal cell migration and proliferation. However, the precise molecular mechanisms underlying MSC-mediated wound healing remain unclear. This review highlights the pivotal role of MSCs and their exosomes in wound repair, with a specific focus on critical signaling pathways, including PI3K/Akt, WNT/β-catenin, Notch, and MAPK. These pathways regulate essential cellular processes such as proliferation, differentiation, and angiogenesis. Moreover, in vitro and in vivo studies reveal that MSCs accelerate wound closure, enhance collagen deposition, and modulate immune responses, contributing to improved tissue regeneration. Understanding these mechanisms provides valuable insights into MSC-based therapeutic strategies for enhancing wound healing.
Collapse
Affiliation(s)
- Maryam Kananivand
- Medical Department, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Nouri
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University (SRBIAU), Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Ali Pajouhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hakimeah Ghorbani
- Department of Sciences, Faculty of Biological Sciences, Tabriz University of Sciences, Tabriz, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran.
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Zi Y, Li J, Qian X, Li J, Jin Y, Zhang Z, Jin Y. Human umbilical cord mesenchymal stem cell exosomes promote elastin production and acute skin wound healing via TGFβ1-Smad pathway. Mol Cell Biochem 2025:10.1007/s11010-025-05264-5. [PMID: 40202710 DOI: 10.1007/s11010-025-05264-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Skin wound healing is a complex physiological process influenced by multiple factors, including the patient's overall health status. Exosomes derived from human umbilical cord mesenchymal stem cells (hUCMSC-Exos) have demonstrated significant potential in enhancing wound repair. This study investigates the mechanisms through which hUCMSC-Exos facilitate skin wound healing and evaluates their potential application in combination with hydrogels for clinical treatment. Human foreskin fibroblasts (HFF-1) were treated with varying concentrations of hUCMSC-Exos to evaluate their impact on cell proliferation, assessed via the CCK-8 assay. Exosome uptake by HFF-1 cells was visualized using PKH-26 dye staining, while flow cytometry was employed to analyze cell cycle changes. Cell migration was evaluated through scratch and Transwell assays. Gene expression levels of Collagen I, Elastin, and Fibronectin were quantified by qRT-PCR, while Elastin secretion was measured by ELISA. Western blotting was used to examine proteins in the TGFβ1-Smad signaling pathway. The role of SP1 in regulating Elastin gene expression was investigated by testing the SP1 inhibitor Plicamycin and examining hUCMSC-Exos ability to counteract its effect. Additionally, a chromatin immunoprecipitation (ChIP) assay was performed to analyze SP1 binding at the Elastin gene promoter. In vivo, the efficacy of hUCMSC-Exos combined with hydrogels in promoting wound healing was assessed using a mouse skin wound model. hUCMSC-Exos significantly enhanced HFF-1 cell proliferation at concentrations exceeding 1 × 10⁹ particles/mL and increased the proportion of cells in the S and G2/M phases. HFF-1 cells readily absorbed these exosomes, leading to improved cell migration. Treatment with hUCMSC-Exos upregulated the gene expression of Collagen I, Fibronectin, and Elastin. The SP1 inhibitor Plicamycin reduced Elastin gene expression, an effect that was reversed by hUCMSC-Exos. In vivo, the combination of hUCMSC-Exos and hydrogels accelerated wound healing, enhanced collagen organization, and promoted the formation of elastic fibers and blood vessels. hUCMSC-Exos facilitate skin wound healing by promoting SP1 binding to the Elastin gene promoter, thereby upregulating Elastin expression and supporting extracellular matrix remodeling. These findings suggest a promising therapeutic role for hUCMSC-Exos in clinical applications for wound healing.
Collapse
Affiliation(s)
- Yi Zi
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - Jie Li
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - XinPing Qian
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - Jian Li
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - Yan Jin
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - ZiBo Zhang
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China
| | - YanHua Jin
- Translational Medicine Research Center, Yanbian University Medical College, Yanbian, 133002, Jilin, China.
| |
Collapse
|
47
|
Rumon MMH. Advances in cellulose-based hydrogels: tunable swelling dynamics and their versatile real-time applications. RSC Adv 2025; 15:11688-11729. [PMID: 40236573 PMCID: PMC11997669 DOI: 10.1039/d5ra00521c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025] Open
Abstract
Cellulose-derived hydrogels have emerged as game-changing materials in biomedical research, offering an exceptional combination of water absorption capacity, mechanical resilience, and innate biocompatibility. This review explores the intricate mechanisms that drive their swelling behaviour, unravelling how molecular interactions and network architectures work synergistically to enable efficient water retention and adaptability. Their mechanical properties are explored in depth, with a focus on innovative chemical modifications and cross-linking techniques that enhance strength, elasticity, and functional versatility. The versatility of cellulose-based hydrogels shines in applications such as wound healing, precision drug delivery, and tissue engineering, where their biodegradability, biocompatibility, and adaptability meet the demands of cutting-edge healthcare solutions. By weaving together recent breakthroughs in their development and application, this review highlights their transformative potential to redefine regenerative medicine and other biomedical fields. Ultimately, it emphasizes the urgent need for continued research to unlock the untapped capabilities of these extraordinary biomaterials, paving the way for new frontiers in healthcare innovation.
Collapse
Affiliation(s)
- Md Mahamudul Hasan Rumon
- Department of Mathematics and Natural Sciences, Brac University 66 Mohakhali Dhaka 1212 Bangladesh
| |
Collapse
|
48
|
Palomeque Chávez JC, McGrath M, O'Connor C, Dervan A, Dixon JE, Kearney CJ, Browne S, O'Brien FJ. Development of a VEGF-activated scaffold with enhanced angiogenic and neurogenic properties for chronic wound healing applications. Biomater Sci 2025; 13:1993-2011. [PMID: 40012508 PMCID: PMC11865941 DOI: 10.1039/d4bm01051e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
Chronic wounds remain in a state of disrupted healing, impeding neurite outgrowth from injured nerves and poor development of new blood vessels by angiogenesis. Current therapeutic approaches primarily focus on the restoration of vascularization and overlook the need of nerve regeneration for complete healing. Vascular endothelial growth factor (VEGF) is a critical growth factor supporting angiogenesis in wound healing, promoting vascularization and has also demonstrated neuro-protective capabilities in both central and peripheral nervous system. While the delivery of pro-regenerative recombinant growth factors has shown promise, gene delivery offers greater stability, reduced off-target side effects, diminished cytotoxicity, and lower production costs. In this context, the overarching goal of this study was to develop a VEGF-activated scaffold with the potential to provide a multifaceted response that enhances both angiogenesis and nerve repair in wound healing through the localized delivery of plasmid encoding VEGF (pVEGF) encapsulated within the GET peptide system. Initially, delivery of pVEGF/GET nanoparticles to dermal fibroblasts led to higher VEGF protein expression without a compromise in cell viability. Transfection of dermal fibroblasts and endothelial cells on the VEGF-activated scaffolds resulted in enhanced VEGF expression, improved endothelial cell migration and organization into vascular-like structures. Finally, the VEGF-activated scaffolds consistently displayed enhanced neurogenic ability through improved neurite outgrowth from neural cells in in vitro and ex vivo models. Taken together, the VEGF-activated scaffold demonstrates multifaceted outcomes through the induction of pro-angiogenic and neurogenic responses from dermal, vascular and neural cells, illustrating the potential of this platform for the healing of chronic wounds.
Collapse
Affiliation(s)
- Juan Carlos Palomeque Chávez
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- Kearney Lab, Department of Biomedical Engineering, University of Massachusetts, Armhest, USA
| | - Matthew McGrath
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Cian O'Connor
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Adrian Dervan
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - James E Dixon
- Regenerative Medicine & Cellular Therapies (RMCT), Biodiscovery Institute (BDI), School of Pharmacy, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Cathal J Kearney
- Kearney Lab, Department of Biomedical Engineering, University of Massachusetts, Armhest, USA
| | - Shane Browne
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), University of Galway, Galway, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
49
|
Javaid A, Sharma KK, Varshney P, Verma A, Mudavath SL. Overcoming drug delivery challenges with lipid-based nanofibers for enhanced wound repair. Biomater Sci 2025; 13:1960-1974. [PMID: 39699201 DOI: 10.1039/d4bm01536c] [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: 12/20/2024]
Abstract
Wound healing is a dynamic, multi-phase process that includes haemostasis, tissue regeneration, cellular proliferation, and matrix modification. Traditional wound care procedures frequently encounter complications such as delayed healing and infection, demanding new therapeutic approaches. In this context, nanomaterial-based devices provide considerable benefits due to their capacity to improve medication delivery and tissue healing. We suggest a lipid-based nanofiber formulation for wound treatment that overcomes the restricted skin penetration of hydrophilic niacin, a strong wound healing agent. Niacin-loaded nanofibers (NLNFs) were manufactured utilizing glyceryl monostearate (GMS) by a self-assembly process, which included high-pressure homogenization and probe sonication for optimum nanostructure creation. The NLNFs were physicochemically characterized using Fourier transform infrared spectroscopy (FTIR), X-ray diffraction, scanning electron microscopy (SEM) and surface profilometry to determine their morphology and homogeneity, and a drop shape analyser was used to determine hydrophobicity. In vitro tests revealed prolonged drug release, great cytocompatibility, and strong antioxidant activity, indicating superior free radical scavenging capacity. Ex vivo tests, such as the Draize skin irritation test, skin permeation test, and drug retention assays, revealed low skin irritation, increased permeability, and efficient drug retention in skin layers. In vivo experiments showed rapid wound closure and positive histological results, which were backed by hemocompatibility tests such as hemolysis and whole blood clot analysis, validating the formulation's safety. ELISA results indicated that the NLNF-treated group had higher levels of critical wound-healing indicators than the controls. Overall, our findings suggest that NLNFs have tremendous potential as a unique and effective treatment alternative for controlling and improving wound healing processes.
Collapse
Affiliation(s)
- Aaqib Javaid
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab, 140306, India
| | - Krishana Kumar Sharma
- Teerthankar Mahaveer University, Delhi Road, NH 24, Bagadpur, Uttar Pradesh 244001, India
| | - Prakhar Varshney
- Teerthankar Mahaveer University, Delhi Road, NH 24, Bagadpur, Uttar Pradesh 244001, India
| | - Anurag Verma
- Teerthankar Mahaveer University, Delhi Road, NH 24, Bagadpur, Uttar Pradesh 244001, India
| | - Shyam Lal Mudavath
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Gachibowli, Hyderabad, 500046, Telangana, India.
| |
Collapse
|
50
|
Wang Z, Liu W, Bai R, Guo Y, Wang Z, Ma X, Yu Z. Transcriptome analysis of regenerated dermis stimulated by mechanical stretch. Gene 2025; 943:149267. [PMID: 39842648 DOI: 10.1016/j.gene.2025.149267] [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: 10/10/2024] [Revised: 12/16/2024] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Mechanical stretch is utilized in the process of tissue expansion to promote skin regeneration, which is crucial for wound healing and organ reconstruction purposes. Enlarged dermal area is one of the significant histological characteristics of the expanded skin. However, the underlying biological processes and molecular pathways associated with dermal regeneration triggered by mechanical stretch are still not well understood. METHODS Twelve male Sprague-Dawley (SD) rats were divided into the expansion group and sham group randomly. Upon creating a rat scalp expansion model, the dermis was isolated from the full-thickness skin in both experimental groups for RNA sequencing. This process led to the identification of differentially expressed genes (DEGs). Subsequently, we conducted Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and Gene Set Enrichment Analysis (GSEA) to identify the essential biological processes associated with dermal regeneration induced by mechanical stretch, leveraging data from the DEGs. A network of protein-protein interactions (PPI) was built to detect the critical modules and central genes. The expression levels of these hub genes were evaluated using quantitative real-time polymerase chain reaction (qPCR). RESULTS Increased expanded skin area and dermal thinning which represent the typical changes of expanded skin were observed in the expansion group. A total of 782 DEGs were identified in the expansion group relative to the sham group. The DEGs were associated with several biological processes, including the organization of the extracellular matrix, the enhancement of macrophage activation, and the promotion of angiogenesis, among others. Cell components encompassing Toll-like receptor 2-Toll-like receptor 6 protein complex, interstitial matrix, extracellular matrix (ECM), and collagen trimer were discovered. Molecular function categories including integrin binding, insulin-like growth factor binding, and fatty acid elongase activity were involved. The KEGG pathway analysis demonstrated the significant enrichment of pathways including the PI3K-Akt signaling pathway, fatty acid metabolism, and extracellular matrix-receptor interactions. GSEA results displayed that mechanical stretch correlated with the regulation of cell activation processes, cytokine-mediated signaling pathways, and immune system processes. PPI network resulted in the identification of 598 nodes along with a total of 5,304 interaction pairs between proteins. And ten hub genes containing Ccl2, Cxcl10, Fasn, Itgad, Cd163, Mmp9, Cd36, Tlr2, Igf1, and Wnt2 were identified by bioinformatics analysis and validated by qPCR. CONCLUSIONS This in vivo study for the first time revealed the DEGs related to mechanical stretch stimulated dermal regeneration and identified the involved pathways and hub genes correlated with macrophage recruitment and polarization, fibroblast proliferation and ECM production and angiogenesis, which may benefit further studies aimed at developing therapeutic strategies for facilitating expanded skin regeneration.
Collapse
Affiliation(s)
- Zhantong Wang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China; Xijing 986 Hospital Department, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| | - Wei Liu
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| | - Ruoxue Bai
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| | - Yaotao Guo
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| | - Zhigang Wang
- Xijing 986 Hospital Department, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| | - Xianjie Ma
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| | - Zhou Yu
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710032, China.
| |
Collapse
|