1
|
Duan L, Liu G, Liao F, Xie C, Shi J, Yang X, Zheng F, Reis RL, Kundu SC, Xiao B. Antheraea pernyi silk nanofibrils with inherent RGD motifs accelerate diabetic wound healing: A novel drug-free strategy to promote hemostasis, regulate immunity and improve re-epithelization. Biomaterials 2025; 318:123127. [PMID: 39879843 DOI: 10.1016/j.biomaterials.2025.123127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/16/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
The chronic inflammation and matrix metalloprotease (MMP)-induced tissue degradation significantly disrupt re-epithelization and delay the healing process of diabetic wounds. To address these issues, we produced nanofibrils from Antheraea pernyi (Ap) silk fibers via a facile and green treatment of swelling and shearing. The integrin receptors on the cytomembrane could specifically bind to the Ap nanofibrils (ApNFs) due to their inherent Arg-Gly-Asp (RGD) motifs, which activated platelets to accelerate coagulation and promoted fibroblast migration, adhesion and spreading. These degradable nanofibrils served as effective competitive substrates to reduce MMP-induced tissue degradation. ApNFs and their enzymatic hydrolysates could modulate macrophage polarization due to their RGD motifs. RNA sequencing further revealed that ApNFs treatment activated the JAK2-STAT5b and PI3K-Akt signaling pathways while suppressed the NF-κB, IL-17 and TNF signaling pathways in macrophages. The full-thickness skin wound experiments confirmed that ApNFs significantly accelerated wound healing in both diabetic and non-diabetic rats. Notably, in diabetic wound, ApNFs and their enzymatic hydrolysates polarized the accumulated M1-type macrophages into M2-type, which promoted the wound to get rid of the inflammatory stage and transition to the following proliferative stage, improving the wound healing percentage on day 14 from 74.9 % to 93.2 % by facilitating collagen deposition, angiogenesis and re-epithelization. These results demonstrate that ApNFs are promising drug-free diabetic wound dressings with favorable inherent immunoregulatory properties for biomedical translation.
Collapse
Affiliation(s)
- Lian Duan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Ga Liu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Fuying Liao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Chunyu Xie
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Jiahao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Xiao Yang
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Fan Zheng
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China.
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, 4800-058, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, 4800-058, Portugal
| | - Bo Xiao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| |
Collapse
|
2
|
Lin Z, Jia J, Liang C, Ma Y, Liu H, Fang K, Hu Y. Shape-recoverable chitosan/sodium alginate aerogels with sustained oxygen release and antibacterial activity for diabetic wound healing. Int J Biol Macromol 2025; 305:141005. [PMID: 39965687 DOI: 10.1016/j.ijbiomac.2025.141005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Wounds in diabetic patients are often inhibited in angiogenesis and extracellular matrix synthesis due to insufficient oxygen supply, leading to prolonged healing time. Although oxygen therapy has been shown to promote the healing of chronic wounds, its therapeutic effectiveness remains limited by high costs, complex procedures, and a lack of sustainability. In this study, we developed an aerogel composed of chitosan (CS), sodium alginate (SA), and calcium peroxide (CPO). These aerogels can absorb wound exudate and release oxygen upon contact, providing continuous oxygen delivery for over five days. The freeze-dried aerogels are soft and exhibit shape recovery properties, which facilitate enhanced oxygen transport. Furthermore, the CS/SA-CPO aerogel demonstrated strong antioxidant activity, excellent biocompatibility, and potent antibacterial properties (exceeding 99.99 %). In vivo experiments indicated that the CS/SA-CPO aerogel promotes wound healing by continuously releasing oxygen, accelerating collagen deposition, and enhancing re-epithelialization, ultimately achieving 99.65 % wound closure within 21 days. This innovative approach offers a promising strategy for managing chronic, hypoxic diabetic wounds.
Collapse
Affiliation(s)
- Zhihao Lin
- Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jiaojiao Jia
- College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China
| | - Chengzhi Liang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Yueyan Ma
- Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Hao Liu
- Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Kuanjun Fang
- College of Textiles and Clothing, Qingdao University, Qingdao 266071, China; Collaborative Innovation Center for Eco-Textiles of Shandong Province and the Ministry of Education, Qingdao University, Qingdao 266071, China.
| | - Yanling Hu
- Department of Orthopedics, Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
3
|
Zhang J, Hou C, Yu H, Newton MAA, Xin B, Li T. Electrospinning Trilayer Dressing of PLA/ZnO with Controlled Released Profiles for Visible pH Monitoring and Effective Exudate Management. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025. [PMID: 40266576 DOI: 10.1021/acs.langmuir.5c01044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
To address persistent challenges in wound care, including exudate management, infection prevention, and healing monitoring, this study developed a multifunctional fiber dressing using electrospinning technology. The system integrates a patterned hydrophobic polylactic acid/zinc oxide (PLA/ZnO) inner layer providing antimicrobial protection and exudate drainage; a polyacrylonitrile/polydopamine (PAN/PDA) intermediate water transport layer enabling unidirectional exudate transfer; and an outer PAN-based functional membrane incorporating sodium polyacrylate (SPA), phenol red (PSP), and tetracycline hydrochloride (TCH) for controlled drug release and visual wound status monitoring. The comprehensive evaluation revealed exceptional performance: rapid exudate removal within 3.35 s, 96 h sustained antibacterial drug release (68.6% cumulative release), pH-responsive chromatic transition (yellow-to-red at pH 5-9), and significant antimicrobial efficacy against Escherichia coli and Staphylococcus aureus (inhibition zone diameters of 10 and 18 mm, respectively). The dressing exhibited a balance of mechanical strength (6.1 MPa tensile strength) and breathability (13.4 mm/s air permeability rate). By integrating three core functions─efficient exudate management, antimicrobial regulation, and intelligent healing monitoring─this multifunctional architecture demonstrates clinical applicability in postoperative care and chronic wound therapy.
Collapse
Affiliation(s)
- Jun Zhang
- Shanghai University of Engineering Science, No.333, Longteng Road, Songjiang District, Shanghai CN 201620, P. R. of China
| | - Chao Hou
- Shanghai University of Engineering Science, No.333, Longteng Road, Songjiang District, Shanghai CN 201620, P. R. of China
| | - He Yu
- Shanghai University of Engineering Science, No.333, Longteng Road, Songjiang District, Shanghai CN 201620, P. R. of China
| | - Md All Amin Newton
- Shanghai University of Engineering Science, No.333, Longteng Road, Songjiang District, Shanghai CN 201620, P. R. of China
| | - Binjie Xin
- Shanghai University of Engineering Science, No.333, Longteng Road, Songjiang District, Shanghai CN 201620, P. R. of China
| | - Tingxiao Li
- Shanghai University of Engineering Science, No.333, Longteng Road, Songjiang District, Shanghai CN 201620, P. R. of China
| |
Collapse
|
4
|
Shi H, Shu P, Wang Z, Meng C, Yu R, Xu Y, Li C. Knowledge mapping and research trends of cathelicidin peptide LL-37 from 1995 to 2024: a bibliometric study. Comput Methods Biomech Biomed Engin 2025:1-14. [PMID: 40083141 DOI: 10.1080/10255842.2025.2477218] [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: 09/28/2024] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND To conduct a comprehensive bibliometric analysis of LL-37, summarize its development trends and patterns, and identify emerging research hotspots. METHODS Bibliometric and Knowledge Graph Analysis of Literature Data Related to LL-37 in the WOSCC Database Using Citespace and Vosviewer. RESULTS A total of 2,814 articles were analyzed, revealing a steady increase in recent publications. The USA and Sweden were the main contributors, with PLOS One publishing the most articles. Research on LL-37 in cancer and mast cells is emerging as a new focus. CONCLUSION The research status and development trends of LL-37 were quantitatively analyzed, providing new directions for future studies.
Collapse
Affiliation(s)
- Hongxin Shi
- Clinical Medical College of Dali University, Dali, China
| | - Peizhou Shu
- Clinical Medical College of Dali University, Dali, China
| | - Zhihao Wang
- Yunnan University of Chinese Medicine, Kunming, China
| | - Chen Meng
- Graduate School of Kunming Medical University, Kunming, China
| | - Rao Yu
- Graduate School of Kunming Medical University, Kunming, China
| | - YongQing Xu
- Department of Orthopedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, China
| | - Chuan Li
- Department of Orthopedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, China
- Department of Orthopaedic Surgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
5
|
Rong Y, Zhao Z, Lv D, Yin R, Lu L, Xu Z, Ren L, Zhao P, Hu Z, Tao J, Cao X, Tang B. Tailored Metal-Phenolic Network with Hypoglycemic Polyphenol for Promoting Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2025; 17:15163-15176. [PMID: 40025657 DOI: 10.1021/acsami.4c22878] [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: 03/04/2025]
Abstract
Diabetic foot ulcer is a common and serious complication of diabetes, with a high risk of amputation, recurrence, and mortality. Aiming at the characteristics of diabetic wounds and based on the result of network pharmacology, a tailored ligand cyanidin-3-O-glucoside (C3G) was selected to construct a metal-phenolic network (CM) through the self-assembly reaction with manganese ions. CM integrates the pharmacological advantages of C3G in antidiabetes and the anti-inflammatory activity of metal-phenolic networks by simulating the metal coordination structure of antioxidant enzymes. Reasonably, the wound areas of db/db mice with CM treatment rapidly decreased to 3.06% at day 14, accompanied by the improvement of tissue microenvironment. Mechanism investigation indicated that CM can not only reduce inflammation activation and immunoreaction but also increase gene transcripts in glucose metabolism, response to hypoxia, and angiogenesis. It is believed that this work opens a way for designing disease-specific metal-phenolic networks, and the CM with high biosafety promotes the clinical treatment of diabetic wounds.
Collapse
Affiliation(s)
- Yanchao Rong
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zirui Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Dongming Lv
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Rong Yin
- Department of Dermatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Ling Lu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhongye Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Lei Ren
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Peng Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhicheng Hu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jia Tao
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Xiaoling Cao
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Bing Tang
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
6
|
Kaczmarek-Szczepańska B, Grabska-Zielińska S. Biopolymeric Scaffolds with Melatonin for Tissue Engineering-A Review. Int J Mol Sci 2025; 26:2520. [PMID: 40141163 PMCID: PMC11942045 DOI: 10.3390/ijms26062520] [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: 01/17/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Melatonin, a natural hormone with antioxidant, anti-inflammatory, and regenerative properties, has gained increasing attention in tissue engineering for its ability to enhance the therapeutic potential of biopolymeric scaffolds. These scaffolds, designed to mimic the extracellular matrix, provide structural support and a bioactive environment for tissue regeneration. By integrating melatonin, researchers aim to create multifunctional scaffolds that promote cell proliferation, modulate inflammatory responses, and improve wound healing outcomes. Challenges in utilizing melatonin include maintaining its stability under light, heat, and oxygen exposure, and optimizing its release profile for sustained therapeutic effects. Innovative fabrication methods, such as electrospinning, 3D printing, and lyophilization, have enabled precise control over scaffold architecture and melatonin delivery. These techniques ensure enhanced interactions with target tissues and tailored regeneration processes. Combining melatonin with growth factors, cytokines, and antimicrobial agents offers the potential for multifunctional applications, from chronic wound management to bone and nerve regeneration. Continued research in this field promises transformative solutions in regenerative medicine, expanding the clinical applicability of melatonin-enriched scaffolds. This review highlights the current progress, challenges, and opportunities associated with harnessing melatonin's therapeutic potential within tissue engineering frameworks.
Collapse
Affiliation(s)
- Beata Kaczmarek-Szczepańska
- Laboratory for Functional Polymeric Materials, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarin 7, 87-100 Toruń, Poland
| | - Sylwia Grabska-Zielińska
- Faculty of Chemical Technology and Engineering, Bydgoszcz University of Science and Technology, Seminaryjna 3, 85-326 Bydgoszcz, Poland;
| |
Collapse
|
7
|
Shahriar SMS, Andrabi SM, Al-Gahmi AM, Yan Z, McCarthy AD, Wang C, Yusuf ZA, Sharma NS, Busquets ME, Nilles MI, Jara CP, Yang K, Carlson MA, Xie J. Bicomponent nano- and microfiber aerogels for effective management of junctional hemorrhage. Nat Commun 2025; 16:2403. [PMID: 40064972 PMCID: PMC11893793 DOI: 10.1038/s41467-025-57836-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Managing junctional hemorrhage is challenging due to ineffective existing techniques, with the groin being the most common site, accounting for approximately 19.2% of potentially survivable field deaths. Here, we report a bicomponent nano- and microfiber aerogel (NMA) for injection into deep, narrow junctional wounds to effectively halt bleeding. The aerogel comprises intertwined poly(lactic acid) nanofibers and poly(ε-caprolactone) microfibers, with mechanical properties tunable through crosslinking. Optimized aerogels demonstrate improved resilience, toughness, and elasticity, enabling rapid re-expansion upon blood contact. They demonstrate superior blood absorption and clotting efficacy compared to commercial products (i.e., QuikClot® Combat Gauze and XStat®). Most importantly, in a lethal swine junctional wound model (Yorkshire swine, both male and female, n = 5), aerogel treatment achieved immediate hemostasis, a 100% survival rate, no rebleeding, hemodynamic stability, and stable coagulation, hematologic, and arterial blood gas testing.
Collapse
Affiliation(s)
- S M Shatil Shahriar
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute for Research in Cancer and Allied Diseases, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Syed Muntazir Andrabi
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Al-Murtadha Al-Gahmi
- Department of Surgery - General Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Zishuo Yan
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Alec D McCarthy
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Chenlong Wang
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Zakariya A Yusuf
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Navatha Shree Sharma
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Milton E Busquets
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mallory I Nilles
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Carlos Poblete Jara
- Department of Surgery - Vascular Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kai Yang
- Department of Surgery - Plastic & Reconstructive Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark A Carlson
- Department of Surgery - General Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Jingwei Xie
- Department of Surgery - Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Mechanical and Materials Engineering, University of Nebraska Lincoln, Lincoln, NE, USA.
| |
Collapse
|
8
|
L J, Kamaraj S, Kandasamy R, Alagarsamy S. Electrospinning: A New Frontier in Peptide Therapeutics. AAPS PharmSciTech 2025; 26:69. [PMID: 40011310 DOI: 10.1208/s12249-025-03054-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
The nanofiber technology has recently undergone an unprecedented transformation, finding widespread utilities across diverse scientific disciplines. It is noteworthy that electrospinning approaches have emerged as an adaptable and successful approach to generate fibers ranging in rapidly as a class of therapeutic agents with a high level of target specificity. Peptides encounter several challenges as drugs, including swift breakdown by the body, rapid elimination from the bloodstream, inadequate stability, and restricted ability to cross cell membranes. This renders it challenging to employ them as drugs. However, electrospun nanofibers might address these problems. This review explores the promising potential of electrospinning nanofibers for peptide delivery. We delve into recent advancements in this technique, highlighting its effectiveness in overcoming challenges associated with peptide drug delivery. It provides an analysis of the trends identified in the use of the electrospinning technique and its role in peptide drug delivery systems, based on a review of data collected over a period of five to seven years.
Collapse
Affiliation(s)
- Jeyanthi L
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, Anna University, BIT Campus, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Sivadharshini Kamaraj
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, Anna University, BIT Campus, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Ruckmani Kandasamy
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, Anna University, BIT Campus, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Shanmugarathinam Alagarsamy
- Centre for Excellence in Nanobio Translational REsearch (CENTRE), Department of Pharmaceutical Technology, Anna University, BIT Campus, Tiruchirappalli, 620024, Tamil Nadu, India.
| |
Collapse
|
9
|
Wang Z, Sun Y, Dong P, Wang J, Wang L, Zhao A, Qu G, Li H, Maheshika Gunarathne KD, Zhang W, Chen Y, Meng X. Thermosensitive-based synergistic antibacterial effects of novel LL37@ZPF-2 loaded poloxamer hydrogel for infected skin wound healing. Int J Pharm 2025; 670:125210. [PMID: 39800001 DOI: 10.1016/j.ijpharm.2025.125210] [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: 11/13/2024] [Revised: 12/29/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Trauma healing is the process of healing after the body has been subjected to an external force and the skin and other tissues have become dissected or defective, showing the synergistic effect of various processes. Therefore, the investigation of innovative wound dressings has significant research and clinical implications. In this study, we constructed a zinc based metal-organic framework (MOF) and loaded with antimicrobial peptide LL37 to prepare LL37@ZPF-2 (ZPF = zeolite pyrimidine backbone), which was subsequently integrated with Poloxamer 407 to fabricate LL37@ZPF-2 thermosensitive hydrogel. Our study showed that in-situ packaging method can achieve encapsulation rate of 98 % and 15 % of drug loading for LL37. LL37@ZPF-2 demonstrated a higher inhibitory potency against S.aureus compared to E.coli. The Poloxamer 407-gel exhibits thermo-responsive sol-to-gel phase transition behaviors with a phase transition temperature (Tsol/gel) of ∼ 28.01℃, making it an appropriate material for wound healing. The composite hydrogel has excellent biocompatibility and hemocompatibility. A full-thickness skin defect model was built to confirm that LL37@ZPF-2 thermosensitive hydrogel dressing could inhibit bacterial growth, reduce the risk of wound infection, and stimulate angiogenesis and collagen deposition, resulting in a wound healing rate of 94.4 % on day 7 and complete healing on day 10. Our findings demonstrate that the novel thermosensitive LL37@ZPF-2 hydrogel confers good antibacterial activity, promoting cell migration and infected-wound healing properties, providing a promising platform for wound healing.
Collapse
Affiliation(s)
- Zixuan Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yingxiao Sun
- Key Laboratory of Biopharmaceutical Preparation and Delivery, State Key Laboratory of Biochemical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Peijie Dong
- Key Laboratory of Biopharmaceutical Preparation and Delivery, State Key Laboratory of Biochemical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jinhu Wang
- Shandong Academy of Pharmaceutical Sciences, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong Engineering Research Center of Percutaneous Drug Delivery Systems, Jinan 250101, China
| | - Lijie Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Aili Zhao
- Shandong Academy of Pharmaceutical Sciences, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong Engineering Research Center of Percutaneous Drug Delivery Systems, Jinan 250101, China
| | - Guangmin Qu
- Shandong Academy of Pharmaceutical Sciences, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong Engineering Research Center of Percutaneous Drug Delivery Systems, Jinan 250101, China
| | - Hang Li
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | | | - Wen Zhang
- Shandong Academy of Pharmaceutical Sciences, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong Engineering Research Center of Percutaneous Drug Delivery Systems, Jinan 250101, China.
| | - Yao Chen
- Shandong Academy of Pharmaceutical Sciences, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong Engineering Research Center of Percutaneous Drug Delivery Systems, Jinan 250101, China; Key Laboratory of Biopharmaceutical Preparation and Delivery, State Key Laboratory of Biochemical Engineering, Chinese Academy of Sciences, Beijing 100190, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University Tianjin 300071 (China) and Haihe Laboratory of Synthetic Biology, Tianjin 300308, China.
| | - Xin Meng
- Shandong Academy of Pharmaceutical Sciences, Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, Shandong Engineering Research Center of Percutaneous Drug Delivery Systems, Jinan 250101, China; Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
10
|
Fan H, Xue B, Lu J, Sun T, Zhao Q, Liu Y, Niu M, Yu S, Yang Y, Zhang L. Recent advances of bioaerogels in medicine: Preparation, property and application. Int J Biol Macromol 2025; 291:139144. [PMID: 39722377 DOI: 10.1016/j.ijbiomac.2024.139144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/14/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Bioaerogels represent a type of three-dimensional porous materials fabricated from natural biopolymers, and show a significant potential for medical application due to their characteristics of extremely low density, high specific surface area, excellent biocompatibility and biodegradability. The preparation method and parameters of bioaerogels are focused on, and their influence on the structure and properties of bioaerogels are discussed in detail. Then, to match the properties of bioaerogels with the medical applications, this work emphasizes the main properties (including biocompatibility, degradability, and mechanical properties), structural parameters (such as suitable porosity, pore size and high specific surface area), and further summarizes the influence of single-component and composite bioaerogels on their properties. Moreover, according to the different applications (wound healing, drug delivery, and tissue engineering and other fields), the function method, mechanism and practical effect of bioaerogels are comprehensively analyzed. Finally, the challenges, future research directions, and solutions for the practical application of bioaerogels in medicine are discussed.
Collapse
Affiliation(s)
- Haoyong Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Baoxia Xue
- College of Textile Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jiaxin Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Tao Sun
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Qinke Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yong Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Mei Niu
- College of Textile Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Shiping Yu
- Department of Interventional Therapy, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China
| | - Yongzhen Yang
- Key Laboratory of Interface Science and Engineering in Advanced Materials, Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China.
| | - Li Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China.
| |
Collapse
|
11
|
Farzan M, Soleimannejad M, Shariat S, Heidari Sureshjani M, Gholipour A, Ashrafi Dehkordi K, Alerasoul Dehkordi SMR, Farzan M. A biomimetic injectable chitosan/alginate hydrogel biocopmosites encapsulating selenium- folic acid nanoparticles for regeneration of spinal cord injury: An in vitro study. Int J Biol Macromol 2025; 288:138682. [PMID: 39672404 DOI: 10.1016/j.ijbiomac.2024.138682] [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: 03/17/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
Spinal cord injury (SCI) poses significant challenges to regenerative medicine due to its limited self-repair capabilities. In this study, we engineered a biomimetic injectable hydrogel using modified chitosan and alginate biopolymers encapsulating selenium-folic acid nanoparticles (Se-FA NPs) to facilitate SCI regeneration. The hydrogel exhibited a unique porous structure attributed to the incorporation of nanofiber fragments, enhancing its biocompatibility and bioactivity. Through a series of in vitro evaluations, including cell viability assays, proliferation studies, gene expression analysis, we assessed the hydrogel's cytocompatibility and its potential for supporting neural cell growth. Our results demonstrate the promising efficacy of the hydrogel in providing a conducive microenvironment for neural tissue regeneration. Moreover, the sustained release of Se-FA NPs from the hydrogel system offers neuroprotective, antioxidative, and anti-inflammatory benefits crucial for SCI therapy. Overall, our biomimetic hydrogel biocomposites hold great potential as a therapeutic strategy for promoting spinal cord regeneration, highlighting their significance in advancing the field of regenerative medicine.
Collapse
Affiliation(s)
- Mahan Farzan
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mostafa Soleimannejad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Saeedeh Shariat
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Heidari Sureshjani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Abolfazl Gholipour
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Korosh Ashrafi Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Mahour Farzan
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
12
|
Mechesso AF, Zhang W, Su Y, Xie J, Wang G. Segment-Based Peptide Design Reveals the Importance of N-Terminal High Cationicity for Antimicrobial Activity Against Gram-Negative Pathogens. Probiotics Antimicrob Proteins 2025; 17:15-34. [PMID: 39377976 DOI: 10.1007/s12602-024-10376-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 02/19/2025]
Abstract
Host defense antimicrobial peptides (AMPs) are recognized candidates to develop a new generation of peptide antibiotics. While high hydrophobicity can be deployed in peptides for eliminating Gram-positive bacteria, high cationicity is usually observed in AMPs against Gram-negative pathogen. This study investigates how the sequence distribution of basic amino acids affects peptide activity. For this purpose, we utilized human cathelicidin LL-37 as a template and designed four highly selective ultrashort peptides with similar length, net charge, and hydrophobic content. LL-10 + , RK-9 + , KR-8 + , and RIK-10 + showed similar activity against methicillin-resistant Staphylococcus aureus in vitro and comparable antibiofilm efficacy in a murine wound model. However, these peptides showed clear activity differences against Gram-negative pathogens with RIK-10 + (i.e., LL-37mini2) being the strongest and LL-10 + the weakest. To understand this activity difference, we characterized peptide toxicity; the effects of salts, pH, and serum on peptide activity; and the mechanism of action and determined the membrane-bound helical structure for RIK-10 + by two-dimensional NMR spectroscopy. By writing an R program, we generated charge density plots for these peptides and uncovered the importance of the N-terminal high-density basic charges for antimicrobial potency. To validate this finding, we reversed the sequences of two peptides. Interestingly, sequence reversal weakened the activity of RIK-10 + but increased the activity of LL-10 + especially against Escherichia coli, Pseudomonas aeruginosa, and Acinetobacter baumannii. Those more active peptides with high cationicity at the N-terminus are also more hydrophobic based on HPLC retention times. A database search found numerous natural sequences that arrange basic amino acids primarily at the N-terminus. Combined, this study not only obtained novel peptide leads but also discovered one useful strategy for designing novel antimicrobials to control drug-resistant Gram-negative pathogens.
Collapse
Affiliation(s)
- Abraham Fikru Mechesso
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Weiwei Zhang
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Guangshun Wang
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
| |
Collapse
|
13
|
Ma D, Su Y, Sharma NS, Hatcher G, Ganguli-Indra G, Indra AK, Gombart AF, Xie J. Prolonged Immunomodulator Delivery Boosts Monocyte Exosome Secretion and Elevates Cathelicidin/LL-37 Content. ACS APPLIED MATERIALS & INTERFACES 2025; 17:799-810. [PMID: 39700070 DOI: 10.1021/acsami.4c20695] [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: 12/21/2024]
Abstract
Human cathelicidin LL-37 offers significant benefits to the immune system and in treating various diseases, but its therapeutic potential is hindered by low activity and instability in physiological environments. Here, we introduce a strategy to boost LL-37 levels in exosomes derived from THP-1 monocytes by incubating cells with electrospun nanofibers containing immunomodulators (e.g., 1α, 25-dihydroxyvitamin D3 and VID400). Notably, the incubation with immunomodulator-loaded nanofibers not only increased LL-37 content in exosomes but also significantly enhanced the production of engineered exosomes. Moreover, these engineered exosomes demonstrated multiple biological activities, including promoting skin cell proliferation and migration, enhancing endothelial cell tube formation, and exhibiting antibacterial properties. Collectively, this study presents an approach to increasing both the yield of engineered exosomes and their LL-37 content, potentially offering a promising therapeutic option for wound healing, tissue regeneration, and infectious disease treatment.
Collapse
Affiliation(s)
- Dezun Ma
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Navatha Shree Sharma
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Grant Hatcher
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon 97331, United States
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon 97331, United States
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97239, United States
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - Adrian F Gombart
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331, United States
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
14
|
Li M, Zhao P, Wang J, Zhang X, Li J. Functional antimicrobial peptide-loaded 3D scaffolds for infected bone defect treatment with AI and multidimensional printing. MATERIALS HORIZONS 2025; 12:20-36. [PMID: 39484845 DOI: 10.1039/d4mh01124d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infection is the most prevalent complication of fractures, particularly in open fractures, and often leads to severe consequences. The emergence of bacterial resistance has significantly exacerbated the burden of infection in clinical practice, making infection control a significant treatment challenge for infectious bone defects. The implantation of a structural stent is necessary to treat large bone defects despite the increased risk of infection. Therefore, there is a need for the development of novel antibacterial therapies. The advancement in antibacterial biomaterials and new antimicrobial drugs offers fresh perspectives on antibacterial treatment. Although antimicrobial 3D scaffolds are currently under intense research focus, relying solely on material properties or antibiotic action remains insufficient. Antimicrobial peptides (AMPs) are one of the most promising new antibacterial therapy approaches. This review discusses the underlying mechanisms behind infectious bone defects and presents research findings on antimicrobial peptides, specifically emphasizing their mechanisms and optimization strategies. We also explore the potential prospects of utilizing antimicrobial peptides in treating infectious bone defects. Furthermore, we propose that artificial intelligence (AI) algorithms can be utilized for predicting the pharmacokinetic properties of AMPs, including absorption, distribution, metabolism, and excretion, and by combining information from genomics, proteomics, metabolomics, and clinical studies with computational models driven by machine learning algorithms, scientists can gain a comprehensive understanding of AMPs' mechanisms of action, therapeutic potential, and optimizing treatment strategies tailored to individual patients, and through interdisciplinary collaborations between computer scientists, biologists, and clinicians, the full potential of AI in accelerating the discovery and development of novel AMPs will be realized. Besides, with the continuous advancements in 3D/4D/5D/6D technology and its integration into bone scaffold materials, we anticipate remarkable progress in the field of regenerative medicine. This review summarizes relevant research on the optimal future for the treatment of infectious bone defects, provides guidance for future novel treatment strategies combining multi-dimensional printing with new antimicrobial agents, and provides a novel and effective solution to the current challenges in the field of bone regeneration.
Collapse
Affiliation(s)
- Mengmeng Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Peizhang Zhao
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jingwen Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xincai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
15
|
Zhao Y, Zhang J, Zhang G, Huang H, Tan WS, Cai H. Injectable Nanocomposite Hydrogel with Synergistic Biofilm Eradication and Enhanced Re-epithelialization for Accelerated Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:69086-69102. [PMID: 39635909 DOI: 10.1021/acsami.4c17855] [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: 12/07/2024]
Abstract
Diabetic wounds remain a critical clinical challenge due to their harsh microenvironment, which impairs cellular function, hinders re-epithelialization and tissue remodeling, and slows healing. Injectable nanocomposite hydrogel dressings offer a promising strategy for diabetic wound repair. In this study, we developed an injectable nanocomposite hydrogel dressing (HDL@W379) using LAP@W379 nanoparticles and an injectable hyaluronic acid-based hydrogel (HA-ADH-ODEX). This dressing provided a sustained, pH-responsive release of W379 antimicrobial peptides, effectively regulating the wound microenvironment to enhance healing. The HDL@W379 hydrogel featured multifunctional properties, including mechanical stability, injectability, self-healing, biocompatibility, and tissue adhesion. In vitro, the HDL@W379 hydrogel achieved synergistic biofilm elimination and subsequent activation of basal cell migration and endothelial cell tube formation. Pathway analysis indicated that the HDL@W379 hydrogel enhances basal cell migration through MEK/ERK pathway activation. In methicillin-resistant Staphylococcus aureus (MRSA)-infected diabetic wounds, the HDL@W379 hydrogel accelerated wound healing by inhibiting bacterial proliferation and promoting re-epithelialization, regenerating the granulation tissue, enhancing collagen deposition, and facilitating angiogenesis. Overall, this strategy of biofilm elimination and basal cell activation to continuously regulate the diabetic wound microenvironment offers an innovative approach to treating chronic wounds.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Jingwei Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Guofeng Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Huimin Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| |
Collapse
|
16
|
Vaseghi A, Sadeghizadeh M, Herb M, Grumme D, Demidov Y, Remmler T, Maleki HH. 3D Printing of Biocompatible and Antibacterial Silica-Silk-Chitosan-Based Hybrid Aerogel Scaffolds Loaded with Propolis. ACS APPLIED BIO MATERIALS 2024; 7:7917-7935. [PMID: 39360961 DOI: 10.1021/acsabm.4c00697] [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: 10/05/2024]
Abstract
The aim of this study is to design a therapeutic enhanced three-dimensional (3D) silk fibroin (SF)-based scaffold containing propolis (Ps)-loaded chitosan (CH) nanocarriers. To this aim, we initially synthesized a hybrid gel-based ink by a synergistic sol-gel and self-assembly approach and then processed the resulting gels by microextrusion-based 3D printing followed by supercritical drying to obtain 3D hybrid aerogel scaffolds. Ps was utilized to enhance the final scaffold's bactericidal efficacy and cell responsiveness. For the synthesis of the scaffold, two Ps loading methods (in preprint and postprinting steps) were investigated in order to optimize the Ps drug quantities in the scaffold and maximize the antibacterial properties of scaffold. In the postprinting Ps loading step, the hybrid silica-oxidized SF (SFO)-CH hydrogel ink was 3D printed into a construct with an interconnected porous structure, and then, Ps was loaded into the printed construct. In the preprint loading method, PS was incorporated into the SF and a hydrolyzed silane solution prior to gelation. The morphological studies demonstrate that the addition of Ps encapsulated CH nanoparticles (NPs) into the hydrogel solution improved the porosity of the developed scaffolds. The rheological analysis of the designed gel ink with and without Ps loading and the release kinetics were studied. The antimicrobial results show that the Ps-loaded scaffolds in the postprinting step exhibited superior antibacterial activity against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) strains compared to a preprinted Ps-loaded scaffold. Direct and indirect in vitro cytotoxicity tests also confirmed the designed Ps-loaded scaffold biocompatibility toward a mouse fibroblast (L929) cell line. We demonstrated that the scaffold formulated by propolis-loaded chitosan NPs can enhance the migration and proliferation of L929 fibroblast cells. The obtained results prove the promise of the designed 3D printed silica-SFO-CH-Ps scaffolds as a potent 3D scaffold to mediate tissue regeneration but also as an antibacterial highly porous matrix to support wound healing.
Collapse
Affiliation(s)
- Akbar Vaseghi
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, 14115 Tehran, Iran
- Department of Chemistry, Institute of Inorganic Chemistry, University of Cologne, Greinstrasse 6, 50939 Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC Research Center, 50931 Cologne, Germany
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, 14115 Tehran, Iran
| | - Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50935 Cologne, Germany
| | - Daniela Grumme
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50935 Cologne, Germany
| | - Yan Demidov
- Department of Chemistry, Institute of Inorganic Chemistry, University of Cologne, Greinstrasse 6, 50939 Cologne, Germany
| | | | - Hajar Homa Maleki
- Department of Chemistry, Institute of Inorganic Chemistry, University of Cologne, Greinstrasse 6, 50939 Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC Research Center, 50931 Cologne, Germany
| |
Collapse
|
17
|
Wu Y, Hu C, Li Y, Wang Y, Gong H, Zheng C, Kong QQ, Yang L, Wang Y. A Versatile Composite Hydrogel with Spatiotemporal Drug Delivery of Mesoporous ZnO and Recombinant Human Collagen for Diabetic Infected Wound Healing. Biomacromolecules 2024; 25:7878-7893. [PMID: 39570390 DOI: 10.1021/acs.biomac.4c01155] [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: 11/22/2024]
Abstract
Diabetic wounds are increasingly common and challenging to treat due to high infection risks in a high-glucose environment. Effective treatment requires wound dressings that combat infections, while promoting angiogenesis and skin regeneration. This study presents a hydrogel-based drug delivery system made from cellulose designed to accelerate diabetic wound healing by eliminating bacterial infections. The hydrogel, formed by linking phenylboronic acid-grafted oxidized methylcellulose (POMC) with poly(vinyl alcohol) (PVA), exhibits self-healing and injectable properties. It is further enhanced by adding type I recombinant human collagen (rhCOL1) to stimulate cell growth and angiogenesis and mesoporous zinc oxide (mZnO) for antibacterial and anti-inflammatory effects. Upon application, the hydrogel degrades under pH/ROS stimuli, releasing mZnO and rhCOL1 in a controlled manner that matches the wound healing stages. In vivo tests show that the hydrogel effectively eliminates bacteria, reduces inflammation, and promotes rapid skin regeneration, making it a promising solution for treating diabetic wounds.
Collapse
Affiliation(s)
- Ye Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yaxing Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Med-X Center for Manufacturing, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yu Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Heng Gong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Cheng Zheng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Qing-Quan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
18
|
Dai X, Nie W, Shen H, Machens HG, Böker K, Taheri S, Lehmann W, Shen Y, Schilling AF. Electrospinning based biomaterials for biomimetic fabrication, bioactive protein delivery and wound regenerative repair. Regen Biomater 2024; 12:rbae139. [PMID: 39803356 PMCID: PMC11723536 DOI: 10.1093/rb/rbae139] [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: 07/28/2024] [Revised: 10/25/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Electrospinning is a remarkably straightforward and adaptable technique that can be employed to process an array of synthetic and natural materials, resulting in the production of nanoscale fibers. It has emerged as a novel technique for biomedical applications and has gained increasing popularity in the research community in recent times. In the context of tissue repair and tissue engineering, there is a growing tendency toward the integration of biomimetic scaffolds and bioactive macromolecules, particularly proteins and growth factors. The design of 'smart' systems provides not merely physical support, but also microenvironmental cues that can guide regenerative tissue repair. Electrospun nanofibrous matrices are regarded as a highly promising tool in this area, as they can serve as both an extracellular matrix (ECM)-mimicking scaffold and a vehicle for the delivery of bioactive proteins. Their highly porous architecture and high surface-to-volume ratio facilitate the loading of drugs and mass transfer. By employing a judicious selection of materials and processing techniques, there is considerable flexibility in efficiently customizing nanofiber architecture and incorporating bioactive proteins. This article presents a review of the strategies employed for the structural modification and protein delivery of electrospun nanofibrous materials, with a focus on the objective of achieving a tailored tissue response. The article goes on to discuss the challenges currently facing the field and to suggest future research directions.
Collapse
Affiliation(s)
- Xinyi Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wei Nie
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, NC 27103, USA
| | - Hua Shen
- Department of Plastic and Reconstructive Surgery, Shanghai First People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, Faculty of Medicine, Technical University of Munich, Munich 81675, Germany
| | - Kai Böker
- Department of Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Göttingen, University of Göttingen, Göttingen 37075, Germany
| | - Shahed Taheri
- Department of Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Göttingen, University of Göttingen, Göttingen 37075, Germany
| | - Wolfgang Lehmann
- Department of Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Göttingen, University of Göttingen, Göttingen 37075, Germany
| | - Yi Shen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Arndt F Schilling
- Department of Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Göttingen, University of Göttingen, Göttingen 37075, Germany
| |
Collapse
|
19
|
Ning X, Wang R, Liu N, You Y, Wang Y, Wang J, Wang Y, Chen Z, Zhao H, Wu T. Three-dimensional structured PLCL/ADM bioactive aerogel for rapid repair of full-thickness skin defects. Biomater Sci 2024. [PMID: 39526449 DOI: 10.1039/d4bm01214c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The failure to treat deep skin wounds can result in significant complications, and the limitations of current clinical treatments highlight the pressing need for the development of new deep wound healing materials. In this study, a series of three-dimensional structured PLCL/ADM composite aerogels were fabricated by electrospinning and subsequently characterized for their microstructure, compression mechanics, exudate absorption, and hemostatic properties. Additionally, the growth of HSFs and HUVECs, which are involved in wound repair, was observed in the aerogels. The composite aerogel was subsequently employed in wound repair experiments on rat full-thickness skin with the objective of observing the wound healing rate and examining histological utilizing H&E, Masson, CD31, and COL-I staining. The findings indicated that the PLCL/ADM composite aerogel with a 10% concentration exhibited uniform pore size distribution, a good three-dimensional structure, and compression properties comparable to those of human skin, which could effectively absorb exudate and exert hemostatic effect. In vivo experiment results demonstrated that the aerogel exhibited superior efficacy to conventional oil-gauze overlay therapy and ADM aerogel in promoting wound healing and could facilitate rapid, high-quality in situ repair of deep wounds, thereby offering a novel approach for skin tissue engineering and clinical wound treatment.
Collapse
Affiliation(s)
- Xuchao Ning
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China.
- Department of Plastic Surgery, Qilu Hospital (Qingdao), Shandong University, Qingdao 266035, China.
| | - Runjia Wang
- School of Stomatology, Shandong University, Jinan 250012, China
| | - Na Liu
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yong You
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China.
| | - Yawen Wang
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, College of Textile & Clothing, Qingdao University, Qingdao 266071, China
| | - Jing Wang
- Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, College of Textile & Clothing, Qingdao University, Qingdao 266071, China
| | - Yuanfei Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China
| | - Zhenyu Chen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China.
| | - Haiguang Zhao
- Department of Plastic Surgery, Qilu Hospital (Qingdao), Shandong University, Qingdao 266035, China.
| | - Tong Wu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, China.
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
- Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, College of Textile & Clothing, Qingdao University, Qingdao 266071, China
| |
Collapse
|
20
|
Zhu Z, Zhou S, Li S, Gong S, Zhang Q. Neutrophil extracellular traps in wound healing. Trends Pharmacol Sci 2024; 45:1033-1045. [PMID: 39419742 DOI: 10.1016/j.tips.2024.09.007] [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: 07/19/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
Wound healing is a complex and orchestrated process that involves hemostasis, inflammation, proliferation, and tissue remodeling. Neutrophil extracellular traps (NETs) are intricate web-like structures released by neutrophils, comprising decondensed chromatin, myeloperoxidase (MPO), and neutrophil elastase (NE), which play vital roles in regulating neutrophil-mediated immune regulation. While NETs contribute to wound healing, excessive activation induced by dysregulated inflammation can hinder the healing process. Understanding the pivotal role of NETs in wound healing and tissue remodeling, as well as their intricate interactions within the wound microenvironment, presents opportunities for innovative wound healing strategies. In this review we discuss the process of NET formation, explore the interactions between NETs and skin cells, and examine therapeutic strategies targeting NETs and drug delivery platforms to accelerate wound healing. Additionally, we discuss current clinical investigations and research challenges towards advancing wound care practices.
Collapse
Affiliation(s)
- Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Shengzhi Zhou
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Sicheng Li
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan 430030, Hubei Province, People's Republic of China.
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China.
| |
Collapse
|
21
|
Kamaraj M, Moghimi N, McCarthy A, Chen J, Cao S, Chethikkattuveli Salih AR, Joshi A, Jucaud V, Panayi A, Shin SR, Noshadi I, Khademhosseini A, Xie J, John JV. Granular Porous Nanofibrous Microspheres Enhance Cellular Infiltration for Diabetic Wound Healing. ACS NANO 2024; 18:28335-28348. [PMID: 39356827 DOI: 10.1021/acsnano.4c10044] [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] [Indexed: 10/04/2024]
Abstract
Diabetic foot ulcers (DFUs) are a significant challenge in the clinical care of diabetic patients, often necessitating limb amputation and compromising the quality of life and life expectancy of this cohort. Minimally invasive therapies, such as modular scaffolds, are at the forefront of current DFU treatment, offering an efficient approach for administering therapeutics that accelerate tissue repair and regeneration. In this study, we report a facile method for fabricating granular nanofibrous microspheres (NMs) with predesigned structures and porosities. The proposed technology combines electrospinning and electrospraying to develop a therapeutic option for DFUs. Specifically, porous NMs were constructed using electrospun poly(lactic-co-glycolic acid) (PLGA):gelatin short nanofibers, followed by gelatin cross-linking. These NMs demonstrated enhanced cell adhesion to human dermal fibroblasts (HDF) during an in vitro cytocompatibility assessment. Notably, porous NMs displayed superior performance owing to their interconnected pores compared to nonporous NMs. Cell-laden NMs demonstrated higher Young's modulus values than NMs without loaded cells, suggesting improved material resiliency attributed to the reinforcement of cells and their secreted extracellular matrix. Dynamic injection studies on cell-laden NMs further elucidated their capacity to safeguard loaded cells under pressure. In addition, porous NMs promoted host cell infiltration, neovascularization, and re-epithelialization in a diabetic mouse wound model, signifying their effectiveness in healing diabetic wounds. Taken together, porous NMs hold significant potential as minimally invasive, injectable treatments that effectively promote tissue integration and regeneration.
Collapse
Affiliation(s)
- Meenakshi Kamaraj
- Terasaki Institute for Biomedical Innovations, Los Angeles, California 91367, United States
| | - Nafiseh Moghimi
- Terasaki Institute for Biomedical Innovations, Los Angeles, California 91367, United States
| | - Alec McCarthy
- Department of Surgery-Transplant and Mary & Dick Holland, Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Junjie Chen
- Terasaki Institute for Biomedical Innovations, Los Angeles, California 91367, United States
| | - Selena Cao
- Terasaki Institute for Biomedical Innovations, Los Angeles, California 91367, United States
| | | | - Akshat Joshi
- Terasaki Institute for Biomedical Innovations, Los Angeles, California 91367, United States
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovations, Los Angeles, California 91367, United States
| | - Adriana Panayi
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Heidelberg 69117, Germany
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Iman Noshadi
- Department of Bioengineering, University of California, Riverside, California 92521, United States
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovations, Los Angeles, California 91367, United States
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland, Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Johnson V John
- Terasaki Institute for Biomedical Innovations, Los Angeles, California 91367, United States
| |
Collapse
|
22
|
Li F, Zhang C, Zhong X, Li B, Zhang M, Li W, Zheng L, Zhu X, Chen S, Zhang Y. A 3D radially aligned nanofiber scaffold co-loaded with LL37 mimetic peptide and PDGF-BB for the management of infected chronic wounds. Mater Today Bio 2024; 28:101237. [PMID: 39315393 PMCID: PMC11419797 DOI: 10.1016/j.mtbio.2024.101237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 09/25/2024] Open
Abstract
Diabetic foot ulcers, pressure ulcers, and bedsores can easily develop into chronic wounds with bacterial infections, complicating wound healing. This work reports a two-step strategy for treating infected chronic wounds. Firstly, LL37 mimetic peptide-W379 peptides were rapidly released to eliminate the bacterial biofilm on the wound. Then, 3D radially aligned nanofiber scaffolds loaded with W379 antimicrobial peptide and PDGF-BB were used to treat the wound to prevent bacterial infection recurrence and promote angiogenesis and granulation tissue regeneration, thereby accelerating wound healing. In the presented study, we found that the combined use of burst and controlled release of W379 antimicrobial peptide effectively clears the bacterial biofilm and prevents the recurrence of bacterial infection. Additionally, we found that the removal of the bacterial biofilm contributed to modulating the local inflammatory response from a pro-inflammatory type to a pro-regenerative type. Furthermore, the use of PDGF-BB significantly promotes neovascularization and granulation tissue regeneration in the wound bed, resulting in accelerating re-epithelialization and wound closure. Our study provides a promising treatment method for the repair of infected chronic wounds.
Collapse
Affiliation(s)
- Fei Li
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Chuwei Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Xiaoping Zhong
- Department of Nursing, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510000, China
| | - Bo Li
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Mengnan Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Wanqian Li
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Lifei Zheng
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Xinghua Zhu
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Yi Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| |
Collapse
|
23
|
Lakshmaiah Narayana J, Mechesso AF, Rather IIG, Zarena D, Luo J, Xie J, Wang G. Origami of KR-12 Designed Antimicrobial Peptides and Their Potential Applications. Antibiotics (Basel) 2024; 13:816. [PMID: 39334990 PMCID: PMC11429261 DOI: 10.3390/antibiotics13090816] [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: 07/31/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
This review describes the discovery, structure, activity, engineered constructs, and applications of KR-12, the smallest antibacterial peptide of human cathelicidin LL-37, the production of which can be induced under sunlight or by vitamin D. It is a moonlighting peptide that shows both antimicrobial and immune-regulatory effects. Compared to LL-37, KR-12 is extremely appealing due to its small size, lack of toxicity, and narrow-spectrum antimicrobial activity. Consequently, various KR-12 peptides have been engineered to tune peptide activity and stability via amino acid substitution, end capping, hybridization, conjugation, sidechain stapling, and backbone macrocyclization. We also mention recently discovered peptides KR-8 and RIK-10 that are shorter than KR-12. Nano-formulation provides an avenue to targeted delivery, controlled release, and increased bioavailability. In addition, KR-12 has been covalently immobilized on biomaterials/medical implants to prevent biofilm formation. These constructs with enhanced potency and stability are demonstrated to eradicate drug-resistant pathogens, disrupt preformed biofilms, neutralize endotoxins, and regulate host immune responses. Also highlighted are the safety and efficacy of these peptides in various topical and systemic animal models. Finaly, we summarize the achievements and discuss future developments of KR-12 peptides as cosmetic preservatives, novel antibiotics, anti-inflammatory peptides, and microbiota-restoring agents.
Collapse
Affiliation(s)
- Jayaram Lakshmaiah Narayana
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore 560078, India
| | - Abraham Fikru Mechesso
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Imran Ibni Gani Rather
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| | - D Zarena
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
- College of Engineering, Jawaharlal Nehru Technological University, Anantapur 515002, India
| | - Jinghui Luo
- Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Guangshun Wang
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
24
|
Shi S, Hu L, Hu D, Ou X, Huang Y. Emerging Nanotherapeutic Approaches for Diabetic Wound Healing. Int J Nanomedicine 2024; 19:8815-8830. [PMID: 39220193 PMCID: PMC11365536 DOI: 10.2147/ijn.s476006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Diabetic wounds pose a significant challenge in modern healthcare due to their chronic and complex nature, often resulting in delayed healing, infections, and, in severe cases, amputations. In recent years, nanotherapeutic approaches have emerged as promising strategies to address the unique pathophysiological characteristics of diabetic wounds. This review paper provides a comprehensive overview of the latest advancements in nanotherapeutics for diabetic wound treatment. We discuss various nanomaterials and delivery systems employed in these emerging therapies. Furthermore, we explore the integration of biomaterials to enhance the efficacy of nanotherapeutic interventions. By examining the current state-of-the-art research, challenges, and prospects, this review aims to offer valuable insights for researchers, clinicians, and healthcare professionals working in the field of diabetic wound care.
Collapse
Affiliation(s)
- Shaoyan Shi
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, 710000, People’s Republic of China
| | - Leiming Hu
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, 710000, People’s Republic of China
| | - Dong Hu
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, 710000, People’s Republic of China
| | - Xuehai Ou
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, 710000, People’s Republic of China
| | - Yansheng Huang
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, 710000, People’s Republic of China
| |
Collapse
|
25
|
Guo Y, Zhang C, Xie B, Xu W, Rao Z, Zhou P, Ma X, Chen J, Cai R, Tao G, He Y. Multifunctional Microneedle Patch Based on Metal-Phenolic Network with Photothermal Antimicrobial, ROS Scavenging, Immunomodulatory, and Angiogenesis for Programmed Treatment of Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:33205-33222. [PMID: 38915205 DOI: 10.1021/acsami.4c07091] [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: 06/26/2024]
Abstract
In diabetic patients with skin injuries, bacterial proliferation, accumulation of reactive oxygen species (ROS) in the tissues, and impaired angiogenesis make wound healing difficult. Therefore, eliminating bacteria, removing ROS, and promoting angiogenesis are necessary for treating acute diabetic wounds. In this study, benefiting from the ability of polyphenols to form a metal-phenolic network (MPN) with metal ions, TA-Eu MPN nanoparticles (TM NPs) were synthesized. The prepared photothermal agent CuS NPs and TM NPs were then loaded onto the supporting base and needle tips of PVA/HA (PH) microneedles, respectively, to obtain PH/CuS/TM microneedles. Antibacterial experiments showed that microneedles loaded with CuS NPs could remove bacteria by the photothermal effect. In vitro experiments showed that the microneedles could effectively scavenge ROS, inhibit macrophage polarization to the M1 type, and induce polarization to the M2 type as well as have the ability to promote vascular endothelial cell migration and angiogenesis. Furthermore, in vivo experiments showed that PH/CuS/TM microneedles accelerated wound healing by inhibiting pro-inflammatory cytokines and promoting angiogenesis in a diabetic rat wound model. Therefore, PH/CuS/TM microneedles have efficient antibacterial, ROS scavenging, anti-inflammatory, immunomodulatory, and angiogenic abilities and hold promise as wound dressings for treating acute diabetic wounds.
Collapse
Affiliation(s)
- Ye Guo
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Chuankai Zhang
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Bingqing Xie
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Wei Xu
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Zihan Rao
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Peirong Zhou
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xuemin Ma
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
| | - Junliang Chen
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Rui Cai
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou 646000, China
| | - Gang Tao
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou 646000, China
| | - Yun He
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
- Institute of Stomatology, Southwest Medical University, Luzhou 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
26
|
Jeong Y, Patel R, Patel M. Biopolymer-Based Biomimetic Aerogel for Biomedical Applications. Biomimetics (Basel) 2024; 9:397. [PMID: 39056838 PMCID: PMC11274548 DOI: 10.3390/biomimetics9070397] [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: 05/29/2024] [Revised: 06/22/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Aerogels are lightweight and highly porous materials that have been found to have great potential in biomedical research because of some of their unique properties, such as their high surface area, tunable porosity, and biocompatibility. Researchers have been exploring ways to use aerogels to create biomimetic scaffolds inspired by natural extracellular matrices (ECMs) for various biomedical applications. Aerogel scaffolds can serve as three-dimensional (3D) templates for cell growth and tissue regeneration, promoting wound healing and tissue repair. Additionally, aerogel-based scaffolds have great potential in controlled drug delivery systems, where their high surface area and porosity enable the efficient loading and release of therapeutic agents. In this review, we discuss biopolymer-based biomimetic aerogel scaffolds for tissue engineering, drug delivery, and biosensors. Finally, we also discuss the potential directions in the development of aerogel-based biomimetic scaffolds.
Collapse
Affiliation(s)
- Yuhan Jeong
- Bio-Convergence, Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdogwahak-ro, Yeonsugu, Incheon 21938, Republic of Korea
| | - Rajkumar Patel
- Energy & Environmental Science and Engineering (EESE), Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdogwahak-ro, Yeonsugu, Incheon 21938, Republic of Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| |
Collapse
|
27
|
Croitoru GA, Pîrvulescu DC, Niculescu AG, Rădulescu M, Grumezescu AM, Nicolae CL. Advancements in Aerogel Technology for Antimicrobial Therapy: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1110. [PMID: 38998715 PMCID: PMC11243751 DOI: 10.3390/nano14131110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024]
Abstract
This paper explores the latest advancements in aerogel technology for antimicrobial therapy, revealing their interesting capacity that could improve the current medical approaches for antimicrobial treatments. Aerogels are attractive matrices because they can have an antimicrobial effect on their own, but they can also provide efficient delivery of antimicrobial compounds. Their interesting properties, such as high porosity, ultra-lightweight, and large surface area, make them suitable for such applications. The fundamentals of aerogels and mechanisms of action are discussed. The paper also highlights aerogels' importance in addressing current pressing challenges related to infection management, like the limited drug delivery alternatives and growing resistance to antimicrobial agents. It also covers the potential applications of aerogels in antimicrobial therapy and their possible limitations.
Collapse
Affiliation(s)
- George-Alexandru Croitoru
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.-A.C.); (C.-L.N.)
| | - Diana-Cristina Pîrvulescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (D.-C.P.); (A.-G.N.); (A.M.G.)
| | - Adelina-Gabriela Niculescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (D.-C.P.); (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Marius Rădulescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (D.-C.P.); (A.-G.N.); (A.M.G.)
| | - Alexandru Mihai Grumezescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (D.-C.P.); (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Carmen-Larisa Nicolae
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.-A.C.); (C.-L.N.)
| |
Collapse
|
28
|
Xiao L, Liu H, Huang H, Wu S, Xue L, Geng Z, Cai L, Yan F. 3D nanofiber scaffolds from 2D electrospun membranes boost cell penetration and positive host response for regenerative medicine. J Nanobiotechnology 2024; 22:322. [PMID: 38849858 PMCID: PMC11162076 DOI: 10.1186/s12951-024-02578-2] [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: 11/10/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
The ideal tissue engineering scaffold should facilitate rapid cell infiltration and provide an optimal immune microenvironment during interactions with the host. Electrospinning can produce two-dimensional (2D) membranes mimicking the extracellular matrix. However, their dense structure hinders cell penetration, and their thin form restricts scaffold utility. In this study, latticed hydrogels were three-dimensional (3D) printed onto electrospun membranes. This technique allowed for layer-by-layer assembly of the membranes into 3D scaffolds, which maintained their resilience impressively under both dry and wet conditions. We assessed the cellular and host responses of these 3D nanofiber scaffolds by comparing random membranes and mesh-like membranes with three different mesh sizes (250, 500, and 750 μm). It was found that scaffolds with a mesh size of 500 μm were superior for M2 macrophage phenotype polarization, vascularization, and matrix deposition. Furthermore, it was confirmed by subsequent experiments such as RNA sequencing that the mesh-like topology may promote polarization to the M2 phenotype by affecting the PI3K/AKT pathway. In conclusion, our work offers a novel method for transforming 2D nanofiber membranes into 3D scaffolds. This method boasts flexibility, allowing for the use of varied electrospun membranes and hydrogels in terms of structure and composition. It has vast potential in tissue repair and regeneration.
Collapse
Affiliation(s)
- Lingfei Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Huifan Liu
- Department of Anesthesiology, Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Huayi Huang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shujuan Wu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Longjian Xue
- The Institute of Technological Science, School of Power and Mechanical Engineering, Wuhan University, Wuhan, 430072, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Feifei Yan
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
29
|
Ghelich P, Samandari M, Hassani Najafabadi A, Tanguay A, Quint J, Menon N, Ghanbariamin D, Saeedinejad F, Alipanah F, Chidambaram R, Krawetz R, Nuutila K, Toro S, Barnum L, Jay GD, Schmidt TA, Tamayol A. Dissolvable Immunomodulatory Microneedles for Treatment of Skin Wounds. Adv Healthc Mater 2024; 13:e2302836. [PMID: 38299437 DOI: 10.1002/adhm.202302836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/21/2023] [Indexed: 02/02/2024]
Abstract
Sustained inflammation can halt or delay wound healing, and macrophages play a central role in wound healing. Inflammatory macrophages are responsible for the removal of pathogens, debris, and neutrophils, while anti-inflammatory macrophages stimulate various regenerative processes. Recombinant human Proteoglycan 4 (rhPRG4) is shown to modulate macrophage polarization and to prevent fibrosis and scarring in ear wound healing. Here, dissolvable microneedle arrays (MNAs) carrying rhPRG4 are engineered for the treatment of skin wounds. The in vitro experiments suggest that rhPRG4 modulates the inflammatory function of bone marrow-derived macrophages. Degradable and detachable microneedles are developed from gelatin methacryloyl (GelMA) attach to a dissolvable gelatin backing. The developed MNAs are able to deliver a high dose of rhPRG4 through the dissolution of the gelatin backing post-injury, while the GelMA microneedles sustain rhPRG4 bioavailability over the course of treatment. In vivo results in a murine model of full-thickness wounds with impaired healing confirm a decrease in inflammatory biomarkers such as TNF-α and IL-6, and an increase in angiogenesis and collagen deposition. Collectively, these results demonstrate rhPRG4-incorporating MNA is a promising platform in skin wound healing applications.
Collapse
Affiliation(s)
- Pejman Ghelich
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Alireza Hassani Najafabadi
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Adam Tanguay
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Nikhil Menon
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Delaram Ghanbariamin
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Farnoosh Saeedinejad
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Fatemeh Alipanah
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ramaswamy Chidambaram
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Roman Krawetz
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Surgery, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Kristo Nuutila
- US Army Institute of Surgical Research, Fort Sam Houston, Texas, 78234, USA
| | - Steven Toro
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Lindsay Barnum
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Gregory D Jay
- Emergency Medicine, Brown University, Providence, RI, 02908, USA
| | - Tannin A Schmidt
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| |
Collapse
|
30
|
Kumar M, Kumar D, Kumar D, Garg Y, Chopra S, Bhatia A. Therapeutic Potential of Nanocarrier Mediated Delivery of Peptides for Wound Healing: Current Status, Challenges and Future Prospective. AAPS PharmSciTech 2024; 25:108. [PMID: 38730090 DOI: 10.1208/s12249-024-02827-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Wound healing presents a complex physiological process that involves a sequence of events orchestrated by various cellular and molecular mechanisms. In recent years, there has been growing interest in leveraging nanomaterials and peptides to enhance wound healing outcomes. Nanocarriers offer unique properties such as high surface area-to-volume ratio, tunable physicochemical characteristics, and the ability to deliver therapeutic agents in a controlled manner. Similarly, peptides, with their diverse biological activities and low immunogenicity, hold great promise as therapeutics in wound healing applications. In this review, authors explore the potential of peptides as bioactive components in wound healing formulations, focusing on their antimicrobial, anti-inflammatory, and pro-regenerative properties. Despite the significant progress made in this field, several challenges remain, including the need for standardized characterization methods, optimization of biocompatibility and safety profiles, and translation from bench to bedside. Furthermore, developing multifunctional nanomaterial-peptide hybrid systems represents promising avenues for future research. Overall, the integration of nanomaterials made up of natural or synthetic polymers with peptide-based formulations holds tremendous therapeutic potential in advancing the field of wound healing and improving clinical outcomes for patients with acute and chronic wounds.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Dikshant Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Devesh Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Yogesh Garg
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Shruti Chopra
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India.
| |
Collapse
|
31
|
Su Y, Shahriar SSM, Andrabi SM, Wang C, Sharma NS, Xiao Y, Wong SL, Wang G, Xie J. It Takes Two to Tangle: Microneedle Patches Co-delivering Monoclonal Antibodies and Engineered Antimicrobial Peptides Effectively Eradicate Wound Biofilms. Macromol Biosci 2024; 24:e2300519. [PMID: 38217528 DOI: 10.1002/mabi.202300519] [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: 11/14/2023] [Revised: 12/20/2023] [Indexed: 01/15/2024]
Abstract
Wound biofilms pose a great clinical challenge. Herein, this work reports a dissolvable microneedle patch for dual delivery of monoclonal antibodies anti-PBP2a and engineers antimicrobial peptides W379. In vitro antibacterial efficacy testing with microneedle patches containing a combination of 250 ng mL-1 W379 and 250 ng mL-1 anti-BPB2a decreases the bacterial count from ≈3.31 × 107 CFU mL-1 to 1.28 × 102 CFU mL-1 within 2 h without eliciting evident cytotoxicity. Ex vivo testing indicates W379 and anti-PBP2a co-loaded microneedle patch displayed a remarkable reduction of bacterial load by ≈7.18 log CFU after administered only once within 48 h. The bacterial count is significantly diminished compared to the treatment by either W379 or anti-PBP2a-loaded alone microneedle patches. When administered twice within 48 h, no bacteria are identified. Further in vivo study also reveals that after two treatments of W379 and anti-PBP2a co-loaded PVP microneedle patches within 48 h, the bacterial colonies are undetectable in a type II diabetic mouse wound biofilm model. Taken together, W379 and anti-PBP2a co-loaded PVP microneedle patches hold great promise in treating wound biofilms.
Collapse
Affiliation(s)
- Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shatil S M Shahriar
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Syed Muntazir Andrabi
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chenlong Wang
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Navatha Shree Sharma
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yizhu Xiao
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shannon L Wong
- Department of Surgery-Plastic Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
32
|
Xi L, Du J, Xue W, Shao K, Jiang X, Peng W, Li W, Huang S. Cathelicidin LL-37 promotes wound healing in diabetic mice by regulating TFEB-dependent autophagy. Peptides 2024; 175:171183. [PMID: 38423213 DOI: 10.1016/j.peptides.2024.171183] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Diabetic patients often experience impaired wound healing. Human cathelicidin LL-37 possesses various biological functions, such as anti-microbial, anti-inflammatory, and pro-wound healing activities. Autophagy has important effects on skin wound healing. However, little is known about whether LL-37 accelerates diabetic wound healing by regulating autophagy. In the study, we aimed to investigate the role of autophagy in LL-37-induced wound healing and uncover the underlying mechanisms involved. A full-thickness wound closure model was established in diabetic mice to evaluate the effects of LL-37 and an autophagy inhibitor (3-MA) on wound healing. The roles of LL-37 and 3-MA in regulating keratinocyte migration were assessed using transwell migration and wound healing assays. The activation of transcription factor EB (TFEB) was measured using western blotting and immunofluorescence (IF) assays of its nuclear translocation. The results showed that LL-37 treatment improved wound healing in diabetic mice, whereas these effects were reversed by 3-MA. In vitro, 3-MA decreased the effects of LL-37 on promoting HaCat keratinocyte migration in the presence of high glucose (HG). Mechanistically, LL-37 promoted TFEB activation and resulted in subsequent activation of autophagy, as evidenced by increased nuclear translocation of TFEB and increased expression of ATG5, ATG7, and beclin 1 (BECN1), whereas these changes were blocked by TFEB knockdown. As expected, TFEB knockdown damaged the effects of LL-37 on promoting keratinocyte migration. Collectively, these results suggest that LL-37 accelerates wound healing in diabetic mice by activating TFEB-dependent autophagy, providing new insights into the mechanism by which LL-37 promotes diabetic wound healing.
Collapse
Affiliation(s)
- Liuqing Xi
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Du
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Xue
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Kan Shao
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Jiang
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenfang Peng
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyi Li
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shan Huang
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
33
|
Kamaraj M, Moghimi N, Chen J, Morales R, Chen S, Khademhosseini A, John JV. New dimensions of electrospun nanofiber material designs for biotechnological uses. Trends Biotechnol 2024; 42:631-647. [PMID: 38158307 PMCID: PMC11065627 DOI: 10.1016/j.tibtech.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024]
Abstract
Electrospinning technology has garnered wide attention over the past few decades in various biomedical applications including drug delivery, cell therapy, and tissue engineering. This technology can create nanofibers with tunable fiber diameters and functionalities. However, the 2D membrane nature of the nanofibers, as well as the rigidity and low porosity of electrospun fibers, lower their efficacy in tissue repair and regeneration. Recently, new avenues have been explored to resolve the challenges associated with 2D electrospun nanofiber membranes. This review discusses recent trends in creating different electrospun nanofiber microstructures from 2D nanofiber membranes by using various post-processing methods, as well as their biotechnological applications.
Collapse
Affiliation(s)
- Meenakshi Kamaraj
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Nafiseh Moghimi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Junjie Chen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Ramon Morales
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Johnson V John
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| |
Collapse
|
34
|
Park SB, Yang Y, Bang SI, Kim TS, Cho D. AESIS-1, a Rheumatoid Arthritis Therapeutic Peptide, Accelerates Wound Healing by Promoting Fibroblast Migration in a CXCR2-Dependent Manner. Int J Mol Sci 2024; 25:3937. [PMID: 38612747 PMCID: PMC11012285 DOI: 10.3390/ijms25073937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
In patients with autoimmune disorders such as rheumatoid arthritis (RA), delayed wound healing is often observed. Timely and effective wound healing is a crucial determinant of a patient's quality of life, and novel materials for skin wound repair, such as bioactive peptides, are continuously being studied and developed. One such bioactive peptide, AESIS-1, has been studied for its well-established anti-rheumatoid arthritis properties. In this study, we attempted to use the anti-RA material AESIS-1 as a therapeutic wound-healing agent based on disease-modifying antirheumatic drugs (DMARDs), which can help restore prompt wound healing. The efficacy of AESIS-1 in wound healing was assessed using a full-thickness excision model in diabetic mice; this is a well-established model for studying chronic wound repair. Initial observations revealed that mice treated with AESIS-1 exhibited significantly advanced wound repair compared with the control group. In vitro studies revealed that AESIS-1 increased the migration activity of human dermal fibroblasts (HDFs) without affecting proliferative activity. Moreover, increased HDF cell migration is mediated by upregulating chemokine receptor expression, such as that of CXC chemokine receptor 2 (CXCR2). The upregulation of CXCR2 through AESIS-1 treatment enhanced the chemotactic reactivity to CXCR2 ligands, including CXC motif ligand 8 (CXCL8). AESIS-1 directly activates the ERK and p38 mitogen-activated protein kinase (MAPK) signaling cascades, which regulate the migration and expression of CXCR2 in fibroblasts. Our results suggest that the AESIS-1 peptide is a strong wound-healing substance that increases the movement of fibroblasts and the expression of CXCR2 by turning on the ERK and p38 MAPK signaling cascades.
Collapse
Affiliation(s)
- Seung Beom Park
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Anam-dong 5-ga, Seongbuk-gu, Seoul 02841, Republic of Korea;
| | - Yoolhee Yang
- Kine Sciences, 6F, 24, Eonju-ro85gil, Gangnam-gu, Seoul 06221, Republic of Korea; (Y.Y.); (D.C.)
| | - Sa Ik Bang
- Department of Plastic Surgery, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Gangnam-gu, Seoul 06351, Republic of Korea;
| | - Tae Sung Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Anam-dong 5-ga, Seongbuk-gu, Seoul 02841, Republic of Korea;
| | - Daeho Cho
- Kine Sciences, 6F, 24, Eonju-ro85gil, Gangnam-gu, Seoul 06221, Republic of Korea; (Y.Y.); (D.C.)
- Institute of Convergence Science, Korea University, Anam-dong 5-ga, Seongbuk-gu, Seoul 02841, Republic of Korea
| |
Collapse
|
35
|
Geyik F, Kaya S, Yılmaz DE, Demirci H, Akmayan İ, Özbek T, Acar S. Propolis-Loaded Poly(lactic- co-glycolic Acid) Nanofibers: An In Vitro Study. ACS OMEGA 2024; 9:14054-14062. [PMID: 38560001 PMCID: PMC10975591 DOI: 10.1021/acsomega.3c09492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 04/04/2024]
Abstract
Nanofibers have high potential through their high porosity, small pore sizes, lightweight materials, and their ability to mimic the extracellular matrix structure for use in the manufacture of wound dressings for wound treatment. In this study, poly(lactic-co-glycolic acid) (PLGA) nanofibers were produced by electrospinning. Propolis was loaded into the PLGA nanofibers by the dropping method. The average diameters and effects of propolis loading on the morphology of 37.5, 50, and 100% propolis-loaded PLGA nanofibers (PLGA-P37.5, PLGA-P50, and PLGA-P100) were evaluated by scanning electron microscopy (SEM). The successful loading of propolis into PLGA nanofibers was confirmed with Fourier transform infrared spectroscopy (FTIR) analysis. In vitro propolis release was examined at physiological pH. The antioxidant activity of propolis-loaded nanofibers was studied with 2,2-diphenyl-1-picrylhydrazyl (DPPH). Antimicrobial activities of the nanofibers against Escherichia coli, Staphylococcus aureus and Candida albicans strains were determined by the disk diffusion method. Consequently, PLGA-P50 and PLGA-P100 showed high antimicrobial activity on S. aureus and C. albicans. Cell viability was tested by 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay, and propolis-loaded PLGA nanofibers were found to be biocompatible with human fibroblast cells. In the wound scratch assay, propolis-loaded nanofibers supported wound closure with cell migration and proliferation. Thus, in vitro wound closure properties of propolis-loaded PLGA nanofibers were evaluated for the first time in the literature.
Collapse
Affiliation(s)
- Fulya Geyik
- Faculty
of Chemical and Metallurgical Engineering, Department of Bioengineering, Yildiz Technical University, Istanbul 34220, Turkey
| | - Seçil Kaya
- Faculty
of Chemical and Metallurgical Engineering, Department of Bioengineering, Yildiz Technical University, Istanbul 34220, Turkey
| | - Duygu Elif Yılmaz
- Department
of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Hasan Demirci
- Institute
of Functional Anatomy, Charité-Universitätsmedizin
Berlin, Berlin 10115, Germany
| | - İlkgül Akmayan
- Faculty
of Arts and Sciences, Department of Molecular Biology and Genetics, Yildiz Technical University, Istanbul 34220, Turkey
| | - Tülin Özbek
- Faculty
of Arts and Sciences, Department of Molecular Biology and Genetics, Yildiz Technical University, Istanbul 34220, Turkey
| | - Serap Acar
- Faculty
of Chemical and Metallurgical Engineering, Department of Bioengineering, Yildiz Technical University, Istanbul 34220, Turkey
| |
Collapse
|
36
|
Karan A, Sharma NS, Darder M, Su Y, Andrabi SM, Shahriar SMS, John JV, Luo Z, DeCoster MA, Zhang YS, Xie J. Copper-Cystine Biohybrid-Embedded Nanofiber Aerogels Show Antibacterial and Angiogenic Properties. ACS OMEGA 2024; 9:9765-9781. [PMID: 38434900 PMCID: PMC10905775 DOI: 10.1021/acsomega.3c10012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/15/2024] [Accepted: 02/01/2024] [Indexed: 03/05/2024]
Abstract
Copper-cystine-based high aspect ratio structures (CuHARS) possess exceptional physical and chemical properties and exhibit remarkable biodegradability in human physiological conditions. Extensive testing has confirmed the biocompatibility and biodegradability of CuHARS under diverse biological conditions, making them a viable source of essential Cu2+. These ions are vital for catalyzing the production of nitric oxide (NO) from the decomposition of S-nitrosothiols (RSNOs) found in human blood. The ability of CuHARS to act as a Cu2+ donor under specific concentrations has been demonstrated in this study, resulting in the generation of elevated levels of NO. Consequently, this dual function makes CuHARS effective as both a bactericidal agent and a promoter of angiogenesis. In vitro experiments have shown that CuHARS actively promotes the migration and formation of complete lumens by redirecting microvascular endothelial cells. To maximize the benefits of CuHARS, they have been incorporated into biomimetic electrospun poly(ε-caprolactone)/gelatin nanofiber aerogels. Through the regulated release of Cu2+ and NO production, these channeled aerogels not only provide antibacterial support but also promote angiogenesis. Taken together, the inclusion of CuHARS in biomimetic scaffolds could hold great promise in revolutionizing tissue regeneration and wound healing.
Collapse
Affiliation(s)
- Anik Karan
- Department
of Surgery-Transplant and Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Navatha Shree Sharma
- Department
of Surgery-Transplant and Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Margarita Darder
- Instituto
de Ciencia de Materiales de Madrid (ICMM), CSIC, Madrid 28049, Spain
| | - Yajuan Su
- Department
of Surgery-Transplant and Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Syed Muntazir Andrabi
- Department
of Surgery-Transplant and Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - S M Shatil Shahriar
- Department
of Surgery-Transplant and Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Johnson V. John
- Terasaki
Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Zeyu Luo
- Division
of Engineering in Medicine, Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Mark A. DeCoster
- Biomedical
Engineering, Louisiana Tech University, Ruston, Louisiana 71272, United States
- Institute
for Micromanufacturing, Louisiana Tech University, Ruston, Louisiana 71272, United States
| | - Yu Shrike Zhang
- Division
of Engineering in Medicine, Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
| | - Jingwei Xie
- Department
of Surgery-Transplant and Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department
of Mechanical and Materials Engineering, University of Nebraska Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
37
|
Shahriar SMS, McCarthy AD, Andrabi SM, Su Y, Polavoram NS, John JV, Matis MP, Zhu W, Xie J. Mechanically resilient hybrid aerogels containing fibers of dual-scale sizes and knotty networks for tissue regeneration. Nat Commun 2024; 15:1080. [PMID: 38316777 PMCID: PMC10844217 DOI: 10.1038/s41467-024-45458-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: 09/24/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
The structure and design flexibility of aerogels make them promising for soft tissue engineering, though they tend to come with brittleness and low elasticity. While increasing crosslinking density may improve mechanics, it also imparts brittleness. In soft tissue engineering, resilience against mechanical loads from mobile tissues is paramount. We report a hybrid aerogel that consists of self-reinforcing networks of micro- and nanofibers. Nanofiber segments physically entangle microfiber pillars, allowing efficient stress distribution through the intertwined fiber networks. We show that optimized hybrid aerogels have high specific tensile moduli (~1961.3 MPa cm3 g-1) and fracture energies (~7448.8 J m-2), while exhibiting super-elastic properties with rapid shape recovery (~1.8 s). We demonstrate that these aerogels induce rapid tissue ingrowth, extracellular matrix deposition, and neovascularization after subcutaneous implants in rats. Furthermore, we can apply them for engineering soft tissues via minimally invasive procedures, and hybrid aerogels can extend their versatility to become magnetically responsive or electrically conductive, enabling pressure sensing and actuation.
Collapse
Affiliation(s)
- S M Shatil Shahriar
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Alec D McCarthy
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Syed Muntazir Andrabi
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Navatha Shree Polavoram
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Johnson V John
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mitchell P Matis
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Mechanical and Materials Engineering, University of Nebraska Lincoln, Lincoln, NE, 68588, USA.
| |
Collapse
|
38
|
Shahriar SMS, Polavoram NS, Andrabi SM, Su Y, Lee D, Tran HQ, Schindler SJ, Xie J. Transforming layered 2D mats into multiphasic 3D nanofiber scaffolds with tailored gradient features for tissue regeneration. BMEMAT 2024; 2:e12065. [PMID: 38586163 PMCID: PMC10997325 DOI: 10.1002/bmm2.12065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/27/2023] [Indexed: 04/09/2024]
Abstract
Multiphasic scaffolds with tailored gradient features hold significant promise for tissue regeneration applications. Herein, this work reports the transformation of two-dimensional (2D) layered fiber mats into three dimensional (3D) multiphasic scaffolds using a 'solids-of-revolution' inspired gas-foaming expansion technology. These scaffolds feature precise control over fiber alignment, pore size, and regional structure. Manipulating nanofiber mat layers and Pluronic F127 concentrations allows further customization of pore size and fiber alignment within different scaffold regions. The cellular response to multiphasic scaffolds demonstrates the number of cells migrated and proliferated onto the scaffolds are mainly dependent on the pore size rather than fiber alignment. In vivo subcutaneous implantation of multiphasic scaffolds to rats reveals substantial cell infiltration, neo tissue formation, collagen deposition, and new vessel formation within scaffolds, greatly surpassing the capabilities of traditional nanofiber mats. Histological examination indicates the importance of optimizing pore size and fiber alignment for promotion of cell infiltration and tissue regeneration. Overall, these scaffolds have potential applications in tissue modeling, studying tissue-tissue interactions, interface tissue engineering, and high-throughput screening for optimized tissue regeneration.
Collapse
Affiliation(s)
- S. M. Shatil Shahriar
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Navatha Shree Polavoram
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Syed Muntazir Andrabi
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yajuan Su
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Donghee Lee
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Huy Quang Tran
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Samantha J. Schindler
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jingwei Xie
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
39
|
Xu K, Sun X, Chong C, Ren L, Tan L, Sun H, Wang X, Li L, Xia J, Zhang R, Wang L. Green Starch-Based Hydrogels with Excellent Injectability, Self-Healing, Adhesion, Photothermal Effect, and Antibacterial Activity for Promoting Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:2027-2040. [PMID: 38183285 DOI: 10.1021/acsami.3c13551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2024]
Abstract
Hydrogel materials have proven valuable in wound healing, but improving the safety of these hydrogels is still challenging. Therefore, designing multifunctional natural polymeric-based hydrogels with excellent mechanical properties to replace toxic or potentially risky, refractory chemical polymer-based hydrogels such as polyacrylamide and polyethylene glycol is of particular significance. Here, a green starch-based hydrogel (Starch@Ca/CGC hydrogel) with injectability, self-healing, and instant adhesion was constructed by coordination interaction, electrostatic interaction, and intramolecular and intermolecular hydrogen bonds. Therein, natural bioactive small molecules gallic acid (GA) and carvacrol (CA) were coordinated with metal ions by the ultrasonic-triggered self-assembly and ionic cross-linking codriven strategy to prepare Cu-gallic acid-carvacrol nanospheres (CGC NPs), which conferred the hydrogel with near-infrared light (NIR)-controlled CA release and photothermal synergistic sterilization properties, as well as antioxidant and anti-infection capabilities. More importantly, the multifunctional hydrogel platforms could completely cover an irregular wound shape to prevent secondary injury and significantly accelerate wound healing under NIR with more skin appendages like hair follicles and blood vessels appearing. Therefore, it is expected that this starch-based hydrogel could serve as a competitive multifunctional dressing in the biomedical field, including bacteria-derived wound infection and other tissue repair.
Collapse
Affiliation(s)
- Ke Xu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Xinyu Sun
- College of Food Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Chuanyu Chong
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Li Ren
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Lili Tan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Haonan Sun
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Lihua Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Junfang Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Ruoxi Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Li Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, School of Chemistry, South China Normal University, Guangzhou, Guangdong 511441, P. R. China
| |
Collapse
|
40
|
Wang X, Li R, Zhao H. Enhancing angiogenesis: Innovative drug delivery systems to facilitate diabetic wound healing. Biomed Pharmacother 2024; 170:116035. [PMID: 38113622 DOI: 10.1016/j.biopha.2023.116035] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
Diabetic wounds (DW) constitute a substantial burden on global healthcare owing to their widespread occurrence as a complication of diabetes. Angiogenesis, a crucial process, plays a pivotal role in tissue recovery by supplying essential oxygen and nutrients to the injury site. Unfortunately, in diabetes mellitus, various factors disrupt angiogenesis, hindering wound healing. While biomaterials designed to enhance angiogenesis hold promise for the treatment of DWs, there is an urgent need for more in-depth investigations to fully unlock their potential in clinical management. In this review, we explore the intricate mechanisms of angiogenesis that are crucial for DW recovery. We introduce a rational design for angiogenesis-enhancing drug delivery systems (DDS) and provide a comprehensive summary and discussion of diverse biomaterials that enhance angiogenesis for facilitating DW healing. Lastly, we address emerging challenges and prospects in angiogenesis-enhancing DDS for facilitating DW healing, aiming to offer a comprehensive understanding of this critical healthcare issue and potential solutions.
Collapse
Affiliation(s)
- Xuan Wang
- Department of foot and ankle surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Runmin Li
- Department of foot and ankle surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China
| | - Hongmou Zhao
- Department of foot and ankle surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an 710054, China.
| |
Collapse
|
41
|
Liang C, Wang H, Lin Z, Zhang C, Liu G, Hu Y. Augmented wound healing potential of photosensitive GelMA hydrogel incorporating antimicrobial peptides and MXene nanoparticles. Front Bioeng Biotechnol 2023; 11:1310349. [PMID: 38179129 PMCID: PMC10764632 DOI: 10.3389/fbioe.2023.1310349] [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: 10/09/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction: Wound healing is a delicate and complex process influenced by many factors. The treatment of skin wounds commonly involves the use of wound dressings, which remain a routine approach. An ideal dressing can provide protection and a suitable environment for wound surfaces by maintaining moisture and exhibiting good biocompatibility, mechanical strength, and antibacterial properties to promote healing and prevent infection. Methods: We encapsulated tick-derived antibacterial polypeptides (Os) as a model drug within a methylacrylyl gelatin (GelMA) hydrogel containing MXene nanoparticles. The prepared composite hydrogels were evaluated for their wound dressing potential by analyzing surface morphology, mechanical properties, swelling behavior, degradation properties, antibacterial activity, and cytocompatibility. Results: The results demonstrated excellent mechanical strength, swelling performance, degradation behavior, and antibacterial activity of the prepared composite hydrogels, effectively promoting cell growth, adhesion, and expression of antibacterial peptide activity. A full-thickness rat wound model then observed the wound healing process and surface interactions between the composite hydrogels and wounds. The composite hydrogel significantly accelerated wound closure, reduced inflammation, and sped epithelial formation and maturation. Discussion: Incorporating antibacterial peptides into GelMA provides a feasible strategy for developing excellent antibacterial wound dressings capable of tissue repair. In conclusion, this study presents a GelMA-based approach for designing antibacterial dressings with strong tissue regenerative ability.
Collapse
Affiliation(s)
- Chengzhi Liang
- Department of Orthopaedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Hongyu Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhihao Lin
- Department of Orthopaedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chengdong Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guoming Liu
- Department of Orthopaedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanling Hu
- Department of Orthopaedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
42
|
Freitas CG, Felipe MS. Candida albicans and Antifungal Peptides. Infect Dis Ther 2023; 12:2631-2648. [PMID: 37940816 PMCID: PMC10746669 DOI: 10.1007/s40121-023-00889-9] [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: 09/15/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023] Open
Abstract
Candida albicans, a ubiquitous opportunistic fungal pathogen, plays a pivotal role in human health and disease. As a commensal organism, it normally resides harmlessly within the human microbiota. However, under certain conditions, C. albicans can transition into a pathogenic state, leading to various infections collectively known as candidiasis. With the increasing prevalence of immunocompromised individuals and the widespread use of invasive medical procedures, candidiasis has become a significant public health concern. The emergence of drug-resistant strains further complicates treatment options, highlighting the urgent need for alternative therapeutic strategies. Antifungal peptides (AFPs) have gained considerable attention as potential candidates for combating Candida spp. infections. These naturally occurring peptides possess broad-spectrum antimicrobial activity, including specific efficacy against C. albicans. AFPs exhibit several advantageous properties, such as rapid killing kinetics, low propensity for resistance development, and diverse mechanisms of action, making them promising alternatives to conventional antifungal agents. In recent years, extensive research has focused on discovering and developing novel AFPs with improved efficacy and selectivity against Candida species. Advances in biotechnology and synthetic peptide design have enabled the modification and optimization of natural peptides, enhancing their stability, bioavailability, and therapeutic potential. Nevertheless, several challenges must be addressed before AFPs can be widely implemented in clinical practice. These include optimizing peptide stability, enhancing delivery methods, overcoming potential toxicity concerns, and conducting comprehensive preclinical and clinical studies. This commentary presents a short overview of candidemia and AFP; articles and reviews published in the last 10 years were searched on The National Library of Medicine (National Center for Biotechnology Information-NIH-PubMed). The terms used were C. albicans infections, antimicrobial peptides, antifungal peptides, antifungal peptides mechanisms of action, candidemia treatments and guidelines, synthetic peptides and their challenges, and antimicrobial peptides in clinical trials as the main ones. Older publications were cited if they brought some relevant concept or helped to bring a perspective into our narrative. Articles older than 20 years and those that appeared in PubMed but did not match our goal to bring updated information about using antifungal peptides as an alternative to C. albicans infections were not considered.
Collapse
Affiliation(s)
- Camila G Freitas
- Higher Education Course in Food Technology, Instituto Federal de Brasília (IFB), Brasília, DF, Brazil
- Genomic Sciences and Biotechnology Graduate Program, Universidade Católica de Brasília (UCB), Brasília, DF, Brazil
| | - Maria Sueli Felipe
- Genomic Sciences and Biotechnology Graduate Program, Universidade Católica de Brasília (UCB), Brasília, DF, Brazil.
- Universidade de Brasília (UNB), Brasília, DF, Brazil.
| |
Collapse
|
43
|
Tan D, Zhu W, Liu L, Pan Y, Xu Y, Huang Q, Li L, Rao L. In situ formed scaffold with royal jelly-derived extracellular vesicles for wound healing. Theranostics 2023; 13:2811-2824. [PMID: 37284440 PMCID: PMC10240823 DOI: 10.7150/thno.84665] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 04/25/2023] [Indexed: 06/08/2023] Open
Abstract
Background: Safe and effective wound healing can be a major clinical challenge. Inflammation and vascular impairment are two main causes of inadequate wound healing. Methods: Here, we developed a versatile hydrogel wound dressing, comprising a straightforward physical mixture of royal jelly-derived extracellular vesicles (RJ-EVs) and methacrylic anhydride modified sericin (SerMA), to accelerate wound healing by inhibiting inflammation and promoting vascular reparation. Results: The RJ-EVs showed satisfactory anti-inflammatory and antioxidant effects, and significantly promoted L929 cell proliferation and migration in vitro. Meanwhile, the photocrosslinked SerMA hydrogel with its porous interior structure and high fluidity made it a good candidate for wound dressing. The RJ-EVs can be gradually released from the SerMA hydrogel at the wound site, ensuring the restorative effect of RJ-EVs. In a full-thickness skin defect model, the SerMA/RJ-EVs hydrogel dressing accelerated wound healing with a healing rate of 96.8% by improving cell proliferation and angiogenesis. The RNA sequencing results further revealed that the SerMA/RJ-EVs hydrogel dressing was involved in inflammatory damage repair-related pathways including recombinational repair, epidermis development, and Wnt signaling. Conclusion: This SerMA/RJ-EVs hydrogel dressing offers a simple, safe and robust strategy for modulating inflammation and vascular impairment for accelerated wound healing.
Collapse
Affiliation(s)
- Dehong Tan
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wenxiang Zhu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
- College of Materials Science and Engineering, Hunan University, Changsha 410082, China
| | - Lujie Liu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yuanwei Pan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yangtao Xu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
- Cancer Center and Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qinqin Huang
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Lingling Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lang Rao
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
44
|
Hao Z, Liu G, Ren L, Liu J, Liu C, Yang T, Wu X, Zhang X, Yang L, Xia J, Li W. A Self-Healing Multifunctional Hydrogel System Accelerates Diabetic Wound Healing through Orchestrating Immunoinflammatory Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:19847-19862. [PMID: 37042619 DOI: 10.1021/acsami.2c23323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Developing an effective treatment strategy of drug delivery to improve diabetic wound healing remains a major challenge in clinical practice nowadays, due to multidrug-resistant bacterial infections, angiopathy, and oxidative damage in the wound microenvironment. Herein, an effective and convenient strategy was designed through a self-healing multiple-dynamic-bond cross-linked hydrogel with interpenetrating networks, which was formed by multiple-dynamic-bond cross-linking of reversible catechol-Fe3+ coordinate bonds, hydrogen bonding, and Schiff base bonds. The excellent autonomous healing of the hydrogel was initiated and accelerated by Schiff bonds with reversible breakage between 3,4-dihydroxybenzaldehyde containing catechol and aldehyde groups and chitosan chains, and further consolidated by the co-optation of other noncovalent interactions contributed of hydrogen bonding and Fe3+ coordinate bonds. Intriguingly, cathelicidin LL-37 was introduced and uniformly dispersed in the dynamic interpenetrating networks of the hydrogel as a bioactive molecular to orchestrate the diabetic wound healing microenvironment. This multifunctional wound dressing can significantly promote diabetic wound healing by antibacterial activity, immunomodulation, anti-inflammation, neovascularization, and antioxidant activity. Therefore, this study provided an effective and safe strategy for guiding the diabetic wound treatment in clinical applications.
Collapse
Affiliation(s)
- Zhichao Hao
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Gen Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Lin Ren
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Jiangchen Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Chuanzi Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Tao Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Xiangnan Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Xinchun Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Ling Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Juan Xia
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| | - Weichang Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510050, China
| |
Collapse
|
45
|
Jang EJ, Patel R, Patel M. Electrospinning Nanofibers as a Dressing to Treat Diabetic Wounds. Pharmaceutics 2023; 15:pharmaceutics15041144. [PMID: 37111630 PMCID: PMC10142830 DOI: 10.3390/pharmaceutics15041144] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/01/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023] Open
Abstract
Globally, diabetic mellitus (DM) is a common metabolic disease that effectively inhibits insulin production, destroys pancreatic β cells, and consequently, promotes hyperglycemia. This disease causes complications, including slowed wound healing, risk of infection in wound areas, and development of chronic wounds all of which are significant sources of mortality. With an increasing number of people diagnosed with DM, the current method of wound healing does not meet the needs of patients with diabetes. The lack of antibacterial ability and the inability to sustainably deliver necessary factors to wound areas limit its use. To overcome this, a new method of creating wound dressings for diabetic patients was developed using an electrospinning methodology. The nanofiber membrane mimics the extracellular matrix with its unique structure and functionality, owing to which it can store and deliver active substances that greatly aid in diabetic wound healing. In this review, we discuss several polymers used to create nanofiber membranes and their effectiveness in the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Eun Jo Jang
- Nano Science and Engineering, Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Rajkumar Patel
- Energy & Environmental Science and Engineering (EESE), Integrated Science and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21938, Republic of Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| |
Collapse
|
46
|
Su Y, Andrabi SM, Shahriar SMS, Wong SL, Wang G, Xie J. Triggered release of antimicrobial peptide from microneedle patches for treatment of wound biofilms. J Control Release 2023; 356:131-141. [PMID: 36858263 PMCID: PMC10073311 DOI: 10.1016/j.jconrel.2023.02.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/01/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Biofilms pose a great challenge for wound management. Herein, this study describes a near-infrared (NIR) light-responsive microneedle patch for on-demand release of antimicrobial peptide for treatment of wound biofilms. IR780 iodide as a photothermal conversion agent and molecularly engineered peptide W379 as an antimicrobial agent are loaded in dissolvable poly(vinylpyrrolidone) (PVP) microneedle patches followed by coating with a phase change material 1-tetradecanol (TD). After placing in an aqueous solution or biofilm containing wounds ex vivo and in vivo, upon exposure to NIR light, the incorporated IR780 induces light-to-heat conversion, causing the melting of TD. This leads to the dissolution of PVP microneedles, enabling the release of loaded W379 peptide from the microneedles into surrounding regions (e.g., solution, biofilm, wound bed). Compared with traditional microneedle patches, NIR light responsive microneedle patches can program the release of antimicrobial peptide and show high antibacterial efficacy in vitro. Meanwhile, this work indicates that NIR light responsive TD-coated, W379-loaded PVP microneedle patches show excellent antibiofilm activities ex vivo and in vivo. Additionally, this microneedle system could be a promising platform for delivering other antimicrobial agents.
Collapse
Affiliation(s)
- Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Syed Muntazir Andrabi
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - S M Shatil Shahriar
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Shannon L Wong
- Department of Surgery-Plastic Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States; Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska Lincoln, Lincoln, NE 68588, United States.
| |
Collapse
|