151
|
Mgwenya TN, Abrahamse H, Houreld NN. Photobiomodulation studies on diabetic wound healing: An insight into the inflammatory pathway in diabetic wound healing. Wound Repair Regen 2025; 33:e13239. [PMID: 39610015 PMCID: PMC11628774 DOI: 10.1111/wrr.13239] [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: 04/03/2024] [Revised: 08/06/2024] [Accepted: 09/02/2024] [Indexed: 11/30/2024]
Abstract
Diabetes mellitus remains a global challenge to public health as it results in non-healing chronic ulcers of the lower limb. These wounds are challenging to heal, and despite the different treatments available to improve healing, there is still a high rate of failure and relapse, often necessitating amputation. Chronic diabetic ulcers do not follow an orderly progression through the wound healing process and are associated with a persistent inflammatory state characterised by the accumulation of pro-inflammatory macrophages, cytokines and proteases. Photobiomodulation has been successfully utilised in diabetic wound healing and involves illuminating wounds at specific wavelengths using predominantly light-emitting diodes or lasers. Photobiomodulation induces wound healing through diminishing inflammation and oxidative stress, among others. Research into the application of photobiomodulation for wound healing is current and ongoing and has drawn the attention of many researchers in the healthcare sector. This review focuses on the inflammatory pathway in diabetic wound healing and the influence photobiomodulation has on this pathway using different wavelengths.
Collapse
Affiliation(s)
- Tintswalo N. Mgwenya
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| | - Nicolette N. Houreld
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgJohannesburgGautengSouth Africa
| |
Collapse
|
152
|
Yan R, Wang Y, Li W, Sun J. Promotion of chronic wound healing by plant-derived active ingredients and research progress and potential of plant polysaccharide hydrogels. CHINESE HERBAL MEDICINES 2025; 17:70-83. [PMID: 39949811 PMCID: PMC11814255 DOI: 10.1016/j.chmed.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/30/2024] [Accepted: 11/19/2024] [Indexed: 02/16/2025] Open
Abstract
Wound healing is a complex biochemical process. The use of herbal medicine in wound healing not only carries forward the wisdom of traditional medicine, with its anti-inflammatory and immune-regulating effects, but also reflects the direction of modern biopharmaceutical technology, such as its potential in developing new biomaterials like hydrogels. This article first outlines the inherent structural properties of healthy skin, along with the physiological characteristics related to chronic wounds in patients with diabetes and burns. Subsequently, the article delves into the latest advancements in clinical and experimental research on the impact of active constituents in herbal medicine on wound tissue regeneration, summarizing existing studies on the mechanisms of various herbal medicines in the healing of diabetic and burn wounds. Finally, the paper thoroughly examines the application and mechanisms of plant polysaccharide hydrogels containing active herbal compounds in chronic wound healing. The primary objective is to provide valuable resources for the clinical application and development of herbal medicine, thereby maximizing its therapeutic potential. It also represents the continuation of traditional medical wisdom, offering new possibilities for advancements in regenerative medicine and wound care.
Collapse
Affiliation(s)
- Ru Yan
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150006, China
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150006, China
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150006, China
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150006, China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150006, China
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150006, China
| | - Jialin Sun
- Department of Medicine, Heilongjiang Minzu College, Harbin 150066, China
- Postdoctoral Research Station, Heilongjiang University of Chinese Medicine, Harbin 150006, China
| |
Collapse
|
153
|
Li P, Li Y, Yao J, Li LL. Peptide-Induced Hydrogelation with Ordered Metal-Organic Framework Nanoparticles Generating Reactive Oxygen Species for Integrated Wound Repair. Adv Healthc Mater 2025; 14:e2403292. [PMID: 39639393 DOI: 10.1002/adhm.202403292] [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: 09/01/2024] [Revised: 11/21/2024] [Indexed: 12/07/2024]
Abstract
Hydrogels, with their high water content and flexible nature, are a promising class of medical dressings for combating bacterial wound infections. However, their development has been hindered by low sterilization efficiency. Here, this issue is addressed by designing a peptide hydrogel that assembles ordered metal-organic framework (MOF) nanoparticles with photocatalytic bactericidal activity. Specifically, a short peptide, Nap-Gly-Phe-Phe-His (Nap-GFFH), is used to induce the assembly of zinc-imidazolate MOF (ZIF-8) into a hydrogel (NHZ gel). This innovative structure integrates three key features: 1) ZIF-8 nanoparticles are encapsulated within the hydrogel, overcoming their inherent brittleness, insolubility, and limited moldability; 2) the ordered ZIF-8 structure enhances charge transfer, enabling efficient generation of reactive oxygen species (ROS); and 3) ZIF-8 simultaneously improves the photocatalytic bactericidal efficiency and mechanical properties of the hydrogel. The NHZ gel demonstrates remarkable antibacterial performance, achieving >99.9% and 99.99% inactivation of Escherichia coli and Staphylococcus aureus, respectively, within 15 min of simulated solar radiation. Additionally, the NHZ gel exhibits excellent biocompatibility, water retention, and exudate absorption, highlighting its broad potential for wound healing.
Collapse
Affiliation(s)
- Ping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
| | - Yiying Li
- Beijing Key Laboratory of Failure, Corrosion, and Protection of Oil/Gas Facilities, New Energy and Material College, China University of Petroleum-Beijing, Beijing, 102249, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
| | - Jiahui Yao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
| | - Li-Li Li
- School of Material Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
| |
Collapse
|
154
|
Zeng Z, Zhang J, Gao Y, Song Y, Liu L, Zhu M, Ma W, Fu J, Miao D, Huang C, Xiong R. Bioadhesive First-Aid Patch with Rapid Hemostasis and High Toughness Designed for Sutureless Sealing of Acute Bleeding Wounds. Adv Healthc Mater 2025; 14:e2403412. [PMID: 39520362 DOI: 10.1002/adhm.202403412] [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: 09/24/2024] [Revised: 10/21/2024] [Indexed: 11/16/2024]
Abstract
The global military and civilian sectors express widespread concern over the significant hemorrhage associated with various acute wounds. Such bleedings lead to numerous casualties in military confrontations, traffic accidents, and surgical injuries. Consequently, the rapid control of the bleedings, particularly for extensive and pressurized wounds, is crucial in first-aid situations. In this work, a double-layered bioadhesive patch that combines a superabsorbent adhesive hydrogel with a highly tough antibacterial polyurethane film, which is called as Bio-Patch, is proposed. The Bio-Patch demonstrates superior mechanical strength and forms robust bioadhesion to acute bleeding wounds. Furthermore, the Bio-Patch enables protecting against external Gram-negative and Gram-positive bacteria. Thanks to the double-layered structures having synergistic functions of stable barrier and robust adhesion, the Bio-Patch provides optimal wound sealing (burst strength exceeding 310 mmHg) both in vitro and in vivo. It also demonstrates superior hemostatic effects (less than 30 s) in vivo. This offers promising opportunities for rapid control of extensive and pressurized hemorrhage in first-aid clinical scenarios.
Collapse
Affiliation(s)
- Ziyuan Zeng
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Jiaming Zhang
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| | - Yige Gao
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| | - Yuanyuan Song
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| | - Luoming Liu
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| | - Miaomiao Zhu
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| | - Wenjing Ma
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| | - Jiajun Fu
- School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Dongyang Miao
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| | - Chaobo Huang
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| | - Ranhua Xiong
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, China
| |
Collapse
|
155
|
Yu Lin MO, Sampath D, Bosykh DA, Wang C, Wang X, Subramaniam T, Han W, Hong W, Chakraborty S. YAP/TAZ Drive Agrin-Matrix Metalloproteinase 12-Mediated Diabetic Skin Wound Healing. J Invest Dermatol 2025; 145:155-170.e2. [PMID: 38810954 DOI: 10.1016/j.jid.2024.05.005] [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: 09/05/2023] [Revised: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 05/31/2024]
Abstract
Macroscopic loss of extracellular matrix can lead to chronic defects in skin wound healing, but supplementation of extracellular matrix holds promise for facilitating wound closure, particularly in diabetic wound healing. We recently showed that the extracellular matrix proteoglycan agrin accelerates cutaneous wound healing by improving mechanoperception of migrating keratinocytes and allowing them to respond to mechanical stresses through matrix metalloproteinase 12 (MMP12). RNA-sequencing analysis revealed that in addition to a disorganized extracellular matrix, agrin-depleted skin cells have impaired YAP/TAZ transcriptional outcomes, leading us to hypothesize that YAP/TAZ, as central mechanosensors, drive the functionality of agrin-MMP12 signaling during cutaneous wound repair. In this study, we demonstrate that agrin activates YAP/TAZ during migration of keratinocytes after wounding in vitro and in vivo. Mechanistically, YAP/TAZ sustain agrin and MMP12 protein expression during migration after wounding through positive feedback. YAP/TAZ silencing abolishes agrin-MMP12-mediated force recognition and geometrical constraints. Importantly, soluble agrin therapy accelerates wound closure in diabetic mouse models by engaging MMP12-YAP. Because patients with diabetic foot ulcers and impaired wound healing have reduced expression of agrin-MMP12 that correlates with YAP/TAZ inactivation, we propose that timely activation of YAP/TAZ by soluble agrin therapy can accentuate mechanobiological microenvironments for efficient wound healing, under normal and diabetic conditions.
Collapse
Affiliation(s)
| | | | - Dmitriy A Bosykh
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Chengchun Wang
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Xiaomeng Wang
- Institute of Molecular and Cell Biology, Singapore, Singapore; Centre for Vision Research, Duke-NUS Medical School, Singapore, Singapore
| | - Tavintharan Subramaniam
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore; Division of Endocrinology, Department of Medicine, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Weiping Han
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Singapore, Singapore.
| | - Sayan Chakraborty
- Institute of Molecular and Cell Biology, Singapore, Singapore; Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA; Program of Developmental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA.
| |
Collapse
|
156
|
Ojeh N, Vecin NM, Pastar I, Volk SW, Wilgus T, Griffiths S, Ramey‐Ward AN, Driver VR, DiPietro LA, Gould LJ, Tomic‐Canic M. The Wound Reporting in Animal and Human Preclinical Studies (WRAHPS) Guidelines. Wound Repair Regen 2025; 33:e13232. [PMID: 39639458 PMCID: PMC11621255 DOI: 10.1111/wrr.13232] [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: 07/19/2024] [Revised: 10/02/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024]
Abstract
Preclinical studies for wound healing disorders are an essential step in translating discoveries into therapies. Also, they are an integral component of initial safety screening and gaining mechanistic insights using an in vivo approach. Given the complexity of the wound healing process, existing guidelines for animal testing do not capture key information due to the inevitable variability in experimental design. Variations in study interpretation are increased by complexities associated with wound aetiology, wounding procedure, multiple treatment conditions, wound assessment, and analysis, as well as lack of acknowledgement of limitation of the model used. Yet, no standards exist to guide reporting crucial experimental information required to interpret results in translational studies of wound healing. Consistency in reporting allows transparency, comparative, and meta-analysis studies and avoids repetition and redundancy. Therefore, there is a critical and unmet need to standardise reporting for preclinical wound studies. To aid in reporting experimental conditions, The Wound Reporting in Animal and Human Preclinical Studies (WRAHPS) Guidelines have now been created by the authors working with the Wound Care Collaborative Community (WCCC) GAPS group to provide a checklist and reporting template for the most frequently used preclinical models in support of development for human clinical trials for wound healing disorders. It is anticipated that the WRAHPS Guidelines will standardise comprehensive methods for reporting in scientific manuscripts and the wound healing field overall. This article is not intended to address regulatory requirements but is intended to provide general guidelines on important scientific considerations for such studies.
Collapse
Affiliation(s)
- Nkemcho Ojeh
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Preclinical and Health Sciences, Faculty of Medical SciencesThe University of the West IndiesBridgetownBarbados
| | - Nicole M. Vecin
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Susan W. Volk
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Traci Wilgus
- Department of PathologyThe Ohio State UniversityColumbusOhioUSA
| | | | | | - Vickie R. Driver
- School of MedicineWashington State UniversitySpokaneWashingtonUSA
| | - Luisa A. DiPietro
- Center for Wound Healing and Tissue RegenerationUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Lisa J. Gould
- South Shore Hospital Center for Wound HealingWeymouthMassachusettsUSA
| | - Marjana Tomic‐Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
157
|
Bayir Y, Erkayman B, Albayrak A, Palabiyik-Yücelik ŞS, Can S, Hanci H, Tunç F, Halici H, Civelek MS, Sevim M, Yurdgülü EE, Metin Ö. Boric acid and zinc borate doped graphene hydrogels designed for burn treatment: In vitro viability-biocompatibility tests and microbiological analysis. J Biomater Appl 2025; 39:592-606. [PMID: 39302915 PMCID: PMC11707965 DOI: 10.1177/08853282241268673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 09/22/2024]
Abstract
Boron, an essential element for human, can be a key factor in wound healing. For this reason, in this study, role of boron products (boric acid and zinc borate) and boron product doped new synthesized graphene hydrogels was investigated for burn healing via in vitro viability-biocompatibility tests and microbiological analysis. It has been determined that boric acid and zinc borate are effective against microbial agents that are frequently seen in burns. In L929 mouse fibroblast cell line, BA, ZB and graphene hydrogels did not show any toxic effects, either alone or doped Graphene Hydrogel forms, except at very high doses. These substances showed antioxidant properties by protecting cells against H2O2 damage. The migration test performed on boron products also confirms the protective effect of boron products. In this study, the synthesis of graphene hydrogels was made for the first time, and their characterization was completed with appropriate instrumental analyses. The results of the biocompatibility tests of graphene hydrogels show that they are at least 96% biocompatible.
Collapse
Affiliation(s)
- Yasin Bayir
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Türkiye
| | - Beyzagül Erkayman
- Department of Pharmacology, Faculty of Pharmacy, Ataturk University, Erzurum, Türkiye
| | - Abdulmecit Albayrak
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Türkiye
| | | | - Sümeyra Can
- Department of Chemistry, Faculty of Science, Ataturk University, Erzurum, Türkiye
| | - Hayrunisa Hanci
- Department of Microbiology, Faculty of Pharmacy, Ataturk University, Erzurum, Türkiye
| | - Fatih Tunç
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Türkiye
| | - Hamza Halici
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Türkiye
| | - Maide Sena Civelek
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Türkiye
| | - Melike Sevim
- Department of Chemistry, Faculty of Science, Ataturk University, Erzurum, Türkiye
| | - Emir Enis Yurdgülü
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Türkiye
| | - Önder Metin
- Department of Chemistry, College of Sciences, Koc University, İstanbul, Türkiye
| |
Collapse
|
158
|
Wu Y, Wang Y, Li W, Li D, Song P, Kang Y, Han X, Wang X, Tian H, Rauf A, Yan J, Zhang H, Li X. Construction of piezoelectric, conductive and injectable hydrogels to promote wound healing through electrical stimulation. Acta Biomater 2025; 191:205-215. [PMID: 39577481 DOI: 10.1016/j.actbio.2024.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Piezoelectric, conductive, and injectable hydrogel (SPG hydrogel) is constructed to rapidly close wounds, efficiently harvest biomechanical energy from animal motion, and generate electrical stimulation for electrotherapy of wound healing. 3-amino-4-methoxybenzoic acid (AMB) monomer was polymerized and grafted onto the gelatin, which was further crosslinked using EDC/NHS and embedded with strontium titanate nanoparticles (80.5 wt%), forming SPG hydrogel. This SPG hydrogel had high tissue adhesion ability, and could generate the output voltage (maximum output voltage 1 V) and current (maximum output current 0.5 nA) upon mechanical bending, promoting NIH-3T3 cell migration and proliferation. Upon application to the mice wound model, the SPG hydrogel rapidly closed the skin wound, smoothed the wound's appearance, reduced the remaining wound size, and increased epidermal thickness, demonstrating remarkable wound healing capabilities. This study suggests that the body motion-promoted electrotherapy offers a promising strategy for wound healing. STATEMENT OF SIGNIFICANCE: Piezoelectric nanomaterials are often incorporated into hydrogels to create piezoelectric hydrogels for wound healing. However, piezoelectric nanomaterials tend to agglomerate within the hydrogel matrix, and the hydrogel's low conductivity hinders efficient electron transfer. Together, both factors significantly reduce the piezoelectric effect. In this study, we developed an SPG hydrogel to improve the homogeneity and conductivity of the piezoelectric hydrogel. We first designed a conductive PG hydrogel and then immoblized piezoelectric STO nanoparticles within its matrix through coordination chemistry. Upon mechanical deformation, the uniformly distributed STO nanoparticles can generate electricity, which can efficiently transfer through the conductive matrix to the hydrogel's surface. This design shows great potential for wound healing applications.
Collapse
Affiliation(s)
- Yunyun Wu
- School of Chemistry and Life Science, Changchun University of Technology, Changchun 130012, China; Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun 130022, China
| | - Yanjing Wang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Weili Li
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry Chinese Academy of Sciences, Changchun 130022, China
| | - Diyi Li
- School of Chemistry and Life Science, Changchun University of Technology, Changchun 130012, China
| | - Panpan Song
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Yaqing Kang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun 130022, China
| | - Xiaoqing Han
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun 130022, China
| | - Xinbo Wang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Hongkun Tian
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry Chinese Academy of Sciences, Changchun 130022, China
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Ambar, Khyber Pakhtunkhwa 23430, Pakistan
| | - Jiao Yan
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Haiyuan Zhang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Xi Li
- School of Chemistry and Life Science, Changchun University of Technology, Changchun 130012, China.
| |
Collapse
|
159
|
Tromp LE, van der Boon TA, de Hilster RH, Bank R, van Rijn P. Modulation of Biomaterial-Associated Fibrosis by Means of Combined Physicochemical Material Properties. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407531. [PMID: 39641386 PMCID: PMC11789587 DOI: 10.1002/advs.202407531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/19/2024] [Indexed: 12/07/2024]
Abstract
Biomaterial-associated fibrosis remains a significant challenge in medical implants. To optimize implant design, understanding the interplay between biomaterials and host cells during the foreign body response (FBR) is crucial. Material properties are known to influence cellular behavior and can be used to manipulate cell responses, but predicting the right combination for the desired outcomes is challenging. This study explores how combined physicochemical material properties impact early myofibroblast differentiation using the Biomaterial Advanced Cell Screening (BiomACS) technology, which assesses hundreds of combinations of surface topography, stiffness, and wettability in a single experiment. Normal human dermal fibroblasts (NHDFs) are screened for cell density, area, and myofibroblast markers α-smooth muscle actin (α-SMA) and Collagen type I (COL1) after 24 h and 7 days of culture, with or without transforming growth factor-beta (TGF-β). Results demonstrated that material properties influence fibroblast behavior after 7 days with TGF-β stimulation, with wettability emerging as the predominant factor, followed by stiffness. The study identified regions with increased cell adhesion while minimizing myofibroblast differentiation, offering the potential for implant surface optimization to prevent fibrosis. This research provides a powerful tool for cell-material studies and represents a critical step toward enhancing implant properties and reducing complications, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Lisa E. Tromp
- Department of Biomaterials and Biomedical TechnologyUniversity of Groningen, University Medical Center GroningenFB‐40, A. Deusinglaan 1Groningen9713 AVthe Netherlands
| | - Torben A.B. van der Boon
- Department of Biomaterials and Biomedical TechnologyUniversity of Groningen, University Medical Center GroningenFB‐40, A. Deusinglaan 1Groningen9713 AVthe Netherlands
| | - Roderick H.J. de Hilster
- Department of Biomaterials and Biomedical TechnologyUniversity of Groningen, University Medical Center GroningenFB‐40, A. Deusinglaan 1Groningen9713 AVthe Netherlands
| | - Ruud Bank
- Department of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenA. Deusinglaan 1Groningen9713 AVthe Netherlands
| | - Patrick van Rijn
- Department of Biomaterials and Biomedical TechnologyUniversity of Groningen, University Medical Center GroningenFB‐40, A. Deusinglaan 1Groningen9713 AVthe Netherlands
| |
Collapse
|
160
|
Lu Y, Huangfu S, Ma C, Ding Y, Zhang Y, Zhou C, Liao L, Li M, You J, Chen Y, Wang D, Chen A, Jiang B. Exosomes derived from umbilical cord mesenchymal stem cells promote healing of complex perianal fistulas in rats. Stem Cell Res Ther 2024; 15:414. [PMID: 39732731 DOI: 10.1186/s13287-024-04028-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/28/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Complex perianal fistulas, challenging to treat and prone to recurrence, often require surgical intervention that may cause fecal incontinence and lower quality of life due to large surgical wounds and potential sphincter damage. Human umbilical cord-derived MSCs (hUC-MSCs) and their exosomes (hUCMSCs-Exo) may promote wound healing. METHODS This study assessed the efficacy, mechanisms, and safety of these exosomes in treating complex perianal fistulas in SD rats. We established a rat model, divided rats with fistulas into the control and the exosome groups. We assessed treatment efficacy through ultrasound, clinical observations, and histopathological analysis. We also evaluated the activation of the HIF-1α/TGF-β/Smad signaling pathway via PCR and Western blot and assessed serological markers for HIF-1α and inflammatory indices through ELISA. We analyzed gut microbiota and the systemic metabolic environment via untargeted metabolomics. RESULTS The hUCMSCs-Exo effectively promoted healing of wound, regulated the immune balance enhanced collagen synthesis and angiogenesis in the perianal fistulas model of rats, and regulated the gut microbiota and metabolomic profiles. Results of PCR and Western blot analyses indicated that the exosomes activated HIF-1α/TGF-β/Smad signaling pathways. To the dosages tested, the 10ug/100ul concentration (medium dose) was found to be the most effective to the treatment of complex perianal fistulas. CONCLUSIONS The hUCMSCs-Exo significantly promoted the healing of wound in perianal fistulas of rats and demonstrated higher safety. The underlying mechanism facilitating the healing process was likely associated with the activation of the HIF-1α/TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Yafei Lu
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Shaohua Huangfu
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Chuanxue Ma
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yan Ding
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
- Department of Biobank, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Chungen Zhou
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Lianming Liao
- Center of Laboratory Medicine, Union Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Ming Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, People's Republic of China
| | - Jia You
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yuting Chen
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Dawei Wang
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Ao Chen
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Bin Jiang
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China.
| |
Collapse
|
161
|
Trimukhe AM, Melo JS, Chaturvedi D, Jain RD, Dandekar P, Deshmukh RR. RF pulsed plasma modified composite scaffold for enhanced anti-microbial activity and accelerated wound healing. Int J Pharm 2024; 667:124864. [PMID: 39461682 DOI: 10.1016/j.ijpharm.2024.124864] [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: 06/11/2024] [Revised: 09/15/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Infected wounds present significant challenges pertaining to healing and often demand administration of strong antibiotics to patients. Also, drug resistant microbes may alter the physiology of wounds to create biofilms, frequently leading to high morbidity and mortality. In this investigation, a biodegradable, microporous composite agarose-chitosan scaffold was fabricated. Furthermore, its surface was modified with diphenyldiselenide deposition, using low pressure pulsed plasma technology. The optimized plasma parameters, viz. 5ON/15OFF (ms) of plasma pulse rate and 80 min of treatment time resulted in scaffolds having enhanced anti-bacterial activity against gram positive microbes like Staphylococcus (S.) aureus and S. epidermidis. The scaffolds were non-toxic to skin cells, as confirmed by the MTT assay. Cell proliferation through plasma treated and untreated scaffolds was assessed by culturing primary human dermal fibroblasts (HdaF) and human keratinocytes (HaCaT) and visualizing via confocal microscopy. Moreover, in-vivo rat model confirmed accelerated wound healing with plasma treated scaffold (100 % on day 14), as compared to the untreated scaffold (100 % on day 16) when compared with over-the-counter (OTC) ointment Betadine (100 % on day 12).
Collapse
Affiliation(s)
- A M Trimukhe
- Department of Physics, Institute of Chemical Technology, Mumbai 400019, India
| | - J S Melo
- Enzyme Microbial Technology Section, Nuclear Agriculture and Biotechnology Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - D Chaturvedi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - R D Jain
- Department of Biological Sciences and Biotechnology, Institute of Chemical Technology, Mumbai 400019, India
| | - P Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - R R Deshmukh
- Department of Physics, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
162
|
Zhang Z, Yang D, Shen F, Xue TT, Jiang JS, Luo Y, Zhang Y, Song JK, Kuai L, Wang MX, Li B, Ru Y. Epidermal keratinocytes-specific PD-L1 knockout causes delayed healing of diabetic wounds. Int Immunopharmacol 2024; 143:113540. [PMID: 39510031 DOI: 10.1016/j.intimp.2024.113540] [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: 03/25/2024] [Revised: 09/14/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Diabetic ulcers (DUs) is a common complication of diabetes, for which the morbidity and mortality increasing annually worldwide. The deficiency of PD-L1 in keratinocytes (KCs) may be linked to the diabetic wound healing impediments. OBJECTIVE Our study utilized transgenic mice to assess the functions of epidermal KCs-specific PD-L1 in DUs treatment. METHODS AND RESULTS Epidermal KCs-specific PD-L1 knockouted mice demonstrate deteriorated healing rates, concomitant with exacerbated inflammatory infiltration and excessive angiogenesis. The streptozotocin-induced murine diabetes model was used to imitate DUs in-vivo context, and the delayed healing was found under diabetic conditions. We then generated transgenic mice overexpressing PD-L1 in the epidermis. PD-L1 overexpression accelerate the DUs healing process accompanied by a reduction in inflammatory infiltration and a corresponding decreasion of angiogenesis. Therefore, overexpression of PD-L1 accelerates the healing process of DUs. CONCLUSION In sum, epidermal KCs-specific PD-L1 plays vital roles in epidermis regeneration, inflammatory infiltration, and angiogenesis during DUs restoration, and would not be easily to format fibrous scar. Our study elucidated a new therapeutic idea for slow-healing wound care.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Dan Yang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Fang Shen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ting-Ting Xue
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jing-Si Jiang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ying Zhang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Jian-Kun Song
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ming-Xia Wang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China.
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China.
| | - Yi Ru
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China.
| |
Collapse
|
163
|
Zhen M, Zhu Y, Wang P, Liu X, Zhu J, Liu H, Li J, Zhao J, Shu B. HMGB1 Accelerates Wound Healing by Promoting the Differentiation of Epidermal Stem Cells via the "HMGB1-TLR4-Wnt/Notch" Axis. Adv Wound Care (New Rochelle) 2024. [PMID: 39694535 DOI: 10.1089/wound.2023.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
Objective: Impairments in the differentiation and migratory capacity of epidermal stem cells (ESCs) are pivotal factors contributing to delayed wound healing. High mobility group box1 (HMGB1) has recently emerged as a potential target for tissue repair. Therefore, we aimed to investigate the role and molecular mechanisms of HMGB1 in ESCs during the wound-healing process. Approach: Initially, we examined the expression of HMGB1 and the differentiation of ESCs in normal skin, normal wounds and chronic wounds. Then, we assessed the ESC migration and differentiation, and the key markers in the Wnt/Notch signaling pathways, after treatment of HMGB1 and inhibitor, and the knockdown of toll-like receptor 4 (TLR4), using scratch assay, qPCR, western blotting, and immunofluorescence. Finally, we conducted mice models to analyze the healing rates and quality in vivo. Results: HMGB1 was decreased across all epidermal layers, and the differentiation of ESCs was hindered in diabetic foot ulcer. In vitro, HMGB1 enhanced both the migration and differentiation of ESCs while stimulating the expression of the Wnt/Notch pathway within ESCs. However, the downregulation of TLR4 negated these effects. Finally, our in vivo experiments provided evidence that HMGB1 facilitates wound healing and epidermis differentiation via TLR4 and Wnt/Notch signaling pathways. Innovation: This study innovatively introduces HMGB1 as a novel target for skin wound healing and elucidates its mechanisms of action. Conclusions: HMGB1 accelerated wound healing by promoting the differentiation of epidermal stem cells through the "HMGB1-TLR4-Wnt/Notch" axis, which reveals a new potential mechanism and target to expedite wound healing.
Collapse
Affiliation(s)
- Miao Zhen
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yongkang Zhu
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Peng Wang
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaogang Liu
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Junyou Zhu
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hengdeng Liu
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingting Li
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingling Zhao
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Shu
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
164
|
Gong R, Yang D, Zhang C, Abbas G, Miao B, Liang Y, Xu J, Fang X, Ding H. NIR-II Light-Driven Multifunctional Nanozymes PS@CS for Efficient Therapy against Melanoma and Post-tumor Surgery Infection. NANO LETTERS 2024; 24:16200-16207. [PMID: 39642288 DOI: 10.1021/acs.nanolett.4c05389] [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: 12/08/2024]
Abstract
Melanoma, the most prevalent form of skin cancer, is primarily treated with surgical intervention. However, complete tumor cell removal is challenging, and surgical wounds are prone to infection, complicating treatment and increasing costs. The successful treatment of melanoma generally requires multifunctional agents that are coordinated in tumor therapy and wound healing. In this study, we developed platinum (Pt)- and selenium (Se)-based nanozymes, Pt-Se@Chitosan (PS@CS), which exhibit synergistic antitumor and bactericidal efficacy attributed to their multienzyme activity and strong photothermal conversion efficiency. Furthermore, we engineered PS@CS hydrogels capable of inhibiting tumor regrowth postsurgery and accelerating healing of infected wounds. The PS@CS and PS@CS hydrogels presented herein incorporate characteristics including catalytic therapy, photothermal therapy, antibacterial properties, and skin damage healing, providing an innovative and comprehensive therapeutic approach for melanoma treatment.
Collapse
Affiliation(s)
- Rui Gong
- Medical Innovation Technology Transformation Center, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518039, China
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518107, China
| | - Decai Yang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Cuijuan Zhang
- Department of Cardiovascular Surgery, First Center of 301 Chinese PLA General Hospital, Beijing 100853, China
| | - Ghulam Abbas
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Beiping Miao
- Department of Otolaryngology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518039, China
| | - Yueyue Liang
- Department of Otolaryngology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518039, China
| | - Jianing Xu
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, Shandong 250012, China
| | - Xueyang Fang
- Medical Innovation Technology Transformation Center, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518039, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Hui Ding
- Medical Innovation Technology Transformation Center, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518039, China
| |
Collapse
|
165
|
Dai W, Wu J, Li K, Xu Y, Wang W, Xiao W. Andrographolide: A promising therapeutic agent against organ fibrosis. Eur J Med Chem 2024; 280:116992. [PMID: 39454221 DOI: 10.1016/j.ejmech.2024.116992] [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: 08/18/2024] [Revised: 09/07/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
Fibrosis is the terminal pathology of chronic illness in many organs, marked by excessive accumulation of extracellular matrix proteins. These changes influence organ function, ultimately resulting in organ failure. Although significant progress has been achieved in comprehending the molecular pathways responsible for fibrosis in the last decades, effective and approved clinical therapies for the condition are still lacking. Andrographolide is a diterpenoid isolated and purified mainly from the aboveground parts of the Andrographis paniculata plant, which possesses good effects of purging heat, detoxifying, antibacterial and anti-inflammatory. In-depth research has gradually confirmed the anticancer, antioxidant, antiviral and other effects of Andro so that it can play a preventive and therapeutic role in various diseases. Over the past few years, an increasing number of research findings have indicated that Andro exerts antifibrotic effects in various organs by acting on transforming growth factor-β/small mother against decapentaplegic protein, mitogen-activated protein kinases, nuclear factor-E2-related factor 2, nuclear factor kappa-B and other signalling molecules to inhibit inflammation, oxidative stress, epithelial-mesenchymal transition, fibroblast activation and collagen buildup. This review presents a compilation of findings regarding the antifibrotic impact of Andro in tissue and cell models in vitro and in vivo. Emphasis is placed on the potential therapeutic benefits of Andro in diseases related to organ fibrosis. Existing studies and cutting-edge technologies on Andro pharmacokinetics, toxicity and bioavailability are briefly discussed to provide evidence for accelerating its clinical conversion and adoption.
Collapse
Affiliation(s)
- Wei Dai
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Jiabin Wu
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Ke Li
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Yingying Xu
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Wenhong Wang
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; Research Institute for Biology and Medicine, Hunan University of Medicine, Huaihua 418000, China.
| | - Weihua Xiao
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
166
|
Basalova NA, Vigovskiy MA, Popov VS, Lagereva EA, Grigorieva OA, Efimenko AY. The Role of Activated Stromal Cells in Fibrotic Foci Formation and Reversion. Cells 2024; 13:2064. [PMID: 39768155 PMCID: PMC11674712 DOI: 10.3390/cells13242064] [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: 10/29/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
Fibrotic focus is a pivotal morphofunctional unit in developing fibrosis in various tissues. For most fibrotic diseases, including progressive forms, the foci are considered unable to remodel and contribute to the worsening of prognosis. Unfortunately, the dynamics of the fibrotic focus formation and resolution remains understudied. A number of data suggest that the key cell type for focus formation are activated stromal cells marked by fibroblast activated protein alpha (FAPα) due to their high capacity for extracellular matrix (ECM) remodeling. We evaluated the dynamics of fibrotic focus formation and the contribution of the main cell types, including FAPα+ cells, in this process using a murine model of bleomycin-induced lung fibrosis. We revealed the very early appearance of FAPα+ cells in lungs after injury and assumed their important involvement to the myofibroblast pool formation. During the first month after bleomycin administration, FAPα+ cells colocalize with CD206+ M2 macrophages. Interestingly, during the reversion stage, we unexpectedly observed the specific structured foci formed by CD90+FAPα+ cells, which we suggested calling "remodeling foci". Our findings highlight the crucial role of activated stromal cells in fibrosis initiation, progression, and reversion and provide emerging issues regarding the novel targets for antifibrotic therapy.
Collapse
Affiliation(s)
- Nataliya Andreevna Basalova
- Centre for Regenerative Medicine, Medical Research and Educational Institute, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.V.); (V.S.P.); (E.A.L.); (O.A.G.); (A.Y.E.)
| | | | | | | | | | | |
Collapse
|
167
|
Wu L, Lu Y, Liu L, Wang J, Bai Y, Song J, Heng BC, Wu T, Ren Q, Li T, Xu M, Deng X, He Y, Liu Y, Zhang X. BaTiO 3 Doping Enhances Ultrasound-Driven Piezoelectric Bactericidal Effects of Fibrous Poly(L-Lactic Acid) Dressings to Accelerate Septic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67477-67490. [PMID: 39601666 DOI: 10.1021/acsami.4c17407] [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: 11/29/2024]
Abstract
Bacterial invasion in infected skin wounds triggers inflammation and impedes healing. Current therapeutic strategies incorporating drug interventions within wound dressings often result in drug resistance and delayed healing. Here, we developed a comprehensive therapeutic modality integrating piezoelectric fibrous dressing with controlled ultrasound stimulation for efficient healing in an infected wound model. The electrospun fibrous dressings composed of barium titanate (BaTiO3) doped poly(L-lactic acid) (PLLA) possess improved piezoelectric properties due to the aligned structure and high crystallinity, which achieved superior bactericidal efficacy upon ultrasound-mechanical-electric conversion that results in the production of reactive oxygen species (ROS). There were 88.72% and 90.43% killing rates of Staphylococcus aureus and Escherichia coli respectively upon ultrasound stimulation without any need for exogenous drugs, and a wound closure rate of 95.5% within 10 days. The in vivo results confirmed that this dressing effectively shortened wound closure time by about 2 days, with a much-improved healing rate of 14% compared with previously reported therapeutic strategies. This was accompanied by reduced inflammation and increased re-epithelialization and angiogenesis. Hence, our synergistic treatment by piezoelectric materials and controlled ultrasound stimulation provides a drug-free alternative approach in regenerative tissue engineering for simultaneously enhancing antibacterial effects and promoting wound healing.
Collapse
Affiliation(s)
- Liping Wu
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Yanhui Lu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Lulu Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Jianfeng Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, PR China
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Jia Song
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Tingting Wu
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| | - Qiaomei Ren
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| | - Tingjun Li
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Xuliang Deng
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Yang Liu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| | - Xuehui Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, PR China
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
- Oral Translational Medicine Research Center, Shanxi Key Laboratory of Oral and Maxillofacial Repair, Reconstruction and Regeneration Joint Training Base, The First People's Hospital of Jinzhong, Jinzhong 030600, Shanxi Province, PR China
| |
Collapse
|
168
|
Liu H, Mei H, Jiang H, Jiang L, Lin K, Jiang M, Ding N, Li X, Gao Z, Liu B, Lin W, Li J, Zhou J. Bioprinted Symbiotic Dressings: A Lichen-Inspired Approach to Diabetic Wound Healing with Enhanced Bioactivity and Structural Integrity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2407105. [PMID: 39663708 DOI: 10.1002/smll.202407105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/13/2024] [Indexed: 12/13/2024]
Abstract
Providing oxygen and preventing infection at wound sites are effective ways to heal diabetic chronic wounds. Inspired by natural lichens, a bioprinted biogenic hydrogel (BBH) containing microalgae and probiotics is developed for diabetic chronic wound therapeutics, which offers prolonged biogenetic oxygen supply by microalgae and infection inhibition by probiotics. The rational design of symbiotic BBH with customizable structure and microorganism composition enhances wound resilience against elevated glucose levels and hypoxia, leading to the increased migration ability of fibroblasts and the angiogenic potential of human umbilical vein endothelial cells. Notably, BBH-treated diabetic wounds exhibit dense vascular distribution, reduced hypoxia levels and inflammatory responses, and enhanced epithelial differentiation and keratinization abilities. Consequently, the BBH achieves rapid tissue repairing within 3 d and restores approximately 90% of the whole skin structure within 12 d. This work presents an engineered platform for regulating biological microenvironment of diabetic wounds and provides insights for developing bioprinted hybrid microorganism systems.
Collapse
Affiliation(s)
- Hai Liu
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Hongxiang Mei
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hejin Jiang
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, 610065, China
| | - Linli Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Kaifeng Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Minwen Jiang
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Ning Ding
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Xiaojie Li
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Ziqi Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Bin Liu
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Advanced Study, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Wei Lin
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, 610065, China
| | - Juan Li
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiajing Zhou
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
169
|
Raghav PK, Mann Z. Nano-Delivery Revolution: Harnessing Mesenchymal Stem Cell-Derived Exosomes' Potential for Wound Healing. Biomedicines 2024; 12:2791. [PMID: 39767697 PMCID: PMC11673788 DOI: 10.3390/biomedicines12122791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/04/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Stem cell transplantation has proven effective in treating acute and chronic wounds, but its limitations, such as low cellular viability and the need for specialized transportation, highlight the necessity for alternative approaches. This review explores the potential of engineered exosomes, containing identified miRNAs/peptides, as a more stable and efficient cell-free therapy for regenerative medicine, particularly in wound healing. The discussion emphasizes the benefits of exosomes, including their stability, reduced damage, and consistent biological activity, paving the way for innovative applications like lyophilized exosomes, mist spray delivery, and exosome-based scaffolds. The exploration of cell-free therapy in this review holds promising implications for advancing wound-healing strategies.
Collapse
Affiliation(s)
- Pawan Kumar Raghav
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco (UCSF), San Francisco, CA 94118, USA
| | | |
Collapse
|
170
|
Mu Y, Ma L, Yao J, Luo D, Ding X. Bioengineered Extracellular Vesicle Hydrogel Modulating Inflammatory Microenvironment for Wound Management. Int J Mol Sci 2024; 25:13093. [PMID: 39684803 DOI: 10.3390/ijms252313093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic wounds, frequently arising from conditions like diabetes, trauma, or chronic inflammation, represent a significant medical challenge due to persistent inflammation, heightened infection risk, and limited treatment solutions. This study presents a novel bioengineered approach to promote tissue repair and improve the healing environment. We developed a bioactive hydrogel patch, encapsulated zeolitic imidazolate framework-8 (ZIF-8) into extracellular vesicles (EVs) derived from anti-inflammatory M2 macrophages, and synthesized ZIF@EV, then embedded it in the sodium alginate matrix. This hydrogel structure enables the controlled release of therapeutic agents directly into the wound site, where it stimulates endothelial cell proliferation and promotes new blood vessel formation. These processes are key components of effective tissue regeneration. Crucially, the EV-infused patch influences the immune response by polarizing macrophages towards an M2 phenotype, shifting the wound environment from inflammation toward regenerative healing. When applied in a murine model of chronic wounds, the EV hydrogel patch demonstrated notable improvements in healing speed, quality, and tissue integration compared to traditional approaches such as growth factor therapies and foam dressings. These promising findings suggest that this bioactive hydrogel patch could serve as a versatile, practical solution for chronic wound management, providing an adaptable platform that addresses both the biological and logistical needs of wound care in clinical settings.
Collapse
Affiliation(s)
- Yunfei Mu
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Liwen Ma
- The First College of Clinical Medicine, Nanjing Medical University, Nanjing 211100, China
| | - Jia Yao
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Dan Luo
- The First College of Clinical Medicine, Nanjing Medical University, Nanjing 211100, China
| | - Xianguang Ding
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| |
Collapse
|
171
|
Chen A, Gong M, Chi J, Wang Z, Dai L. Exploring the potential mechanisms of the ethyl acetate fraction of Hippophae rhamnoides L. seeds as a natural healing agent for wound repair. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118688. [PMID: 39142622 DOI: 10.1016/j.jep.2024.118688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sea buckthorn (Hippophae rhamnoides L.) has been designated a "medicine food homology" fruit by the National Health Commission of China due to its nutritional value. In traditional Chinese ethnomedicine, Hippophae rhamnoides L. is commonly used to treat nonhealing wounds such as burns, sores, and gastric ulcers. The aim of this study was to explore the healing effects of the ethyl acetate extract of sea buckthorn seeds (SBS-EF) on burn wounds. AIM OF THE STUDY The primary objectives of this research were to determine the most effective medicinal site of action for treating burns with sea buckthorn seeds (SBS) and to investigate the underlying material basis and mechanisms of their therapeutic effects. MATERIALS AND METHODS The effects of different components of SBS-EF on the proliferation and migration of human skin fibroblasts (HSFs) were evaluated via MTT assays, scratch assays, transwell assays, and hydroxyproline secretion analysis. SBS-EF displayed the greatest activity amongst the extracts. Subsequent analyses included network pharmacology methodology, molecular docking studies, ultraperformance liquid chromatography UPLC-Orbitrap-Exploris-120-MS and a severe second-degree burn rat model to investigate the chemical constituents and potential therapeutic mechanisms of the SBS-EF. RESULTS In vitro studies demonstrated the efficacy of SBS-EF in promoting HSF growth and migration. UPLC-Orbitrap-Exploris-120-MS analysis revealed that SBS-EF had ten major constituents, with flavonoids being the predominant compounds, especially catechin, quercetin, and kaempferol derivatives. Network pharmacology and molecular docking analyses indicated that SBS-EF may exert its healing effects by modulating the Wnt/β-catenin signalling pathway. Subsequent in vivo experiments demonstrated that SBS-EF accelerated burn wound healing in rats, increased hydroxyproline expression in skin tissue, facilitated skin structure repair, and enhanced collagen production and organisation over a 21 d period. Additionally, exposure to SBS-EF upregulated WNT3a and β-catenin while downregulating GSK-3β levels in rat skin tissue. CONCLUSIONS The wound healing properties of SBS-EF were attributed to its ability to enhance HSF growth and migration, increase hydroxyproline levels in the skin, promote collagen accumulation, reduce scarring, and decrease the skin water content. SBS-EF may also provide therapeutic benefits for burns by modulating the Wnt/β-catenin signalling pathway, as evidenced by its effective site and likely mechanism of action in the treatment of burned rats.
Collapse
Affiliation(s)
- Anying Chen
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Henan, 450046, China; Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials from Henan, Henan, 450046, China.
| | - Man Gong
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Henan, 450046, China; Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials from Henan, Henan, 450046, China.
| | - Jun Chi
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Henan, 450046, China; Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials from Henan, Henan, 450046, China.
| | - Zhimin Wang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Henan, 450046, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Liping Dai
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Henan, 450046, China; Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials from Henan, Henan, 450046, China.
| |
Collapse
|
172
|
Alqabandi JA, David R, Abdel-Motal UM, ElAbd RO, Youcef-Toumi K. An innovative cellular medicine approach via the utilization of novel nanotechnology-based biomechatronic platforms as a label-free biomarker for early melanoma diagnosis. Sci Rep 2024; 14:30107. [PMID: 39627312 PMCID: PMC11615046 DOI: 10.1038/s41598-024-79154-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 11/06/2024] [Indexed: 12/06/2024] Open
Abstract
Innovative cellular medicine (ICM) is an exponentially emerging field with a promising approach to combating complex and ubiquitous life-threatening diseases such as multiple sclerosis (MS), arthritis, Parkinson's disease, Alzheimer's, heart disease, and cancer. Together with the advancement of nanotechnology and bio-mechatronics, ICM revolutionizes cellular therapy in understanding the essence and nature of the disease initiated at a single-cell level. This paper focuses on the intricate nature of cancer that requires multi-disciplinary efforts to characterize it well in order to achieve the objectives of modern world contemporary medicine in the early detection of the disease at a cellular level and potentially arrest its proliferation mechanism. This justifies the multidisciplinary research backgrounds of the authors of this paper in advancing cellular medicine by bridging the gap between experimental biology and the engineering field. Thus, in pursuing this approach, two novel miniaturized and highly versatile biomechatronic platforms with dedicated operating software and microelectronics are designed, modeled, nanofabricated, and tested in numerous in vitro experiments to investigate a hypothesis and arrive at a proven theorem in carcinogenesis by interrelating cellular contractile force, membrane potential, and cellular morphology for early detection and characterization of melanoma cancer cells. The novelties that flourished within this work are manifested in sixfold: (1) developing a mathematical model that utilizes a Heaviside step function, as well as a pin-force model to compute the contractile force of a living cell, (2) deriving an expression of cell-membrane potential based on Laplace and Fourier Transform and their Inverse Transform functions by encountering Warburg diffusion impedance factor, (3) nano-fabricating novel biomechatronic platforms with associated microelectronics and customized software that extract cellular physics and mechanics, (4) developing a label-free biomarker, (5) arrive at a proved theorem in developing a mathematical expression in relating cancer cell mechanobiology to its biophysics in connection to the stage of the disease, and (6) to the first time in literature, and to the best of the authors' knowledge, discriminating different stages and morphology of cancer cell melanoma based on their cell-membrane potentials, and associated contractile forces that could introduce a new venue of cellular therapeutic modalities, preclinical early cancer diagnosis, and a novel approach in immunotherapy drug development. The proposed innovative technology-based versatile bio-mechatronic platforms shall be extended for future studies, investigating the role of electrochemical signaling of the nervous system in cancer formation that will significantly impact modern oncology by pursuing a targeted immunotherapy approach. This work also provides a robust platform for immunotherapy practitioners in extending the study of cellular biophysics in stalling neural-cancer interactions, of which the FDA-approved chimeric antigen receptor (CAR)-T cell therapies can be enhanced (genetically engineered) in a lab by improving its receptors to capture cancer antigens. This work amplifies the importance of studying neurotransmitters and electrochemical signaling molecules in shaping the immune T-cell function and its effectiveness in arresting cancer proliferation rate (mechanobiology mechanism).
Collapse
Affiliation(s)
- Jassim A Alqabandi
- Mechatronics Research Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
- Mechatronics in Medicine Laboratory, Imperial College London, London, UK.
- Department of Manufacturing Engineering Technology (Bio-Mechatronics) Department, PAAET, Kuwait, State of Kuwait.
| | - Rhiannon David
- Division of Computational and Systems Medicine (CSM), Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, London, UK
| | - Ussama M Abdel-Motal
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Rawan O ElAbd
- McGill University Health Center, Montreal, QC, Canada
| | - Kamal Youcef-Toumi
- Mechatronics Research Laboratory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| |
Collapse
|
173
|
Lyu X, Wu H, Chen Y, Sun Y, Cai X, Li S, Lin Y. A Multifunctional Nanocomplex as miRNA/Antibiotic Co-Delivery System Based on Tetrahedral Framework DNA: Application to Infected Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2406629. [PMID: 39279370 DOI: 10.1002/smll.202406629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/28/2024] [Indexed: 09/18/2024]
Abstract
Infected wounds are a complex disease involving bacterial infections and dysregulated inflammation. However, current research has mostly focused on bacterial inhibition rather than on inflammation. Thus, combined therapeutic strategies with anti-bacterial and anti-inflammation efficacies are urgently needed. Antibiotics are the main treatment strategy for infections. However, the excessive use of antibiotics throughout the body can cause serious side effects. In addition, miRNA-based therapeutics are superior for the treatment of wounds, but their rapid degradation and poor cellular uptake limit their clinical application. Tetrahedral framework DNA (tFNA) is an ideal drug delivery system owing to its excellent stability and remarkable transport ability. Herein, a novel multi-functional miRNA and antibiotic co-delivery system based on tFNA is presented for the first time, called B/L. B/L has heightened resistance to serum and excellent codelivery ability. After transdermal administration, B/L can specifically target TNF receptor-associated factor 6(TRAF6) and IL-1receptor-associated kinase 1(IRAK1), thereby regulating nuclear factor kappa-B (NF-𝜿B) and thus effectively reducing inflammation and promoting the healing of infected wounds. This novel multi-functional co-delivery system provides a versatile, simple, biocompatible, and powerful platform for the personalized and combined treatment of multiple diseases.
Collapse
Affiliation(s)
- Xiaoying Lyu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Haoyan Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ye Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yue Sun
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| | - Songhang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
174
|
Costa ADSD, Jeong H, Subbiah R, Park K, Choi IS, Shin JH. Intercellular junction-driven stromal cell stacking in a confined 3D microcavity. APL Bioeng 2024; 8:046109. [PMID: 39525363 PMCID: PMC11549968 DOI: 10.1063/5.0197187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Understanding the detailed mechanisms driving fibroblast migration within native tissue settings during pathophysiological events presents a critical research challenge. In this study, we elucidate how stromal cells migrate and contribute to the development of three-dimensional (3D) cellular aggregates within confined microcavities. Integrin α5β1 and β-catenin (β-cat) are central in guiding this collective migration and achieving optimal filling of the microcavity. When β-cat is suppressed, cells tend to migrate more sporadically, leading to less efficient cellular organization. Furthermore, we also detail the pivotal roles of Cx43 and N-cadherin (N-cad) in orchestrating collective migration and in shaping efficient cellular stacking. Suppressing gap junctions, especially Cx43, significantly impacts the extracellular matrix expression, integrin α5 and β1, and other elements in the 3D construct, emphasizing the importance of physicochemical cell-cell interactions. The distribution patterns of N-cad and focal adhesion kinase (FAK) further corroborate the essential roles in forming cell-cell junctions and FAK in establishing the foundational layer that underpins the cell stacking within the microcavity. Interestingly, neither Rho-associated protein kinase (ROCK) nor RhoA significantly alter the cell migration pattern toward microcavity. These findings provide fresh perspectives on fibroblast activities in 3D space, enriching our understanding and offering implications for advancements in wound healing and tissue engineering.
Collapse
Affiliation(s)
| | - Hyuntae Jeong
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Ramesh Subbiah
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University (OHSU), Portland, Oregon 97201, USA
| | | | - In-Suk Choi
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jennifer H. Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
175
|
Wei W, Qu ZL, Lei L, Zhang P. TREM2-mediated Macrophage Glycolysis Promotes Skin Wound Angiogenesis via the Akt/mTOR/HIF-1α Signaling Axis. Curr Med Sci 2024; 44:1280-1292. [PMID: 39672999 DOI: 10.1007/s11596-024-2946-3] [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: 08/20/2024] [Accepted: 09/30/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE The trigger receptor expressed on myeloid cells-2 (TREM2) pathway in myeloid cells is a key disease-inducing immune signaling hub that is essential for detecting tissue damage and limiting its pathological spread. However, the role and potential mechanisms of TREM2 in wound repair remain unclear. The purpose of this study was to determine the role and mechanism of TREM2 in skin wound healing in mice. METHODS Immunofluorescence staining was used to determine the expression and cellular localization of TREM2 and test the effects of TREM2 knockout on angiogenesis, glycolysis, and lactylation in skin tissue. Western blotting was used to analyze the expression of the Akt/mTOR/HIF-1α signaling pathway in the wounded skin tissues of wild-type (WT) and TREM2 knockout mice. A coimmunoprecipitation assay was used to determine whether HIF-1α, which mediates angiogenesis, is modified by lactylation. RESULTS The number of TREM2+ macrophages was increased, and TREM2+ macrophages mediated angiogenesis after skin injury. TREM2 promoted glycolysis and lactylation in macrophages during wound healing. Mechanistically, TREM2 promoted macrophage glycolysis and angiogenesis in wounded skin tissues by activating the Akt/mTOR/HIF-1α signaling pathway. HIF-1α colocalized with Klac to mediate lactylation in macrophages, and lactate could stabilize the expression of the HIF-1α protein through lactylation. Lactate treatment ameliorated the impaired angiogenesis and delayed wound healing in wounded skin in TREM2 knockout mice. CONCLUSION TREM2+ macrophage-mediated glycolysis can promote angiogenesis and wound healing. Our findings provide an effective strategy and target for promoting skin wound healing.
Collapse
Affiliation(s)
- Wei Wei
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Wuhan, 430022, China
| | - Zi-Lu Qu
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Wuhan, 430022, China
| | - Li Lei
- Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210000, China
| | - Ping Zhang
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Wuhan, 430022, China.
| |
Collapse
|
176
|
Nepon H, Julien C, Petrecca S, Kalashnikov N, Safran T, Murphy A, Dionisopoulos T, Davison P, Vorstenbosch J. The cellular and molecular properties of capsule surrounding silicone implants in humans vary uniquely according to the tissue type adjacent to the implant. J Biomed Mater Res A 2024; 112:2055-2070. [PMID: 38864257 DOI: 10.1002/jbm.a.37762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/15/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024]
Abstract
The foreign body reaction (FBR) to biomaterials results in fibrous encapsulation. Excessive capsule fibrosis (capsular contracture) is a major challenge to the long-term stability of implants. Clinical data suggests that the tissue type in contact with silicone breast implants alters susceptibility to developing capsular contracture; however, the tissue-specific inflammatory and fibrotic characteristics of capsule have not been well characterized at the cellular and molecular level. In this study, 60 breast implant capsule samples are collected from patients and stratified by the adjacent tissue type including subcutaneous tissue, glandular breast tissue, or muscle tissue. Capsule thickness, collagen organization, immune and fibrotic cellular populations, and expression of inflammatory and fibrotic markers is quantified with histological staining, immunohistochemistry, and real-time PCR. The findings suggest there are significant differences in M1-like macrophages, CD4+ T cells, CD26+ fibroblasts, and expression of IL-1β, IL-6, TGF-β, and collagen type 1 depending on the tissue type abutting the implant. Subglandular breast implant capsule displays a significant increase in inflammatory and fibrotic markers. These findings suggest that the tissue microenvironment contributes uniquely to the FBR. This data could provide new avenues for research and clinical applications to improve the site-specific biocompatibility and longevity of implantable devices.
Collapse
Affiliation(s)
- Hillary Nepon
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
- Division of Surgical and Interventional Sciences, McGill University, Montreal General Hospital, Quebec, Canada
| | - Cedric Julien
- McGill University Hospital Centre Research Institute, Montreal General Hospital, Quebec, Canada
| | - Sarah Petrecca
- Faculty of Medicine and Health Sciences, McGill University, Quebec, Canada
| | - Nikita Kalashnikov
- Division of Surgical and Interventional Sciences, McGill University, Montreal General Hospital, Quebec, Canada
- Faculty of Medicine and Health Sciences, McGill University, Quebec, Canada
| | - Tyler Safran
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
| | - Amanda Murphy
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
| | - Tassos Dionisopoulos
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
| | - Peter Davison
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
| | - Joshua Vorstenbosch
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
- McGill University Hospital Centre Research Institute, Montreal General Hospital, Quebec, Canada
| |
Collapse
|
177
|
Staianov J, Struz JMR, Vieira RV, Luiz RM, Zarpelon‐Schutz AC, Teixeira KN, Bernardi‐Wenzel J. Histomorphometric analysis of excisional cutaneous wounds with different diameters in an animal model. Int J Exp Pathol 2024; 105:235-245. [PMID: 39439085 PMCID: PMC11576331 DOI: 10.1111/iep.12520] [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: 07/18/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
The skin wound model in rats is a fundamental stage in preclinical trials, but there is a lack of standardization in these trials regarding the initial wound area, making analysis and comparison between studies difficult. Therefore, this study evaluates the healing progression of excisional skin lesions of varying diameters in Wistar rats, aiming to identify the optimal wound size for monitoring treatment effects on wound healing. Excisions of 0.8, 1.5, 2.0 and 3.0 cm in diameter were made on the back of the animals. Thirty animals were used per treatment and evaluated on days 3, 7, 10, 14 and 21 after surgery. The lesions were cleaned daily with saline solution until they were completely closed. The 0.8 cm group showed complete repair on D14, while in the other groups, the wounds persisted until day 21, with a reddened surface and no complete epidermal coverage, but with greater keratinization and presence of appendages in the 1.5 cm lesions. Therefore, as a standardization model for creating skin wounds, we suggest using 1.5 or 2.0 cm excisions, considering that 0.8 cm wounds close very early and 3.0 cm wounds, although behaving similarly to 2.0 cm wounds, are more invasive for the animals. The 1.5 cm model proved to be suitable for closure within 21 days. When evaluating a product intended to accelerate wound healing, 2.0 cm lesions are recommended to assess the effectiveness of the treatment.
Collapse
Affiliation(s)
- Janiele Staianov
- Curso de MedicinaCampus Toledo, Universidade Federal do ParanáToledoBrazil
| | | | | | | | - Ana Carla Zarpelon‐Schutz
- Curso de MedicinaCampus Toledo, Universidade Federal do ParanáToledoBrazil
- Programa de Pós‐graduação em BiotecnologiaSetor Palotina, Universidade Federal do ParanáPalotinaBrazil
| | - Kádima Nayara Teixeira
- Curso de MedicinaCampus Toledo, Universidade Federal do ParanáToledoBrazil
- Programa Multicêntrico de Pós‐graduação em Bioquímica e Biologia MolecularSetor Palotina, Universidade Federal do ParanáPalotinaBrazil
| | - Juliana Bernardi‐Wenzel
- Curso de MedicinaCampus Toledo, Universidade Federal do ParanáToledoBrazil
- Programa de Pós‐graduação em BiotecnologiaSetor Palotina, Universidade Federal do ParanáPalotinaBrazil
| |
Collapse
|
178
|
Pallod S, Aguilera Olvera R, Ghosh D, Rai L, Brimo S, DeCambra W, Sant HG, Ristich E, Singh V, Abedin MR, Chang N, Yarger JL, Lee JK, Kilbourne J, Yaron JR, Haydel SE, Rege K. Skin repair and infection control in diabetic, obese mice using bioactive laser-activated sealants. Biomaterials 2024; 311:122668. [PMID: 38908232 PMCID: PMC11562812 DOI: 10.1016/j.biomaterials.2024.122668] [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: 04/08/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Conventional wound approximation devices, including sutures, staples, and glues, are widely used but risk of wound dehiscence, local infection, and scarring can be exacerbated in these approaches, including in diabetic and obese individuals. This study reports the efficacy and quality of tissue repair upon photothermal sealing of full-thickness incisional skin wounds using silk fibroin-based laser-activated sealants (LASEs) containing copper chloride salt (Cu-LASE) or silver nanoprisms (AgNPr-LASE), which absorb and convert near-infrared (NIR) laser energy to heat. LASE application results in rapid and effective skin sealing in healthy, immunodeficient, as well as diabetic and obese mice. Although lower recovery of epidermal structure and function was seen with AgNPr-LASE sealing, likely because of the hyperthermia induced by laser and presence of this material in the wound space, this approach resulted in higher enhancement in recovery of skin biomechanical strength compared to sutures and Cu-LASEs in diabetic, obese mice. Histological and immunohistochemical analyses revealed that AgNPr-LASEs resulted in significantly lower neutrophil migration to the wound compared to Cu-LASEs and sutures, indicating a more muted inflammatory response. Cu-LASEs resulted in local tissue toxicity likely because of effects of copper ions as manifested in the form of a significant epidermal gap and a 'depletion zone', which was a region devoid of viable cells proximal to the wound. Compared to sutures, LASE-mediated sealing, in later stages of healing, resulted in increased angiogenesis and diminished myofibroblast activation, which can be indicative of lower scarring. AgNPr-LASE loaded with vancomycin, an antibiotic drug, significantly lowered methicillin-resistant Staphylococcus aureus (MRSA) load in a pathogen challenge model in diabetic and obese mice and also reduced post-infection inflammation of tissue compared to antibacterial sutures. Taken together, these attributes indicate that AgNPr-LASE demonstrated a more balanced quality of tissue sealing and repair in diabetic and obese mice and can be used for combating local infections, that can result in poor healing in these individuals.
Collapse
Affiliation(s)
- Shubham Pallod
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA
| | - Rodrigo Aguilera Olvera
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, USA
| | - Deepanjan Ghosh
- Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA
| | - Lama Rai
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; College of Health Solutions, Arizona State University, USA
| | - Souzan Brimo
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Arizona State University, USA
| | | | - Harsh Girish Sant
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA
| | - Eron Ristich
- School of Molecular Sciences, Arizona State University, USA; School of Computing and Augmented Intelligence, Arizona State University, USA
| | - Vanshika Singh
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Arizona State University, USA
| | - Muhammad Raisul Abedin
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA
| | - Nicolas Chang
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Arizona State University, USA
| | | | - Jung Keun Lee
- Departments of Pathology and Population Medicine, Midwestern University, College of Veterinary Medicine, 5725 West Utopia Rd., Glendale, AZ, 85308, USA
| | | | - Jordan R Yaron
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA
| | - Shelley E Haydel
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, USA; School of Life Sciences, Arizona State University, 501 E. Tyler Mall ECG 303, Tempe, AZ, 85287-6106, USA
| | - Kaushal Rege
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA.
| |
Collapse
|
179
|
Song YT, Liu PC, Zhou XL, Chen YM, Wu W, Zhang JY, Li-Ling J, Xie HQ. Extracellular matrix-based biomaterials in burn wound repair: A promising therapeutic strategy. Int J Biol Macromol 2024; 283:137633. [PMID: 39549816 DOI: 10.1016/j.ijbiomac.2024.137633] [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: 07/05/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024]
Abstract
Burns are common traumatic injuries affecting many people worldwide. Development of specialized burn units, advances in acute care modalities, and burn prevention programs have successfully reduced the mortality rate of severe burns. Autologous skin grafting has been considered as the gold standard for wound coverage after the removal of burned skin. For full-thickness burns of a larger scale, however, the autograft donor site may be quickly exhausted, so that alternative skin coverage is necessary. Although rapid progress has been made in the development of skin substitutes for burn wounds during the last decade, no skin substitute has fulfilled the criteria as a perfect replacement for the damaged skin. Extracellular matrix (ECM) derived components have emerged as a source for the engineering of biomaterials capable of inducing desirable cell-specific responses and one of the most promising biomaterials for burn wound healing. Among these, acellular dermal matrix, small intestinal submucosa, and amniotic membrane have been applied to treat burn wounds with acceptable outcomes. This review has explored the use of biomaterials derived from naturally occurring ECM and their derivatives for approaches aiming to promote burn wound healing, and summarized the ECM-based wound dressings products applicable in burn wound and postburn scar contracture to date.
Collapse
Affiliation(s)
- Yu-Ting Song
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Peng-Cheng Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xing-Li Zhou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Dermatology, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan-Ming Chen
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wu Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ji-Ye Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China.
| |
Collapse
|
180
|
Le Z, Ramos MC, Shou Y, Li RR, Cheng HS, Jang CJ, Liu L, Xue C, Li X, Liu H, Lim CT, Tan NS, White AD, Charles CJ, Chen Y, Liu Z, Tay A. Bioactive sucralfate-based microneedles promote wound healing through reprogramming macrophages and protecting endogenous growth factors. Biomaterials 2024; 311:122700. [PMID: 38996671 DOI: 10.1016/j.biomaterials.2024.122700] [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: 04/03/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024]
Abstract
Impaired wound healing due to insufficient cell proliferation and angiogenesis is a significant physical and psychological burden to patients worldwide. Therapeutic delivery of exogenous growth factors (GFs) at high doses for wound repair is non-ideal as GFs have poor stability in proteolytic wound environments. Here, we present a two-stage strategy using bioactive sucralfate-based microneedle (SUC-MN) for delivering interleukin-4 (IL-4) to accelerate wound healing. In the first stage, SUC-MN synergistically enhanced the effect of IL-4 through more potent reprogramming of pro-regenerative M2-like macrophages via the JAK-STAT pathway to increase endogenous GF production. In the second stage, sucralfate binds to GFs and sterically disfavors protease degradation to increase bioavailability of GFs. The IL-4/SUC-MN technology accelerated wound healing by 56.6 % and 46.5 % in diabetic mice wounds and porcine wounds compared to their respective untreated controls. Overall, our findings highlight the innovative use of molecular simulations to identify bioactive ingredients and their incorporation into microneedles for promoting wound healing through multiple synergistic mechanisms.
Collapse
Affiliation(s)
- Zhicheng Le
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore; Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Mayk Caldas Ramos
- Department of Chemical Engineering, University of Rochester, 14627, USA
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Renee R Li
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore; Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, 119228, Singapore
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Clarisse Jm Jang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Ling Liu
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore
| | - Chencheng Xue
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore; Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Hong Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore; Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Andrew D White
- Department of Chemical Engineering, University of Rochester, 14627, USA
| | - Christopher John Charles
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore; Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, 119228, Singapore; Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhijia Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore; Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore.
| |
Collapse
|
181
|
Kim H, Shin HY, Park M, Ahn K, Kim SJ, An SH. Exosome-Like Vesicles from Lithospermum erythrorhizon Callus Enhanced Wound Healing by Reducing LPS-Induced Inflammation. J Microbiol Biotechnol 2024; 35:e2410022. [PMID: 39848679 PMCID: PMC11813354 DOI: 10.4014/jmb.2410.10022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 01/25/2025]
Abstract
Lithospermum erythrorhizon (LE), a medicinal plant from the Boraginaceae family, is traditionally used in East Asia for its therapeutic effects on skin conditions, including infections, inflammation, and wounds. Recently, the role of extracellular vesicles (EVs) as mediators of intercellular communication that regulate inflammation and promote tissue regeneration has garnered increasing attention in the field of regenerative medicine. This study investigates exosome-like vesicles derived from LE callus (LELVs) and their potential in enhancing wound healing. In vitro studies using normal human dermal fibroblasts (NHDFs) demonstrated that LELVs significantly improved cell viability, proliferation, and wound closure, while also enhancing collagen type I synthesis, indicating anti-inflammatory and regenerative properties. For in vivo analysis, LELVs were applied to lipopolysaccharide (LPS)-induced wounds in mice, where wound healing progression was monitored over 14 days. LELV-treated wounds exhibited accelerated re-epithelialization, reduced inflammation, and improved tissue remodeling, with histological analysis revealing enhanced collagen deposition and reduced inflammatory cell infiltration. These results highlight the ability of LELVs to modulate the inflammatory response and promote wound healing. With their natural origin, low immunogenicity, and ease of production, LELVs represent a promising alternative to synthetic treatments for inflammation-associated skin injuries and hold significant potential for clinical applications in wound care.
Collapse
Affiliation(s)
- Hyeonoh Kim
- Preclinical Research Center, Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu 41061, Republic of Korea
| | - Hyun-young Shin
- Research Institute, Sphebio Co., Ltd., Seoul 04796, Republic of Korea
| | - Mira Park
- Research Institute, Sphebio Co., Ltd., Seoul 04796, Republic of Korea
| | - Keunsun Ahn
- Research Institute, Sphebio Co., Ltd., Seoul 04796, Republic of Korea
| | - Seung-Jin Kim
- Research Institute, Sphebio Co., Ltd., Seoul 04796, Republic of Korea
| | - Sang-Hyun An
- Preclinical Research Center, Daegu Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu 41061, Republic of Korea
| |
Collapse
|
182
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Kleb SS, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Fibroblast-Mediated Macrophage Recruitment Supports Acute Wound Healing. J Invest Dermatol 2024:S0022-202X(24)02956-7. [PMID: 39581458 DOI: 10.1016/j.jid.2024.10.609] [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: 05/08/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single-nuclei RNA sequencing, we defined changes in gene expression associated with inflammation 1 day after wounding in mouse skin. Compared with those in keratinocytes and myeloid cells, we detected enriched expression of proinflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL-33, compared with SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound-bed macrophages and monocytes during injury-induced inflammation, with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M Amuso
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - MaryEllen R Haas
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Paula O Cooper
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Sana Hafiz
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Shatha Salameh
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Miguel F Mazumder
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Violet Josephson
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Sarah S Kleb
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Khatereh Khorsandi
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Anelia Horvath
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Brett A Shook
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA; The Department of Dermatology, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA.
| |
Collapse
|
183
|
Wang K, Song B, Zhu Y, Dang J, Wang T, Song Y, Shi Y, You S, Li S, Yu Z, Song B. Peripheral nerve-derived CSF1 induces BMP2 expression in macrophages to promote nerve regeneration and wound healing. NPJ Regen Med 2024; 9:35. [PMID: 39572537 PMCID: PMC11582358 DOI: 10.1038/s41536-024-00379-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
The precise mechanisms regulating inflammatory and prorepair macrophages have not been fully elucidated, despite the pivotal role played by innate immunity in wound healing. We first employed a denervation wound model to validate the crosstalk between neurons and macrophages. Compared to normal wound healing, the denervation wound healing process involved fewer macrophages, decreased angiogenesis, and delayed wound healing. Consistent with the results of the scRNA-seq libraries, the number of early-phase wound proinflammatory and late-phase wound prorepair macrophages were decreased during the denervation wound healing process. We profiled early-phase and late-phase skin wounds in mice at the transcriptional and functional levels and compared them to those of normal wounds. We revealed a neuroimmune regulatory pathway driven by peripheral nerve-derived CSF1 that induces BMP2 expression in prorepair macrophages and enhances nerve regeneration. Crosstalk between neurons and macrophages facilitates the healing process of wounds and provides a potential strategy for wound healing therapy.
Collapse
Affiliation(s)
- Kai Wang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Binyu Song
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuhan Zhu
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Juanli Dang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tong Wang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yajuan Song
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yi Shi
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuang You
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Sijia Li
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhou Yu
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Baoqiang Song
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
184
|
Jing G, Hu C, Fang K, Li Y, Wang L. How Nanoparticles Help in Combating Chronic Wound Biofilms Infection? Int J Nanomedicine 2024; 19:11883-11921. [PMID: 39563901 PMCID: PMC11575445 DOI: 10.2147/ijn.s484473] [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: 07/06/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
Chronic wound infection has become a global health problem, with bacterial biofilms, which are difficult to penetrate using traditional antibiotics, considered the primary cause of recurrent infection and delayed healing in chronic wounds. In recent years, the outstanding performance of nanomaterials in controlling biofilm infections has been widely acknowledged, and these materials are regarded as highly promising for chronic wound infection management. The formation and structure of chronic wound biofilms undergo complex dynamic changes. Therefore, a deep understanding of the underlying causes of repeated wound infections and the specific antibacterial mechanisms of nanomaterials at different stages of biofilm formation is crucial for effective "chronic wound infection management". This review first reveals the relationship between biofilms, wound chronicity, and recurrent infections. Secondly, it focuses on the four stages of chronic wound biofilm formation: (1) adhesion stage, (2) aggregation and promotion stage, (3) maturation stage, and (4) regeneration and dissemination stage. It also comprehensively summarizes the specific antibacterial mechanisms of nanomaterials. This study analyzes essential factors affecting the control of chronic wound biofilms by nanoparticles from various perspectives, such as the material itself, the local wound environment, and the systemic host response. Finally, the limitations and potential future trends in current research are discussed. In summary, nanoparticles represent a promising strategy for combating chronic wound biofilm infections, and this review provides new insights for alternative adjuvant therapies in managing bacterial biofilm infections in chronic wounds.
Collapse
Affiliation(s)
- Gang Jing
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People's Republic of China
| | - Chen Hu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Keyi Fang
- School of Stomatology, Hainan Medical University, Haikou, Hainan, People's Republic of China
| | - Yingying Li
- School of Stomatology, Hainan Medical University, Haikou, Hainan, People's Republic of China
| | - Linlin Wang
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People's Republic of China
| |
Collapse
|
185
|
Cheng L, Zhuang Z, Yin M, Lu Y, Liu S, Zhan M, Zhao L, He Z, Meng F, Tian S, Luo L. A microenvironment-modulating dressing with proliferative degradants for the healing of diabetic wounds. Nat Commun 2024; 15:9786. [PMID: 39532879 PMCID: PMC11557877 DOI: 10.1038/s41467-024-54075-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetic wounds are usually entangled in a disorganized and self-perpetuating microenvironment and accompanied by a prolonged delay in tissue repair. Sustained and coordinated microenvironment regulation and tissue regeneration are key to the healing process of diabetic wounds, yet they continue to pose a formidable challenge. Here we report a rational double-layered dressing design based on chitosan and a degradable conjugated polymer polydiacetylene, poly(deca-4,6-diynedioic acid) (PDDA), that can meet this intricate requirement. With an alternating ene-yne backbone, PDDA degrades when reacting with various types of reactive oxygen species (ROS), and more importantly, generates proliferative succinic acid as a major degradant. Inheriting from PDDA, the developed PDDA-chitosan double layer dressing (PCD) can eliminate ROS in the microenvironment of diabetic wounds, alleviate inflammation, and downregulate gene expression of innate immune receptors. PCD degradation also triggers simultaneous release of succinic acid in a sustainable manner, enabling long-term promotion on tissue regeneration. We have validated the biocompatibility and excellent performance of PCD in expediting the wound healing on both diabetic mouse and porcine models, which underscores the significant translational potential of this microenvironment-modulating, growth-promoting wound dressing in diabetic wounds care.
Collapse
Affiliation(s)
- Lianghui Cheng
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Zhuang
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Mingming Yin
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Lu
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Sujuan Liu
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Minle Zhan
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Liyuan Zhao
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenyan He
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Fanling Meng
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinformatics and Molecular Imaging, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, China.
| | - Sidan Tian
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinformatics and Molecular Imaging, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
| | - Liang Luo
- National Engineering Research Center for Nanomedicine and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinformatics and Molecular Imaging, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
186
|
Liu J, Chen S, Zhang Z, Song X, Hou Z, Wang Z, Liu T, Yang L, Liu Y, Luo Z. The oxidized hyaluronic acid hydrogels containing paeoniflorin microspheres regulates the polarization of M1/M2 macrophages to promote wound healing. Int J Biol Macromol 2024; 282:137107. [PMID: 39515704 DOI: 10.1016/j.ijbiomac.2024.137107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Controlling excessive inflammation of acute wound is an effective means to shorten the healing time. Therefore, targeted control of the inflammatory response of the wound is a promising therapeutic strategy. In this study, paeoniflorin (Pae) was encapsulated in microspheres and combined with oxidized hyaluronic acid hydrogels to prepare the hydrogel loaded with Pae microspheres (Pae-MPs@OHA) to promote the healing of acute wounds in rats. The results demonstrated that the particle size of the Pae-MPs was 6.84 ± 0.51 μm, and the positive charge was 26.87 ± 1.51 mV. The uniform spherical structure of the Pae-MPs was observed by TEM. The Pae-MPs@OHA can maintain colloidal state in the range of 0.1-3.16 Hz. FTIR suggested that Pae could be effectively wrapped in MPs, and SEM indicated that the Pae-MPs@OHA had a uniform network pore structure. The Pae-MPs@OHA can realize the sustained release of Pae for 96 h. Biocompatibility experiments showed that the Pae-MPs@OHA hydrogels were safe and available. The Pae-MPs@OHA hydrogels can accelerate wound healing in rats. HE and masson staining suggested that the Pae-MPs@OHA could reduce inflammatory cell infiltration, promote re-epithelialization and collagen formation. The Pae-MPs@OHA could decrease the number of M1 and increase the number of M2 in macrophages, thus regulating the release of inflammatory factor TNF-α and IL-1β. The results of molecular docking and western blot results also confirmed that the Pae-MPs@OHA could reduce the expression of NF-κB, pNF-κB, NLRP3, ASC and pro-caspase-1. These findings suggest that the Pae-MPs@OHA has great potential for application in the treatment of inflammatory wound.
Collapse
Affiliation(s)
- Jiarui Liu
- Shuren International College, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Siqi Chen
- School of Public Health, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Zijing Zhang
- Graduate School, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Xitong Song
- Graduate School, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Zhiquan Hou
- Graduate School, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Ziyi Wang
- Graduate School, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China
| | - Tao Liu
- University of Michigan, Ann Arbor, School of Pharmacy, Integrated Pharmaceutical Sciences, 428 Church St, Ann Arbor, MI 48109, United States of America
| | - Liqun Yang
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yunen Liu
- Shuren International College, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China.
| | - Zhonghua Luo
- Shuren International College, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China.
| |
Collapse
|
187
|
Khattak S, Ullah I, Sohail M, Akbar MU, Rauf MA, Ullah S, Shen J, Xu H. Endogenous/exogenous stimuli‐responsive smart hydrogels for diabetic wound healing. AGGREGATE 2024. [DOI: 10.1002/agt2.688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
AbstractDiabetes significantly impairs the body's wound‐healing capabilities, leading to chronic, infection‐prone wounds. These wounds are characterized by hyperglycemia, inflammation, hypoxia, variable pH levels, increased matrix metalloproteinase activity, oxidative stress, and bacterial colonization. These complex conditions complicate effective wound management, prompting the development of advanced diabetic wound care strategies that exploit specific wound characteristics such as acidic pH, high glucose levels, and oxidative stress to trigger controlled drug release, thereby enhancing the therapeutic effects of the dressings. Among the solutions, hydrogels emerge as promising due to their stimuli‐responsive nature, making them highly effective for managing these wounds. The latest advancements in mono/multi‐stimuli‐responsive smart hydrogels showcase their superiority and potential as healthcare materials, as highlighted by relevant case studies. However, traditional wound dressings fall short of meeting the nuanced needs of these wounds, such as adjustable adhesion, easy removal, real‐time wound status monitoring, and dynamic drug release adjustment according to the wound's specific conditions. Responsive hydrogels represent a significant leap forward as advanced dressings proficient in sensing and responding to the wound environment, offering a more targeted approach to diabetic wound treatment. This review highlights recent advancements in smart hydrogels for wound dressing, monitoring, and drug delivery, emphasizing their role in improving diabetic wound healing. It addresses ongoing challenges and future directions, aiming to guide their clinical adoption.
Collapse
Affiliation(s)
- Saadullah Khattak
- The Fifth Affiliated Hospital of Wenzhou Medical University Lishui China
| | - Ihsan Ullah
- Zhejiang Engineering Research Center for Tissue Repair Materials Wenzhou Institute University of Chinese Academy of Sciences Wenzhou China
| | - Mohammad Sohail
- The Fifth Affiliated Hospital of Wenzhou Medical University Lishui China
| | - Muhammad Usman Akbar
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou China
| | - Mohd Ahmar Rauf
- Department of Internal Medicine, Heme Oncology Unit, University of Michigan Ann Arbor Michigan USA
| | - Salim Ullah
- The Fifth Affiliated Hospital of Wenzhou Medical University Lishui China
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry Eye Hospital Wenzhou Medical University Wenzhou China
- Wenzhou Institute University of Chinese Academy of Sciences Wenzhou China
| | - Hong‐Tao Xu
- The Fifth Affiliated Hospital of Wenzhou Medical University Lishui China
| |
Collapse
|
188
|
Lotfollahi Z, Tan JTM, Nankivell VA, Sandeman L, Liyanage S, Solly EL, Stretton L, Williamson AE, Dawson J, Nicholls SJ, Psaltis PJ, Fitridge R, Bursill CA. Topical Reconstituted High-Density Lipoproteins Elicit Anti-Inflammatory Effects in Diabetic Wounds. Adv Wound Care (New Rochelle) 2024. [PMID: 39505368 DOI: 10.1089/wound.2023.0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Objective: Reconstituted high-density lipoproteins (rHDL) improve wound healing in diabetes. We aimed to determine if rHDL elicit anti-inflammatory effects in diabetic wounds, as a mechanism to explain their wound healing benefits. Approach: Diabetes was induced using streptozotocin in C57Bl6/J mice. Two full-thickness wounds were placed on the subflanks of diabetic and nondiabetic (ND) mice. Phosphate-buffered saline (PBS) or rHDL (50 µg/wound/day) were applied topically. Wound closure was assessed daily. Inflammatory gene transcripts were measured by qPCR and proteins by Western blotting and enzyme-linked immunosorbent assay in wounds collected at baseline, 24 h, and 3 days postwounding. Wound macrophages were assessed by flow cytometry 7 days postwounding. The fate of fluorescent 3,3-dioctadecyloxacarbocyanine, perchlorate (DiO)-labeled rHDL was tracked by flow cytometry, fluorescent imaging, and microscopy. Results: In diabetic mice, rHDL increased wound closure rates at days 6 (+288%, p < 0.01) and 7 (+639%, p < 0.0001) postwounding, compared with PBS controls. After 3 days, rHDL-treated diabetic wounds had lower Rela (-65%) and C-C motif chemokine ligand 2 (Ccl2) (-59%) mRNA levels and CCL2 protein (29%) than PBS controls, p < 0.05 for all. Wound macrophage content was higher in diabetic than ND wounds, but rHDL did not change macrophage content or polarity. DiO-rHDL were taken up by key wound cells including fibroblasts, macrophages, keratinocytes and endothelial cells, and retained in wounds for at least 48 h. Innovation: rHDL exerts anti-inflammatory effects in diabetic wounds early postwounding, which may contribute to its wound healing properties. Conclusion: The anti-inflammatory properties of rHDL in diabetic wounds present topical rHDL as a novel treatment option for improving healing in patients with diabetic foot ulcers.
Collapse
Affiliation(s)
- Zahra Lotfollahi
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Victoria A Nankivell
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Lauren Sandeman
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sanuri Liyanage
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Emma L Solly
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Liam Stretton
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anna E Williamson
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Joseph Dawson
- Discipline of Surgery, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Vascular and Endovascular Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Clayton, Victoria, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - Robert Fitridge
- Discipline of Surgery, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Vascular and Endovascular Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Christina A Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Faculty of Health and Medical Science, The University of Adelaide, Adelaide, South Australia, Australia
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
189
|
Stelling-Férez J, Puente-Cuadrado JM, Álvarez-Yepes V, Alcaraz S, Tristante E, Hernández-Mármol I, Mompeán-Egea I, García-Hernández AM, Nicolás FJ. Refrigerated amniotic membrane maintains its therapeutic qualities for 48 hours. Front Bioeng Biotechnol 2024; 12:1455397. [PMID: 39569163 PMCID: PMC11576280 DOI: 10.3389/fbioe.2024.1455397] [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: 06/26/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
During wound healing, the migration of keratinocytes is critical for wound closure. The application of amniotic membrane (AM) on wounds with challenging contexts (e.g., chronification and diabetic foot ulcer) has proven very successful. However, the use of AM for clinical practice has several restraints when applied to patients; the most important restriction is preserving AM's therapeutic properties between its thawing and application onto the patient's wound. Moreover, AM collection and processing requires a cleanroom, together with specialized staff and equipment, and facilities that are not usually available in many hospitals and healthcare units. In this publication, we kept previously cryopreserved AM at different temperatures (37°C, 20°C, and 4°C) in different media (DMEM high glucose and saline solution with or without human albumin) and for long incubation time periods after thawing (24 h and 48 h). HaCaT keratinocytes and TGF-β1-chronified HaCaT keratinocytes were used to measure several parameters related to wound healing: migration, cell cycle arrest rescue, and the expression of key genes and migration-related proteins. Our findings indicate that AM kept in physiological saline solution at 4°C for 24 h or 48 h performed excellently in promoting HaCaT cell migration compared to AM that had been immediately thawed (0 h). Indeed, key proteins, extracellular signal-regulated kinase (ERK) and c-Jun, were induced by AM at 4°C in saline solution. Similarly, cell proliferation and different genes related to survival, inflammation, and senescence had, in all cases, the same response as to standard AM. These data suggest that the handling method in saline solution at 4°C does not interfere with AM's therapeutic properties.
Collapse
Affiliation(s)
- J Stelling-Férez
- Soluciones de Biología Molecular y Celular en Medicina Regenerativa, Health Sciences PhD Program, Universidad Católica de Murcia (UCAM), Murcia, Spain
- Regeneración Oncología Molecular y TGF-β, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - J M Puente-Cuadrado
- Regeneración Oncología Molecular y TGF-β, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - V Álvarez-Yepes
- Plataforma Sala Blanca Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
| | - S Alcaraz
- Soluciones de Biología Molecular y Celular en Medicina Regenerativa, Health Sciences PhD Program, Universidad Católica de Murcia (UCAM), Murcia, Spain
- Regeneración Oncología Molecular y TGF-β, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - E Tristante
- Plataforma Sala Blanca Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
| | - I Hernández-Mármol
- Regeneración Oncología Molecular y TGF-β, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - I Mompeán-Egea
- Plataforma Sala Blanca Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
| | - A M García-Hernández
- Plataforma Sala Blanca Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, Murcia, Spain
- Hematopoietic Transplant and Cellular Therapy Unit, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - F J Nicolás
- Regeneración Oncología Molecular y TGF-β, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| |
Collapse
|
190
|
Rouzaire M, Blanchon L, Sapin V, Gallot D. Application of Fetal Membranes and Natural Materials for Wound and Tissue Repair. Int J Mol Sci 2024; 25:11893. [PMID: 39595963 PMCID: PMC11594142 DOI: 10.3390/ijms252211893] [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: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The human fetal membrane is a globally accepted biological biomaterial for wound and tissue repair and regeneration in numerous fields, including dermatology, ophthalmology, and more recently orthopedics, maxillofacial and oral surgery, and nerve regeneration. Both cells and matrix components of amnion and chorion are beneficial, releasing a diverse range of growth factors, cytokines, peptides, and soluble extracellular matrix components. Beside fetal membranes, numerous natural materials have also been reported to promote wound healing. The biological properties of these materials may potentiate the pro-healing action of fetal membranes. Comparison of such materials with fetal membranes has been scant, and their combined use with fetal membranes has been underexplored. This review presents an up-to-date overview of (i) clinical applications of human fetal membranes in wound healing and tissue regeneration; (ii) studies comparing human fetal membranes with natural materials for promoting wound healing; and (iii) the literature on the combined use of fetal membranes and natural pro-healing materials.
Collapse
Affiliation(s)
- Marion Rouzaire
- Obstetrics and Gynaecology Department, Centre Hospitalier Universitaire Clermont-Ferrand, 63000 Clermont-Ferrand, France;
| | - Loïc Blanchon
- “Translational Approach to Epithelial Injury and Repair” Team, Auvergne University, CNRS 6293, Inserm 1103, iGReD, 63000 Clermont-Ferrand, France; (L.B.); (V.S.)
| | - Vincent Sapin
- “Translational Approach to Epithelial Injury and Repair” Team, Auvergne University, CNRS 6293, Inserm 1103, iGReD, 63000 Clermont-Ferrand, France; (L.B.); (V.S.)
- Biochemistry and Molecular Genetic Department, Centre Hospitalier Universitaire Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Denis Gallot
- Obstetrics and Gynaecology Department, Centre Hospitalier Universitaire Clermont-Ferrand, 63000 Clermont-Ferrand, France;
- “Translational Approach to Epithelial Injury and Repair” Team, Auvergne University, CNRS 6293, Inserm 1103, iGReD, 63000 Clermont-Ferrand, France; (L.B.); (V.S.)
| |
Collapse
|
191
|
Mottaghitalab F, Farokhi M. Stimulus-responsive biomacromolecule wound dressings for enhanced drug delivery in chronic wound healing: A review. Int J Biol Macromol 2024; 281:136496. [PMID: 39419149 DOI: 10.1016/j.ijbiomac.2024.136496] [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: 04/30/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Addressing the challenge of poor wound healing in chronic wounds remains complex, as the underlying physiological mechanisms are still not fully understood. Traditional wound dressings often fail to meet the specific needs of the chronic wound healing process. Recently, considerable interest has shifted toward employing biomacromolecule-based smart wound dressings to facilitate wound healing. These stimuli-responsive dressings have undergone substantial development to manage local drug delivery, demonstrating promising therapeutic effects in treating chronic wound defects. They have displayed improved drug release profiles both in vitro and in vivo. Recently, there have been advancements in the development of innovative dual and multi-stimuli responsive dressings that react to combinations of signals including pH-temperature, pH-enzyme, pH-ROS, pH-glucose, pH-NIR, and multiple stimuli. This paper offers an in-depth review of recent progress in responsive wound dressings based on biomacromolecules, with a specific focus on their design, drug release capabilities, and therapeutic advantages.
Collapse
Affiliation(s)
- Fatemeh Mottaghitalab
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
192
|
Nagaraj A, Subramaniyan Y, Surya S, Rekha PD. Burn Wound Healing Abilities of a Uronic Acid Containing Exopolysaccharide Produced by the Marine Bacterium Halomonas malpeensis YU-PRIM-29 T. Appl Biochem Biotechnol 2024; 196:8190-8213. [PMID: 38700619 DOI: 10.1007/s12010-024-04966-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 12/14/2024]
Abstract
Bacterial exopolysaccharides (EPS) are an emerging class of biopolymers with extensive applications in different fields due to their versatile physico-chemical and biological properties. The role of EPS in healing of different wound types is gaining interest in the tissue engineering sector. Burn is one of the devitalizing injuries that causes greater physical harm and can be fatal. Appropriate treatment modalities have to be followed for faster healing outcomes and to minimize the risk. In this study, a bacterial EPS (EPS-H29) from the marine bacterium Halomonas malpeensis YU-PRIM-29 T was used to treat the burn wound in vivo. The biochemical and structural characterizations of EPS-H29 were carried out using standard methods. In addition, FE-SEM, conformational, rheological, and HP-GPC analyses were carried out. In vitro biocompatibility of EPS-H29 was studied in human dermal fibroblasts (HDFs) and keratinocytes (HaCaT). Scratch assay was used to study the wound healing in vitro. For in vivo evaluation, burn wound (second-degree) was created on Wistar albino rats and treated with EPS-H29 along with appropriate control groups. The total sugar and protein contents of EPS-H29 were 72.0 ± 1.4% and 4.0 ± 0.5%, respectively, with a molecular weight of 5.2 × 105 Da. The lyophilized samples exhibited porous surface features, and in solution, it showed triple helical conformation and shear thickening behavior. In vitro cell-based assays showed biocompatibility of EPS-H29 up to 200 μg/mL concentration. At a concentration up to 50 μg/mL, EPS-H29 promoted cell proliferation. Significant increase in the HDF cell migration was evident with EPS-H29 (15 μg/mL) treatment in vitro and induced significantly higher (p ≤ 0.0001) closure of the scratch area (90.3 ± 1.1%), compared to the control (84.3 ± 1.3%) at 24 h. Enhanced expression of Ki-67 was associated with the cell proliferative activities of EPS-H29. The animals treated with EPS-H29 showed improved healing of burn wounds with significantly higher wound contraction rate (80.6 ± 9.4%) compared to the positive control (54.6 ± 8.0%) and untreated group (49.2 ± 3.7%) with histopathological evidence of epidermal tissue formation at 15 days of treatment. These results demonstrate the biocompatibility and burn wound healing capability of EPS-H29 and its potential as an effective topical agent for the burn wound care.
Collapse
Affiliation(s)
- Athmika Nagaraj
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Yuvarajan Subramaniyan
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India
| | - Suprith Surya
- Advanced Surgical Skill ENhancement Division (ASSEND), Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, India
| | - Punchappady Devasya Rekha
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India.
| |
Collapse
|
193
|
Mukai K, Nakatani T. Comparison of different modern wound dressings on full-thickness murine cutaneous wound healing with wild-type and type-2 diabetes db/db mice. J Tissue Viability 2024; 33:616-624. [PMID: 39349341 DOI: 10.1016/j.jtv.2024.09.011] [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: 12/26/2023] [Revised: 04/02/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND To evaluate the process of cutaneous wound healing, experiments have been conducted. However, to date, what modern wound dressings are suitable remains unclear. Therefore, this study aimed to compare the healing process in different modern wound dressings to determine their suitability in experimental acute wound and chronic diabetic wound. MATERIALS AND METHODS Twelve C57BL/6J mice and eleven db/db mice were subjected to full-thickness wound injuries. The mice were divided into the following four groups: hydrocolloid, form, film, and gauze groups in both wild-type and db/db mice. Wound healing was assessed until day 14. RESULTS In the wild-type groups, all wounds were healed and completed re-epithelialization by day 14. However, the wound surface was dry, and the periwound was hypercontracted in the wild-type-form and wild-type-gauze groups. In the db/db groups, wounds were not healed until day 14. Wound exudates in the db/db-hydrocolloid group were abundant and gradually increased until day 14. Wound exudates in the db/db-film group were present until day 14. Conversely, in the db/db-form and db/db-gauze groups, the wound surface was dry, and the periwound was hypercontracted. CONCLUSION These results showed that hydrocolloid and film dressings are suitable modern wound dressings for the mice wound models of acute wound and chronic diabetic wound. Moreover, using either hydrocolloid or film dressing depending on the purpose of the study on cutaneous wound healing in diabetes is necessary.
Collapse
Affiliation(s)
- Kanae Mukai
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| | - Toshio Nakatani
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
194
|
Dinić M, Verpile R, Burgess JL, Ming J, Marjanovic J, Beliz CN, Plano L, Hower S, Thaller SR, Banerjee S, Lev‐Tov H, Tomic‐Canic M, Pastar I. Multi-drug resistant Staphylococcus epidermidis from chronic wounds impair healing in human wound model. Wound Repair Regen 2024; 32:799-810. [PMID: 39439244 PMCID: PMC11584363 DOI: 10.1111/wrr.13231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Venous leg ulcers (VLUs) represent one of the most prevalent types of chronic wounds characterised by perturbed microbiome and biofilm-forming bacteria. As one of the most abundant skin-commensal, Staphylococcus epidermidis is known as beneficial for the host, however, some strains can form biofilms and hinder wound healing. In this study, S. epidermidis distribution in VLUs and associated resistome were analysed in ulcer tissue from patients. Virulence of S. epidermidis isolates from VLUs were evaluated by whole genome sequencing, antimicrobial susceptibility testing, in vitro biofilm and binding assays, and assessment of biofilm-forming capability and pro-inflammatory potential using human ex vivo wound model. We demonstrated that S. epidermidis isolates from VLUs inhibit re-epithelialization through biofilm-dependent induction of IL-1β, IL-8, and IL-6 which was in accordance with impaired healing outcomes observed in patients. High extracellular matrix binding ability of VLU isolates was associated with antimicrobial resistance and expression levels of the embp and sdrG, responsible for bacterial binding to fibrinogen and fibrin, respectively. Finally, we showed that S. epidermidis from VLUs demonstrate pathogenic features with ability to impair healing which underscores the emergence of treatment-resistant virulent lineages in patients with chronic ulcers.
Collapse
Affiliation(s)
- Miroslav Dinić
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Group for Probiotics and Microbiota‐Host Interaction, Institute of Molecular Genetics and Genetic EngineeringUniversity of BelgradeBelgradeSerbia
| | - Rebecca Verpile
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jamie L. Burgess
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jingjing Ming
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jelena Marjanovic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Carmen Nicole Beliz
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Lisa Plano
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Suzanne Hower
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Seth R. Thaller
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Santanu Banerjee
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Hadar Lev‐Tov
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Marjana Tomic‐Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
195
|
Lothstein KE, Chen F, Mishra P, Smyth DJ, Wu W, Lemenze A, Kumamoto Y, Maizels RM, Gause WC. Helminth protein enhances wound healing by inhibiting fibrosis and promoting tissue regeneration. Life Sci Alliance 2024; 7:e202302249. [PMID: 39179288 PMCID: PMC11342954 DOI: 10.26508/lsa.202302249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
Skin wound healing due to full thickness wounds typically results in fibrosis and scarring, where parenchyma tissue is replaced with connective tissue. A major advance in wound healing research would be to instead promote tissue regeneration. Helminth parasites express excretory/secretory (ES) molecules, which can modulate mammalian host responses. One recently discovered ES protein, TGF-β mimic (TGM), binds the TGF-β receptor, though likely has other activities. Here, we demonstrate that topical administration of TGM under a Tegaderm bandage enhanced wound healing and tissue regeneration in an in vivo wound biopsy model. Increased restoration of normal tissue structure in the wound beds of TGM-treated mice was observed during mid- to late-stage wound healing. Both accelerated re-epithelialization and hair follicle regeneration were observed. Further analysis showed differential expansion of myeloid populations at different wound healing stages, suggesting recruitment and reprogramming of specific macrophage subsets. This study indicates a role for TGM as a potential therapeutic option for enhanced wound healing.
Collapse
Affiliation(s)
- Katherine E Lothstein
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Fei Chen
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Pankaj Mishra
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Danielle J Smyth
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Wenhui Wu
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Alexander Lemenze
- Center for Immunity and Inflammation, Department of Pathology, Immunology, and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Yosuke Kumamoto
- Center for Immunity and Inflammation, Department of Pathology, Immunology, and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - William C Gause
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
196
|
Xiao Y, Martinez L, Zigmond Z, Woltmann D, Singer DV, Singer HA, Vazquez-Padron RI, Salman LH. Functions for platelet factor 4 (PF4/CXCL4) and its receptors in fibroblast-myofibroblast transition and fibrotic failure of arteriovenous fistulas (AVFs). J Vasc Access 2024; 25:1911-1924. [PMID: 37589266 PMCID: PMC10998683 DOI: 10.1177/11297298231192386] [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] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Over 60% of End Stage Renal Disease (ESRD) patients are relying on hemodialysis (HD) to survive, and the arteriovenous fistula (AVF) is the preferred vascular access method for HD. However approximately half of all newly created AVF fail to mature and cannot be used without a salvage procedure. We have recently demonstrated an association between AVF maturation failure and post-operative fibrosis, while our RNA-seq study also revealed that veins that ultimately failed during AVF maturation had elevated levels of platelet factor 4 (PF4/CXCL4). However, a link between these two findings was yet to be established. METHODS In this study, we investigated potential mechanisms between PF4 levels and fibrotic remodeling in veins. We compared the local expression of PF4 and fibrosis marker integrin β6 (ITGB6) in veins that successfully underwent maturation with that in veins that ultimately failed to mature. We also measured the changes of expression level of α-smooth muscle actin (αSMA/ACTA2) and collagen (Col1/COL1A1) in venous fibroblasts upon various treatments, such as PF4 pharmacological treatment, alteration of PF4 expression, and blocking of PF4 receptors. RESULTS We found that PF4 is expressed in veins and co-localizes with αSMA. In venous fibroblasts, PF4 stimulates expression of αSMA and Col1 via different pathways. The former requires integrins αvβ5 and α5β1, while chemokine receptor CXCR3 is needed for the latter. Interestingly, we also discovered that the expression of PF4 is associated with that of ITGB6, the β subunit of integrin αvβ6. This integrin is critical for the activation of the major fibrosis factor TGFβ, and overexpression of PF4 promotes activation of the TGFβ pathway. CONCLUSIONS These results indicate that upregulation of PF4 may cause venous fibrosis both directly by stimulating fibroblast differentiation and expression of extracellular matrix (ECM) molecules and indirectly by facilitating the activation of the TGFβ pathway.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Zachary Zigmond
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Daniel Woltmann
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Diane V Singer
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Harold A Singer
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Loay H Salman
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
- Division of Nephrology & Hypertension, Albany Medical College, Albany, NY, USA
| |
Collapse
|
197
|
Wang HJ, Sin CH, Yang SH, Hsueh HM, Lo WY. miR-200b-3p accelerates diabetic wound healing through anti-inflammatory and pro-angiogenic effects. Biochem Biophys Res Commun 2024; 731:150388. [PMID: 39024974 DOI: 10.1016/j.bbrc.2024.150388] [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: 04/01/2024] [Revised: 06/20/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024]
Abstract
The poor healing characteristics of diabetic foot ulcers are partially attributed to diabetes-induced pro-inflammatory wounds. Our previous study reported that both miR-146a-5p and miR-200b-3p decrease endothelial inflammation in human aortic endothelial cells and db/db diabetic mice. Although miR-146a-5p has been reported to improve diabetic wound healing, the role of miR-200b-3p is not clear. This study compared the roles of these miRNAs in diabetic wound healing. Two 8-mm full-thickness wounds were created in 12-week-old male db/db mice on the left and right back. After surgery, 100 ng miR-146a-5p, miR-200b-3p, or miR-negative control (NC) was injected in each wound. Full-thickness skin samples were harvested from mice at the 14th day for real-time polymerase chain reaction and immunohistochemistry analyses. At the 14th day, the miR-200b-3p group showed better wound healing and greater granulation tissue thickness than the miR-146a-5p group. The miR-200b-3p group showed a significant decrease of IL-6 and IL-1β gene expression and a significant increase of Col3α1 gene expression compared to those in the miR-NC group. The miR-200b-3p group had the lowest gene expression of TGF-β1, followed by the miR-146a-5p and miR-NC groups. Our findings suggest that the miR-200b-3p group had better healing characteristics than the other two groups. Immunohistochemical staining revealed that CD68 immunoreactivity was significantly decreased in both the miR-146a-5p and miR-200b-3p groups compared with that in the miR-NC group. In addition, CD31 immunoreactivity was significantly higher in the miR-200b-3p group than in the miR-146a-5p group. In conclusion, these results suggest that miR-200b-3p is more effective than miR-146a-5p in promoting diabetic wound healing through its anti-inflammatory and pro-angiogenic effects.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Animals
- Wound Healing/genetics
- Male
- Mice
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/genetics
- Diabetic Foot/genetics
- Diabetic Foot/metabolism
- Diabetic Foot/pathology
- Neovascularization, Physiologic/genetics
- Interleukin-6/metabolism
- Interleukin-6/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Interleukin-1beta/metabolism
- Interleukin-1beta/genetics
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Skin/metabolism
- Skin/pathology
- Inflammation/genetics
- Inflammation/pathology
- Inflammation/metabolism
- Mice, Inbred C57BL
- CD68 Molecule
Collapse
Affiliation(s)
- Huang-Joe Wang
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung City 404327, Taiwan; School of Medicine, China Medical University, No. 91, Xueshi Rd., North Dist., Taichung City 404328, Taiwan
| | - Cian-Huei Sin
- Department of Life Science, National Chung Hsing University, No. 145, Xingda Rd., South Dist., Taichung City 402202, Taiwan
| | - Shang-Hsuan Yang
- Shiny Brands Group, 7F, No. 311, Fuxing N. Rd., Songshan Dist., Taipei, 10544, Taiwan
| | - Hsiang-Ming Hsueh
- Shiny Brands Group, 7F, No. 311, Fuxing N. Rd., Songshan Dist., Taipei, 10544, Taiwan
| | - Wan-Yu Lo
- Cardiovascular & Translational Medicine Laboratory, Department of Food Science and Technology, Hungkuang University, No. 1018, Sec. 6, Taiwan Blvd., Shalu Dist., Taichung City 43302, Taiwan.
| |
Collapse
|
198
|
Ye Z, Sun L, Xiang Q, Hao Y, Liu H, He Q, Yang X, Liao W. Advancements of Biomacromolecular Hydrogel Applications in Food Nutrition and Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23689-23708. [PMID: 39410660 DOI: 10.1021/acs.jafc.4c05903] [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: 11/01/2024]
Abstract
Hydrogels exhibit remarkable degradability, biocompatibility and functionality, which position them as highly promising materials for applications within the food and pharmaceutical industries. Although many relevant studies on hydrogels have been reported in the chemical industry, materials, and other fields, there have been few reviews on their potential applications in food nutrition and human health. This study aims to address this gap by reviewing the functional properties of hydrogels and assessing their value in terms of food nutrition and human health. The use of hydrogels in preserving bioactive ingredients, food packaging and food distribution is delved into specifically in this review. Hydrogels can serve as cutting-edge materials for food packaging and delivery, ensuring the preservation of nutritional activity within food products, facilitating targeted delivery of bioactive compounds and regulating the digestion and absorption processes in the human body, thereby promoting human health. Moreover, hydrogels find applications in in vitro cell and tissue culture, human tissue repair, as well as chronic disease prevention and treatment. These broad applications have attracted great attention in the fields of human food nutrition and health. Ultimately, this paper serves as a valuable reference for further utilization and exploration of hydrogels in these respective fields.
Collapse
Affiliation(s)
- Zichong Ye
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Linye Sun
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Qianru Xiang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Hongji Liu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Qi He
- Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, P. R. China
| | - Xingfen Yang
- Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, P. R. China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| |
Collapse
|
199
|
Huang J, Sati S, Murphy C, Spencer CA, Rapp E, Prouty SM, Korte S, Ahart O, Sheng E, Jones P, Kersh AE, Leung D, Leung TH. Granulocyte colony stimulating factor promotes scarless tissue regeneration. Cell Rep 2024; 43:114742. [PMID: 39306847 PMCID: PMC11574610 DOI: 10.1016/j.celrep.2024.114742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/19/2024] [Accepted: 08/27/2024] [Indexed: 10/26/2024] Open
Abstract
Mammals typically heal with fibrotic scars, and treatments to regenerate human skin and hair without a scar remain elusive. We discovered that mice lacking C-X-C motif chemokine receptor 2 (CXCR2 knockout [KO]) displayed robust and complete tissue regeneration across three different injury models: skin, hair follicle, and cartilage. Remarkably, wild-type mice receiving plasma from CXCR2 KO mice through parabiosis or injections healed wounds scarlessly. A comparison of circulating proteins using multiplex ELISA revealed a 24-fold higher plasma level of granulocyte colony stimulating factor (G-CSF) in CXCR2 KO blood. Local injections of G-CSF into wild-type (WT) mouse wound beds reduced scar formation and increased scarless tissue regeneration. G-CSF directly polarized macrophages into an anti-inflammatory phenotype, and both CXCR2 KO and G-CSF-treated mice recruited more anti-inflammatory macrophages into injured areas. Modulating macrophage activation states at early time points after injury promotes scarless tissue regeneration and may offer a therapeutic approach to improve healing of human skin wounds.
Collapse
Affiliation(s)
- Jianhe Huang
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Satish Sati
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Christina Murphy
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Casey A Spencer
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Emmanuel Rapp
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Stephen M Prouty
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Scott Korte
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Olivia Ahart
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Emily Sheng
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Parker Jones
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Anna E Kersh
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Denis Leung
- Singapore Management University, Singapore, Singapore
| | - Thomas H Leung
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA; Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA.
| |
Collapse
|
200
|
Wang X, Zheng Z, Zhang Y, Sun J, Liu J, Liu Y, Ding G. Application of hydrogel-loaded dental stem cells in the field of tissue regeneration. Hum Cell 2024; 38:2. [PMID: 39436502 DOI: 10.1007/s13577-024-01134-2] [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: 07/05/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Mesenchymal stem cells (MSCs) are highly favored in clinical trials due to their unique characteristics, which have isolated from various human tissues. Derived from dental tissues, dental stem cells (DSCs) are particularly notable for their applications in tissue repair and regenerative medicine, attributed to their readily available sources, absence of ethical controversies, and minimal immunogenicity. Hydrogel-loaded stem cell therapy is widespread across a variety of injuries and diseases, and has good repair capabilities for both soft and hard tissues. This review comprehensively summarizes the regenerative and differentiation potential of various DSCs encapsulated in hydrogels across different tissues. In addition, the existing problems and future direction are also addressed. The application of hydrogel-DSCs composite has gained substantial progress in the field of tissue regeneration and need in-depth study in the future.
Collapse
Affiliation(s)
- Xiaolan Wang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Ying Zhang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Jinmeng Sun
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Jian Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Yunxia Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China.
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China.
| |
Collapse
|