1
|
Chansoria P, Winkelbauer M, Zhang S, Janiak J, Liu H, Boev D, Morandi A, Grange R, Zenobi-Wong M. Structured Light Projection Using Image Guide Fibers for In Situ Photo-biofabrication. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2419350. [PMID: 40297914 DOI: 10.1002/adma.202419350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/30/2025] [Indexed: 04/30/2025]
Abstract
Light-based biofabrication techniques have revolutionized the field of tissue engineering and regenerative medicine. Specifically, the projection of structured light, where the spatial distribution of light is controlled at both macro and microscale, has enabled precise fabrication of complex three dimensional structures with high resolution and speed. However, despite tremendous progress, biofabrication processes are mostly limited to benchtop devices which limit the flexibility in terms of where the fabrication can occur. Here, a Fiber-assisted Structured Light (FaSt-Light) projection apparatus for rapid in situ crosslinking of photoresins is demonstrated. This approach uses image-guide fiber bundles which can project bespoke images at multiple wavelengths, enabling flexibility and spatial control of different photoinitiation systems and crosslinking chemistries and also the location of fabrication. Coupling of different sizes of fibers and different lenses attached to the fibers to project small (several mm) or large (several cm) images for material crosslinking is demonstrated. FaSt-Light allows control over the cross-section of the crosslinked resins and enables the introduction of microfilaments which can further guide cellular infiltration, differentiation, and anisotropic matrix production. The proposed approach can lead to a new range of in situ biofabrication techniques which improve the translational potential of photofabricated tissues and grafts.
Collapse
Affiliation(s)
- Parth Chansoria
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Michael Winkelbauer
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Shipin Zhang
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Jakub Janiak
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Hao Liu
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Dimitar Boev
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| | - Andrea Morandi
- Department of Physics, Institute for Quantum Electronics, Optical Nanomaterial Group, ETH Zürich, 8093, Switzerland
| | - Rachel Grange
- Department of Physics, Institute for Quantum Electronics, Optical Nanomaterial Group, ETH Zürich, 8093, Switzerland
| | - Marcy Zenobi-Wong
- Department of Health Sciences and Technology, Institute for Biomechanics, Tissue Engineering and Biofabrication Group, ETH Zürich, 8093, Switzerland
| |
Collapse
|
2
|
Yaron JR, Pallod S, Grigaitis-Esman N, Singh V, Rhodes S, Patel DM, Ghosh D, Rege K. Histamine receptor agonism differentially induces immune and reparative healing responses in biomaterial-facilitated tissue repair. Biomaterials 2025; 315:122967. [PMID: 39586217 DOI: 10.1016/j.biomaterials.2024.122967] [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: 05/19/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
Tissue repair is a highly regulated process involving immune, stromal, vascular, and parenchymal cell responses. Mediators of cellular responses at different phases of the healing process stimulate transitions through the continuum of repair. Histamine is an early mediator of healing, which, in skin, is released by resident cells (e.g., mast cells) after cutaneous injury, and acts to stimulate diverse responses in multiple cell populations. Histamine signaling is regulated by four distinct cell surface G-protein coupled receptors (HRH1-4 in humans, Hrh1-4 in mice) which initiate different downstream signaling cascades upon activation, but the specific effect of each receptor on tissue repair is poorly understood. Here, we systematically investigated the effect of selective histamine receptor agonism in laser-activated sealing and tissue repair of incisional skin wounds in immunocompetent mice. Although all four histamine receptors exhibited wound responsiveness in the epidermis, we find that activation of Hrh1, Hrh2, and Hrh4 stimulate a pro-healing immune response characterized by increased pro-resolution macrophages, reduced pro-inflammatory macrophages, and suppressed neutrophil responses. Further, activation of Hrh1 and Hrh4 stimulate angiogenesis after injury. Lastly, although Hrh1 activation resulted in enhanced epidermal epithelial-to-mesenchymal transition (EMT) in vivo and epithelialization in vitro, activation of Hrh2 suppressed both epidermal EMT and epithelialization. Activation of Hrh3, primarily found on neuronal cells, had no effect on any measure in our study. Selective histamine receptor agonism, specifically of histamine receptors Hrh-1 and 4, is a potential reparative approach to promote the efficacy of biomaterial-mediated repair of tissues, including skin.
Collapse
Affiliation(s)
- Jordan R Yaron
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Shubham Pallod
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Nicole Grigaitis-Esman
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Vanshika Singh
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Samantha Rhodes
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Dirghau Manishbhai Patel
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Deepanjan Ghosh
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Kaushal Rege
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
3
|
Kim Y, Kim SE, Park KD, Park KM. Bioadhesives and bioactive hydrogels for wound management. J Control Release 2025; 379:285-302. [PMID: 39788376 DOI: 10.1016/j.jconrel.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/25/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Delayed wound healing remains a major challenge in biomedical research, often leading to complications such as scarring, acute trauma, and chronic diseases. Effective wound management is crucial for enhancing treatment outcomes, preventing complications, and promoting tissue regeneration. In response to this need, a variety of polymeric biomaterials have been developed. A growing focus in the field involves the design of bioadhesives and bioactive materials, which offer promising solutions for wound management. Recent advances in materials engineering have led to the development of polymer biomaterials with excellent biocompatibility, strong adhesion to biological surfaces, and bioactive properties that support rapid wound closure and tissue repair. This review discusses the latest progress in the development and application of bioadhesives and bioactive hydrogels for wound management and tissue regeneration, highlighting potential directions for future biomaterial research.
Collapse
Affiliation(s)
- Yeonjeong Kim
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
| | - Sung Eun Kim
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
| | - Ki Dong Park
- Department of Molecular Science and Technology, Ajou University, 5 Woncheon, Yeongtong, Suwon 443-749, Republic of Korea.
| | - Kyung Min Park
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea; Research Center for Bio Materials & Process Development, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea.
| |
Collapse
|
4
|
Zhang Z, Yang S, Mi F, Yang Y, Song Q, Gao Y, Wu C, Wen W. Nanoparticle-Reinforced Hydrogel with a Well-Defined Pore Structure for Sustainable Drug Release and Effective Wound Healing. ACS APPLIED BIO MATERIALS 2025; 8:1406-1417. [PMID: 39916309 PMCID: PMC11836925 DOI: 10.1021/acsabm.4c01659] [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: 11/07/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
Impaired chronic wounds are a common complication of diabetes. Inhibited angiogenesis and dysfunctional inflammation render diabetic wound healing a critical challenge. Herein, a sustainable therapeutic composite hydrogel is presented for diabetic wound healing, consisting of a cocktail formulation of anti-inflammatory and local anesthetic nanoparticles incorporated into a composite hydrogel. The surface-modified drug nanoparticles are loaded into the biocompatible hydrogels and cross-linked with a gel precursor to enhance the structure. The sustainable delivery system achieves more than 90% drug release, with a total therapy duration tunable from 4 to 72 h. Through the long-lasting anti-inflammatory and analgesic effects of the composite hydrogel, diabetic wounds are swiftly transitioned into the proliferation phase, augmenting the survival and migration of keratinocytes and facilitating neovascularization and collagen alignment in diabetic wounds. These effects significantly improve the wound healing rate and skin regeneration process, achieving a healing rate that is 17 times that of untreated wounds. This study demonstrates that the hydrogel platform loaded with cocktail drug nanoparticles is promising for the rapid healing of diabetic wounds.
Collapse
Affiliation(s)
- Ziyi Zhang
- Division
of Emerging Interdisciplinary Areas, The
Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 000000, China
- Thrust
of Advanced Materials, The Hong Kong University
of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
| | - Siyu Yang
- Department
of Physics, The Hong Kong University of
Science and Technology, Clear Water Bay,
Kowloon, Hong Kong 000000, China
| | - Feixue Mi
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen 518055, China
| | - Yicheng Yang
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen 518055, China
| | - Qi Song
- Shenzhen
Shineway Technology Corporation, Shenzhen 518048, China
| | - Yibo Gao
- Shenzhen
Shineway Technology Corporation, Shenzhen 518048, China
| | - Changfeng Wu
- Department
of Biomedical Engineering, Southern University
of Science and Technology, Shenzhen 518055, China
| | - Weijia Wen
- Division
of Emerging Interdisciplinary Areas, The
Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 000000, China
- Thrust
of Advanced Materials, The Hong Kong University
of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, China
- Department
of Physics, The Hong Kong University of
Science and Technology, Clear Water Bay,
Kowloon, Hong Kong 000000, China
| |
Collapse
|
5
|
Tian Z, Chen H, Zhao P. Compliant immune response of silk-based biomaterials broadens application in wound treatment. Front Pharmacol 2025; 16:1548837. [PMID: 40012629 PMCID: PMC11861559 DOI: 10.3389/fphar.2025.1548837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025] Open
Abstract
The unique properties of sericin and silk fibroin (SF) favor their widespread application in biopharmaceuticals, particularly in wound treatment and bone repair. The immune response directly influences wound healing cycle, and the extensive immunomodulatory functions of silk-based nanoparticles and hydrogels have attracted wide attention. However, different silk-processing methods may trigger intense immune system resistance after implantation into the body. In this review, we elaborate on the inflammation and immune responses caused by the implantation of sericin and SF and also explore their anti-inflammatory properties and immune regulatory functions. More importantly, we describe the latest research progress in enhancing the immunotherapeutic and anti-inflammatory effects of composite materials prepared from silk from a mechanistic perspective. This review will provide a useful reference for using the correct processes to exploit silk-based biomaterials in different wound treatments.
Collapse
Affiliation(s)
- Zhiqiang Tian
- Biological Science Research Center, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Southwest University, Chongqing, China
| | - Hong Chen
- Department of Orthopedics, 903 Hospital of Joint Logistic Support Force of The People’s Liberation Army, Hangzhou, China
| | - Ping Zhao
- Biological Science Research Center, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Southwest University, Chongqing, China
| |
Collapse
|
6
|
Guo Z, Liu X, Xia Y, Wang J, Li J, Wang L, Li Y, Jia S, Sun Y, Feng J, Huang J, Dong Y, Wang L, Li X. Assembly of Recombinant Proteins into β-Sheet Fibrillating Peptide-Driven Supramolecular Hydrogels for Enhanced Diabetic Wound Healing. ACS Biomater Sci Eng 2025; 11:228-238. [PMID: 39651554 DOI: 10.1021/acsbiomaterials.4c01723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Supramolecular hydrogels offer a noncovalent binding platform that preserves the bioactivity of structural molecules while enhancing their stability, particularly in the context of diabetic wound repair. In this study, we developed protein-peptide-based supramolecular hydrogels by assembling β-sheet fibrillizing peptides (designated Q11) with β-tail fused recombinant proteins. The Q11 peptides have the ability to drive the gradated assembly of N- or C-terminal β-sheet structure (β-tail) fused recombinant proteins. We first investigated the assembly properties of Q11 and assessed its stability under varying pH and temperature conditions by combining Q11 with two β-tail fused fluorescent proteins. The results showed that Q11 enhanced the tolerance of the fluorescent proteins to changes in pH and temperature. Building upon these findings, we designed collagen-like proteins and Sonic Hedgehog-fused recombinant proteins (CLP-Shh) that could be assembled with Q11 to form peptide-protein supramolecular hydrogels. These hydrogels demonstrated the ability to improve cell viability and migration and upregulate key markers of cell growth. Further in vivo studies revealed that the Q11-driven supramolecular hydrogel effectively enhances diabetic wound healing and epidermal regeneration by promoting the expression of epidermal-related proteins and immune factors. This study highlights the potential of supramolecular hydrogels for clinical applications and their promise in the development of biofunctional hydrogels for therapeutic use.
Collapse
Affiliation(s)
- Zhao Guo
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Xing Liu
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Yan Xia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Jie Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Jiaqi Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Liping Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Yimiao Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Shuang Jia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Yinan Sun
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Jian Feng
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Jingxia Huang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Yuxin Dong
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Liyao Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| | - Xinyu Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot 010020, PR China
| |
Collapse
|
7
|
Chen X, Tian P, Chai W, Zhang L, Qin M, Fan M, Liang N, Kim J, Wang Y, Lu WW, Wang D, Cui X, Pan H. A Multisynergistic Strategy for Bone Tumor Treatment: Orchestrating Oxidative Stress and Autophagic Flux Inhibition by Environmental-Response Nanoparticle. Adv Healthc Mater 2025; 14:e2402872. [PMID: 39663711 DOI: 10.1002/adhm.202402872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Tumor therapy has advanced significantly in recent years, but tumor cells can still evade and survive the treatment through various mechanisms. Notably, tumor cells use autophagy to sustain viability by removing impaired mitochondria and clearing excess reactive oxygen species (ROS). In this study, the aim is to amplify intracellular oxidative stress by inhibiting mitochondrial autophagic flux. Multisynergistic environmental-response nanoparticles (ERNs) are engineered by integrating gold nanoparticles and copper peroxide with borosilicate bioactive glass. The controlled release of copper and inhibition of autophagy flux triggered an overabundance and accumulation of oxidative stress within the tumor cells. This stress triggered immunogenic tumor cell death, believed to initiate a systemic immune response. The tumor microenvironment (TME) transitioned back to a normal physiological state as tumor cells are ablated. ERNs responded to the microenvironment changes by depositing hydroxyapatite on the surface and spontaneously enhancing bone regeneration. This innovative formulation facilitates the functional transition of ERNs from "anti-tumor therapy" to "biomineralization" that kills cancers and induces new bone formation. Overall, it is shown that the ERNs effectively eradicate cancers by utilizing chemodynamic therapy, starvation therapy, and immunotherapy.
Collapse
Affiliation(s)
- Xiaochen Chen
- School of materials science and engineering, Tongji University, Shanghai, 201804, P.R. China
| | - Pengfei Tian
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Wenwen Chai
- School of materials science and engineering, Tongji University, Shanghai, 201804, P.R. China
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Liyan Zhang
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Muyan Qin
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Mengke Fan
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Na Liang
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Jua Kim
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Yansong Wang
- Department of Orthopedics, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150070, P.R. China
| | - Weijia William Lu
- Department of Orthopaedics and Traumatology, Li Ka Shing faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, P.R. China
| | - Deping Wang
- School of materials science and engineering, Tongji University, Shanghai, 201804, P.R. China
| | - Xu Cui
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
- Shenzhen Healthemes Biotechnology Co. Ltd., Shenzhen, 518120, P.R. China
| |
Collapse
|
8
|
Liu M, Liu X, Zhang J, Liang S, Gong Y, Shi S, Yuan X. Single-cell RNA sequencing reveals the heterogeneity of myofibroblasts in wound repair. Genomics 2025; 117:110982. [PMID: 39706310 DOI: 10.1016/j.ygeno.2024.110982] [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: 03/20/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Skin wound repair involves myofibroblasts crucial for tissue integrity. This study utilized single-cell RNA sequencing to explore myofibroblast diversity in various wound healing scenarios. Analysis of 89,148 cells from skin ulcers, keloids, and normal scars identified 13 cell clusters. Myofibroblast subcluster analysis unveiled 11 subsets, with subclusters 1 and 9 predominant in ulcers. Subcluster 1 exhibited heightened matrix metalloproteinase expression and involvement in bacterial response and angiogenesis, crucial in inflammation. Tissue validation confirmed subcluster 1 significance., while animal models supported upregulated CA12, TDO2, and IL-7R in chronic ulcers. These findings illuminate myofibroblast heterogeneity and their impact on wound healing, offering insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Miaonan Liu
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxuan Liu
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingchi Zhang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shaocong Liang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Gong
- Department of Burns and Wound Repairing, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shengjun Shi
- Department of Burns and Wound Repairing, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Xiaopeng Yuan
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Department of Laboratory Medicine, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University; Shenzhen 518020, Guangdong China..
| |
Collapse
|
9
|
Feng J, Gao W, Ge P, Chang S, Wang T, Zhao Q, He B, Pan S. Poly(thioctic acid) Hydrogels Integrated with Self-Healing, Bioadhesion, Antioxidation, and Antibiosis for Infected Wound Treatment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:65877-65889. [PMID: 39574373 DOI: 10.1021/acsami.4c14752] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Bacterial infections pose significant challenges in wound healing and are a serious threat to human health. Hydrogels have emerged as an ideal wound dressing due to their three-dimensional network, which facilitates exudate absorption and maintains a moist environment conducive to healing. Herein, we developed integrated hydrogels composed of poly(thioctic acid) (PTA), polydopamine (PDA), and curcumin (Cur). The formation of covalent and hydrogen bonds among PTA, PDA, and Cur endowed the hydrogels with excellent self-healing and bioadhesion properties. These hydrogels were utilized as dressings for healing Staphylococcus aureus-infected wounds. The PDA-PTA-Cur 16 hydrogel showed the best overall performance in stability, bioadhesion, antioxidant activity, and antibacterial effectiveness. The in vivo results revealed that the PDA-PTA-Cur 16 hydrogel accelerated infected wound healing by inhibiting bacterial growth, alleviating inflammation, promoting collagen deposition, and inducing angiogenesis. This multifunctional hydrogel not only enhances wound healing but also presents a promising strategy for combating bacterial infections in clinical settings.
Collapse
Affiliation(s)
- Juan Feng
- School of Pharmacy, Chengdu University, Chengdu 610106, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Wenxia Gao
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Pengjin Ge
- Chengdu Baili-Biopharm. LTD, Chengdu 610041, China
| | - Shuhua Chang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Ting Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
| | - Quan Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Shengsheng Pan
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
10
|
Wang Y, Zhou C, Li Z, Li G, Zou Y, Li X, Gu P, Liu J, Bai L, Yan H, Liang J, Zhang X, Fan Y, Sun Y. Injectable immunoregulatory hydrogels sequentially drive phenotypic polarization of macrophages for infected wound healing. Bioact Mater 2024; 41:193-206. [PMID: 39149597 PMCID: PMC11326493 DOI: 10.1016/j.bioactmat.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 08/17/2024] Open
Abstract
Regulating macrophage phenotypes to reconcile the conflict between bacterial suppression and tissue regeneration is ideal for treating infectious skin wounds. Here, an injectable immunoregulatory hydrogel (SrmE20) that sequentially drives macrophage phenotypic polarization (M0 to M1, then to M2) was constructed by integrating anti-inflammatory components and proinflammatory solvents. In vitro experiments demonstrated that the proinflammatory solvent ethanol stabilized the hydrogel structure, maintained the phenolic hydroxyl group activity, and achieved macrophages' proinflammatory transition (M0 to M1) to enhance antibacterial effects. With ethanol depletion, the hydrogel's cations and phenolic hydroxyl groups synergistically regulated macrophages' anti-inflammatory transition (M1 to M2) to initiate regeneration. In the anti-contraction full-thickness wound model with infection, this hydrogel effectively eliminated bacteria and even achieved anti-inflammatory M2 macrophage accumulation at three days post-surgery, accelerated angiogenesis and collagen deposition. By sequentially driving macrophage phenotypic polarization, this injectable immunoregulatory hydrogel will bring new guidance for the care and treatment of infected wounds.
Collapse
Affiliation(s)
- Yuxiang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Chen Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Zhulian Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Gong Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Yaping Zou
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Xing Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Peiyang Gu
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Jingyi Liu
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital of Sichuan University, 37# Guoxue Lane, Chengdu, Sichuan, 610041, China
| | - Hong Yan
- Department of Plastic, Aesthetic, Reparative and Reconstructive Surgery/Wound Repair Center, West China Second University Hospital of Sichuan University, 20# Section 3, South Renmin Road, Chengdu, Sichuan, 610041, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29# Wangjiang Road, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan, 610064, China
| |
Collapse
|
11
|
Li Y, Xia Y, Liu X, Wang J, Sun Y, Huang J, Guo Z, Jia S, Chen Y, Wang J, Wang L, Li J, Feng J, Wang L, Li X. Rational design of bioengineered recombinant collagen-like protein enhances GelMA hydrogel for diabetic wound healing. Int J Biol Macromol 2024; 280:136012. [PMID: 39326607 DOI: 10.1016/j.ijbiomac.2024.136012] [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: 07/13/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Gelatin methacryloyl (GelMA) holds significant potential in tissue engineering; however, its clinical applications are often constrained by its lack of functional groups. To overcome this limitation, recombinant proteins with multiple biofunctional domains present a promising strategy for GelMA functionalization, enhancing its biological properties. In this study, we developed a rationally designed recombinant collagen-like protein (RC) engineered with multiple biofunctional domains, which demonstrated the ability to upregulate collagen 1α (COL-1α) expression in NIH-3 T3 cells. By utilizing EDC/NHS chemistry, the purified RC was conjugated to GelMA, resulting in GelMA-RC hydrogels that significantly improved cell viability and migration compared to unmodified GelMA. Subsequent in vivo studies showed that RC-modified GelMA exhibited superior wound healing efficacy, largely attributed to enhanced expression of cytokeratin-14 (CK-14) and COL-1α. These findings underscore the potential of RC-functionalized GelMA in promoting diabetic wound repair and suggest broader applicability for functionalizing other biomaterials.
Collapse
Affiliation(s)
- Yimiao Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Yan Xia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Xing Liu
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jieqi Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Yinan Sun
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jinxia Huang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Zhao Guo
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Shuang Jia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Yufang Chen
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jie Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Liping Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jiaqi Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jian Feng
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Liyao Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Xinyu Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China.
| |
Collapse
|
12
|
Wang M, Deng Y, Huang C, Javeed A, Wang Y, Han B, Jiang G. A chitosan-based hydrogel loaded with fenofibrate for diabetic wound healing. Biomater Sci 2024; 12:4682-4694. [PMID: 39077924 DOI: 10.1039/d4bm00499j] [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: 07/31/2024]
Abstract
Diabetic wounds represent a common chronic condition, posing significant challenges in the treatment process due to bacterial infections, increased generation of reactive oxygen species (ROS) and exacerbated inflammation. Fenofibrate (FEN) is a clinical medication used for lipid regulation. In this study, it was utilized for the first time as an effective component of wound dressings for treating diabetic ulcers, exploring its novel applications further. Therefore, we prepared a polyvinyl alcohol/chitosan/FEN (PCF) hydrogel using a freeze-thaw method and conducted physicochemical characterization of the PCF hydrogel to further elucidate its biological functions. In vitro studies demonstrated that the PCF hydrogel exhibits excellent biocompatibility along with significant antimicrobial, pro-angiogenic, ROS-scavenging, and anti-inflammatory properties. Subsequent animal experiments indicated that the PCF hydrogel has the ability to promote blood vessel formation and collagen deposition. Additionally, the PCF hydrogel showed a significant inhibitory effect on the inflammatory response, as evidenced by the reductions in the levels of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). These compelling findings accentuate the promising application of the PCF hydrogel in the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Miaofeng Wang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Laboratory of Antiallergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China.
| | - Yaping Deng
- Department of Clinical Pharmacology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang 311202, China
| | - Cancan Huang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Laboratory of Antiallergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China.
| | - Ansar Javeed
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Laboratory of Antiallergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China.
| | - Yifan Wang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Laboratory of Antiallergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China.
| | - Bingnan Han
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Laboratory of Antiallergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China.
| | - Guojun Jiang
- Department of Pharmacy, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou 311200, Zhejiang, China.
| |
Collapse
|
13
|
Zhang L, Huang J, Li L, Zhang H, Li S, Chai W, Chen X, Liu C, Honiball JR, Li B, Ren Y, Chu L, Luo X, Pan H, Cui X. Exploratory Investigation of Zinc-Modified Borosilicate Bioactive Glass: A New Methodology for Its Biocompatibility, Immunoregulation, and Pro-Angiogenic Property Evaluation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:46016-46034. [PMID: 39167416 DOI: 10.1021/acsami.4c08487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
The assessment of biodegradable materials, such as bioactive glass, under the existing ISO 10993 standard test methods poses a significant challenge due to potential cell viability impairment caused by the accumulation of degraded products in a static environment. Therefore, innovative methodologies are urgently needed to tailor the unique biodegradation characteristics of these materials, providing more precise and scientific insights into biosafety and efficacy verification. Motivation by its bidirectional regulation of angiogenesis and immunity, zinc (Zn) was incorporated into sol-gel-derived borosilicate bioactive glasses (SBSGs) to fabricate Zn-incorporated borosilicate bioactive glasses (SBSG-Zn) to complement the tissue repair capabilities of bioactive glasses. Both SBSG and SBSG-Zn glasses consist of nanosized particles, slit mesoporous pores, high specific surface areas, and bioreactivity. In vitro comparative analysis, conducted according to ISO 10993 standards, demonstrates that only at suitable dilution rates─such as the 8-fold dilution employed in this study─do extracts of SBSG and SBSG-Zn glasses exhibit low cytotoxicity when cultured with human umbilical vein endothelial cells (HUVECs). Notably, SBSG-Zn glasses show optimal promotion of angiogenic gene expression in HUVECs. Furthermore, within an appropriate concentration range of released ions, SBSG-Zn glass extracts not only promote cell survival but also modulate the expression of anti-inflammatory genes while simultaneously inhibiting pro-inflammatory genes concurrently. After being implanted in rat subcutaneous defect models, both SBSG and SBSG-Zn glasses demonstrated the local immunoregulation and angiogenic effects. SBSG-Zn stands out by demonstrating superior modulation of M1/M2 polarization in macrophages as validated by altered secretion of key factors in macrophages and expression of relevant growth factors in HUVECs. These findings underscore the potential for convenient manipulation of localized angiogenic and immunoregulation through the incorporation of zinc into bioactive glass, emphasizing the importance of ensuring the appropriate ion doses are applied for achieving optimal therapeutic efficiency.
Collapse
Affiliation(s)
- Liyan Zhang
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Jing Huang
- Department of Orthopedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, P. R. China
| | - Li Li
- Department of Orthopedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, P. R. China
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR 999077, P. R. China
| | - Hao Zhang
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Shuaijie Li
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Wenwen Chai
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Xiaochen Chen
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Chunyu Liu
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - John Robert Honiball
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR 999077, P. R. China
| | - Bing Li
- Department of Orthopedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, P. R. China
| | - Youliang Ren
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, P. R. China
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Lei Chu
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, P. R. China
| | - Xuegang Luo
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Xu Cui
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| |
Collapse
|
14
|
Kamal R, Awasthi A, Pundir M, Thakur S. Healing the diabetic wound: Unlocking the secrets of genes and pathways. Eur J Pharmacol 2024; 975:176645. [PMID: 38759707 DOI: 10.1016/j.ejphar.2024.176645] [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: 03/04/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Diabetic wounds (DWs) are open sores that can occur anywhere on a diabetic patient's body. They are often complicated by infections, hypoxia, oxidative stress, hyperglycemia, and reduced growth factors and nucleic acids. The healing process involves four phases: homeostasis, inflammation, proliferation, and remodeling, regulated by various cellular and molecular events. Numerous genes and signaling pathways such as VEGF, TGF-β, NF-κB, PPAR-γ, MMPs, IGF, FGF, PDGF, EGF, NOX, TLR, JAK-STAT, PI3K-Akt, MAPK, ERK, JNK, p38, Wnt/β-catenin, Hedgehog, Notch, Hippo, FAK, Integrin, and Src pathways are involved in these events. These pathways and genes are often dysregulated in DWs leading to impaired healing. The present review sheds light on the pathogenesis, healing process, signaling pathways, and genes involved in DW. Further, various therapeutic strategies that target these pathways and genes via nanotechnology are also discussed. Additionally, clinical trials on DW related to gene therapy are also covered in the present review.
Collapse
Affiliation(s)
- Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Mandeep Pundir
- School of Pharmaceutical Sciences, RIMT University, Punjab, 142001, India; Chitkara College of Pharmacy, Chitkara University, Punjab, 142001, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
15
|
Chansoria P, Chaudhari A, Etter EL, Bonacquisti EE, Heavey MK, Le J, Maruthamuthu MK, Kussatz CC, Blackwell J, Jasiewicz NE, Sellers RS, Maile R, Wallet SM, Egan TM, Nguyen J. Instantly adhesive and ultra-elastic patches for dynamic organ and wound repair. Nat Commun 2024; 15:4720. [PMID: 38830847 PMCID: PMC11148085 DOI: 10.1038/s41467-024-48980-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
Bioadhesive materials and patches are promising alternatives to surgical sutures and staples. However, many existing bioadhesives do not meet the functional requirements of current surgical procedures and interventions. Here, we present a translational patch material that exhibits instant adhesion to tissues (2.5-fold stronger than Tisseel, an FDA-approved fibrin glue), ultra-stretchability (stretching to >300% its original length without losing elasticity), compatibility with rapid photo-projection (<2 min fabrication time/patch), and ability to deliver therapeutics. Using our established procedures for the in silico design and optimization of anisotropic-auxetic patches, we created next-generation patches for instant attachment to tissues while conforming to a broad range of organ mechanics ex vivo and in vivo. Patches coated with extracellular vesicles derived from mesenchymal stem cells demonstrate robust wound healing capability in vivo without inducing a foreign body response and without the need for patch removal that can cause pain and bleeding. We further demonstrate a single material-based, void-filling auxetic patch designed for the treatment of lung puncture wounds.
Collapse
Affiliation(s)
- Parth Chansoria
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ameya Chaudhari
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Emma L Etter
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Emily E Bonacquisti
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mairead K Heavey
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jiayan Le
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Murali Kannan Maruthamuthu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Caden C Kussatz
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - John Blackwell
- Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Natalie E Jasiewicz
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rani S Sellers
- Pathology and Laboratory Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Robert Maile
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shannon M Wallet
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Thomas M Egan
- Division of Cardiothoracic Surgery, Department of Surgery, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- North Carolina State University, Raleigh, NC, 27695, USA
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
16
|
Zhou Y, Li K, Adelson DL. An unmet need for pharmacology: Treatments for radiation-induced gastrointestinal mucositis. Biomed Pharmacother 2024; 175:116767. [PMID: 38781863 DOI: 10.1016/j.biopha.2024.116767] [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: 03/04/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Gastrointestinal mucositis (GIM) continues to be a significant issue in the management of abdominal cancer radiation treatments and chemotherapy, causing significant patient discomfort and therapy interruption or even cessation. This review will first focus on radiotherapy induced GIM, providing an understanding of its clinical landscape. Subsequently, the aetiology of GIM will be reviewed, highlighting diverse contributing factors. The cellular and tissue damage and associated molecular responses in GIM will be summarised in the context of the underlying complex biological processes. Finally, available drugs and pharmaceutical therapies will be evaluated, underscoring their insufficiency, and highlighting the need for further research and innovation. This review will emphasize the urgent need for improved pharmacologic therapeutics for GIM, which is a key research priority in oncology.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; Zhendong Australia China Centre for Molecular Chinese Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia.
| | - Kun Li
- Beijing Zhendong Guangming Pharmaceutical Research Institute, Beijing 100120, China.
| | - David L Adelson
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; Zhendong Australia China Centre for Molecular Chinese Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia.
| |
Collapse
|
17
|
Liu X, Guo Z, Wang J, Shen W, Jia Z, Jia S, Li L, Wang J, Wang L, Li J, Sun Y, Chen Y, Zhang M, Bai J, Wang L, Li X. Thiolation-Based Protein-Protein Hydrogels for Improved Wound Healing. Adv Healthc Mater 2024; 13:e2303824. [PMID: 38303578 DOI: 10.1002/adhm.202303824] [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: 11/02/2023] [Revised: 01/28/2024] [Indexed: 02/03/2024]
Abstract
The limitations of protein-based hydrogels, including their insufficient mechanical properties and restricted biological functions, arise from the highly specific functions of proteins as natural building blocks. A potential solution to overcome these shortcomings is the development of protein-protein hydrogels, which integrate structural and functional proteins. In this study, a protein-protein hydrogel formed by crosslinking bovine serum albumin (BSA) and a genetically engineered intrinsically disordered collagen-like protein (CLP) through Ag─S bonding is introduced. The approach involves thiolating lysine residues of BSA and crosslinking CLP with Ag+ ions, utilizing thiolation of BSA and the free-cysteines of CLP. The resulting protein-protein hydrogels exhibit exceptional properties, including notable plasticity, inherent self-healing capabilities, and gel-sol transition in response to redox conditions. In comparison to standalone BSA hydrogels, these protein-protein hydrogels demonstrate enhanced cellular viability, and improved cellular migration. In vivo experiments provide conclusive evidence of accelerated wound healing, observed not only in murine models with streptozotocin (Step)-induced diabetes but also in zebrafish models subjected to UV-burn injuries. Detailed mechanistic insights, combined with assessments of proinflammatory cytokines and the expression of epidermal differentiation-related proteins, robustly validate the protein-protein hydrogel's effectiveness in promoting wound repair.
Collapse
Affiliation(s)
- Xing Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Zhao Guo
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Jie Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Wenting Shen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, P. R. China
| | - Zhenzhen Jia
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, P. R. China
| | - Shuang Jia
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Limiao Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Jieqi Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Liping Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Jiaqi Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Yinan Sun
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Yufang Chen
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Min Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Jia Bai
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Liyao Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Xinyu Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| |
Collapse
|
18
|
Xu C, Cao JF, Pei Y, Kim Y, Moon H, Fan CQ, Liao MC, Wang XY, Yao F, Zhang YJ, Zhang SH, Zhang J, Li JZ, Kim JS, Ma L, Xie ZJ. Injectable hydrogel harnessing foreskin mesenchymal stem cell-derived extracellular vesicles for treatment of chronic diabetic skin wounds. J Control Release 2024; 370:339-353. [PMID: 38685383 DOI: 10.1016/j.jconrel.2024.04.049] [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: 11/14/2023] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Chronic skin wounds are a serious complication of diabetes with a high incidence rate, which can lead to disability or even death. Previous studies have shown that mesenchymal stem cells derived extracellular vesicles (EVs) have beneficial effects on wound healing. However, the human foreskin mesenchymal stem cell (FSMSCs)-derived extracellular vesicle (FM-EV) has not yet been isolated and characterized. Furthermore, the limited supply and short lifespan of EVs also hinder their practical use. In this study, we developed an injectable dual-physical cross-linking hydrogel (PSiW) with self-healing, adhesive, and antibacterial properties, using polyvinylpyrrolidone and silicotungstic acid to load FM-EV. The EVs were evenly distributed in the hydrogel and continuously released. In vivo and vitro tests demonstrated that the synergistic effect of EVs and hydrogel could significantly promote the repair of diabetic wounds by regulating macrophage polarization, promoting angiogenesis, and improving the microenvironment. Overall, the obtained EVs-loaded hydrogels developed in this work exhibited promising applicability for the repair of chronic skin wounds in diabetes patients.
Collapse
Affiliation(s)
- Chang Xu
- Institute of Pediatrics, Shenzhen Children's Hospital, Clinical Medical College of Southern University of Science and Technology, Shenzhen 518038, China
| | - Jin-Feng Cao
- Beijing Key Laboratory of Wood Science and Engineering, Beijing Forestry University, Beijing 100083, China; Key Laboratory of Wood Material Science and Application (Beijing Forestry University), Ministry of Education, Beijing 100083, China
| | - Yue Pei
- Institute of Pediatrics, Shenzhen Children's Hospital, Clinical Medical College of Southern University of Science and Technology, Shenzhen 518038, China
| | - Yujin Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Huiyeon Moon
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Chui-Qin Fan
- Institute of Pediatrics, Shenzhen Children's Hospital, Clinical Medical College of Southern University of Science and Technology, Shenzhen 518038, China
| | - Mao-Chuan Liao
- Institute of Pediatrics, Shenzhen Children's Hospital, Clinical Medical College of Southern University of Science and Technology, Shenzhen 518038, China
| | - Xing-Yu Wang
- Department of Emergency, ChangYang Tujia Autonomous County People's Hospital, Yichang 443000, China
| | - Fei Yao
- Eye Center of Xiangya Hospital, Central South University, Changsha 410000, China
| | - Yu-Jun Zhang
- Institute of Pediatrics, Shenzhen Children's Hospital, Clinical Medical College of Southern University of Science and Technology, Shenzhen 518038, China
| | - Shao-Hui Zhang
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jian Zhang
- Institute of Materials Science and Devices, School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jian-Zhang Li
- Beijing Key Laboratory of Wood Science and Engineering, Beijing Forestry University, Beijing 100083, China; Key Laboratory of Wood Material Science and Application (Beijing Forestry University), Ministry of Education, Beijing 100083, China.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Lian Ma
- Institute of Pediatrics, Shenzhen Children's Hospital, Clinical Medical College of Southern University of Science and Technology, Shenzhen 518038, China; Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen 518038, China; Department of Pediatrics, The Third Affifiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.
| | - Zhong-Jian Xie
- Institute of Pediatrics, Shenzhen Children's Hospital, Clinical Medical College of Southern University of Science and Technology, Shenzhen 518038, China; Shenzhen International Institute for Biomedical Research, Shenzhen 518116, Guangdong, China.
| |
Collapse
|
19
|
Yaron JR, Gosangi M, Pallod S, Rege K. In situ light-activated materials for skin wound healing and repair: A narrative review. Bioeng Transl Med 2024; 9:e10637. [PMID: 38818119 PMCID: PMC11135152 DOI: 10.1002/btm2.10637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/22/2023] [Accepted: 12/12/2023] [Indexed: 06/01/2024] Open
Abstract
Dermal wounds are a major global health burden made worse by common comorbidities such as diabetes and infection. Appropriate wound closure relies on a highly coordinated series of cellular events, ultimately bridging tissue gaps and regenerating normal physiological structures. Wound dressings are an important component of wound care management, providing a barrier against external insults while preserving the active reparative processes underway within the wound bed. The development of wound dressings with biomaterial constituents has become an attractive design strategy due to the varied functions intrinsic in biological polymers, such as cell instructiveness, growth factor binding, antimicrobial properties, and tissue integration. Using photosensitive agents to generate crosslinked or photopolymerized dressings in situ provides an opportunity to develop dressings rapidly within the wound bed, facilitating robust adhesion to the wound bed for greater barrier protection and adaptation to irregular wound shapes. Despite the popularity of this fabrication approach, relatively few experimental wound dressings have undergone preclinical translation into animal models, limiting the overall integrity of assessing their potential as effective wound dressings. Here, we provide an up-to-date narrative review of reported photoinitiator- and wavelength-guided design strategies for in situ light activation of biomaterial dressings that have been evaluated in preclinical wound healing models.
Collapse
Affiliation(s)
- Jordan R. Yaron
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State UniversityTempeArizonaUSA
- School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State UniversityTempeArizonaUSA
| | - Mallikarjun Gosangi
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State UniversityTempeArizonaUSA
| | - Shubham Pallod
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State UniversityTempeArizonaUSA
| | - Kaushal Rege
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State UniversityTempeArizonaUSA
- School for Engineering of Matter, Transport, and Energy, Ira A. Fulton Schools of Engineering, Arizona State UniversityTempeArizonaUSA
- Chemical Engineering, Arizona State UniversityTempeArizonaUSA
| |
Collapse
|
20
|
Zhu Z, Ding J, Qin M, Wang L, Jiang D, Zhao J, Wang D, Jia W. Enhanced ·OH-Scavenging Activity of Cu-CeO x Nanozyme via Resurrecting Macrophage Nrf2 Transcriptional Activity Facilitates Diabetic Wound Healing. Adv Healthc Mater 2024; 13:e2303229. [PMID: 38298062 DOI: 10.1002/adhm.202303229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/23/2024] [Indexed: 02/02/2024]
Abstract
Diabetic wounds are a prevalent and devastating complication of diabetes, which may impede their healing and regeneration. In diabetic wounds, excess reactive oxygen species (ROS) activate the nuclear factor kappa-B pathway, leading to transcriptional silencing of nuclear factor erythroid 2-related factor 2 (Nrf2), resulting in a vicious cycle of oxidative stress and inflammation. Conventional nanozymes have limitations in preventing the continuous production of ROS, including the most oxidizing reactive hydroxyl radical (·OH), although they can remove pre-existing ROS. Herein, a novel antioxidant nanoplatform addresses this challenge by incorporating JSH-23 into the mesoporous of cupric-doped cerium oxide nanozymes. Additionally, for rapid wound adaptability and durable tissue adhesion, a nanozyme hydrogel spray consisting of oxidized sodium alginate and methacrylate gelatin is constructed, named OG@CCJs. This platform resurrects Nrf2 transcriptional activity of macrophages in vitro, curbing the production of ROS at its source, particularly ·OH, while enabling the nanozymes to scavenge previously generated ROS. OG@CCJs significantly alleviate oxidative stress in diabetic wounds in vivo, promoting wound healing. Overall, the proposed nanozyme-hydrogel spray with enhanced ·OH-scavenging activity uses a "two-track" antioxidant strategy to rebuild the antioxidant defense barrier of macrophages. This pioneering approach highlights the tremendous potential of OG@CCJs for facilitating diabetic wound healing.
Collapse
Affiliation(s)
- Ziyang Zhu
- Postgraduate Training Base of Jinzhou Medical University in Shanghai Sixth People's Hospital, Jinzhou Medical University, Jinzhou, 121001, China
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Jingxin Ding
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Muyan Qin
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Lingtian Wang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Dajun Jiang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Jinhui Zhao
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Deping Wang
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Weitao Jia
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| |
Collapse
|
21
|
Xu C, Hu L, Zeng J, Wu A, Deng S, Zhao Z, Geng K, Luo J, Wang L, Zhou X, Huang W, Long Y, Song J, Zheng S, Wu J, Chen Q. Gynura divaricata (L.) DC. promotes diabetic wound healing by activating Nrf2 signaling in diabetic rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117638. [PMID: 38135237 DOI: 10.1016/j.jep.2023.117638] [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: 08/08/2023] [Revised: 12/08/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
THE ETHNOPHARMACOLOGICAL SIGNIFICANCE Diabetic chronic foot ulcers pose a significant therapeutic challenge as a result of the oxidative stress caused by hyperglycemia. Which impairs angiogenesis and delays wound healing, potentially leading to amputation. Gynura divaricata (L.) DC. (GD), a traditional Chinese herbal medicine with hypoglycemic effects, has been proposed as a potential therapeutic agent for diabetic wound healing. However, the underlying mechanisms of its effects remain unclear. AIM OF THE STUDY In this study, we aimed to reveal the effect and potential mechanisms of GD on accelerating diabetic wound healing in vitro and in vivo. MATERIALS AND METHODS The effects of GD on cell proliferation, apoptosis, reactive oxygen species (ROS) production, migration, mitochondrial membrane potential (MMP), and potential molecular mechanisms were investigated in high glucose (HG) stimulated human umbilical vein endothelial cells (HUVECs) using CCK-8, flow cytometry assay, wound healing assay, immunofluorescence, DCFH-DA staining, JC-1 staining, and Western blot. Full-thickness skin defects were created in STZ-induced diabetic rats, and wound healing rate was tracked by photographing them every day. HE staining, immunohistochemistry, and Western blot were employed to investigate the effect and molecular mechanism of GD on wound healing in diabetic rats. RESULTS GD significantly improved HUVEC survival, decreased apoptosis, lowered ROS production, restored MMP, improved migration ability, and raised VEGF expression. The use of Nrf2-siRNA completely abrogated these effects. Topical application of GD promoted angiogenesis and granulation tissue growth, resulting in faster healing of diabetic wounds. The expression of VEGF, CD31, and VEGFR was elevated in the skin tissue of diabetic rats after GD treatment, which upregulated HO-1, NQO-1, and Bcl-2 expression while downregulating Bax expression via activation of the Nrf2 signaling pathway. CONCLUSION The findings of this study indicate that GD has the potential to serve as a viable alternative treatment for diabetic wounds. This potential arises from its ability to mitigate the negative effects of oxidative stress on angiogenesis, which is regulated by the Nrf2 signaling pathway. The results of our study offer valuable insights into the therapeutic efficacy of GD in the treatment of diabetic wounds, emphasizing the significance of directing interventions towards the Nrf2 signaling pathway to mitigate oxidative stress and facilitate the process of angiogenesis.
Collapse
Affiliation(s)
- Caimin Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; School of Nursing, Southwest Medical University, Luzhou, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University
| | - Lixin Hu
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; School of Nursing, Southwest Medical University, Luzhou, China
| | - Jing Zeng
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Shilong Deng
- Department of Nursing, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Wound Healing Basic Research and Clinical Application Key Laboratory, School of Nursing, Southwest Medical University, LuZhou, China
| | - Zijuan Zhao
- Department of Nursing, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Wound Healing Basic Research and Clinical Application Key Laboratory, School of Nursing, Southwest Medical University, LuZhou, China
| | - Kang Geng
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jiesi Luo
- School of Basic Medicine Sciences, Southwest Medical University, Luzhou, China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaogang Zhou
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University
| | - Jianying Song
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; School of Nursing, Southwest Medical University, Luzhou, China
| | - Silin Zheng
- Department of Nursing, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jianming Wu
- School of Basic Medicine Sciences, Southwest Medical University, Luzhou, China.
| | - Qi Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, China; School of Nursing, Southwest Medical University, Luzhou, China; Department of Nursing, The Affiliated Hospital, Southwest Medical University, Luzhou, China; Wound Healing Basic Research and Clinical Application Key Laboratory, School of Nursing, Southwest Medical University, LuZhou, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Southwest Medical University.
| |
Collapse
|
22
|
Zhang H, Lin X, Cao X, Wang Y, Wang J, Zhao Y. Developing natural polymers for skin wound healing. Bioact Mater 2024; 33:355-376. [PMID: 38282639 PMCID: PMC10818118 DOI: 10.1016/j.bioactmat.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/02/2023] [Accepted: 11/16/2023] [Indexed: 01/30/2024] Open
Abstract
Natural polymers are complex organic molecules that occur in the natural environment and have not been subjected to artificial synthesis. They are frequently encountered in various creatures, including mammals, plants, and microbes. The aforementioned polymers are commonly derived from renewable sources, possess a notable level of compatibility with living organisms, and have a limited adverse effect on the environment. As a result, they hold considerable significance in the development of sustainable and environmentally friendly goods. In recent times, there has been notable advancement in the investigation of the potential uses of natural polymers in the field of biomedicine, specifically in relation to natural biomaterials that exhibit antibacterial and antioxidant characteristics. This review provides a comprehensive overview of prevalent natural polymers utilized in the biomedical domain throughout the preceding two decades. In this paper, we present a comprehensive examination of the components and typical methods for the preparation of biomaterials based on natural polymers. Furthermore, we summarize the application of natural polymer materials in each stage of skin wound repair. Finally, we present key findings and insights into the limitations of current natural polymers and elucidate the prospects for their future development in this field.
Collapse
Affiliation(s)
- Han Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiang Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xinyue Cao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yu Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jinglin Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518038, China
| |
Collapse
|
23
|
Zhu D, Wei W, Zhang J, Zhao B, Li Q, Jin P. Mechanism of damage of HIF-1 signaling in chronic diabetic foot ulcers and its related therapeutic perspectives. Heliyon 2024; 10:e24656. [PMID: 38318060 PMCID: PMC10839564 DOI: 10.1016/j.heliyon.2024.e24656] [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: 10/18/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
Diabetic foot ulcer (DFU) is a chronic complication of diabetes. Wound healing in patients with DFU is generally very slow, with a high recurrence rate even after the ulcer healed. The DFU remains a major clinical challenge due to a lack of understanding of its pathogenesis. Given the significant impact of DFU on patient health and medical costs, enhancing our understanding of pathophysiological alterations and wound healing in DFU is critical. A growing body of research has shown that impaired activation of the HIF-1 pathway in diabetics, which weakens HIF-1 mediated responses to hypoxia and leads to down-regulation of its downstream target genes, leading to incurable diabetic foot ulcers. By analyzing and summarizing the literature in recent years, this review summarizes the mechanism of HIF-1 signaling pathway damage in the development of DFU, analyzes and compares the application of PHD inhibitors, VHL inhibitors, biomaterials and stem cell therapy in chronic wounds of diabetes, and proposes a new treatment scheme mediated by participation in the HIF-1 signaling pathway, which provides new ideas for the treatment of DFU.
Collapse
Affiliation(s)
- Dong Zhu
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wuhan Wei
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jingyu Zhang
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bingkun Zhao
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qiang Li
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Peisheng Jin
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
24
|
Wang HY, Zhang Y, Zhang M, Zhang YQ. Functional modification of silk fibroin from silkworms and its application to medical biomaterials: A review. Int J Biol Macromol 2024; 259:129099. [PMID: 38176506 DOI: 10.1016/j.ijbiomac.2023.129099] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/26/2023] [Accepted: 12/26/2023] [Indexed: 01/06/2024]
Abstract
Silk fibroin (SF) from the silkworm Bombyx mori is a fibrous protein identified as a widely suitable biomaterial due to its biocompatibility, tunable degradation, and mechanical strength. Various modifications of SF protein can give SF fibers new properties and functions, broadening their applications in textile and biomedical industries. A diverse array of functional modifications on various forms of SF has been reported. In order to provide researchers with a more systematic understanding of the types of functional modifications of SF protein, as well as the corresponding applications, we comprehensively review the different types of functional modifications, including transgenic modification, modifications with chemical groups or biologically active substance, cross-linking and copolymerization without chemical reactions, their specific modification methods and applications. Furthermore, recent applications of SF in various medical biomaterials are briefly discussed.
Collapse
Affiliation(s)
- Hai-Yan Wang
- Obstetrical department, The People's Hospital of Suzhou New District, Suzhou, China
| | - Yun Zhang
- Obstetrical department, The People's Hospital of Suzhou New District, Suzhou, China
| | - Meng Zhang
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Yu-Qing Zhang
- Silk Biotechnology Laboratory, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
25
|
Zhao Y, Ran B, Lee D, Liao J. Photo-Controllable Smart Hydrogels for Biomedical Application: A Review. SMALL METHODS 2024; 8:e2301095. [PMID: 37884456 DOI: 10.1002/smtd.202301095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/28/2023] [Indexed: 10/28/2023]
Abstract
Nowadays, smart hydrogels are being widely studied by researchers because of their advantages such as simple preparation, stable performance, response to external stimuli, and easy control of response behavior. Photo-controllable smart hydrogels (PCHs) are a class of responsive hydrogels whose physical and chemical properties can be changed when stimulated by light at specific wavelengths. Since the light source is safe, clean, simple to operate, and easy to control, PCHs have broad application prospects in the biomedical field. Therefore, this review timely summarizes the latest progress in the PCHs field, with an emphasis on the design principles of typical PCHs and their multiple biomedical applications in tissue regeneration, tumor therapy, antibacterial therapy, diseases diagnosis and monitoring, etc. Meanwhile, the challenges and perspectives of widespread practical implementation of PCHs are presented in biomedical applications. This study hopes that PCHs will flourish in the biomedical field and this review will provide useful information for interested researchers.
Collapse
Affiliation(s)
- Yiwen Zhao
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Bei Ran
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Dashiell Lee
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
26
|
Xiang T, Guo Q, Jia L, Yin T, Huang W, Zhang X, Zhou S. Multifunctional Hydrogels for the Healing of Diabetic Wounds. Adv Healthc Mater 2024; 13:e2301885. [PMID: 37702116 DOI: 10.1002/adhm.202301885] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/10/2023] [Indexed: 09/14/2023]
Abstract
The healing of diabetic wounds is hindered by various factors, including bacterial infection, macrophage dysfunction, excess proinflammatory cytokines, high levels of reactive oxygen species, and sustained hypoxia. These factors collectively impede cellular behaviors and the healing process. Consequently, this review presents intelligent hydrogels equipped with multifunctional capacities, which enable them to dynamically respond to the microenvironment and accelerate wound healing in various ways, including stimuli -responsiveness, injectable self-healing, shape -memory, and conductive and real-time monitoring properties. The relationship between the multiple functions and wound healing is also discussed. Based on the microenvironment of diabetic wounds, antibacterial, anti-inflammatory, immunomodulatory, antioxidant, and pro-angiogenic strategies are combined with multifunctional hydrogels. The application of multifunctional hydrogels in the repair of diabetic wounds is systematically discussed, aiming to provide guidelines for fabricating hydrogels for diabetic wound healing and exploring the role of intelligent hydrogels in the therapeutic processes.
Collapse
Affiliation(s)
- Tao Xiang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Qianru Guo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Lianghao Jia
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Tianyu Yin
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Wei Huang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xinyu Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
27
|
Wang X, Li R, Zhao H. Enhancing angiogenesis: Innovative drug delivery systems to facilitate diabetic wound healing. Biomed Pharmacother 2024; 170:116035. [PMID: 38113622 DOI: 10.1016/j.biopha.2023.116035] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
Diabetic wounds (DW) constitute a substantial burden on global healthcare owing to their widespread occurrence as a complication of diabetes. Angiogenesis, a crucial process, plays a pivotal role in tissue recovery by supplying essential oxygen and nutrients to the injury site. Unfortunately, in diabetes mellitus, various factors disrupt angiogenesis, hindering wound healing. While biomaterials designed to enhance angiogenesis hold promise for the treatment of DWs, there is an urgent need for more in-depth investigations to fully unlock their potential in clinical management. In this review, we explore the intricate mechanisms of angiogenesis that are crucial for DW recovery. We introduce a rational design for angiogenesis-enhancing drug delivery systems (DDS) and provide a comprehensive summary and discussion of diverse biomaterials that enhance angiogenesis for facilitating DW healing. Lastly, we address emerging challenges and prospects in angiogenesis-enhancing DDS for facilitating DW healing, aiming to offer a comprehensive understanding of this critical healthcare issue and potential solutions.
Collapse
Affiliation(s)
- Xuan Wang
- Department of foot and ankle surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Runmin Li
- Department of foot and ankle surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Hongmou Zhao
- Department of foot and ankle surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China.
| |
Collapse
|
28
|
Jia S, Wang J, Wang X, Liu X, Li S, Li Y, Li J, Wang J, Man S, Guo Z, Sun Y, Jia Z, Wang L, Li X. Genetically encoded in situ gelation redox-responsive collagen-like protein hydrogel for accelerating diabetic wound healing. Biomater Sci 2023; 11:7748-7758. [PMID: 37753880 DOI: 10.1039/d3bm01010d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Genetically encoded collagen-like protein-based hydrogels have demonstrated remarkable efficacy in promoting the healing process in diabetic patients. However, the current methods for preparing these hydrogels pose significant challenges due to harsh reaction conditions and the reliance on chemical crosslinkers. In this study, we present a genetically encoded approach that allows for the creation of protein hydrogels without the need for chemical additives. Our design involves the genetic encoding of paired-cysteine residues at the C- and N-terminals of a meticulously engineered collagen-like recombination protein. The protein-based hydrogel undergoes a gel-sol transition in response to redox stimulation, achieving a gel-sol transition. We provide evidence that the co-incubation of the protein hydrogel with 3T3 cells not only enhances cell viability but also promotes cell migration. Moreover, the application of the protein hydrogel significantly accelerates the healing of diabetic wounds by upregulating the expression of collagen-1α (COL-1α) and Cytokeratin 14 (CK-14), while simultaneously reducing oxidant stress in the wound microenvironment. Our study highlights a straightforward strategy for the preparation of redox-responsive protein hydrogels, removing the need for additional chemical agents. Importantly, our findings underscore the potential of this hydrogel system for effectively treating diabetic wounds, offering a promising avenue for future therapeutic applications.
Collapse
Affiliation(s)
- Shuang Jia
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Jie Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Xiaojie Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Xing Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Shubin Li
- Department of Geriatric Medical Center, Inner Mongolia people's Hospital, 20 Zhaowuda Road, Hohhot, 010021, Inner Mongolia, China
| | - Yimiao Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Jiaqi Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Jieqi Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Shad Man
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Zhao Guo
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Yinan Sun
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Zhenzhen Jia
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, PR China
| | - Liyao Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
| | - Xinyu Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, PR China.
- Institutes of Biomedical Sciences, Inner Mongolia University, Inner Mongolia University, Hohhot, 010020, PR China
| |
Collapse
|
29
|
Chen X, Zhang L, Chai W, Tian P, Kim J, Ding J, Zhang H, Liu C, Wang D, Cui X, Pan H. Hypoxic Microenvironment Reconstruction with Synergistic Biofunctional Ions Promotes Diabetic Wound Healing. Adv Healthc Mater 2023; 12:e2301984. [PMID: 37740829 DOI: 10.1002/adhm.202301984] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/14/2023] [Indexed: 09/25/2023]
Abstract
Chronic hypoxia and ischemia make diabetic wounds non-healing. Cellular functions of diabetic chronic wounds are inhibited under a pathological environment. Therefore, this work develops a composite hydrogel system to promote diabetic wound healing. The composite hydrogel system consists of ε-poly-lysine (EPL), calcium peroxide (CP), and borosilicate glass (BG). The hydrogel supplies continuous dissolved oxygen molecules to the wound that can penetrate the skin tissue to restore normal cellular function and promote vascular regeneration. Biofunctional ions released from BGs can recruit more macrophages through neovascularization and modulate macrophage phenotypic transformation. Combining oxygen-mediated vascular regeneration and ion-mediated inflammatory regulation significantly accelerated diabetic wound healing. These findings indicate that this composite hydrogel system holds promise as a novel tissue engineering material.
Collapse
Affiliation(s)
- Xiaochen Chen
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Liyan Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Wenwen Chai
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Pengfei Tian
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jua Kim
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jingxin Ding
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Hao Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Chunyu Liu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Deping Wang
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Xu Cui
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd, Shenzhen, 518071, P. R. China
| |
Collapse
|
30
|
Wen W, Yang L, Wang X, Zhang H, Wu F, Xu K, Chen S, Liao Z. Fucoidan promotes angiogenesis and accelerates wound healing through AKT/Nrf2/HIF-1α signalling pathway. Int Wound J 2023; 20:3606-3618. [PMID: 37203309 PMCID: PMC10588368 DOI: 10.1111/iwj.14239] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023] Open
Abstract
After skin injury, wound repair involves a complex process in which angiogenesis plays a crucial role. Previous research has indicated that fucoidan may aid in wound healing; we therefore hypothesised that fucoidan may speed up the process by promoting angiogenesis. In this study, we investigated the potential molecular mechanism underlying fucoidan's ability to accelerate wound healing by promoting angiogenesis. Using a full-cut wound model, we observed that fucoidan significantly intensified wound closure and promoted granulation formation and collagen deposition. Immunofluorescence staining revealed that fucoidan also promoted wound angiogenesis, specifically by accelerating the migration of new blood vessels to the middle area of the wound. Furthermore, fucoidan demonstrated the ability to enhance the proliferation of human umbilical vein endothelial cells (HUVECs) damaged by hydrogen peroxide (H2 O2 ) and to improve the formation of endothelial tubes. Mechanistic studies revealed that fucoidan upregulated the protein levels of the AKT/Nrf2/HIF-1α signalling pathway, which plays a crucial role in angiogenesis. This was further confirmed using the inhibitor LY294002, which reversed the promotion of endothelial tube formation by fucoidan. Overall, our findings suggest that fucoidan can promote angiogenesis via the AKT/Nrf2/HIF-1α signalling pathway and accelerate wound healing.
Collapse
Affiliation(s)
- Wenting Wen
- College of Life and Environmental SciencesWenzhou UniversityZhejiangChina
| | - Liangliang Yang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research InstituteWenzhou Medical UniversityZhejiangChina
| | - Xin Wang
- Dpartment of Plastic and Reconstructive Surgery, Hand and MicrosurgeryNingbo NO.6 HospitalZhejiangChina
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research InstituteWenzhou Medical UniversityZhejiangChina
| | - Fangfang Wu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's HospitalWenzhou Medical UniversityWenzhouChina
| | - Ke Xu
- College of Life and Environmental SciencesWenzhou UniversityZhejiangChina
| | - Shaodong Chen
- Department of OrthopaedicsLishui People's HospitalZhejiangChina
| | - Zhiyong Liao
- College of Life and Environmental SciencesWenzhou UniversityZhejiangChina
| |
Collapse
|
31
|
Han H, Xing L, Chen BT, Liu Y, Zhou TJ, Wang Y, Zhang LF, Li L, Cho CS, Jiang HL. Progress on the pathological tissue microenvironment barrier-modulated nanomedicine. Adv Drug Deliv Rev 2023; 200:115051. [PMID: 37549848 DOI: 10.1016/j.addr.2023.115051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Imbalance in the tissue microenvironment is the main obstacle to drug delivery and distribution in the human body. Before penetrating the pathological tissue microenvironment to the target site, therapeutic agents are usually accompanied by three consumption steps: the first step is tissue physical barriers for prevention of their penetration, the second step is inactivation of them by biological molecules, and the third step is a cytoprotective mechanism for preventing them from functioning on specific subcellular organelles. However, recent studies in drug-hindering mainly focus on normal physiological rather than pathological microenvironment, and the repair of damaged physiological barriers is also rarely discussed. Actually, both the modulation of pathological barriers and the repair of damaged physiological barriers are essential in the disease treatment and the homeostasis maintenance. In this review, we present an overview describing the latest advances in the generality of these pathological barriers and barrier-modulated nanomedicine. Overall, this review holds considerable significance for guiding the design of nanomedicine to increase drug efficacy in the future.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
32
|
Tian P, Zhao L, Kim J, Li X, Liu C, Cui X, Liang T, Du Y, Chen X, Pan H. Dual stimulus responsive borosilicate glass (BSG) scaffolds promote diabetic alveolar bone defectsrepair by modulating macrophage phenotype. Bioact Mater 2023; 26:231-248. [PMID: 36936808 PMCID: PMC10020664 DOI: 10.1016/j.bioactmat.2023.02.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
The regeneration of alveolar bone is still clinical challenge, particularly accompanied with diabetes, causing metabolic disorder with a protracted low-grade inflammatory phenotype. As a result, the anticipated loading of biomaterials is highly suspicious in spontaneous modulation of cells function, which is mostly disturbed by constant inflammation. In this study, we developed glucose and hydrogen peroxide dual-responsive borosilicate glass (BSG) scaffolds loaded with epigallocatechin gallate (EGCG) to synergistically modulate the abnormal inflammation of diabetic alveolar bone defects. It was found that the release of EGCG by BSG could directly regulate the shift of macrophages from M1 to the M2 phenotype by promoting autophagy and lessening the inhibition of autophagic flux. Moreover, EGCG can also indirectly regulate the polarization phenotype of macrophages by reducing the activation of NF-κb in stem cells and restoring its immunoregulatory capacity. Therefore, the addition of EGCG to BSG scaffold in diabetes allows for a more striking modulation of the macrophage phenotype in a timely manner. The altered macrophage phenotype reduces local inflammation and thus increases the ability to repair diabetic alveolar bone, showing promise for the treatment of alveolar defect in diabetic patients.
Collapse
Affiliation(s)
- Pengfei Tian
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Limin Zhao
- Shenzhen Longhua District Central Hospital, Shenzhen, 518000, PR China
| | - Jua Kim
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Xian Li
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Chunyu Liu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Xu Cui
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Tao Liang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Yunbo Du
- Shenzhen Longhua District Central Hospital, Shenzhen, 518000, PR China
| | - Xiehui Chen
- Shenzhen Longhua District Central Hospital, Shenzhen, 518000, PR China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Shenzhen Healthemes Biotechnology Co. Ltd, Shenzhen, 518102, PR China
- Corresponding author. Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China.
| |
Collapse
|
33
|
Tian H, Yan J, Zhang W, Li H, Jiang S, Qian H, Chen X, Dai X, Wang X. Cu-GA-coordination polymer nanozymes with triple enzymatic activity for wound disinfection and accelerated wound healing. Acta Biomater 2023:S1742-7061(23)00313-6. [PMID: 37270076 DOI: 10.1016/j.actbio.2023.05.048] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/21/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023]
Abstract
During the past few years, bacterial infection and oxidative stress have become important issues for wound healing. However, the emergence of numerous drug-resistant superbugs has had a serious impact on the treatment of infected wounds. Presently, the development of new nanomaterials has become one of the most important approaches to the treatment of drug-resistant bacterial infections. Herein, coordination polymer copper-gallic acid (Cu-GA) nanorods with multi-enzyme activity is successfully prepared for efficient wound treatment of bacterial infection, which can effectively promote wound healing. Cu-GA can be efficiently prepared by a simple solution method and had good physiological stability. Interestingly, Cu-GA shows enhanced multienzyme activity (peroxidase, glutathione peroxidase, and superoxide dismutase), which can produce a large number of reactive oxygen species (ROS) under acidic conditions while scavenging ROS under neutral conditions. In acidic environment, Cu-GA possesses POD (peroxidase)-like and glutathione peroxidase (GSH-Px)-like catalytic activities that is capable of killing bacteria; but in neutral environment, Cu-GA exhibits superoxide dismutase (SOD)-like catalytic activity that can scavenge ROS and promote wound healing. In vivo studies show that Cu-GA can promote wound infection healing and have good biosafety. Cu-GA contributes to the healing of infected wounds by inhibiting bacterial growth, scavenging reactive oxygen species, and promoting angiogenesis. STATEMENT OF SIGNIFICANCE: Cu-GA-coordinated polymer nanozymes with multienzyme activity were successfully prepared for efficient wound treatment of bacterial infection, which could effectively promote wound healing. Interestingly, Cu-GA exhibited enhanced multienzyme activity (peroxidase, glutathione peroxidase, and superoxide dismutase), which could produce a large number of reactive oxygen species (ROS) under acidic conditions and scavenge ROS under neutral conditions. In vitro and in vivo studies demonstrated that Cu-GA was capable of killing bacteria, controlling inflammation, and promoting angiogenesis.
Collapse
Affiliation(s)
- Haotian Tian
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, P.R. China; School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, P. R. China
| | - Wei Zhang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| | - Huaixu Li
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, P.R. China
| | - Shouwei Jiang
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, P.R. China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China
| | - Xulin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Xingliang Dai
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, P.R. China.
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, P. R. China; College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, P. R. China.
| |
Collapse
|
34
|
Guo Z, Yan L, Zhou B, Zhao P, Wang W, Dong S, Cheng B, Yang J, Li B, Wang X. In situ photo-crosslinking silk fibroin based hydrogel accelerates diabetic wound healing through antibacterial and antioxidant. Int J Biol Macromol 2023:125028. [PMID: 37244328 DOI: 10.1016/j.ijbiomac.2023.125028] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/13/2023] [Accepted: 05/20/2023] [Indexed: 05/29/2023]
Abstract
Bacterial infection and excessive reactive oxygen species (ROS) in diabetic wounds lead to a prolonged inflammatory phase, and injuries are highly susceptible to developing into chronic wounds. Improving the poor microenvironment is vital to achieving effective diabetic wound healing. In this work, methacrylated silk fibroin (SFMA) was combined with ε-polylysine (EPL) and manganese dioxide nanoparticles (BMNPs) to form an SF@(EPL-BM) hydrogel with in situ forming, antibacterial and antioxidant properties. EPL imparted high antibacterial activity (>96 %) to the hydrogel. BMNPs and EPL showed good scavenging activity against a variety of free radicals. SF@(EPL-BM) hydrogel had low cytotoxicity and could alleviate H2O2-induced oxidative stress in L929 cells. In diabetic wounds infected with Staphylococcus aureus (S. aureus), the SF@(EPL-BM) hydrogel exhibited better antibacterial properties and reduced wound ROS levels more significantly than that of the control in vivo. In this process, the pro-inflammatory factor TNF-α was down-regulated, and the vascularization marker CD31 was up-regulated. H&E and Masson staining showed a rapid transition from the inflammatory to the proliferative phase of the wounds, with significant new tissue and collagen deposition. These results confirm that this multifunctional hydrogel dressing holds well potential for chronic wound healing.
Collapse
Affiliation(s)
- Zhendong Guo
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu hydrogen Valley, Foshan 528200, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China
| | - Lisi Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China
| | - Bo Zhou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China
| | - Peiwen Zhao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China
| | - Wenyuan Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China
| | - Siyan Dong
- Biotechnology Institute WUT-AMU School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, PR China; School of Biosciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Bo Cheng
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430060, PR China
| | - Jing Yang
- School of Foreign Languages, Wuhan University of Technology, Wuhan 430070, PR China
| | - Binbin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Hainan Institute, Wuhan University of Technology, Sanya 572000, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China; Shenzhen Research Institute of Wuhan University of Technology, Shenzhen 518000, PR China.
| | - Xinyu Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu hydrogen Valley, Foshan 528200, PR China; Hainan Institute, Wuhan University of Technology, Sanya 572000, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China; Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430060, PR China.
| |
Collapse
|
35
|
Yuan N, Shao K, Huang S, Chen C. Chitosan, alginate, hyaluronic acid and other novel multifunctional hydrogel dressings for wound healing: A review. Int J Biol Macromol 2023; 240:124321. [PMID: 37019198 DOI: 10.1016/j.ijbiomac.2023.124321] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
Wound healing is a complex project, and effectively promoting skin repair is a huge clinical challenge. Hydrogels have great prospect in the field of wound dressings because their physical properties are very similar to those of living tissue and have excellent properties such as high water content, oxygen permeability and softness. However, the single performance of traditional hydrogels limits their application as wound dressings. Therefore, natural polymers such as chitosan, alginate and hyaluronic acid, which are non-toxic and biocompatible, are individually or combined with other polymer materials, and loaded with typical drugs, bioactive molecules or nanomaterials. Then, the development of novel multifunctional hydrogel dressings with good antibacterial, self-healing, injectable and multi-stimulation responsiveness by using advanced technologies such as 3D printing, electrospinning and stem cell therapy has become a hot topic of current research. This paper focuses on the functional properties of novel multifunctional hydrogel dressings such as chitosan, alginate and hyaluronic acid, which lays the foundation for the research of novel hydrogel dressings with better performance.
Collapse
|
36
|
Solanki D, Vinchhi P, Patel MM. Design Considerations, Formulation Approaches, and Strategic Advances of Hydrogel Dressings for Chronic Wound Management. ACS OMEGA 2023; 8:8172-8189. [PMID: 36910992 PMCID: PMC9996804 DOI: 10.1021/acsomega.2c06806] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Wound healing is a complex and dynamic physiological process consisting of a series of cellular and molecular events that initiate immediately after a tissue lesion, to reconstruct the skin layer. It is indubitable that patients with chronic wounds, severely infected wounds, or any metabolic disorder of the wound microenvironment always endure severe pain and discomfort that affect their quality of life. It is essential to treat chronic wounds for conserving the physical as well as mental well-being of affected patients and for convalescing to improve their quality of life. For supporting and augmenting the healing process, the selection of pertinent wound dressing is essential. A substantial reduction in healing duration, disability, associated cost, and risk of recurrent infections can be achieved via engineering wound dressings. Hydrogels play a leading role in the path of engineering ideal wound dressings. Hydrogels, comprising water to a large extent, providing a moist environment, being comfortable to patients, and having biocompatible and biodegradable properties, have found their success as suitable wound dressings in the market. The exploitation of hydrogels is increasing perpetually after substantiation of their broader therapeutic actions owing to their resemblance to dermal tissues, their capability to stimulate partial skin regeneration, and their ability to incorporate therapeutic moieties promoting wound healing. This review entails properties of hydrogel supporting wound healing, types of hydrogels, cross-linking mechanisms, design considerations, and formulation strategies of hydrogel engineering. Various categories of hydrogel wound dressing fabricated recently are discussed based on their gel network composition, degradability, and physical and chemical cross-linking mechanisms, which provide an outlook regarding the importance of tailoring the physicochemical properties of hydrogels. The examples of marketed hydrogel wound dressings are also incorporated along with the future perspectives and challenges associated with them.
Collapse
|
37
|
Carboxyl Functionalization of N-MWCNTs with Stone-Wales Defects and Possibility of HIF-1α Wave-Diffusive Delivery. Int J Mol Sci 2023; 24:ijms24021296. [PMID: 36674808 PMCID: PMC9866222 DOI: 10.3390/ijms24021296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Nitrogen-doped multi-walled carbon nanotubes (N-MWCNTs) are widely used for drug delivery. One of the main challenges is to clarify their interaction with hypoxia-inducible factor 1 alpha (HIF-1α), the lack of which leads to oncological and cardiovascular diseases. In the presented study, N-MWCNTs were synthesized by catalytic chemical vapor deposition and irradiated with argon ions. Their chemical state, local structure, interfaces, Stone-Wales defects, and doping with nitrogen were analyzed by high resolution transmission electron microscopy (HRTEM), Raman spectroscopy, X-ray photoelectron spectroscopy (XPS), and near-edge X-ray absorption fine structure (NEXAFS) spectroscopy. Using experimental data, supercells of functionalized N-MWCNTs with an oxygen content of 2.7, 4 and 6 at. % in carboxyl groups were built by quantum chemical methods. Our analysis by the self-consistent charge density functional tight-binding (SCC DFTB) method shows that a key role in the functionalization of CNTs with carboxyl groups belongs to Stone-Wales defects. The results of research in the decoration of CNTs with HIF-1α demonstrate the possibility of wave-diffusion drug delivery. The nature of hybridization and relaxation determines the mechanism of oxygen regulation with HIF-1α molecules, namely, by OH-(OH-C) and OH-(O=C) chemical bonds. The concentration dependence of drug release in the diffusion mode suggests that the best pattern for drug delivery is provided by the tube with a carboxylic oxygen content of 6 at. %.
Collapse
|
38
|
Ge P, Chang S, Wang T, Zhao Q, Wang G, He B. An antioxidant and antibacterial polydopamine-modified thermo-sensitive hydrogel dressing for Staphylococcus aureus-infected wound healing. NANOSCALE 2023; 15:644-656. [PMID: 36515078 DOI: 10.1039/d2nr04908b] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Bacteria-infected wound healing is a complex and chronic process that poses a great threat to human health. A thermo-sensitive hydrogel that undergoes a sol-gel transition at body temperature is an attractive wound dressing for healing acceleration and infection prevention. In this paper, we present a thermo-sensitive and reactive oxygen species (ROS)-scavenging hydrogel based on polydopamine modified poly(ε-caprolactone-co-glycolide)-b-poly(ethylene glycol)-b-poly(ε-caprolactone-co-glycolide) (PDA/P2) triblock copolymer. The PDA/P2 solution at a concentration of 30 wt% could form a gel at 34-38 °C. The ROS-scavenging ability of PDA/P2 was demonstrated by DPPH and ABTS assays and intracellular ROS downregulation in RAW264.7 cells. Furthermore, silver nanoparticles were encapsulated in the hydrogel (PDA/P2-4@Ag gel) to provide antibacterial activity against E. coli and S. aureus. An in vivo S. aureus-infected rat model demonstrated that the PDA/P2-4@Ag hydrogel dressing could promote wound healing via inhibiting bacterial growth, alleviating the inflammatory response, and inducing angiogenesis and collagen deposition. This study provides a new strategy to prepare temperature-sensitive hydrogel-based multifunctional wound dressings.
Collapse
Affiliation(s)
- Pengjin Ge
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
| | - Shuhua Chang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
| | - Ting Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China
| | - Quan Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
| | - Gang Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
39
|
Yuan Z, Zhang L, Jiang S, Shafiq M, Cai Y, Chen Y, Song J, Yu X, Ijima H, Xu Y, Mo X. Anti-inflammatory, antibacterial, and antioxidative bioactive glass-based nanofibrous dressing enables scarless wound healing. SMART MATERIALS IN MEDICINE 2023; 4:407-426. [DOI: 10.1016/j.smaim.2023.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
40
|
Fu M, Zhao Y, Wang Y, Li Y, Wu M, Liu Q, Hou Z, Lu Z, Wu K, Guo J. On-Demand Removable Self-Healing and pH-Responsive Europium-Releasing Adhesive Dressing Enables Inflammatory Microenvironment Modulation and Angiogenesis for Diabetic Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205489. [PMID: 36319477 DOI: 10.1002/smll.202205489] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/08/2022] [Indexed: 06/16/2023]
Abstract
Current diabetic wound treatments remain unsatisfactory due to the lack of a comprehensive strategy that can integrate strong applicability (tissue adhesiveness, shape adaptability, fast self-healability, and facile dressing change) with the initiation and smooth connection of the cascade wound healing processes. Herein, benefiting from the multifaceted bonding ability of tannic acid to metal ions and various polymers, a family of tannin-europium coordination complex crosslinked citrate-based mussel-inspired bioadhesives (TE-CMBAs) are specially developed for diabetic wound healing. TE-CMBAs can gel instantly (< 60 s), possess favorable shape-adaptability, considerable mechanical strengths, high elasticity, considerable wet tissue adhesiveness (≈40 kPa), favorable photothermal antimicrobial activity, excellent anti-oxidant activity, biocompatibility, and angiogenetic property. The reversible hydrogen bond crosslinking and sensitive metal-phenolic coordination also confers TE-CMBAs with self-healability, pH-responsive europium ion and TA releasing properties and on-demand removability upon mixing with borax solution, enabling convenient painless dressing change and the smooth connection of inflammatory microenvironment modulation, angiogenesis promotion, and effective extracellular matrix production leveraging the acidic pH condition of diabetic wounds. This adhesive dressing provides a comprehensive regenerative strategy for diabetic wound management and can be extended to other complicated tissue healing scenarios.
Collapse
Affiliation(s)
- Meimei Fu
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yitao Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yue Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yue Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, P. R. China
| | - Min Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Qi Liu
- Regenerative Medicine and Tissue Repair Material Research Center, Huangpu Institute of Materials, Guangzhou, 510530, P. R. China
| | - Zhiguo Hou
- Regenerative Medicine and Tissue Repair Material Research Center, Huangpu Institute of Materials, Guangzhou, 510530, P. R. China
| | - Zhihui Lu
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
- Regenerative Medicine and Tissue Repair Material Research Center, Huangpu Institute of Materials, Guangzhou, 510530, P. R. China
| | - Keke Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jinshan Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
41
|
Zhang H, Cui Y, Zhuo X, Kim J, Li H, Li S, Yang H, Su K, Liu C, Tian P, Li X, Li L, Wang D, Zhao L, Wang J, Cui X, Li B, Pan H. Biological Fixation of Bioactive Bone Cement in Vertebroplasty: The First Clinical Investigation of Borosilicate Glass (BSG) Reinforced PMMA Bone Cement. ACS APPLIED MATERIALS & INTERFACES 2022; 14:51711-51727. [PMID: 36354323 DOI: 10.1021/acsami.2c15250] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
PMMA bone cement has been clinically used for decades in vertebroplasty due to its high mechanical strength and satisfactory injectability. However, the interface between bone and PMMA is fragile and more prone to refracture in situ because PMMA lacks a proper biological response from the host bone with minimal bone integration and dense fibrous tissue formation. Here, we modified PMMA by incoporating borosilicate glass (BSG) with a dual glass network of [BO3] and [SiO4], which spontaneously modulates immunity and osteogenesis. In particular, the BSG modified PMMA bone cement (abbreviated as BSG/PMMA cement) provided an alkaline microenvironment that spontaneously balanced the activities between osteoclasts and osteoblasts. Furthermore, the trace elements released from the BSGs enhanced the osteogenesis to strengthen the interface between the host bone and the implant. This study shows the first clinical case after implantation of BSG/PMMA for three months using the dual-energy CT, which found apatite nucleation around PMMA instead of fibrous tissues, indicating the biological interface was formed. Therefore, BSG/PMMA is promising as a biomaterial in vertebroplasty, overcoming the drawback of PMMA by improving the biological response from the host bone.
Collapse
Affiliation(s)
- Hao Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yinglin Cui
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xianglong Zhuo
- Department of Orthopaedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, China
| | - Jua Kim
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Honglong Li
- Shenzhen Healthemes Biotechnology Co., Ltd, Shenzhen 518120, China
| | - Shuaijie Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Hongsheng Yang
- Shenzhen Healthemes Biotechnology Co., Ltd, Shenzhen 518120, China
| | - Kun Su
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Chunyu Liu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Pengfei Tian
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xian Li
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Li Li
- Department of Orthopaedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, China
| | - Deping Wang
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Limin Zhao
- Shenzhen Longhua District Central Hospital/The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen 518110, China
| | - Jianyun Wang
- Shenzhen Healthemes Biotechnology Co., Ltd, Shenzhen 518120, China
| | - Xu Cui
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Bing Li
- Department of Orthopaedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
42
|
Skin-adaptive film dressing with smart-release of growth factors accelerated diabetic wound healing. Int J Biol Macromol 2022; 222:2729-2743. [DOI: 10.1016/j.ijbiomac.2022.10.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022]
|
43
|
Ma H, Peng Y, Zhang S, Zhang Y, Min P. Effects and Progress of Photo-Crosslinking Hydrogels in Wound Healing Improvement. Gels 2022; 8:609. [PMID: 36286110 PMCID: PMC9601727 DOI: 10.3390/gels8100609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 09/18/2023] Open
Abstract
Wound healing is a dynamic physiological process, including three stages: inflammation, tissue formation, and remodeling. The quality of wound healing is affected by many topical and systemic factors, while any small factor may affect the process. Therefore, improving the quality of wound healing is a complex and arduous challenge. Photo-crosslinking reaction using visible light irradiation is a novel method for hydrogel preparation. Photo-crosslinking hydrogels can be controlled in time and space, and are not interfered by temperature conditions, which have been widely used in the fields of medicine and engineering. This review aims to summarize the application of photo-crosslinking hydrogels in improving the quality of wound healing, mainly including the material design, application mechanism, and effect of photo-crosslinking hydrogels applied in wound healing, followed by the applicable animal models for experimental research. Finally, this review analyzes the clinical application prospects of photo-crosslinking hydrogels in the field of wound healing.
Collapse
Affiliation(s)
| | | | | | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Peiru Min
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| |
Collapse
|
44
|
Chiral Supramolecular Hydrogel Loaded with Dimethyloxalyglycine to Accelerate Chronic Diabetic Wound Healing by Promoting Cell Proliferation and Angiogenesis. Gels 2022; 8:gels8070437. [PMID: 35877522 PMCID: PMC9321917 DOI: 10.3390/gels8070437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic refractory wounds are one of the most serious complications of diabetes, and the effects of common treatments are limited. Chiral hydrogel combined with dimethyloxalyglycine (DMOG) as a dressing is a promising strategy for the treatment of chronic wounds. In this research, we have developed a DMOG-loaded supramolecular chiral amino-acid-derivative hydrogel for wound dressings for full-thickness skin regeneration of chronic wounds. The properties of the materials, the ability of sustained release drugs, and the ability to promote angiogenesis were tested in vitro, and the regeneration rate and repair ability of full-thickness skin were tested in vivo. The chiral hydrogel had the ability to release drugs slowly. It can effectively promote cell migration and angiogenesis in vitro, and promote full-thickness skin regeneration and angiogenesis in vivo. This work offers a new approach for repairing chronic wounds completely through a supramolecular chiral hydrogel loaded with DMOG.
Collapse
|
45
|
Relevance of NLRP3 Inflammasome-Related Pathways in the Pathology of Diabetic Wound Healing and Possible Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9687925. [PMID: 35814271 PMCID: PMC9262551 DOI: 10.1155/2022/9687925] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
Wound healing is a major secondary complication in type 2 diabetes, which results in significant disability and mortality, imposing a significant clinical and social burden. Sustained activation of the Nod-like receptor protein (NLRP) inflammasome in wounds is responsible for excessive inflammatory responses and aggravates wound damage. The activation of the NLRP3 inflammasome is regulated by a two-step process: the priming/licensing (signal 1) step involved in transcription and posttranslation and the protein complex assembly (signal 2) step triggered by danger molecules. This review focuses on the advances made in understanding the pathophysiological mechanisms underlying wound healing in the diabetic microenvironment. Simultaneously, this review summarizes the molecular mechanisms of the main regulatory pathways associated with signal 1 and signal 2, which trigger the NLRP3 inflammasome complex assembly in the development of diabetic wounds (DW). Activation of the NLRP3 inflammasome-related pathway, involving the disturbance in Nrf2 and the NF-κB/NLRP3 inflammasome, TLR receptor-mediated activation of the NF-κB/NLRP3 inflammasome, and various stimuli inducing NLRP3 inflammasome assembly play a pivotal role in DW healing. Furthermore, therapeutics targeting the NLRP3 inflammasome-related pathways may promote angiogenesis, reprogram immune cells, and improve DW healing.
Collapse
|
46
|
Long S, Xie C, Lu X. Natural polymer‐based adhesive hydrogel for biomedical applications. BIOSURFACE AND BIOTRIBOLOGY 2022. [DOI: 10.1049/bsb2.12036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Siyu Long
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| | - Chaoming Xie
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| | - Xiong Lu
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| |
Collapse
|
47
|
Multifunctional polysaccharide hydrogels for skin wound healing prepared by photoinitiator-free crosslinking. Carbohydr Polym 2022; 285:119254. [DOI: 10.1016/j.carbpol.2022.119254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/16/2022] [Accepted: 02/11/2022] [Indexed: 12/14/2022]
|
48
|
Li R, Liu K, Huang X, Li D, Ding J, Liu B, Chen X. Bioactive Materials Promote Wound Healing through Modulation of Cell Behaviors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105152. [PMID: 35138042 PMCID: PMC8981489 DOI: 10.1002/advs.202105152] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/24/2021] [Indexed: 05/13/2023]
Abstract
Skin wound repair is a multistage process involving multiple cellular and molecular interactions, which modulate the cell behaviors and dynamic remodeling of extracellular matrices to maximize regeneration and repair. Consequently, abnormalities in cell functions or pathways inevitably give rise to side effects, such as dysregulated inflammation, hyperplasia of nonmigratory epithelial cells, and lack of response to growth factors, which impedes angiogenesis and fibrosis. These issues may cause delayed wound healing or even non-healing states. Current clinical therapeutic approaches are predominantly dedicated to preventing infections and alleviating topical symptoms rather than addressing the modulation of wound microenvironments to achieve targeted outcomes. Bioactive materials, relying on their chemical, physical, and biological properties or as carriers of bioactive substances, can affect wound microenvironments and promote wound healing at the molecular level. By addressing the mechanisms of wound healing from the perspective of cell behaviors, this review discusses how bioactive materials modulate the microenvironments and cell behaviors within the wounds during the stages of hemostasis, anti-inflammation, tissue regeneration and deposition, and matrix remodeling. A deeper understanding of cell behaviors during wound healing is bound to promote the development of more targeted and efficient bioactive materials for clinical applications.
Collapse
Affiliation(s)
- Ruotao Li
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Kai Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Xu Huang
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Di Li
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Bin Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
49
|
Qin J, Chen F, Wu P, Sun G. Recent Advances in Bioengineered Scaffolds for Cutaneous Wound Healing. Front Bioeng Biotechnol 2022; 10:841583. [PMID: 35299645 PMCID: PMC8921732 DOI: 10.3389/fbioe.2022.841583] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/04/2022] [Indexed: 12/14/2022] Open
Abstract
Wound healing is an evolved dynamic biological process. Though many research and clinical approaches have been explored to restore damaged or diseased skin, the current treatment for deep cutaneous injuries is far from being perfect, and the ideal regenerative therapy remains a significant challenge. Of all treatments, bioengineered scaffolds play a key role and represent great progress in wound repair and skin regeneration. In this review, we focus on the latest advancement in biomaterial scaffolds for wound healing. We discuss the emerging philosophy of designing biomaterial scaffolds, followed by precursor development. We pay particular attention to the therapeutic interventions of bioengineered scaffolds for cutaneous wound healing, and their dual effects while conjugating with bioactive molecules, stem cells, and even immunomodulation. As we review the advancement and the challenges of the current strategies, we also discuss the prospects of scaffold development for wound healing.
Collapse
Affiliation(s)
- Jianghui Qin
- College of Chemistry and Environmental Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Fang Chen
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Pingli Wu
- College of Chemistry and Environmental Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Guoming Sun
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Institute of Life Science and Green Development, Hebei University, Baoding, China
| |
Collapse
|
50
|
Tang X, Gu X, Huang T, Chen X, Zhou Z, Yang Y, Ling J. Anisotropic Silk-Inspired Nerve Conduit with Peptides Improved the Microenvironment for Long-Distance Peripheral Nerve Regeneration. ACS Macro Lett 2021; 10:1501-1509. [PMID: 35549152 DOI: 10.1021/acsmacrolett.1c00533] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A lack of effective bioactivity to create a desirable microenvironment for peripheral nerve regeneration has been challenging in successful treatment of long-distance injuries using nerve guidance conduits (NGCs) clinically. Herein, we developed a silk-inspired phototriggered gelation system combining dual therapeutic cues of anisotropic topography and adhesive ligands for improving peripheral nerve regeneration. Importantly, enhanced cell recruitment and myelination of Schwann cells were successfully achieved by the Arg-Gly-Asp (RGD)-peptide-immobilized hydrogel scaffolds to promote axon growth. Moreover, as the orientated growth of Schwann cells and rapid axon growth were facilitated by aligned grooved micropatterns, this multifunctional bioactive system provides remarkable nerve regeneration with function recovery for long-distance nerve injury. Therefore, this bioengineered silk-inspired nerve guidance conduit delivers a platform for desirable peripheral nerve repair.
Collapse
Affiliation(s)
- Xiaoxuan Tang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226001, PR China
- Medical School of Nantong University, Nantong University, Nantong 226001, PR China
| | - Xinyi Gu
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226001, PR China
| | - Tingting Huang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226001, PR China
| | - Xiaoli Chen
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226001, PR China
| | - Zhihao Zhou
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226001, PR China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226001, PR China
| | - Jue Ling
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong 226001, PR China
| |
Collapse
|