1
|
Ding Y, Jia Q, Su Z, Chen H, Ye J, Xie D, Wu Y, He H, Peng Y, Ni Y. Homologous cell membrane-based hydrogel creates spatiotemporal niches to improve outcomes of dysregulated chronic wound healing. Mater Today Bio 2024; 28:101243. [PMID: 39315394 PMCID: PMC11419813 DOI: 10.1016/j.mtbio.2024.101243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/25/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
The (M2M + TGF-β)@HAMA hydrogel dressing improves the outcomes of dysregulated chronic wound healing by protecting the open wound from repeated bacterial infections, reprogramming endogenous monocytes and M1 macrophages into an M2-phenotype, as well as enhancing fibroblastic proliferation and migration for matrix remodeling and granulation tissue formation.Image 1.
Collapse
Affiliation(s)
| | | | - Ziwen Su
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Heying Chen
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jialing Ye
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Dafeng Xie
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yubo Wu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Haiyan He
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yanlin Peng
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yilu Ni
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| |
Collapse
|
2
|
Anggelia MR, Cheng HY, Lin CH. Thermosensitive Hydrogels as Targeted and Controlled Drug Delivery Systems: Potential Applications in Transplantation. Macromol Biosci 2024; 24:e2400064. [PMID: 38991045 DOI: 10.1002/mabi.202400064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/27/2024] [Indexed: 07/13/2024]
Abstract
Drug delivery in transplantation plays a vital role in promoting graft survival, preventing rejection, managing complications, and contributing to positive patient outcomes. Targeted and controlled drug delivery can minimize systemic effects. Thermosensitive hydrogels, due to their unique sol-gel transition properties triggered by thermo-stimuli, have attracted significant research interest as a potential drug delivery system in transplantation. This review describes the current status, characteristics, and recent applications of thermosensitive hydrogels for drug delivery. Studies aimed at improving allotransplantation outcomes using thermosensitive hydrogels are then elaborated on. Finally, the challenges and opportunities associated with their use are discussed. Understanding the progress of research will serve as a guide for future improvements in their application as a means of targeted and controlled drug delivery in translational therapeutic applications for transplantation.
Collapse
Affiliation(s)
- Madonna Rica Anggelia
- Center for Vascularized Composite Allotransplantation, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung Medical College and Chang Gung University, Taoyuan, 333, Taiwan
| | - Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung Medical College and Chang Gung University, Taoyuan, 333, Taiwan
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung Medical College and Chang Gung University, Taoyuan, 333, Taiwan
| |
Collapse
|
3
|
Cong Z, Li Y, Xie L, Chen Q, Tang M, Thongpon P, Jiao Y, Wu S. Engineered Microrobots for Targeted Delivery of Bacterial Outer Membrane Vesicles (OMV) in Thrombus Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400847. [PMID: 38801399 DOI: 10.1002/smll.202400847] [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: 02/01/2024] [Revised: 05/06/2024] [Indexed: 05/29/2024]
Abstract
In the realm of thrombosis treatment, bioengineered outer membrane vesicles (OMVs) offer a novel and promising approach, as they have rich content of bacterial-derived components. This study centers on OMVs derived from Escherichia coli BL21 cells, innovatively engineered to encapsulate the staphylokinase-hirudin fusion protein (SFH). SFH synergizes the properties of staphylokinase (SAK) and hirudin (HV) to enhance thrombolytic efficiency while reducing the risks associated with re-embolization and bleeding. Building on this foundation, this study introduces two cutting-edge microrobotic platforms: SFH-OMV@H for venous thromboembolism (VTE) treatment, and SFH-OMV@MΦ, designed specifically for cerebral venous sinus thrombosis (CVST) therapy. These platforms have demonstrated significant efficacy in dissolving thrombi, with SFH-OMV@H showcasing precise vascular navigation and SFH-OMV@MΦ effectively targeting cerebral thrombi. The study shows that the integration of these bioengineered OMVs and microrobotic systems marks a significant advancement in thrombosis treatment, underlining their potential to revolutionize personalized medical approaches to complex health conditions.
Collapse
Affiliation(s)
- Zhaoqing Cong
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
- South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Yangyang Li
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
| | - Leiming Xie
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
| | - Qiwei Chen
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
| | - Menghuan Tang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Phonpilas Thongpon
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Yanxiao Jiao
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Song Wu
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
- South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| |
Collapse
|
4
|
Kuan CH, Chang L, Ho CY, Tsai CH, Liu YC, Huang WY, Wang YN, Wang WH, Wang TW. Immunomodulatory hydrogel orchestrates pro-regenerative response of macrophages and angiogenesis for chronic wound healing. Biomaterials 2024; 314:122848. [PMID: 39342917 DOI: 10.1016/j.biomaterials.2024.122848] [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: 06/15/2024] [Revised: 08/22/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
Chronic wound healing often encounters challenges characterized by prolonged inflammation and impaired angiogenesis. While the immune response plays a pivotal role in orchestrating the intricate process of wound healing, excessive inflammation can hinder tissue repair. In this study, a bilayer alginate hydrogel system encapsulating polyelectrolyte complex nanoparticles (PCNs) loaded with anti-inflammatory cytokines and angiogenic growth factors is developed to address the challenges of chronic wound healing. The alginate hydrogel is designed using two distinct crosslinking methods to achieve differential degradation, thereby enabling precise spatial and temporal controlled release of PCNs. Initially, interleukin-10 (IL-10) is released to mitigate inflammation, while unsaturated PCNs bind and remove accumulated pro-inflammatory cytokines at the wound site. Subsequently, angiogenic growth factors, including vascular endothelial growth factor and platelet-derived growth factor, are released to promote vascularization and vessel maturation. Our results demonstrate that the bilayer hydrogel exhibits distinct degradation kinetics between the two layers, facilitating the staged release of multiple signaling molecules. In vitro experiments reveal that IL-10 can activate the Jak1/STAT3 pathway, thereby suppressing pro-inflammatory cytokines and chemokines while down-regulating inflammation-related genes. In vivo studies demonstrate that application of the hydrogel in chronic wounds using diabetic murine model promotes healing by positively modulating multiple integral reparative mechanisms. These include reducing inflammation, promoting macrophage polarization towards a pro-regenerative phenotype, enhancing keratinocyte migration, stimulating angiogenesis, and expediting wound closure. In conclusion, our hydrogel system effectively mitigates inflammatory responses and provides essential physiological cues by inducing a synergistic angiogenic effect, thus offering a promising approach for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Chen-Hsiang Kuan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taiwan.
| | - Ling Chang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Yu Ho
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan; Department of Bioengineering, Rice University, Houston, USA
| | - Chia-Hsuan Tsai
- Division of Plastic Surgery, Department of Surgery, Chang Gung Memorial Hospital, Keelung Branch, Taiwan
| | - Yu-Chung Liu
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan; Department of Biomedical Engineering, University of Michigan-Ann Arbor, Michigan, USA
| | - Wei-Yuan Huang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ning Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Wei-Hung Wang
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Tzu-Wei Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
5
|
Gao J, Zhu D, Fan Y, Liu H, Shen Z. Human Umbilical Cord Mesenchymal Stem Cells-Derived Extracellular Vesicles for Rat Jawbone Regeneration in Periapical Periodontitis. ACS Biomater Sci Eng 2024; 10:5784-5795. [PMID: 39164977 DOI: 10.1021/acsbiomaterials.4c00622] [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: 08/22/2024]
Abstract
Extracellular vesicles derived from mesenchymal stem cells (MSCs-EVs) have great potential for bone remodeling and anti-inflammatory therapy. For the repair and reconstruction of inflammatory jawbone defects caused by periapical periodontitis, bone meal filling after debridement is commonly used in the clinic. However, this treatment has disadvantages such as large individual differences and the need for surgical operation. Therefore, it is of great significance to search for other bioactive substances that can promote jawbone regeneration in periapical periodontitis. Herein, it is found that CT results showed that local injection of human umbilical cord mesenchymal stem cells-derived extracellular vesicles (HUC-MSCs-EVs) and bone meal filling into the alveolar bone defect area could promote bone tissue regeneration using a rat model of a jawbone defect in periapical periodontitis. Histologically, the new periodontal tissue in the bone defect area was thicker, and the number of blood vessels was higher by local injection of HUC-MSCs-EVs, and fewer inflammatory cells and osteoclasts were formed compared to bone meal filling. In vitro, HUC-MSCs-EVs can be internalized by rat bone marrow mesenchymal stem cells (BMSCs), enhancing the ability for proliferation and migration of BMSCs. Additionally, 20 μg/mL HUC-MSCs-EVs can facilitate the expression of osteogenic genes and proteins including runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), and osteopontin (OPN). In summary, in vivo and in vitro experiments showed that HUC-MSCs-EVs can promote bone regeneration in periapical periodontitis, and the effect of tissue regeneration is better than that of traditional bone meal treatment. Therefore, local injection of HUC-MSCs-EVs may be an effective method to promote jawbone regeneration in periapical periodontitis.
Collapse
Affiliation(s)
- Jiahui Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei 230001, China
| | - Dongao Zhu
- Department of Stomatology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yue Fan
- Department of Stomatology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Honghong Liu
- Department of Stomatology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zuojun Shen
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei 230001, China
| |
Collapse
|
6
|
Yang Y, Suo D, Xu T, Zhao S, Xu X, Bei HP, Wong KKY, Li Q, Zheng Z, Li B, Zhao X. Sprayable biomimetic double mask with rapid autophasing and hierarchical programming for scarless wound healing. SCIENCE ADVANCES 2024; 10:eado9479. [PMID: 39141725 PMCID: PMC11323895 DOI: 10.1126/sciadv.ado9479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
Current sprayable hydrogel masks lack the stepwise protection, cleansing, and nourishment of extensive wounds, leading to delayed healing with scarring. Here, we develop a sprayable biomimetic double wound mask (BDM) with rapid autophasing and hierarchical programming for scarless wound healing. The BDMs comprise hydrophobic poly (lactide-co-propylene glycol-co-lactide) dimethacrylate (PLD) as top layer and hydrophilic gelatin methacrylate (GelMA) hydrogel as bottom layer, enabling swift autophasing into bilayered structure. After photocrosslinking, BDMs rapidly solidify with strong interfacial bonding, robust tissue adhesion, and excellent joint adaptiveness. Upon implementation, the bottom GelMA layer could immediately release calcium ion for rapid hemostasis, while the top PLD layer could maintain a moist, breathable, and sterile environment. These traits synergistically suppress the inflammatory tumor necrosis factor-α pathway while coordinating the cyclic guanosine monophosphate/protein kinase G-Wnt/calcium ion signaling pathways to nourish angiogenesis. Collectively, our BDMs with self-regulated construction of bilayered structure could hierarchically program the healing progression with transformative potential for scarless wound healing.
Collapse
Affiliation(s)
- Yuhe Yang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Di Suo
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Tianpeng Xu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Shuai Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Xiaoxiao Xu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, Guangdong 518057, China
| | - Ho-Pan Bei
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, Guangdong 518057, China
| | - Kenneth Kak-yuen Wong
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Qibin Li
- Research Center for Intelligent Aesthetic Medicine, PolyU-Hangzhou Technology and Innovation Research Institute, Hangzhou, Zhejiang 310016, China
- Hangzhou Industrial Investment Group Co., Ltd., Hangzhou, Zhejiang, 310025, China
| | - Zijian Zheng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Research Institute for Intelligent Wearable Systems, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Bin Li
- Medical 3D Printing Center, Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, Guangdong 518057, China
- Research Center for Intelligent Aesthetic Medicine, PolyU-Hangzhou Technology and Innovation Research Institute, Hangzhou, Zhejiang 310016, China
- Research Institute for Intelligent Wearable Systems, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| |
Collapse
|
7
|
Ding H, Hao L, Mao H. Magneto-responsive biocomposites in wound healing: from characteristics to functions. J Mater Chem B 2024; 12:7463-7479. [PMID: 38990160 DOI: 10.1039/d4tb00743c] [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: 07/12/2024]
Abstract
The number of patients with non-healing wounds continuously increases, and has become a prominent societal issue that imposes a heavy burden on both patients and the entire healthcare system. Although traditional dressings play an important role in wound healing, the complexity and diversity of the healing process pose serious challenges in this field. Magneto-responsive biocomposites, with their excellent biocompatibility, remote spatiotemporal controllability, and unique convenience, demonstrate enticing advantages in the field of wound dressings. However, current research on magneto-responsive biocomposites as wound dressings lacks comprehensive and in-depth reviews, which to some extent, restricts the deeper understanding and further development of this field. Based on this, this paper reviews the latest advances in magnetic responsive wound dressings for wound healing. First, we review the process of skin wound healing and parameters for assessing repair progress. Then, we systematically discuss the preparation strategies and unique characteristics of magneto-responsive biocomposites, focusing on magneto-induced orientation, magneto-induced mechanical stimulation, and magnetocaloric effect. Subsequently, this review elaborates the multiple mechanisms of magneto-responsive biocomposites in promoting wound healing, including regulating cell behavior, enhancing electrical signal, controlling drug release, and accelerating tissue reconstruction. Finally, we further propose the development direction and future challenges of magnetic responsive biomaterials as wound dressings in clinical application.
Collapse
Affiliation(s)
- Haoyang Ding
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Lili Hao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
8
|
Li XL, Fan W, Fan B. Dental pulp regeneration strategies: A review of status quo and recent advances. Bioact Mater 2024; 38:258-275. [PMID: 38745589 PMCID: PMC11090883 DOI: 10.1016/j.bioactmat.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/18/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024] Open
Abstract
Microorganisms, physical factors such as temperature or mechanical injury, and chemical factors such as free monomers from composite resin are the main causes of dental pulp diseases. Current clinical treatment methods for pulp diseases include the root canal therapy, vital pulp therapy and regenerative endodontic therapy. Regenerative endodontic therapy serves the purpose of inducing the regeneration of new functional pulp tissues through autologous revascularization or pulp tissue engineering. This article first discusses the current clinical methods and reviews strategies as well as the research outcomes regarding the pulp regeneration. Then the in vivo models, the prospects and challenges for regenerative endodontic therapy were further discussed.
Collapse
Affiliation(s)
- Xin-Lu Li
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, 430079, Wuhan, China
| | - Wei Fan
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, 430079, Wuhan, China
| | - Bing Fan
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, 430079, Wuhan, China
| |
Collapse
|
9
|
Hao R, Ye X, Chen X, Du J, Tian F, Zhang L, Ma G, Rao F, Xue J. Integrating Bioactive Graded Hydrogel with Radially Aligned Nanofibers to Dynamically Manipulate Wound Healing Process. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37770-37782. [PMID: 38987992 DOI: 10.1021/acsami.4c09204] [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: 07/12/2024]
Abstract
Skin wound healing is a complex process that requires appropriate treatment and management. Using a single scaffold to dynamically manipulate angiogenesis, cell migration and proliferation, and tissue reconstruction during skin wound healing is a great challenge. We developed a hybrid scaffold platform that integrates the spatiotemporal delivery of bioactive cues with topographical cues to dynamically manipulate the wound-healing process. The scaffold comprised gelatin methacryloyl hydrogels and electrospun poly(ε-caprolactone)/gelatin nanofibers. The hydrogels had graded cross-linking densities and were loaded with two different functional bioactive peptides. The nanofibers comprised a radially aligned nanofiber array layer and a layer of random fibers. During the early stages of wound healing, the KLTWQELYQLKYKGI peptide, which mimics vascular endothelial growth factor, was released from the inner layer of the hydrogel to accelerate angiogenesis. During the later stages of wound healing, the IKVAVS peptide, which promotes cell migration, synergized with the radially aligned nanofiber membrane to promote cell migration, while the nanofiber membrane also supported further cell proliferation. In an in vivo rat skin wound-healing model, the hybrid scaffold significantly accelerated wound healing and collagen deposition, and the ratio of type I to type III collagen at the wound site resembled that of normal skin. The prepared scaffold dynamically regulated the skin tissue regeneration process in stages to achieve rapid wound repair with clinical application potential, providing a strategy for skin wound repair.
Collapse
Affiliation(s)
- Ruinan Hao
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xilin Ye
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Xiaofeng Chen
- Trauma Center, Peking University People's Hospital, Beijing 100044, P.R. China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, National Trauma Medical Center, Peking University, Beijing 100044, P.R. China
| | - Jinzhi Du
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Feng Tian
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Liqun Zhang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Feng Rao
- Trauma Center, Peking University People's Hospital, Beijing 100044, P.R. China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, National Trauma Medical Center, Peking University, Beijing 100044, P.R. China
| | - Jiajia Xue
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
10
|
Xia Y, Yan S, Wei H, Zhang H, Hou K, Chen G, Cao R, Zhu M. Multifunctional Porous Bilayer Artificial Skin for Enhanced Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:34578-34590. [PMID: 38946497 DOI: 10.1021/acsami.4c05074] [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: 07/02/2024]
Abstract
Meeting the exacting demands of wound healing encompasses rapid coagulation, superior exudate absorption, high antibacterial efficacy, and imperative support for cell growth. In this study, by emulating the intricate structure of natural skin, we prepare a multifunctional porous bilayer artificial skin to address these critical requirements. The bottom layer, mimicking the dermis, is crafted through freeze-drying a gel network comprising carboxymethyl chitosan (CMCs) and gelatin (GL), while the top layer, emulating the epidermis, is prepared via electrospinning poly(l-lactic acid) (PLLA) nanofibers. With protocatechuic aldehyde and gallium ion complexation (PA@Ga) as cross-linking agents, the bottom PA@Ga-CMCs/GL layer featured an adjustable pore size (78-138 μm), high hemostatic performance (67s), and excellent bacterial inhibition rate (99.9%), complemented by an impressive liquid-absorbing capacity (2000% swelling rate). The top PLLA layer, with dense micronanostructure and hydrophobic properties, worked as a shield to effectively thwarted liquid or bacterial penetration. Furthermore, accelerated wound closure, reduced inflammatory responses, and enhanced formation of hair follicles and blood vessels are achieved by the porous artificial skin covered on the surface of wound. Bilayer artificial skin integrates the advantages of nanofibers and freeze-drying porous materials to effectively replicate the protective properties of the epidermal layer of the skin, as well as the cell migration and tissue regeneration of the dermis. This bioabsorbable artificial skin demonstrates structural and functional comparability to real skin, which would advance the field of wound care through its multifaceted capabilities.
Collapse
Affiliation(s)
- Yuhan Xia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Sai Yan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Huidan Wei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Han Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Kai Hou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Guoyin Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Ran Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, P. R. China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
11
|
Teng X, Liu T, Zhao G, Liang Y, Li P, Li F, Li Q, Fu J, Zhong C, Zou X, Li L, Qi L. A novel exosome-based multifunctional nanocomposite platform driven by photothermal-controlled release system for repair of skin injury. J Control Release 2024; 371:258-272. [PMID: 38815704 DOI: 10.1016/j.jconrel.2024.05.049] [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: 03/09/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Currently, exosomes showed appropriate potential in the repair of skin injury. However, the functions of the exosomes could be compromised rapidly due to their short half-life and high clearance rate in vivo. In addition, the controlled release of effective concentrations of exosomes could increase the utilization efficiency of exosomes in wound healing. Accordingly, the design of an effective system for the controlled delivery of exosomes during the wound treatment period was necessary. In this contribution, we designed a novel exosome-based multifunctional nanocomposite platform with photothermal-controlled release performance for the repair of skin injury. Based on the agarose hydrogel, two-dimensional Ti3C2 (Ti3C2 MXene) and human umbilical cord mesenchymal stem cell (hucMSC)-derived exosomes, the as-prepared platform (i.e., hucMSC-derived exosome/Ti3C2 MXene hydrogel) was synthesized for the first time. Apart from possessing injectability, the hucMSC-derived exosome/Ti3C2 MXene hydrogel utilized the excellent photothermal effect of Ti3C2 MXene and proper phase transition performance of agarose hydrogel to provide a photothermal-controlled release system for the hucMSC-derived exosomes, which was beneficial for the personalized on-demand drug delivery. Importantly, the hucMSC-derived exosomes maintained their inherent structure and activity after being released from the Ti3C2 MXene hydrogel. Additionally, the as-prepared hydrogel with multifunctional performance also presented remarkable biocompatibility and photothermal-antibacterial property, and could efficiently accelerate wound healing by promoting cell proliferation, angiogenesis, collagen deposition, and reducing the level of inflammation at the wound site. The results suggested that the exosome-based multifunctional nanocomposite platform with great potential for wound healing would make significant advances in the revolution of traditional treatment methods in skin injury.
Collapse
Affiliation(s)
- Xu Teng
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China.
| | - Tao Liu
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China; DALI University, Dali 671000, China
| | - Guifang Zhao
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China; Department of Pathology, Jilin Medical University, Jilin 130013, China
| | - Yaru Liang
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Pengdong Li
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Fengjin Li
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Qiguang Li
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Jiacai Fu
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China; DALI University, Dali 671000, China
| | - Chengming Zhong
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Xiaohui Zou
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Linhai Li
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China.
| | - Ling Qi
- Institute of Digestive Disease, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China.
| |
Collapse
|
12
|
Li N, Zhang W, Wu S, Shafiq M, Xie P, Zhang L, Jiang S, Bi Y. Mesoporous Silicon with Strontium-Powered Poly(Lactic-Co-Glycolic acid)/Gelatin-Based Dressings Facilitate Skin Tissue Repair. Int J Nanomedicine 2024; 19:6449-6462. [PMID: 38946883 PMCID: PMC11214017 DOI: 10.2147/ijn.s460177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/22/2024] [Indexed: 07/02/2024] Open
Abstract
Purpose Functional inorganic nanomaterials (NMs) are widely exploited as bioactive materials and drug depots. The lack of a stable form of application of NMs at the site of skin injury, may impede the removal of the debridement, elevate pH, induce tissue toxicity, and limit their use in skin repair. This necessitates the advent of innovative wound dressings that overcome the above limitations. The overarching objective of this study was to exploit strontium-doped mesoporous silicon particles (PSiSr) to impart multifunctionality to poly(lactic-co-glycolic acid)/gelatin (PG)-based fibrous dressings (PG@PSiSr) for excisional wound management. Methods Mesoporous silicon particles (PSi) and PSiSr were synthesized using a chemo-synthetic approach. Both PSi and PSiSr were incorporated into PG fibers using electrospinning. A series of structure, morphology, pore size distribution, and cumulative pH studies on the PG@PSi and PG@PSiSr membranes were performed. Cytocompatibility, hemocompatibility, transwell migration, scratch wound healing, and delineated angiogenic properties of these composite dressings were tested in vitro. The biocompatibility of composite dressings in vivo was assessed by a subcutaneous implantation model of rats, while their potential for wound healing was discerned by implantation in a full-thickness excisional defect model of rats. Results The PG@PSiSr membranes can afford the sustained release of silicon ions (Si4+) and strontium ions (Sr2+) for up to 192 h as well as remarkably promote human umbilical vein endothelial cells (HUVECs) and NIH-3T3 fibroblasts migration. The PG@PSiSr membranes also showed better cytocompatibility, hemocompatibility, and significant formation of tubule-like networks of HUVECs in vitro. Moreover, PG@PSiSr membranes also facilitated the infiltration of host cells and promoted the deposition of collagen while reducing the accumulation of inflammatory cells in a subcutaneous implantation model in rats as assessed for up to day 14. Further evaluation of membranes transplanted in a full-thickness excisional wound model in rats showed rapid wound closure (PG@SiSr vs control, 96.1% vs 71.7%), re-epithelialization, and less inflammatory response alongside skin appendages formation (eg, blood vessels, glands, hair follicles, etc.). Conclusion To sum up, we successfully fabricated PSiSr particles and prepared PG@PSiSr dressings using electrospinning. The PSiSr-mediated release of therapeutic ions, such as Si4+ and Sr2+, may improve the functionality of PLGA/Gel dressings for an effective wound repair, which may also have implications for the other soft tissue repair disciplines.
Collapse
Affiliation(s)
- Naijing Li
- Department of Orthopedic Oncology, Yantai Shan Hospital, Yantai, People’s Republic of China
| | - Weiying Zhang
- Health Management Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Siyuan Wu
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Muhammad Shafiq
- Innovation Center of Nanomedicines, Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Peihan Xie
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Lixiang Zhang
- Department of Health Management, Characteristic Medical Center of Chinese People’s Armed Police Force, Tianjin, People’s Republic of China
| | - Shichao Jiang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Yue Bi
- Department of Orthopedic Oncology, Yantai Shan Hospital, Yantai, People’s Republic of China
| |
Collapse
|
13
|
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
|
14
|
Fan W, Yang X, Hu X, Huang R, Shi H, Liu G. A novel conductive microtubule hydrogel for electrical stimulation of chronic wounds based on biological electrical wires. J Nanobiotechnology 2024; 22:258. [PMID: 38755644 PMCID: PMC11097419 DOI: 10.1186/s12951-024-02524-2] [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: 01/09/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Electrical stimulation (ES) is considered a promising therapy for chronic wounds via conductive dressing. However, the lack of a clinically suitable conductive dressing is a serious challenge. In this study, a suitable conductive biomaterial with favorable biocompatibility and conductivity was screened by means of an inherent structure derived from the body based on electrical conduction in vivo. Ions condensed around the surface of the microtubules (MTs) derived from the cell's cytoskeleton are allowed to flow in the presence of potential differences, effectively forming a network of biological electrical wires, which is essential to the bioelectrical communication of cells. We hypothesized that MT dressing could improve chronic wound healing via the conductivity of MTs applied by ES. We first developed an MT-MAA hydrogel by a double cross-linking method using UV and calcium chloride to improve chronic wound healing by ES. In vitro studies showed good conductivity, mechanical properties, biocompatibility, and biodegradability of the MT-MAA hydrogel, as well as an elevated secretion of growth factors with enhanced cell proliferation and migration ability in response to ES. The in vivo experimental results from a full-thickness diabetic wound model revealed rapid wound closure within 7d in C57BL/6J mice, and the wound bed dressed by the MT-MAA hydrogel was shown to have promoted re-epithelization, enhanced angiogenesis, accelerated nerve growth, limited inflammation phases, and improved antibacterial effect under the ES treatment. These preclinical findings suggest that the MT-MAA hydrogel may be an ideal conductive dressing for chronic wound healing. Furthermore, biomaterials based on MTs may be also promising for treating other diseases.
Collapse
Affiliation(s)
- Weijing Fan
- Department of Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Zhangheng Street, Pu Dong New District, Shanghai, 201203, China
| | - Xiao Yang
- Department of Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Zhangheng Street, Pu Dong New District, Shanghai, 201203, China.
| | - Xiaoming Hu
- Department of Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Zhangheng Street, Pu Dong New District, Shanghai, 201203, China
| | - Renyan Huang
- Department of Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Zhangheng Street, Pu Dong New District, Shanghai, 201203, China
| | - Hongshuo Shi
- Department of Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Zhangheng Street, Pu Dong New District, Shanghai, 201203, China.
| | - Guobin Liu
- Department of Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Zhangheng Street, Pu Dong New District, Shanghai, 201203, China.
| |
Collapse
|
15
|
Sun H, Dong J, Fu Z, Lu X, Chen X, Lei H, Xiao X, Chen S, Lu J, Su D, Xiong Y, Fang Z, Mao J, Chen L, Wang X. TSG6-Exo@CS/GP Attenuates Endometrium Fibrosis by Inhibiting Macrophage Activation in a Murine IUA Model. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308921. [PMID: 38588501 DOI: 10.1002/adma.202308921] [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: 09/01/2023] [Revised: 04/04/2024] [Indexed: 04/10/2024]
Abstract
Intrauterine adhesion (IUA) is characterized by the formation of fibrous scar tissue within the uterine cavity, which significantly impacts female reproductive health and even leads to infertility. Unfortunately, severe cases of IUA currently lack effective treatments. This study presents a novel approach that utilizes tumor necrosis factor-(TNF) stimulated gene 6 (TSG6)-modified exosomes (Exos) in conjunction with an injectable thermosensitive hydrogel (CS/GP) to mitigate the occurrence of IUA by reducing endometrium fibrosis in a mouse IUA model. This study demonstrate that TSG6-modified Exos effectively inhibits the activation of inflammatory M1-like macrophages during the initial stages of inflammation and maintains the balance of macrophage phenotypes (M1/M2) during the repair phase. Moreover, TSG6 inhibits the interaction between macrophages and endometrial stromal fibroblasts, thereby preventing the activation of stromal fibroblasts into myofibroblasts. Furthermore, this research indicates that CS/GP facilitates the sustained release of TSG6-modified Exos, leading to a significant reduction in both the manifestations of IUA and the extent of endometrium fibrosis. Collectively, through the successful construction of CS/GP loaded with TSG6-modified Exos, a reduction in the occurrence and progression of IUA is achieved by mitigating endometrium fibrosis. Consequently, this approach holds promise for the treatment of IUA.
Collapse
Affiliation(s)
- Huijun Sun
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Jie Dong
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Zhaoyue Fu
- Department of Immunology, School of Basic Medicine, Air Force Medical University, No.169 West Changle Road, Xi'an, 710038, China
| | - Xueyan Lu
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Xutao Chen
- Department of Immunology, School of Basic Medicine, Air Force Medical University, No.169 West Changle Road, Xi'an, 710038, China
| | - Hui Lei
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Xifeng Xiao
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Shuqiang Chen
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Jie Lu
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Danjie Su
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Yujing Xiong
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
- Department of Immunology, School of Basic Medicine, Air Force Medical University, No.169 West Changle Road, Xi'an, 710038, China
| | - Zheng Fang
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Jiaqin Mao
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| | - Lihua Chen
- Department of Immunology, School of Basic Medicine, Air Force Medical University, No.169 West Changle Road, Xi'an, 710038, China
| | - Xiaohong Wang
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Clinical Research Center for Reproductive Medicine and Gynecological Endocrine Diseases of Shaanxi Province, Air Force Medical University, No.1 Xinsi Road, Xi'an, 710038, China
| |
Collapse
|
16
|
Zheng J, Yang B, Liu S, Xu Z, Ding Z, Mo M. Applications of Exosomal miRNAs from Mesenchymal Stem Cells as Skin Boosters. Biomolecules 2024; 14:459. [PMID: 38672475 PMCID: PMC11048182 DOI: 10.3390/biom14040459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The skin is the outer layer of the human body, and it is crucial in defending against injuries and damage. The regenerative capacity of aging and damaged skin caused by exposure to external stimuli is significantly impaired. Currently, the rise in average life expectancy and the modern population's aesthetic standards have sparked a desire for stem-cell-based therapies that can address skin health conditions. In recent years, mesenchymal stem cells (MSCs) as therapeutic agents have provided a promising and effective alternative for managing skin regeneration and rejuvenation, attributing to their healing capacities that can be applied to damaged and aged skin. However, it has been established that the therapeutic effects of MSC may be primarily mediated by paracrine mechanisms, particularly the release of exosomes (Exos). Exosomes are nanoscale extracellular vesicles (EVs) that have lipid bilayer and membrane structures and can be naturally released by different types of cells. They influence the physiological and pathological processes of recipient cells by transferring a variety of bioactive molecules, including lipids, proteins, and nucleic acids such as messenger RNAs (mRNAs) and microRNAs (miRNAs) between cells, thus playing an important role in intercellular communication and activating signaling pathways in target cells. Among them, miRNAs, a type of endogenous regulatory non-coding RNA, are often incorporated into exosomes as important signaling molecules regulating protein biosynthesis. Emerging evidence suggests that exosomal miRNAs from MSC play a key role in skin regeneration and rejuvenation by targeting multiple genes and regulating various biological processes, such as participating in inflammatory responses, cell migration, proliferation, and apoptosis. In this review, we summarize the recent studies and observations on how MSC-derived exosomal miRNAs contribute to the regeneration and rejuvenation of skin tissue, with particular attention to the applications of bioengineering methods for manipulating the miRNA content of exosome cargo to improve their therapeutic potential. This review can provide new clues for the diagnosis and treatment of skin damage and aging, as well as assist investigators in exploring innovative therapeutic strategies for treating a multitude of skin problems with the aim of delaying skin aging, promoting skin regeneration, and maintaining healthy skin.
Collapse
Affiliation(s)
- Jinmei Zheng
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Beibei Yang
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Siqi Liu
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Zhenfeng Xu
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Zhimeng Ding
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
| | - Miaohua Mo
- Department of Biotechnology, Guangdong Medical University, Dongguan 523808, China; (J.Z.); (B.Y.); (S.L.); (Z.X.); (Z.D.)
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
17
|
Liu X, Sun Y, Wang J, Kang Y, Wang Z, Cao W, Ye J, Gao C. A tough, antibacterial and antioxidant hydrogel dressing accelerates wound healing and suppresses hypertrophic scar formation in infected wounds. Bioact Mater 2024; 34:269-281. [PMID: 38261887 PMCID: PMC10794931 DOI: 10.1016/j.bioactmat.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Wound management is an important issue that places enormous pressure on the physical and mental health of patients, especially in cases of infection, where the increased inflammatory response could lead to severe hypertrophic scars (HSs). In this study, a hydrogel dressing was developed by combining the high strength and toughness, swelling resistance, antibacterial and antioxidant capabilities. The hydrogel matrix was composed of a double network of polyvinyl alcohol (PVA) and agarose with excellent mechanical properties. Hyperbranched polylysine (HBPL), a highly effective antibacterial cationic polymer, and tannic acid (TA), a strong antioxidant molecule, were added to the hydrogel as functional components. Examination of antibacterial and antioxidant properties of the hydrogel confirmed the full play of the efficacy of HBPL and TA. In the in vivo studies of methicillin-resistant Staphylococcus aureus (MRSA) infection, the hydrogel had shown obvious promotion of wound healing, and more profoundly, significant suppression of scar formation. Due to the common raw materials and simple preparation methods, this hydrogel can be mass produced and used for accelerating wound healing while preventing HSs in infected wounds.
Collapse
Affiliation(s)
- Xiaoqing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yiming Sun
- Eye Center, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, China
| | - Jie Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yongyuan Kang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Zhaolong Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Wangbei Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030000, China
| |
Collapse
|
18
|
He Y, Cen Y, Tian M. Immunomodulatory hydrogels for skin wound healing: cellular targets and design strategy. J Mater Chem B 2024; 12:2435-2458. [PMID: 38284157 DOI: 10.1039/d3tb02626d] [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: 01/30/2024]
Abstract
Skin wounds significantly impact the global health care system and represent a significant burden on the economy and society due to their complicated dynamic healing processes, wherein a series of immune events are required to coordinate normal and sequential healing phases, involving multiple immunoregulatory cells such as neutrophils, macrophages, keratinocytes, and fibroblasts, since dysfunction of these cells may impede skin wound healing presenting persisting inflammation, impaired vascularization, and excessive collagen deposition. Therefore, cellular target-based immunomodulation is promising to promote wound healing as cells are the smallest unit of life in immune response. Recently, immunomodulatory hydrogels have become an attractive avenue to promote skin wound healing. However, a detailed and comprehensive review of cellular targets and related hydrogel design strategies remains lacking. In this review, the roles of the main immunoregulatory cells participating in skin wound healing are first discussed, and then we highlight the cellular targets and state-of-the-art design strategies for immunomodulatory hydrogels based on immunoregulatory cells that cover defect, infected, diabetic, burn and tumor wounds and related scar healing. Finally, we discuss the barriers that need to be addressed and future prospects to boost the development and prosperity of immunomodulatory hydrogels.
Collapse
Affiliation(s)
- Yinhai He
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Cen
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Tian
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Wang P, Cai F, Li Y, Yang X, Feng R, Lu H, Bai X, Han J. Emerging trends in the application of hydrogel-based biomaterials for enhanced wound healing: A literature review. Int J Biol Macromol 2024; 261:129300. [PMID: 38216016 DOI: 10.1016/j.ijbiomac.2024.129300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/01/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Currently, there is a rising global incidence of diverse acute and chronic wounds, underscoring the immediate necessity for research and treatment advancements in wound repair. Hydrogels have emerged as promising materials for wound healing due to their unique physical and chemical properties. This review explores the classification and characteristics of hydrogel dressings, innovative preparation strategies, and advancements in delivering and releasing bioactive substances. Furthermore, it delves into the functional applications of hydrogels in wound healing, encompassing areas such as infection prevention, rapid hemostasis and adhesion adaptation, inflammation control and immune regulation, granulation tissue formation, re-epithelialization, and scar prevention and treatment. The mechanisms of action of various functional hydrogels are also discussed. Finally, this article also addresses the current limitations of hydrogels and provides insights into their potential future applications and upcoming innovative designs.
Collapse
Affiliation(s)
- Peng Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Feiyu Cai
- Department of Burns and Plastic Surgery & Wound Repair Surgery, the Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yu Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Rongqin Feng
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - He Lu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
20
|
Du S, Zhou X, Zheng B. Beyond Traditional Medicine: EVs-Loaded Hydrogels as a Game Changer in Disease Therapeutics. Gels 2024; 10:162. [PMID: 38534580 DOI: 10.3390/gels10030162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 03/28/2024] Open
Abstract
Extracellular vesicles (EVs), especially exosomes, have shown great therapeutic potential in the treatment of diseases, as they can target cells or tissues. However, the therapeutic effect of EVs is limited due to the susceptibility of EVs to immune system clearance during transport in vivo. Hydrogels have become an ideal delivery platform for EVs due to their good biocompatibility and porous structure. This article reviews the preparation and application of EVs-loaded hydrogels as a cell-free therapy strategy in the treatment of diseases. The article also discusses the challenges and future outlook of EVs-loaded hydrogels.
Collapse
Affiliation(s)
- Shutong Du
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Xiaohu Zhou
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Bo Zheng
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
21
|
Liu X, Liu Y, Zhou J, Yu X, Wan J, Wang J, Lei S, Zhang Z, Zhang L, Wang S. Porous Collagen Sponge Loaded with Large Efficacy-Potentiated Exosome-Mimicking Nanovesicles for Diabetic Skin Wound Healing. ACS Biomater Sci Eng 2024; 10:975-986. [PMID: 38236143 DOI: 10.1021/acsbiomaterials.3c01282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Diabetic skin wounds are difficult to heal quickly due to insufficient angiogenesis and prolonged inflammation, which is an urgent clinical problem. To address this clinical problem, it becomes imperative to develop a dressing that can promote revascularization and reduce inflammation during diabetic skin healing. Herein, a multifunctional collagen dressing (CTM) was constructed by loading large efficacy-potentiated exosome-mimicking nanovesicles (L-Meseomes) into a porous collagen sponge with transglutaminase (TGase). L-Meseomes were constructed in previous research with the function of promoting cell proliferation, migration, and angiogenesis and inhibiting inflammation. CTM has a three-dimensional porous network structure with good biocompatibility, swelling properties, and degradability and could release L-Meseome slowly. In vitro experiments showed that CTM could promote the proliferation of fibroblasts and the polarization of macrophages to the anti-inflammatory phenotype. For in vivo experiments, on the 21st day after surgery, the wound healing rates of the control and CTM were 83.026 ± 4.17% and 93.12 ± 2.16%, respectively; the epidermal maturation and dermal differentiation scores in CTM were approximately four times that of the control group, and the skin epidermal thickness of the CTM group was approximately 20 μm, which was closest to that of normal rats. CTM could significantly improve wound healing in diabetic rats by promoting anti-inflammation, angiogenesis, epidermal recovery, and dermal collagen deposition. In summary, the multifunctional collagen dressing CTM could significantly promote the healing of diabetic skin wounds, which provides a new strategy for diabetic wound healing in the clinic.
Collapse
Affiliation(s)
- Xiangsheng Liu
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yufei Liu
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jie Zhou
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xinyi Yu
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jinpeng Wan
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jie Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shaojin Lei
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | | | - Lin Zhang
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan Shandong 250022, China
| | - Shufang Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
- Nankai International Advanced Research Institute (SHENZHEN FUTIAN), Binglang Road 3#, Futian District, Shenzhen 518045, China
| |
Collapse
|
22
|
Zheng Y, Pan C, Xu P, Liu K. Hydrogel-mediated extracellular vesicles for enhanced wound healing: the latest progress, and their prospects for 3D bioprinting. J Nanobiotechnology 2024; 22:57. [PMID: 38341585 PMCID: PMC10858484 DOI: 10.1186/s12951-024-02315-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Extracellular vesicles have shown promising tissue recovery-promoting effects, making them increasingly sought-after for their therapeutic potential in wound treatment. However, traditional extracellular vesicle applications suffer from limitations such as rapid degradation and short maintenance during wound administration. To address these challenges, a growing body of research highlights the role of hydrogels as effective carriers for sustained extracellular vesicle release, thereby facilitating wound healing. The combination of extracellular vesicles with hydrogels and the development of 3D bioprinting create composite hydrogel systems boasting excellent mechanical properties and biological activity, presenting a novel approach to wound healing and skin dressing. This comprehensive review explores the remarkable mechanical properties of hydrogels, specifically suited for loading extracellular vesicles. We delve into the diverse sources of extracellular vesicles and hydrogels, analyzing their integration within composite hydrogel formulations for wound treatment. Different composite methods as well as 3D bioprinting, adapted to varying conditions and construction strategies, are examined for their roles in promoting wound healing. The results highlight the potential of extracellular vesicle-laden hydrogels as advanced therapeutic tools in the field of wound treatment, offering both mechanical support and bioactive functions. By providing an in-depth examination of the various roles that these composite hydrogels can play in wound healing, this review sheds light on the promising directions for further research and development. Finally, we address the challenges associated with the application of composite hydrogels, along with emerging trends of 3D bioprinting in this domain. The discussion covers issues such as scalability, regulatory considerations, and the translation of this technology into practical clinical settings. In conclusion, this review underlines the significant contributions of hydrogel-mediated extracellular vesicle therapy to the field of 3D bioprinting and wound healing and tissue regeneration. It serves as a valuable resource for researchers and practitioners alike, fostering a deeper understanding of the potential benefits, applications, and challenges involved in utilizing composite hydrogels for wound treatment.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Chuqiao Pan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China
| | - Peng Xu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China.
| | - Kai Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, China.
| |
Collapse
|
23
|
Holkar K, Kale V, Pethe P, Ingavle G. The symbiotic effect of osteoinductive extracellular vesicles and mineralized microenvironment on osteogenesis. J Biomed Mater Res A 2024; 112:155-166. [PMID: 37671776 DOI: 10.1002/jbm.a.37600] [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: 05/19/2023] [Revised: 07/29/2023] [Accepted: 08/16/2023] [Indexed: 09/07/2023]
Abstract
The increasing prevalence of bone-related diseases has raised concern about the need for an osteoinductive and mechanically stronger scaffold-based bone tissue engineering (BTE) alternative. A mineralized microenvironment, similar to the native bone microenvironment, is required in the scaffold to recruit and differentiate local mesenchymal stem cells at the bone defect site. Further, extracellular vesicles (EVs), pre-osteoblasts' secretome, contain osteoinductive cargo and have recently been exploited in bone regeneration. This work developed a cell-free and mechanically strong interpenetrating network-based scaffold for BTE by combining the action of osteoinductive EVs with a mineralized microenvironment. The MC3T3 (a pre-osteoblast cell line) is used as a source of EVs and as the target population. The optimal concentration of MC3T3-EVs was first determined to induce osteogenesis in target cells. The osteoinductive potential of the scaffold was estimated in vitro by osteogenesis-related markers like the alkaline phosphatase (ALP) enzyme and calcium content. The MC3T3-EVs cargo was also studied for osteoinductive signals such as ALP, calcium, and mRNA. The findings of this work indicated that MC3T3-EVs at a 90 μg/mL dose had significantly higher ALP activity than 0 μg/mL (1.47-fold), 10 μg/mL (1.41-fold), and 30 μg/mL (1.39-fold) EV-concentration on day 14. Further combination of the optimum dose of EVs with a mineralized microenvironment significantly enhanced ALP activity (1.5-fold) and mineralization (3.36-fold) as compared to the control group on day 7. EV cargo analysis revealed the presence of calcium, the ALP enzyme, and the mRNAs necessary for osteogenesis and angiogenesis. ALP activity was significantly boosted in the EV-containing target cells as early as day 1, and mineralization began on day 7 because MC3T3-EVs carry ALP enzymes and calcium as cargo. When osteoinductive EVs were combined with an osteoconductive mineralized microenvironment, osteogenesis was significantly enhanced in target cells at early time points. The interaction between osteoinductive EVs and the mineralized milieu facilitates the process of osteogenesis in the target cells and suggests a potential cell-free strategy for in vivo bone repair.
Collapse
Affiliation(s)
- Ketki Holkar
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University), Pune, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University), Pune, India
| | - Prasad Pethe
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University), Pune, India
| | - Ganesh Ingavle
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University), Pune, India
| |
Collapse
|
24
|
Ghosh S, Ghosh S, Sharma H, Bhaskar R, Han SS, Sinha JK. Harnessing the power of biological macromolecules in hydrogels for controlled drug release in the central nervous system: A review. Int J Biol Macromol 2024; 254:127708. [PMID: 37923043 DOI: 10.1016/j.ijbiomac.2023.127708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
Hydrogels have immense potential in revolutionizing central nervous system (CNS) drug delivery, improving outcomes for neurological disorders. They serve as promising tools for controlled drug delivery to the CNS. Available hydrogel types include natural macromolecules (e.g., chitosan, hyaluronic acid, alginate), as well as hybrid hydrogels combining natural and synthetic polymers. Each type offers distinct advantages in terms of biocompatibility, mechanical properties, and drug release kinetics. Design and engineering considerations encompass hydrogel composition, crosslinking density, porosity, and strategies for targeted drug delivery. The review emphasizes factors affecting drug release profiles, such as hydrogel properties and formulation parameters. CNS drug delivery applications of hydrogels span a wide range of therapeutics, including small molecules, proteins and peptides, and nucleic acids. However, challenges like limited biodegradability, clearance, and effective CNS delivery persist. Incorporating 3D bioprinting technology with hydrogel-based CNS drug delivery holds the promise of highly personalized and precisely controlled therapeutic interventions for neurological disorders. The review explores emerging technologies like 3D bioprinting and nanotechnology as opportunities for enhanced precision and effectiveness in hydrogel-based CNS drug delivery. Continued research, collaboration, and technological advancements are vital for translating hydrogel-based therapies into clinical practice, benefiting patients with CNS disorders. This comprehensive review article delves into hydrogels for CNS drug delivery, addressing their types, design principles, applications, challenges, and opportunities for clinical translation.
Collapse
Affiliation(s)
- Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India; ICMR - National Institute of Nutrition, Tarnaka, Hyderabad, Telangana 500007, India
| | - Soumya Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Hitaishi Sharma
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea.
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea.
| | | |
Collapse
|
25
|
Chen Z, Sun Z, Fan Y, Yin M, Jin C, Guo B, Yin Y, Quan R, Zhao S, Han S, Cheng X, Liu W, Chen B, Xiao Z, Dai J, Zhao Y. Mimicked Spinal Cord Fibers Trigger Axonal Regeneration and Remyelination after Injury. ACS NANO 2023; 17:25591-25613. [PMID: 38078771 DOI: 10.1021/acsnano.3c09892] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Spinal cord injury (SCI) causes tissue structure damage and composition changes of the neural parenchyma, resulting in severe consequences for spinal cord function. Mimicking the components and microstructure of spinal cord tissues holds promise for restoring the regenerative microenvironment after SCI. Here, we have utilized electrospinning technology to develop aligned decellularized spinal cord fibers (A-DSCF) without requiring synthetic polymers or organic solvents. A-DSCF preserves multiple types of spinal cord extracellular matrix proteins and forms a parallel-oriented structure. Compared to aligned collagen fibers (A-CF), A-DSCF exhibits stronger mechanical properties, improved enzymatic stability, and superior functionality in the adhesion, proliferation, axonal extension, and myelination of differentiated neural progenitor cells (NPCs). Notably, axon extension or myelination has been primarily linked to Agrin (AGRN), Laminin (LN), or Collagen type IV (COL IV) proteins in A-DSCF. When transplanted into rats with complete SCI, A-DSCF loaded with NPCs improves the survival, maturation, axon regeneration, and motor function of the SCI rats. These findings highlight the potential of structurally and compositionally biomimetic scaffolds to promote axonal extension and remyelination after SCI.
Collapse
Affiliation(s)
- Zhenni Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Jin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rui Quan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuaijing Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyu Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaokang Cheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
26
|
Kim MJ, Ko H, Kim JY, Kim HJ, Kim HY, Cho HE, Cho HD, Seo WS, Kang HC. Improvement in Yield of Extracellular Vesicles Derived from Edelweiss Callus Treated with LED Light and Enhancement of Skin Anti-Aging Indicators. Curr Issues Mol Biol 2023; 45:10159-10178. [PMID: 38132480 PMCID: PMC10742862 DOI: 10.3390/cimb45120634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/09/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
The process of skin aging is currently recognized as a disease, and extracellular vesicles (EVs) are being used to care for it. While various EVs are present in the market, there is a growing need for research on improving skin conditions through microbial and plant-derived EVs. Edelweiss is a medicinal plant and is currently an endangered species. Callus culture is a method used to protect rare medicinal plants, and recently, research on EVs using callus culture has been underway. In this study, the researchers used LED light to increase the productivity of Edelweiss EVs and confirmed that productivity was enhanced by LED exposure. Additionally, improvements in skin anti-aging indicators were observed. Notably, M-LED significantly elevated callus fresh and dry weight, with a DW/FW ratio of 4.11%, indicating enhanced proliferation. Furthermore, M-LED boosted secondary metabolite production, including a 20% increase in total flavonoids and phenolics. The study explores the influence of M-LED on EV production, revealing a 2.6-fold increase in concentration compared to darkness. This effect is consistent across different plant species (Centella asiatica, Panax ginseng), demonstrating the universality of the phenomenon. M-LED-treated EVs exhibit a concentration-dependent inhibition of reactive oxygen species (ROS) production, surpassing dark-cultured EVs. Extracellular melanin content analysis reveals M-LED-cultured EVs' efficacy in reducing melanin production. Additionally, the expression of key skin proteins (FLG, AQP3, COL1) is significantly higher in fibroblasts treated with M-LED-cultured EVs. These results are expected to provide valuable insights into research on improving the productivity of plant-derived EVs and enhancing skin treatment using plant-derived EVs.
Collapse
Affiliation(s)
- Mi-Jung Kim
- Human & Microbiome Communicating Laboratory, GFC Co., Ltd., Hwaseong 18471, Republic of Korea; (M.-J.K.); (J.-Y.K.); (H.-J.K.)
| | - Hoon Ko
- Creative Innovation Research Center, Cosmecca Korea Co., Ltd., Seongnam 13488, Republic of Korea; (H.K.); (H.-Y.K.); (H.-E.C.); (H.-D.C.)
| | - Ji-Young Kim
- Human & Microbiome Communicating Laboratory, GFC Co., Ltd., Hwaseong 18471, Republic of Korea; (M.-J.K.); (J.-Y.K.); (H.-J.K.)
| | - Hye-Jin Kim
- Human & Microbiome Communicating Laboratory, GFC Co., Ltd., Hwaseong 18471, Republic of Korea; (M.-J.K.); (J.-Y.K.); (H.-J.K.)
| | - Hwi-Yeob Kim
- Creative Innovation Research Center, Cosmecca Korea Co., Ltd., Seongnam 13488, Republic of Korea; (H.K.); (H.-Y.K.); (H.-E.C.); (H.-D.C.)
| | - Hang-Eui Cho
- Creative Innovation Research Center, Cosmecca Korea Co., Ltd., Seongnam 13488, Republic of Korea; (H.K.); (H.-Y.K.); (H.-E.C.); (H.-D.C.)
| | - Hyun-Dae Cho
- Creative Innovation Research Center, Cosmecca Korea Co., Ltd., Seongnam 13488, Republic of Korea; (H.K.); (H.-Y.K.); (H.-E.C.); (H.-D.C.)
| | - Won-Sang Seo
- Human & Microbiome Communicating Laboratory, GFC Co., Ltd., Hwaseong 18471, Republic of Korea; (M.-J.K.); (J.-Y.K.); (H.-J.K.)
| | - Hee-Cheol Kang
- Human & Microbiome Communicating Laboratory, GFC Co., Ltd., Hwaseong 18471, Republic of Korea; (M.-J.K.); (J.-Y.K.); (H.-J.K.)
| |
Collapse
|
27
|
Meng S, Hu H, Qiao Y, Wang F, Zhang BN, Sun D, Zhou L, Zhao L, Xie L, Zhang H, Zhou Q. A Versatile Hydrogel with Antibacterial and Sequential Drug-Releasing Capability for the Programmable Healing of Infectious Keratitis. ACS NANO 2023; 17:24055-24069. [PMID: 38044579 DOI: 10.1021/acsnano.3c09034] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Hydrogels have attracted tremendous attention as favorable corneal substitutes for treating severe infectious keratitis (IK). However, current hydrogel-based corneal substitutes were majorly designed to promote the single stage of corneal regeneration, which falls short in meeting the clinical management needs of severe IK including the multiple phases of corneal wound healing. Herein, we introduce a versatile hybrid hydrogel (SQPV) composed of silk fibroin and chitosan, which exhibits spatiotemporal properties for drug release. The SQPV is fabricated by incorporating verteporfin-loaded poly(lactic-co-glycolic)-polyethylene glycol-o-nitrobenzene micelles into a hydrogel network, which is formed from methacrylate silk fibroin and glycidyl methacrylate functionalized quaternized chitosan containing polydeoxyribonucleotide. This double network approach results in a material with exceptional anti-inflammatory, antibacterial, and proliferative stimulation and tissue remodeling regulation capabilities. Furthermore, SQPV showcases mechanical strength and transparency akin to those of native cornea. Extensive in vitro and in vivo studies validate SQPV's ability to effectively eliminate residual bacteria, mitigate inflammation, foster regeneration of corneal epithelium and stroma, prevent corneal scarring, and ultimately expedite wound healing. In summary, the SF/CS-based hybrid hydrogel may represent a promising substitute for comprehensive corneal repair and regeneration in severe IK.
Collapse
Affiliation(s)
- Shuqin Meng
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, P.R. China
| | - Yujie Qiao
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Fuyan Wang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Bi Ning Zhang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Difang Sun
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Longfang Zhou
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Long Zhao
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Lixin Xie
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Hengrui Zhang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao 266071, P.R. China
| |
Collapse
|
28
|
Yan Z, Zhang T, Wang Y, Xiao S, Gao J. Extracellular vesicle biopotentiated hydrogels for diabetic wound healing: The art of living nanomaterials combined with soft scaffolds. Mater Today Bio 2023; 23:100810. [PMID: 37810755 PMCID: PMC10550777 DOI: 10.1016/j.mtbio.2023.100810] [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: 07/31/2023] [Revised: 09/08/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023] Open
Abstract
Diabetic wounds (DWs) pose a major challenge for the public health system owing to their high incidence, complex pathogenesis, and long recovery time; thus, there is an urgent need to develop innovative therapies to accelerate the healing process of diabetic wounds. As natural nanovesicles, extracellular vesicles (EVs) are rich in sources with low immunogenicity and abundant nutritive molecules and exert potent therapeutic effects on diabetic wound healing. To avoid the rapid removal of EVs, a suitable delivery system is required for their controlled release. Owing to the advantages of high porosity, good biocompatibility, and adjustable physical and chemical properties of hydrogels, EV biopotentiated hydrogels can aid in achieving precise and favorable therapy against diabetic wounds. This review highlights the different design strategies, therapeutic effects, and mechanisms of EV biopotentiated hydrogels. We also discussed the future challenges and opportunities of using EV biopotentiated hydrogels for diabetic wound healing.
Collapse
Affiliation(s)
- Zhenzhen Yan
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Yuxiang Wang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| |
Collapse
|
29
|
Zhang D, Li Z, Yang L, Ma H, Chen H, Zeng X. Architecturally designed sequential-release hydrogels. Biomaterials 2023; 303:122388. [PMID: 37980822 DOI: 10.1016/j.biomaterials.2023.122388] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/23/2023] [Accepted: 11/04/2023] [Indexed: 11/21/2023]
Abstract
Drug synergy has made significant strides in clinical applications in recent decades. However, achieving a platform that enables "single administration, multi-stage release" by emulating the natural physiological processes of the human body poses a formidable challenge in the field of molecular pharmaceutics. Hydrogels, as the novel generation of drug delivery systems, have gained widespread utilization in drug platforms owing to their exceptional biocompatibility and modifiability. Sequential drug delivery hydrogels (SDDHs), which amalgamate the advantages of hydrogel and sequential release platforms, offer a promising solution for effectively navigating the intricate human environment and accomplishing drug sequential release. Inspired by architectural design, this review establishes connections between three pivotal factors in SDDHs construction, namely mechanisms, carrier spatial structure, and stimuli-responsiveness, and three aspects of architectural design, specifically building materials, house structures, and intelligent interactive furniture, aiming at providing insights into recent developments in SDDHs. Furthermore, the dual-drug collocation and cutting-edge hydrogel preparation technologies as well as the prevailing challenges in the field were elucidated.
Collapse
Affiliation(s)
- Dan Zhang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zimu Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Li Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Hualin Ma
- Department of Nephrology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, China.
| | - Hongzhong Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Xiaowei Zeng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
30
|
Wang S, Wu S, Yang Y, Zhang J, Wang Y, Zhang R, Yang L. Versatile Hydrogel Dressings That Dynamically Regulate the Healing of Infected Deep Burn Wounds. Adv Healthc Mater 2023; 12:e2301224. [PMID: 37657086 DOI: 10.1002/adhm.202301224] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/15/2023] [Indexed: 09/03/2023]
Abstract
Severe burns threaten patient lives due to pain, inflammation, bacterial infection, and scarring. Most burn dressings that are commonly used perform a single function and are not well suited for the management of deep burns. Therefore, a multifunctional antimicrobial peptide- and stem cell-loaded macroporous hydrogel that can fight bacterial infection and regulate wound healing progression by temporally regulating cytokine production by internal stem cells is developed. The macroporous skeletal hydrogel is manufactured via the cryogenic gelation of hyaluronic acid (cryogel). Based on the oxidative polymerization reaction of dopamine, the antimicrobial peptide DP7 is immobilized on the surface of the cryogel (DA7CG). Placental mesenchymal stem cells (PMSCs) are then packaged inside the macroporous hydrogel (DA7CG@C). According to the results of in vitro and in vivo experiments, during the inflammatory phase, DP7 inhibits infection and modulates inflammation; during the proliferative phase, DA7CG@C accelerates the regeneration of skin, blood vessels, and hair follicles via internal stem cells; and during the remodeling phase, DA7CG@C contributes to extracellular matrix remodeling due to the ability of DP7 to regulate the paracrine secretion of PMSCs, synergistically promoting scar-free healing. DA7CG@C can participate in all phases of wound healing; therefore, it is a promising dressing for burn treatment.
Collapse
Affiliation(s)
- Shihan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Siwen Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuling Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiani Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
31
|
Gao Y, Jiang Z, Xu B, Mo R, Li S, Jiang Y, Zhao D, Cao W, Chen B, Tian M, Tan Q. Evaluation of topical methylene blue nanoemulsion for wound healing in diabetic mice. PHARMACEUTICAL BIOLOGY 2023; 61:1462-1473. [PMID: 37691404 PMCID: PMC10496548 DOI: 10.1080/13880209.2023.2254341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
CONTEXT Diabetic wounds (DW) are a complication of diabetes and slow wound healing is the main manifestation. Methylene blue (MB) has been shown to exhibit therapeutic effects on diabetes-related diseases. OBJECTIVE To investigate the mechanisms of action of MB-nanoemulsion (NE) in the treatment of DW. MATERIALS AND METHODS The concentration of MB-NE used in the in vivo and in vitro experiments was 0.1 mg/mL. Streptozocin-induced diabetic mice were used as models. The mice were separated into nondiabetic, diabetic, MB-NE treated, and NE-treated groups. Intervention of high glucose-induced human umbilical vein endothelial cells using MB-NE. The mechanism by which MB-NE promotes DW healing is investigated by combining histological analysis, immunofluorescence analysis, TUNEL and ROS assays and western blotting. RESULTS In diabetic mice, the MB-NE accelerated DW healing (p < 0.05), promoted the expression of endothelial cell markers (α-SMA, CD31 and VEGF) (p < 0.05), and reduced TUNEL levels. In vitro, MB accelerated the migration rate of cells (p < 0.05); promoted the expression of CD31, VEGF, anti-apoptotic protein Bcl2 (p < 0.05) and decreased the expression of the pro-apoptotic proteins cleaved caspase-3 and Bax (p < 0.05). MB upregulated the expression of Nrf2, catalase, HO-1 and SOD2 (p < 0.05). In addition, MB reduced the immunofluorescence intensity of TUNEL and ROS in cells and reduced apoptosis. The therapeutic effect of MB was attenuated after treatment with an Nrf2 inhibitor (ML385). DISCUSSION AND CONCLUSION This study provides a foundation for the application of MB-NE in the treatment of DW.
Collapse
Affiliation(s)
- Yu Gao
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Zhounan Jiang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Xu
- Hubei Xiangyang Central Hospital, Xiangyang, China
| | - Ran Mo
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shiyan Li
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yanan Jiang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Demei Zhao
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wangbin Cao
- Nanjing Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Bin Chen
- Institute of Plant Resources and Chemistry, Nanjing Research Institute for Comprehensive Utilization of Wild Plants, Nanjing, China
| | - Meng Tian
- Department of Plastic Surgery, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qian Tan
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
32
|
Cui C, Mei L, Wang D, Jia P, Zhou Q, Liu W. A self-stabilized and water-responsive deliverable coenzyme-based polymer binary elastomer adhesive patch for treating oral ulcer. Nat Commun 2023; 14:7707. [PMID: 38001112 PMCID: PMC10673908 DOI: 10.1038/s41467-023-43571-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Oral ulcer can be treated with diverse biomaterials loading drugs or cytokines. However, most patients do not benefit from these materials because of poor adhesion, short-time retention in oral cavity and low drug therapeutic efficacy. Here we report a self-stabilized and water-responsive deliverable coenzyme salt polymer poly(sodium α-lipoate) (PolyLA-Na)/coenzyme polymer poly(α-lipoic acid) (PolyLA) binary synergistic elastomer adhesive patch, where hydrogen bonding cross-links between PolyLA and PolyLA-Na prevents PolyLA depolymerization and slow down the dissociation of PolyLA-Na, thus allowing water-responsive sustainable delivery of bioactive LA-based small molecules and durable adhesion to oral mucosal wound due to the adhesive action of PolyLA. In the model of mice and mini-pig oral ulcer, the adhesive patch accelerates the healing of the ulcer by regulating the damaged tissue inflammatory environment, maintaining the stability of oral microbiota, and promoting faster re-epithelialization and angiogenesis. This binary synergistic patch provided a therapeutic strategy to treat oral ulcer.
Collapse
Affiliation(s)
- Chunyan Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Li Mei
- Department of Stomatology, Qingdao University, Qingdao, 266021, China
| | - Danyang Wang
- Department of Stomatology, Qingdao University, Qingdao, 266021, China
| | - Pengfei Jia
- Department of Stomatology, Qingdao University, Qingdao, 266021, China
| | - Qihui Zhou
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266071, China.
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China.
| |
Collapse
|
33
|
Shu QH, Zuo RT, Chu M, Shi JJ, Ke QF, Guan JJ, Guo YP. Fiber-reinforced gelatin/β-cyclodextrin hydrogels loaded with platelet-rich plasma-derived exosomes for diabetic wound healing. BIOMATERIALS ADVANCES 2023; 154:213640. [PMID: 37804684 DOI: 10.1016/j.bioadv.2023.213640] [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: 05/26/2023] [Revised: 09/19/2023] [Accepted: 09/24/2023] [Indexed: 10/09/2023]
Abstract
Diabetic complications with high-glucose status (HGS) cause the dysregulated autophagy and excessive apoptosis of multiple-type cells, leading to the difficulty in wound self-healing. Herein, we firstly developed fiber-reinforced gelatin (GEL)/β-cyclodextrin (β-CD) therapeutic hydrogels by the modification of platelet-rich plasma exosomes (PRP-EXOs). The GEL fibers that were uniformly dispersed within the GEL/β-CD hydrogels remarkably enhanced the compression strengths and viscoelasticity. The PRP-EXOs were encapsulated in the hydrogels via the covalent crosslinking between the PRP-EXOs and genipin. The diabetic rat models demonstrated that the GEL/β-CD hydrogels and PRP-EXOs cooperatively promoted diabetic wound healing. On the one hand, the GEL/β-CD hydrogels provided the biocompatible microenvironments and active components for cell adhesion, proliferation and skin tissue regeneration. On the other hand, the PRP-EXOs in the therapeutic hydrogels significantly activated the autophagy and inhibited the apoptosis of human umbilical vein endothelial cells (HUVECs) and human skin fibroblasts (HSFs). The activation of autophagy and inhibition of apoptosis in HUVECs and HSFs induced the blood vessel creation, collagen formation and re-epithelialization. Taken together, this work proved that the incorporation of PRP-EXOs in a wound dressing was an effective strategy to regulate autophagy and apoptosis, and provide a novel therapeutic platform for diabetic wound healing.
Collapse
Affiliation(s)
- Qiu-Hao Shu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China
| | - Rong-Tai Zuo
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Min Chu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China
| | - Jing-Jing Shi
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China
| | - Qin-Fei Ke
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China
| | - Jun-Jie Guan
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Ya-Ping Guo
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China.
| |
Collapse
|
34
|
Si L, Guo X, Bera H, Chen Y, Xiu F, Liu P, Zhao C, Abbasi YF, Tang X, Foderà V, Cun D, Yang M. Unleashing the healing potential: Exploring next-generation regenerative protein nanoscaffolds for burn wound recovery. Asian J Pharm Sci 2023; 18:100856. [PMID: 38204470 PMCID: PMC10777420 DOI: 10.1016/j.ajps.2023.100856] [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: 06/18/2023] [Revised: 09/15/2023] [Accepted: 10/07/2023] [Indexed: 01/12/2024] Open
Abstract
Burn injury is a serious public health problem and scientists are continuously aiming to develop promising biomimetic dressings for effective burn wound management. In this study, a greater efficacy in burn wound healing and the associated mechanisms of α-lactalbumin (ALA) based electrospun nanofibrous scaffolds (ENs) as compared to other regenerative protein scaffolds were established. Bovine serum albumin (BSA), collagen type I (COL), lysozyme (LZM) and ALA were separately blended with poly(ε-caprolactone) (PCL) to fabricate four different composite ENs (LZM/PCL, BSA/PCL, COL/PCL and ALA/PCL ENs). The hydrophilic composite scaffolds exhibited an enhanced wettability and variable mechanical properties. The ALA/PCL ENs demonstrated higher levels of fibroblast proliferation and adhesion than the other composite ENs. As compared to PCL ENs and other composite scaffolds, the ALA/PCL ENs also promoted a better maturity of the regenerative skin tissues and showed a comparable wound healing effect to Collagen spongeⓇ on third-degree burn model. The enhanced wound healing activity of ALA/PCL ENs compared to other ENs could be attributed to their ability to promote serotonin production at wound sites. Collectively, this investigation demonstrated that ALA is a unique protein with a greater potential for burn wound healing as compared to other regenerative proteins when loaded in the nanofibrous scaffolds.
Collapse
Affiliation(s)
- Liangwei Si
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Xiong Guo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Hriday Bera
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
- Dr. B. C. Roy College of Pharmacy and Allied Health Sciences, Durgapur, 713206, India
| | - Yang Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Fangfang Xiu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Peixin Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Chunwei Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Yasir Faraz Abbasi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Vito Foderà
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen O, Denmark
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang l10016, China
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen O, Denmark
| |
Collapse
|
35
|
Liu W, Jin M, Chen Q, Li Q, Xing X, Luo Y, Sun X. Insight into extracellular vesicles in vascular diseases: intercellular communication role and clinical application potential. Cell Commun Signal 2023; 21:310. [PMID: 37907962 PMCID: PMC10617214 DOI: 10.1186/s12964-023-01304-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/02/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Cells have been increasingly known to release extracellular vesicles (EVs) to the extracellular environment under physiological and pathological conditions. A plethora of studies have revealed that EVs contain cell-derived biomolecules and are found in circulation, thereby implicating them in molecular trafficking between cells. Furthermore, EVs have an effect on physiological function and disease development and serve as disease biomarkers. MAIN BODY Given the close association between EV circulation and vascular disease, this review aims to provide a brief introduction to EVs, with a specific focus on the EV cargoes participating in pathological mechanisms, diagnosis, engineering, and clinical potential, to highlight the emerging evidence suggesting promising targets in vascular diseases. Despite the expansion of research in this field, some noticeable limitations remain for clinical translational research. CONCLUSION This review makes a novel contribution to a summary of recent advances and a perspective on the future of EVs in vascular diseases. Video Abstract.
Collapse
Affiliation(s)
- Wenxiu Liu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Meiqi Jin
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Qiuyan Chen
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Qiaoyu Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Xiaoyan Xing
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China.
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China.
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
| |
Collapse
|
36
|
Ma J, Zhang L, Lei B. Multifunctional MXene-Based Bioactive Materials for Integrated Regeneration Therapy. ACS NANO 2023; 17:19526-19549. [PMID: 37804317 DOI: 10.1021/acsnano.3c01913] [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: 10/09/2023]
Abstract
The reconstruction engineering of tissue defects accompanied by major diseases including cancer, infection, and inflammation is one of the important challenges in clinical medicine. The development of innovative tissue engineering strategies such as multifunctional bioactive materials presents a great potential to overcome the challenge of disease-impaired tissue regeneration. As the major representative of two-dimensional nanomaterials, MXenes have shown multifunctional physicochemical properties and have been diffusely studied as multimodal nanoplatforms in the field of biomedicine. This review summarized the recent advances in the multifunctional properties of MXenes and integrated regeneration-therapy applications of MXene-based biomaterials, including tissue regeneration-tumor therapy, tissue regeneration-infection therapy, and tissue regeneration-inflammation therapy. MXenes have been recognized as good candidates for promoting tissue regeneration and treating diseases through photothermal therapy, regulating cell behavior, and drug and gene delivery. The current challenges and future perspectives of MXene-based biomaterials in integrated regeneration-therapy are also discussed well in this review. In summary, MXene-based biomaterials have shown promising potential for integrated tissue regeneration and disease treatment due to their favorable physicochemical properties and bioactive functions. However, there are still many obstacles and challenges that must be addressed for the regeneration-therapy applications of MXene-based biomaterials, including understanding the bioactive mechanism, ensuring long-term biosafety, and improving their targeting therapy capacity.
Collapse
Affiliation(s)
- Junping Ma
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China
| | - Long Zhang
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Bo Lei
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710000, China
| |
Collapse
|
37
|
Kumawat A, Jasuja K, Ghoroi C. TiB 2-Derived Nanosheets Enhance the Tensile Strength and Controlled Drug Release of Biopolymeric Films Used in Wound Healing. ACS APPLIED BIO MATERIALS 2023; 6:4111-4126. [PMID: 37796555 DOI: 10.1021/acsabm.3c00101] [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: 10/06/2023]
Abstract
Wound healing using an alginate-based biopolymeric film is one of the most preferred treatments. However, these films lack mechanical strength (elasticity and tensile strength), show higher initial burst release, and exhibit high vapor permeability. The present study reports the development of nanosheets derived from titanium diboride (10 nm) (NTB)-incorporated biopolymeric films (0.025, 0.05, and 0.1% w/v) using sodium alginate (SA) and carboxymethyl cellulose (CMC) to overcome the shortfalls. The surface properties of the film, nanosheet distribution within the film, and possible interactions with the film are explored by using scanning electron microscopy (SEM), energy-dispersive spectroscopy (EDS), Fourier transform infrared (FTIR), and X-ray diffraction (XRD). These analyses confirm that nanosheets are uniformly distributed in the film and introduce unevenness on the film's surface. The tensile strength of the nanosheet-incorporated film (0.1% NTB film) using UTM is found to be 24.30 MPa (six times higher compared to the blank film), equivalent to human skin. The water vapor transmission rate of the film is also found to be in the desired range (i.e., 2000-2500 g/m2 day). The biocompatibility of the NTB film is confirmed by the MTT assay test using NIH/3T3 cells and HEK 293 cells. Furthermore, the scratch assay shows that the developed films promote cell migration and proliferation. The antibacterial activity of the film is also demonstrated using a model drug, tetracycline hydrochloride (TCl). Besides, the film exhibits the sustained release of TCl and follows the Korsmeyer-Peppas model for drug release. Overall, the 0.1% w/v NTB film is easy to fabricate, biocompatible and shows superior mechanical properties.
Collapse
Affiliation(s)
- Akshant Kumawat
- DryProTech Lab. and BoRN Research Lab, Chemical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382055, Gujarat, India
| | - Kabeer Jasuja
- DryProTech Lab. and BoRN Research Lab, Chemical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382055, Gujarat, India
| | - Chinmay Ghoroi
- DryProTech Lab. and BoRN Research Lab, Chemical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382055, Gujarat, India
| |
Collapse
|
38
|
Zhang J, Luo Q, Hu Q, Zhang T, Shi J, Kong L, Fu D, Yang C, Zhang Z. An injectable bioactive dressing based on platelet-rich plasma and nanoclay: Sustained release of deferoxamine to accelerate chronic wound healing. Acta Pharm Sin B 2023; 13:4318-4336. [PMID: 37799395 PMCID: PMC10547914 DOI: 10.1016/j.apsb.2022.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Delayed diabetic wound healing has placed an enormous burden on society. The key factors limiting wound healing include unresolved inflammation and impaired angiogenesis. Platelet-rich plasma (PRP) gel, a popular biomaterial in the field of regeneration, has limited applications due to its non-injectable properties and rapid release and degradation of growth factors. Here, we prepared an injectable hydrogel (DPLG) based on PRP and laponite by a simple one-step mixing method. Taking advantages of the non-covalent interactions, DPLG could overcome the limitations of PRP gels, which is injectable to fill irregular injures and could serve as a local drug reservoir to achieve the sustained release of growth factors in PRP and deferoxamine (an angiogenesis promoter). DPLG has an excellent ability in accelerating wound healing by promoting macrophage polarization and angiogenesis in a full-thickness skin defect model in type I diabetic rats and normal rats. Taken together, this study may provide the ingenious and simple bioactive wound dressing with a superior ability to promote wound healing.
Collapse
Affiliation(s)
- Jiao Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Luo
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Hu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tiantian Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingyu Shi
- Liyuan Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dehao Fu
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Center for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
39
|
Yang Y, Chu C, Liu L, Wang C, Hu C, Rung S, Man Y, Qu Y. Tracing immune cells around biomaterials with spatial anchors during large-scale wound regeneration. Nat Commun 2023; 14:5995. [PMID: 37752124 PMCID: PMC10522601 DOI: 10.1038/s41467-023-41608-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Skin scarring devoid of dermal appendages after severe trauma has unfavorable effects on aesthetic and physiological functions. Here we present a method for large-area wound regeneration using biodegradable aligned extracellular matrix scaffolds. We show that the implantation of these scaffolds accelerates wound coverage and enhances hair follicle neogenesis. We perform multimodal analysis, in combination with single-cell RNA sequencing and spatial transcriptomics, to explore the immune responses around biomaterials, highlighting the potential role of regulatory T cells in mitigating tissue fibrous by suppressing excessive type 2 inflammation. We find that immunodeficient mice lacking mature T lymphocytes show the typical characteristic of tissue fibrous driven by type 2 macrophage inflammation, validating the potential therapeutic effect of the adaptive immune system activated by biomaterials. These findings contribute to our understanding of the coordination of immune systems in wound regeneration and facilitate the design of immunoregulatory biomaterials in the future.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chenyu Chu
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Li Liu
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chenbing Wang
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chen Hu
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shengan Rung
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yi Man
- Department of Oral Implantology & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yili Qu
- Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
40
|
Wang H, Jin J, Zhang C, Gong F, Hu B, Wu X, Guan M, Xia D. Multifunctional Drugs-Loaded Carbomol Hydrogel Promotes Diabetic Wound Healing via Antimicrobial and Immunoregulation. Gels 2023; 9:761. [PMID: 37754442 PMCID: PMC10530860 DOI: 10.3390/gels9090761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023] Open
Abstract
Diabetic wound healing poses a significant clinical dilemma. Bacterial infection and immune dysregulation are the predominant reasons. However, conventional wound dressings with a single treatment approach often limit therapeutic efficacy and continue working with difficulty. These limitations cause high treatment failure for diabetic wounds. In this study, we developed a multiple drug-loaded carbomer hydrogel containing Que/Van/Rif (QVR-CBMG) for the simultaneous treatment of infection and immune dysregulation. Honeycomb-like QVR-CBMG hydrogel exhibits excellent abilities to eliminate bacterial infection and biofilms in vitro. Moreover, QVR-CBMG hydrogel possesses an immunomodulatory capacity via affecting the Sirt3/SOD2 signaling pathway to promote M2 macrophages. Furthermore, QVR-CBMG hydrogel effectively promotes wound healing in diabetic rats through several mechanisms. The multidrug-loaded wound dressing not only eliminates bacterial infection and facilitated angiogenesis but also promotes collagen deposition and remodulates the local immune microenvironment in the areas of wounds. In summary, this synthetic strategy to eliminate infection and regulate immune disorders has potential translational value for the prevention and management of diabetic wounds.
Collapse
Affiliation(s)
- Hehui Wang
- Department of Orthopedics, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; (H.W.); (C.Z.); (F.G.); (B.H.); (X.W.)
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China;
| | - Jiale Jin
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China;
| | - Chi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; (H.W.); (C.Z.); (F.G.); (B.H.); (X.W.)
| | - Fangyi Gong
- Department of Orthopedics, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; (H.W.); (C.Z.); (F.G.); (B.H.); (X.W.)
| | - Baiwen Hu
- Department of Orthopedics, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; (H.W.); (C.Z.); (F.G.); (B.H.); (X.W.)
| | - Xiaochuan Wu
- Department of Orthopedics, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; (H.W.); (C.Z.); (F.G.); (B.H.); (X.W.)
| | - Ming Guan
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China;
| | - Dongdong Xia
- Department of Orthopedics, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China; (H.W.); (C.Z.); (F.G.); (B.H.); (X.W.)
| |
Collapse
|
41
|
Pourtalebi Jahromi L, Rothammer M, Fuhrmann G. Polysaccharide hydrogel platforms as suitable carriers of liposomes and extracellular vesicles for dermal applications. Adv Drug Deliv Rev 2023; 200:115028. [PMID: 37517778 DOI: 10.1016/j.addr.2023.115028] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/26/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Lipid-based nanocarriers have been extensively investigated for their application in drug delivery. Particularly, liposomes are now clinically established for treating various diseases such as fungal infections. In contrast, extracellular vesicles (EVs) - small cell-derived nanoparticles involved in cellular communication - have just recently sparked interest as drug carriers but their development is still at the preclinical level. To drive this development further, the methods and technologies exploited in the context of liposome research should be applied in the domain of EVs to facilitate and accelerate their clinical translation. One of the crucial steps for EV-based therapeutics is designing them as proper dosage forms for specific applications. This review offers a comprehensive overview of state-of-the-art polysaccharide-based hydrogel platforms designed for artificial and natural vesicles with application in drug delivery to the skin. We discuss their various physicochemical and biological properties and try to create a sound basis for the optimization of EV-embedded hydrogels as versatile therapeutic avenues.
Collapse
Affiliation(s)
- Leila Pourtalebi Jahromi
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Biology, Pharmaceutical Biology, Staudtstr. 5, 91058 Erlangen, Germany
| | - Markus Rothammer
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Biology, Pharmaceutical Biology, Staudtstr. 5, 91058 Erlangen, Germany
| | - Gregor Fuhrmann
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Biology, Pharmaceutical Biology, Staudtstr. 5, 91058 Erlangen, Germany; FAU NeW, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany.
| |
Collapse
|
42
|
Yu Y, Xiao H, Tang G, Wang H, Shen J, Sun Y, Wang S, Kong W, Chai Y, Liu X, Wang X, Wen G. Biomimetic hydrogel derived from decellularized dermal matrix facilitates skin wounds healing. Mater Today Bio 2023; 21:100725. [PMID: 37483381 PMCID: PMC10359665 DOI: 10.1016/j.mtbio.2023.100725] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/15/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Cutaneous wound healing affecting millions of people worldwide represents an unsolvable clinical issue that is frequently challenged by scar formation with dramatical pain, impaired mobility and disfigurement. Herein, we prepared a kind of light-sensitive decellularized dermal extracellular matrix-derived hydrogel with fast gelling performance, biomimetic porous microstructure and abundant bioactive functions. On account of its excellent cell biocompatibility, this ECM-derived hydrogel could induce a marked cellular infiltration and enhance the tube formation of HUVECs. In vivo experiments based upon excisional wound splinting model showed that the hydrogel prominently imparted skin wound healing, as evidenced by notably increased skin appendages and well-organized collagen expression, coupled with significantly enhanced angiogenesis. Moreover, the skin regeneration mediated by this bioactive hydrogel was promoted by an accelerated M1-to-M2 macrophage phenotype transition. Consequently, the decellularized dermal matrix-derived bioactive hydrogel orchestrates the entire skin healing microenvironment to promote wound healing and will be of high value in treatment of cutaneous wound healing. As such, this biomimetic ddECMMA hydrogel provides a promising versatile opinion for the clinical translation.
Collapse
Affiliation(s)
- Yaling Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Huimin Xiao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Guoke Tang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China
| | - Hongshu Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Junjie Shen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yi Sun
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xuanzhe Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Gen Wen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
43
|
Zhu Y, Liao ZF, Mo MH, Xiong XD. Mesenchymal Stromal Cell-Derived Extracellular Vesicles for Vasculopathies and Angiogenesis: Therapeutic Applications and Optimization. Biomolecules 2023; 13:1109. [PMID: 37509145 PMCID: PMC10377109 DOI: 10.3390/biom13071109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Extracellular vesicles (EVs), as part of the cellular secretome, have emerged as essential cell-cell communication regulators in multiple physiological and pathological processes. Previous studies have widely reported that mesenchymal stromal cell-derived EVs (MSC-EVs) have potential therapeutic applications in ischemic diseases or regenerative medicine by accelerating angiogenesis. MSC-EVs also exert beneficial effects on other vasculopathies, including atherosclerosis, aneurysm, vascular restenosis, vascular calcification, vascular leakage, pulmonary hypertension, and diabetic retinopathy. Consequently, the potential of MSC-EVs in regulating vascular homeostasis is attracting increasing interest. In addition to native or naked MSC-EVs, modified MSC-EVs and appropriate biomaterials for delivering MSC-EVs can be introduced to this area to further promote their therapeutic applications. Herein, we outline the functional roles of MSC-EVs in different vasculopathies and angiogenesis to elucidate how MSC-EVs contribute to maintaining vascular system homeostasis. We also discuss the current strategies to optimize their therapeutic effects, which depend on the superior bioactivity, high yield, efficient delivery, and controlled release of MSC-EVs to the desired regions, as well as the challenges that need to be overcome to allow their broad clinical translation.
Collapse
Affiliation(s)
- Ying Zhu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Zhao-Fu Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Miao-Hua Mo
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Xing-Dong Xiong
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
44
|
Near-infrared light-responsive multifunctional hydrogel releasing peptide-functionalized gold nanorods sequentially for diabetic wound healing. J Colloid Interface Sci 2023; 639:369-384. [PMID: 36812853 DOI: 10.1016/j.jcis.2023.02.078] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/16/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Treatment for chronic diabetic wounds remains a clinical challenge. Wound healing process occurs in three phases: inflammation, proliferation and remodeling. Several factors including bacterial infection, decreased local angiogenesis and diminished blood supply delay wound healing. There is an urgent need to develop wound dressings with multiple biological effects for different stages of diabetic wound healing. Here, we develop a multifunctional hydrogel with two-stage sequential release upon near-infrared (NIR) stimulation, antibacterial activity and pro-angiogenic efficacy. This hydrogel consists of covalently crosslinked bilayer structure, with the lower thermoresponsive poly(N-isopropylacrylamide)/gelatin methacrylate (NG) layer and the upper highly stretchable alginate/polyacrylamide (AP) layer embedding different peptide-functionalized gold nanorods (AuNRs) in each layer. Antimicrobial peptide-functionalized AuNRs released from NG layer exert antibacterial effects. After NIR irradiation, the photothermal transition efficacy of AuNRs synergistically enhances bactericidal efficacy. The contraction of thermoresponsive layer also promotes the release of embedded cargos during early stage. The pro-angiogenic peptide-functionalized AuNRs released from AP layer promote angiogenesis and collagen deposition by accelerating fibroblast and endothelial cell proliferation, migration and tube formation during the subsequent healing phases. Therefore, the multifunctional hydrogel with effective antibacterial activity, pro-angiogenic efficacy and sequential release behaviors is a potential biomaterial for diabetic chronic wound healing.
Collapse
|
45
|
Debnath K, Heras KL, Rivera A, Lenzini S, Shin JW. Extracellular vesicle-matrix interactions. NATURE REVIEWS. MATERIALS 2023; 8:390-402. [PMID: 38463907 PMCID: PMC10919209 DOI: 10.1038/s41578-023-00551-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 03/12/2024]
Abstract
The extracellular matrix in microenvironments harbors a variety of signals to control cellular functions and the materiality of tissues. Most efforts to synthetically reconstitute the matrix by biomaterial design have focused on decoupling cell-secreted and polymer-based cues. Cells package molecules into nanoscale lipid membrane-bound extracellular vesicles and secrete them. Thus, extracellular vesicles inherently interact with the meshwork of the extracellular matrix. In this Review, we discuss various aspects of extracellular vesicle-matrix interactions. Cells receive feedback from the extracellular matrix and leverage intracellular processes to control the biogenesis of extracellular vesicles. Once secreted, various biomolecular and biophysical factors determine whether extracellular vesicles are locally incorporated into the matrix or transported out of the matrix to be taken up by other cells or deposited into tissues at a distal location. These insights can be utilized to develop engineered biomaterials where EV release and retention can be precisely controlled in host tissue to elicit various biological and therapeutic outcomes.
Collapse
Affiliation(s)
- Koushik Debnath
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kevin Las Heras
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU)
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Ambar Rivera
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
| | - Stephen Lenzini
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
46
|
Zhao K, Kong C, Shi N, Jiang J, Li P. Potential angiogenic, immunomodulatory, and antifibrotic effects of mesenchymal stem cell-derived extracellular vesicles in systemic sclerosis. Front Immunol 2023; 14:1125257. [PMID: 37251412 PMCID: PMC10213547 DOI: 10.3389/fimmu.2023.1125257] [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: 12/16/2022] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Systemic sclerosis (SSc) is an intricate systemic autoimmune disease with pathological features such as vascular injury, immune dysregulation, and extensive fibrosis of the skin and multiple organs. Treatment options are limited; however, recently, mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been acknowledged in preclinical and clinical trials as being useful in treating autoimmune diseases and are likely superior to MSCs alone. Recent research has also shown that MSC-EVs can ameliorate SSc and the pathological changes in vasculopathy, immune dysfunction, and fibrosis. This review summarizes the therapeutic effects of MSC-EVs on SSc and the mechanisms that have been discovered to provide a theoretical basis for future studies on the role of MSC-EVs in treating SSc.
Collapse
Affiliation(s)
- Kelin Zhao
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Chenfei Kong
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Naixu Shi
- Department of Stomatology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
47
|
Yan T, Huang L, Yan Y, Zhong Y, Xie H, Wang X. Bone marrow mesenchymal stem cell-derived exosome miR-29b-3p alleviates UV irradiation-induced photoaging in skin fibroblast. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2023; 39:235-245. [PMID: 35950642 DOI: 10.1111/phpp.12827] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/17/2022] [Accepted: 08/08/2022] [Indexed: 05/10/2023]
Abstract
BACKGROUND Mesenchymal stem cells-derived exosome (MSCs-exo) was identified to reduce photoaging. The purpose of this study was to investigate the potential role of microRNA (miR)-29b-3p derived from bone marrow MSCs-exo (BMSCs-exo) in photoaging. METHODS Exosomes were isolated from BMSCs and verified by Western blot. A photoaging cell model was constructed by UVB irradiation of human dermal fibroblasts (HDFs). Quantitative real-time PCR (RT-qPCR) was performed to detect the mRNA levels of miR-29b-3p, collagen type I and matrix metalloproteinases (MMPs). CCK-8, Transwell and flow cytometry were applicated to examine cell viability, migration and apoptosis. Commercial kits are used to measure levels of oxidative stress indicators. Finally, a dual-luciferase reporter assay was applied to validate the target of miR-29b-3p. RESULTS Extracted exosomes were positive for HSP70 and CD9. Survival of HDFs increased in an exosome concentration-dependent manner. UVB irradiation inhibited miR-29b-3p levels compared with controls, but BMSCs-exo treatment restored miR-29b-3p levels (p < .05). Additionally, BMSCs-exo-miR-29b-3p reversed the inhibition of HDFs migration and oxidative stress by UVB irradiation, as well as the promotion of apoptosis. However, this reversal was attenuated by the suppression of miR-29b-3p (p < .05). Furthermore, BMSCs-exo-miR-29b-3p also inhibited the degradation of collagen type I and the production of MMPs in photoaging, and they were also eliminated by the reduced miR-29b-3p. Finally, MMP-2 was the target gene of miR-29b-3p. CONCLUSION Our study presented a novel role for BMSCs-exo-miR-29b-3p in improving skin photoaging function, and these findings may provide new insights into the targeted treatment of skin photoaging.
Collapse
Affiliation(s)
- Tingting Yan
- Department of Medical Cosmetology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Lining Huang
- Department of Medical Cosmetology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yunling Yan
- Department of Medical Cosmetology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yiping Zhong
- Department of Medical Cosmetology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Heng Xie
- Department of Medical Cosmetology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Xiaohua Wang
- Department of Medical Cosmetology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
48
|
Zhang E, Phan P, Zhao Z. Cellular nanovesicles for therapeutic immunomodulation: A perspective on engineering strategies and new advances. Acta Pharm Sin B 2023; 13:1789-1827. [PMID: 37250173 PMCID: PMC10213819 DOI: 10.1016/j.apsb.2022.08.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023] Open
Abstract
Cellular nanovesicles which are referred to as cell-derived, nanosized lipid bilayer structures, have emerged as a promising platform for regulating immune responses. Owing to their outstanding advantages such as high biocompatibility, prominent structural stability, and high loading capacity, cellular nanovesicles are suitable for delivering various immunomodulatory molecules, such as small molecules, nucleic acids, peptides, and proteins. Immunomodulation induced by cellular nanovesicles has been exploited to modulate immune cell behaviors, which is considered as a novel cell-free immunotherapeutic strategy for the prevention and treatment of diverse diseases. Here we review emerging concepts and new advances in leveraging cellular nanovesicles to activate or suppress immune responses, with the aim to explicate their applications for immunomodulation. We overview the general considerations and principles for the design of engineered cellular nanovesicles with tailored immunomodulatory activities. We also discuss new advances in engineering cellular nanovesicles as immunotherapies for treating major diseases.
Collapse
Affiliation(s)
- Endong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
- Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
49
|
Zhou Z, Mei X, Hu K, Ma M, Zhang Y. Nanohybrid Double Network Hydrogels Based on a Platinum Nanozyme Composite for Antimicrobial and Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2023; 15:17612-17626. [PMID: 37010097 DOI: 10.1021/acsami.3c00459] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Along with hypoxia, severe bacterial infection, and abnormal pH, continuous inflammatory response hinders diabetic wounds from healing. It leads to the accumulation of large amounts of reactive oxygen species (ROS) and therefore prevents the transition of diabetic wounds from the inflammatory phase to the proliferative phase. In this work, a nanohybrid double network hydrogel with injectable, self-healing, and tissue adhesion properties based on a platinum nanozyme composite (PFOB@PLGA@Pt) was constructed to manage diabetic wound healing. PFOB@PLGA@Pt exhibited oxygen supply capacity and enzyme catalytic performance accompanied by pH self-regulation in the entire phases of wound healing. In the first stage, the oxygen carried by perfluorooctyl bromide (PFOB) can ameliorate the hypoxia and boost the glucose oxidase-like catalyzed reaction of Pt NPs, leading to a lowered pH environment with gluconic acid. As a result, the NADH oxidase-like, peroxidase-like, and oxidase-like multiple enzyme activities were activated successively, leading to synergistic antibacterial effects through the production of ROS. After the bacterial infection had cleared, the catalase-like and superoxide dismutase-like activities of Pt NPs reshaped the redox microenvironment by scavenging the excess ROS, which transitioned the wound from the inflammatory phase to the proliferative phase. The microenvironmentally adaptive hydrogel treatment can cover all phases of wound healing, showing the significant promoting effect in the repair of diabetic infected wounds.
Collapse
Affiliation(s)
- Ziying Zhou
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Xiuming Mei
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
- Key Laboratory of Biotoxin Analysis & Assessment for State Market Regulation, Nanjing Institute of Product Quality Inspection & Testing, Nanjing 210019, P. R. China
| | - Ke Hu
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 210009, Jiangsu, P. R. China
| | - Ming Ma
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
50
|
Cheng S, Pan M, Hu D, Han R, Li L, Bei Z, Li Y, Sun A, Qian Z. Adhesive chitosan-based hydrogel assisted with photothermal antibacterial property to prompt mice infected skin wound healing. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|