1
|
Huang L, Chen H, Nie J, Zhao Y, Miao J. Advanced dressings based on novel biological targets for diabetic wound healing: A review. Eur J Pharmacol 2025; 987:177201. [PMID: 39667426 DOI: 10.1016/j.ejphar.2024.177201] [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: 07/15/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
The diabetic wound is one of the most common complications of diabetes in clinic. The existing diabetic wound dressings all have bottlenecks in decreasing inflammation, stopping peripheral neuropathy, relieving local ischemia and hypoxia in diabetic wounds. These challenges are intricately linked to the roles of various growth factors, as well as matrix metalloproteinases. Thus, a comprehensive understanding of growth factors-particularly their dynamic interactions with the extracellular matrix (ECM) and cellular components-is essential. Cells and proteins that influence the synthesis of growth factors and matrix metalloproteinases emerge as potential therapeutic targets for diabetic wound management. This review discusses the latest advancements in the pathophysiology of diabetic wound healing, highlights novel biological targets, and evaluates new wound dressing strategies designed for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Lantian Huang
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310052, China; Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hangbo Chen
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310052, China; Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jing Nie
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310052, China; Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yingzheng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China.
| | - Jing Miao
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310052, China; Research Center for Clinical Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Tibatan MA, Katana D, Yin CM. The emerging role of nanoscaffolds in chronic diabetic wound healing: a new horizon for advanced therapeutics. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-32. [PMID: 39291361 DOI: 10.1080/09205063.2024.2402148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
Non-healing or chronic wounds in extremities that lead to amputations in patients with Type II diabetes (hyperglycemia) are among the most serious and common health problems in the modern world. Over the past decade, more efficient solutions for diabetic ulcers have been developed. Nanofibers and/or composite materials capable of drug delivery, moisture control, and antibacterial effectiveness are increasingly utilized in the formulation of wound dressings, with a particular focus on the biofunctionalization of polymeric and hydrogel materials. Natural products, including plant extracts, honey, antibacterial agents, nanozymes, and metal nanoparticles, are now commonly and effectively implemented to enhance the functionality of wound dressings. Due to the complicated and dysfunctional physiological structure of the chronic wound sites in the extremities of diabetic patients, formulated nanoscaffold or hydrogel components are becoming more intricate and versatile. This study aimed to investigate the development of wound dressing materials over the years while demonstrating their progressively enhanced complexity in effectively targeting, treating, and managing chronic wounds. The mechanisms of action and bio-functionality of wound dressing technologies were elucidated based on findings from 290 studies conducted over the last decade. A notable observation that emerged from these studies is the evolution of wound dressing development technology, which has led to significant advancements in the operational range of smart systems. These include, but are not limited to, self-healing, self-oxygenation, and adaptable mimicry of human tissue.
Collapse
Affiliation(s)
| | - Dzana Katana
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Casey M Yin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Rehan IF, Elnagar A, Zigo F, Sayed-Ahmed A, Yamada S. Biomimetic strategies for the deputization of proteoglycan functions. Front Cell Dev Biol 2024; 12:1391769. [PMID: 39170918 PMCID: PMC11337302 DOI: 10.3389/fcell.2024.1391769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Proteoglycans (PGs), which have glycosaminoglycan chains attached to their protein cores, are essential for maintaining the morphology and function of healthy body tissues. Extracellular PGs perform various functions, classified into the following four categories: i) the modulation of tissue mechanical properties; ii) the regulation and protection of the extracellular matrix; iii) protein sequestration; and iv) the regulation of cell signaling. The depletion of PGs may significantly impair tissue function, encompassing compromised mechanical characteristics and unregulated inflammatory responses. Since PGs play critical roles in the function of healthy tissues and their synthesis is complex, the development of PG mimetic molecules that recapitulate PG functions for tissue engineering and therapeutic applications has attracted the interest of researchers for more than 20 years. These approaches have ranged from semisynthetic graft copolymers to recombinant PG domains produced by cells that have undergone genetic modifications. This review discusses some essential extracellular PG functions and approaches to mimicking these functions.
Collapse
Affiliation(s)
- Ibrahim F. Rehan
- Department of Husbandry and Development of Animal Wealth, Faculty of Veterinary Medicine, Menoufia University, Shebin Alkom, Egypt
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Asmaa Elnagar
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - František Zigo
- Department of Animal Nutrition and Husbandry, University of Veterinary Medicine and Pharmacy, Košice, Slovakia
| | - Ahmed Sayed-Ahmed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Menoufia University, Shebin Alkom, Egypt
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| |
Collapse
|
4
|
Pal D, Das P, Mukherjee P, Roy S, Chaudhuri S, Kesh SS, Ghosh D, Nandi SK. Biomaterials-Based Strategies to Enhance Angiogenesis in Diabetic Wound Healing. ACS Biomater Sci Eng 2024; 10:2725-2741. [PMID: 38630965 DOI: 10.1021/acsbiomaterials.4c00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Amidst the present healthcare issues, diabetes is unique as an emerging class of affliction with chronicity in a majority of the population. To check and control its effects, there have been huge turnover and constant development of management strategies, and though a bigger part of the health care area is involved in achieving its control and the related issues such as the effect of diabetes on wound healing and care and many of the works have reached certain successful outcomes, still there is a huge lack in managing it, with maximum effect yet to be attained. Studying pathophysiology and involvement of various treatment options, such as tissue engineering, application of hydrogels, drug delivery methods, and enhancing angiogenesis, are at constantly developing stages either direct or indirect. In this review, we have gathered a wide field of information and different new therapeutic methods and targets for the scientific community, paving the way toward more settled ideas and research advances to cure diabetic wounds and manage their outcomes.
Collapse
Affiliation(s)
- Debajyoti Pal
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Pratik Das
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Prasenjit Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Subhasis Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Shubhamitra Chaudhuri
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Shyam Sundar Kesh
- Department of Veterinary Clinical Complex, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Debaki Ghosh
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal & Fishery Sciences, Kolkata 700037, India
| |
Collapse
|
5
|
Mari W, Younes S, Sheehan E, Oroszi TL, Cool DR, Suliman R, Simman R. Wound Fluid Extracellular Microvesicles: A Potential Innovative Biomarker for Wound Healing. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e5781. [PMID: 38706469 PMCID: PMC11068147 DOI: 10.1097/gox.0000000000005781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/12/2024] [Indexed: 05/07/2024]
Abstract
Background Extracellular vesicles, or microvesicles, are a large family of membrane-bound fluid-filled sacs that cells release into the extracellular environment. Extracellular microvesicles (EMVs) are essential for cell-to-cell communications that promote wound healing. We hypothesize a correlation between the concentration of EMVs in wound fluid and the percentage of wound healing in treated chronic, nonhealing, wounds. A prospective, multicenter, randomized, single-blind clinical trial was conducted to evaluate EMV concentration in relation to wound healing percentages. Methods Wound fluid samples were obtained from 16 patients with stage IV trunk pressure ulcers. Patients were divided equally into two groups: (1) control group on negative pressure wound therapy (NPWT) alone and (2) study group with NPWT plus porcine extracellular matrix dressing. NPWT was replaced two times a week, and porcine extracellular matrix applied once weekly for all subjects. An NPWT canister device, called a wound vacuum-assisted closure, containing wound fluid was collected from each patient every 4 weeks. EMVs were isolated and the concentration measured by nanoparticle tracking analysis. Results The study group's total healing percentage was around 89% after 12 weeks compared with the control group's percentage of about 52% (P ≤ 0.05). Using R programming software, simple linear regression was carried out to investigate the hypothesis. Data demonstrated significant positive correlation (R2 = 0.70; P = 0.05) between EMV concentrations and the healing percentage. Conclusions There is a positive correlation between EMV concentration and wound healing percentages. Results propose that the EMVs in wound fluid could serve as a biomarker for healing.
Collapse
Affiliation(s)
- Walid Mari
- From the Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Sara Younes
- From the Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Erin Sheehan
- College of Medicine and Life Science, University of Toledo, Toledo, Ohio
| | - Terry L Oroszi
- From the Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - David R Cool
- From the Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Rajab Suliman
- Department of Information, Operation and Technology Management, John B. and Lillian E. Neff College of Business and Innovation, University of Toledo, Toledo, Ohio
| | - Richard Simman
- Department of Surgery, College of Medicine and Life Science, University of Toledo, Toledo, Ohio
- Wound Care Program, ProMedica Health Network, Jobst Vascular Institute, Toledo, Ohio
| |
Collapse
|
6
|
Sadeghi-Ardebili M, Hasannia S, Dabirmanesh B, Khavari-Nejad RA. Functional characterization of the dimeric form of PDGF-derived fusion peptide fabricated based on theoretical arguments. Sci Rep 2024; 14:1003. [PMID: 38200288 PMCID: PMC10781716 DOI: 10.1038/s41598-024-51707-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/08/2024] [Indexed: 01/12/2024] Open
Abstract
A skin wound leads to the loss of skin integrity and the influx of pathogens into the tissue. Platelet-derived growth factors (PDGFs) are cytokines released from alpha granules during wound healing and interact with their cell surface receptors and activate signals involved in chemotaxis, growth, proliferation, and differentiation pathways. Due to the low stability of growth factors (GFs), a new peptide-derived PDGF-BB was designed, expressed in the Shuffle strain of E. coli, and purified by Ni-NTA agarose affinity column chromatography. The effect of fusion peptide was then evaluated on L929 fibroblast cells and animal models with skin lesions. In vitro, studies showed that the peptide led to an increase in the migration of fibroblast cells in the scratch assay. Its positive effect on wound healing was also observed in the skin-injured rats after 3, 7, and 12 days. A significant rise in neutrophils and granular tissue formation, re-epithelialization, angiogenesis, and collagen formation was exhibited on the third day of treatment when compared to the control group. The results showed that, despite reducing PDGF size, the fusion peptide was able to maintain at least some of the known functions attributed to full-length PDGF and showed positive results in wound healing.
Collapse
Affiliation(s)
- Maryam Sadeghi-Ardebili
- Department of Biology, Science and Research Branch, Islamic Azad University, PO BoX 14515-775, Tehran, Iran
| | - Sadegh Hasannia
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, PO Box 14115-175, Tehran, Iran.
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, PO Box 14115-175, Tehran, Iran
| | - Ramazan Ali Khavari-Nejad
- Department of Biology, Science and Research Branch, Islamic Azad University, PO BoX 14515-775, Tehran, Iran
| |
Collapse
|
7
|
Matwiejuk M, Myśliwiec H, Chabowski A, Flisiak I. An Overview of Growth Factors as the Potential Link between Psoriasis and Metabolic Syndrome. J Clin Med 2023; 13:109. [PMID: 38202116 PMCID: PMC10780265 DOI: 10.3390/jcm13010109] [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: 10/31/2023] [Revised: 11/26/2023] [Accepted: 12/05/2023] [Indexed: 01/12/2024] Open
Abstract
Psoriasis is a chronic, complex, and immunologically mediated systemic disease that not only affects the skin, but also the joints and nails. It may coexist with various other disorders, such as depression, psoriatic arthritis, cardiovascular diseases, diabetes mellitus, and metabolic syndrome. In particular, the potential link between psoriasis and metabolic syndrome is an issue worthy of attention. The dysregulation of growth factors could potentially contribute to the disturbances of keratinocyte proliferation, inflammation, and itch severity. However, the pathophysiology of psoriasis and its comorbidities, such as metabolic syndrome, remains incompletely elucidated. Growth factors and their abnormal metabolism may be a potential link connecting these conditions. Overall, the objective of this review is to analyze the role of growth factor disturbances in both psoriasis and metabolic syndrome.
Collapse
Affiliation(s)
- Mateusz Matwiejuk
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Hanna Myśliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Iwona Flisiak
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
8
|
Valdivia A, Avalos AM, Leyton L. Thy-1 (CD90)-regulated cell adhesion and migration of mesenchymal cells: insights into adhesomes, mechanical forces, and signaling pathways. Front Cell Dev Biol 2023; 11:1221306. [PMID: 38099295 PMCID: PMC10720913 DOI: 10.3389/fcell.2023.1221306] [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: 05/12/2023] [Accepted: 09/25/2023] [Indexed: 12/17/2023] Open
Abstract
Cell adhesion and migration depend on the assembly and disassembly of adhesive structures known as focal adhesions. Cells adhere to the extracellular matrix (ECM) and form these structures via receptors, such as integrins and syndecans, which initiate signal transduction pathways that bridge the ECM to the cytoskeleton, thus governing adhesion and migration processes. Integrins bind to the ECM and soluble or cell surface ligands to form integrin adhesion complexes (IAC), whose composition depends on the cellular context and cell type. Proteomic analyses of these IACs led to the curation of the term adhesome, which is a complex molecular network containing hundreds of proteins involved in signaling, adhesion, and cell movement. One of the hallmarks of these IACs is to sense mechanical cues that arise due to ECM rigidity, as well as the tension exerted by cell-cell interactions, and transduce this force by modifying the actin cytoskeleton to regulate cell migration. Among the integrin/syndecan cell surface ligands, we have described Thy-1 (CD90), a GPI-anchored protein that possesses binding domains for each of these receptors and, upon engaging them, stimulates cell adhesion and migration. In this review, we examine what is currently known about adhesomes, revise how mechanical forces have changed our view on the regulation of cell migration, and, in this context, discuss how we have contributed to the understanding of signaling mechanisms that control cell adhesion and migration.
Collapse
Affiliation(s)
- Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
9
|
Wang F, Wang P, Yang H, Cai R, Tan W. Self-Powered Biosensing System with Multivariate Signal Amplification for Real-Time Amplified Detection of PDGF-BB. Anal Chem 2023; 95:16359-16365. [PMID: 37889605 DOI: 10.1021/acs.analchem.3c03662] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
A self-powered biosensing system with multivariate signal amplification is designed for the ultrasensitive, highly efficient, rapid-response, and real-time detection of platelet-derived growth factor-BB (PDGF-BB). The biosensing system is composed of enzymatic biofuel cells (EBFCs), a capacitor, a digital multimeter (DMM), and a computer. Using the hybridization chain reaction (HCR), a few single DNA chains are transformed into abundant double-helix chains, which stimulates the reduction of [Ru(NH3)6]3+ to [Ru(NH3)6]2+ by electrostatic interaction, corresponding to the "on" state for HCR. As a result, the open-circuit voltage (EOCV) is significantly increased in this self-powered biosensing system. When PDGF-BB is present, a binding interaction between the target and the aptamer, i.e., PDGF-BB/Apt, corresponding to the "off" state for HCR, results in a decrease of EOCV. The PDGF-BB concentration is inversely proportional to EOCV, allowing readable, effective, and precise real-time detection of PDGF-BB. The detection limit of the biosensing system is 0.031 pg/mL (S/N = 3). This strategy provides a promising and powerful tool for the early clinical diagnosis of related colorectal cancer markers.
Collapse
Affiliation(s)
- Futing Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Material Science and Engineering, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Peng Wang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Hongfen Yang
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Ren Cai
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Material Science and Engineering, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Material Science and Engineering, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- School of Medicine, and College of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
10
|
Sligar AD, Howe G, Goldman J, Felli P, Gómez-Hernández A, Takematsu E, Veith A, Desai S, Riley WJ, Singeetham R, Mei L, Callahan G, Ashirov D, Smalling R, Baker AB. Syndecan-4 Proteoliposomes Enhance Revascularization in a Rabbit Hind Limb Ischemia Model of Peripheral Ischemia. Acta Biomater 2023:S1742-7061(23)00331-8. [PMID: 37321528 DOI: 10.1016/j.actbio.2023.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023]
Abstract
Regenerative therapeutics for treating peripheral arterial disease are an appealing strategy for creating more durable solutions for limb ischemia. In this work, we performed preclinical testing of an injectable formulation of syndecan-4 proteoliposomes combined with growth factors as treatment for peripheral ischemia delivered in an alginate hydrogel. We tested this therapy in an advanced model of hindlimb ischemia in rabbits with diabetes and hyperlipidemia. Our studies demonstrate enhancement in vascularity and new blood vessel growth with treatment with syndecan-4 proteoliposomes in combination with FGF-2 or FGF-2/PDGF-BB. The effects of the treatments were particularly effective in enhancing vascularity in the lower limb with a 2-4 increase in blood vessels in the treatment group in comparison to the control group. In addition, we demonstrate that the syndecan-4 proteoliposomes have stability for at least 28 days when stored at 4°C to allow transport and use in the hospital environment. In addition, we performed toxicity studies in the mice and found no toxic effects even when injected at high concentration. Overall, our studies support that syndecan-4 proteoliposomes markedly enhance the therapeutic potential of growth factors in the context of disease and may be promising therapeutics for inducing vascular regeneration in peripheral ischemia. STATEMENT OF SIGNIFICANCE: Peripheral ischemia is a common condition in which there is a lack of blood flow to the lower limbs. This condition can lead to pain while walking and, in severe cases, critical limb ischemia and limb loss. In this study, we demonstrate the safety and efficacy of a novel injectable therapy for enhancing revascularization in peripheral ischemia using an advanced large animal model of peripheral vascular disease using rabbits with hyperlipidemia and diabetes.
Collapse
Affiliation(s)
- Andrew D Sligar
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Gretchen Howe
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Texas Medical School at Houston, TX
| | - Julia Goldman
- Center for Laboratory Animal Medicine and Care, UT Health Science Center at Houston
| | - Patricia Felli
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Texas Medical School at Houston, TX
| | - Almudena Gómez-Hernández
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Eri Takematsu
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Austin Veith
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Shubh Desai
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - William J Riley
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Rohan Singeetham
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Lei Mei
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Gregory Callahan
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - David Ashirov
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Richard Smalling
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Texas Medical School at Houston, TX; Memorial Hermann Heart and Vascular Institute, Houston, TX
| | - Aaron B Baker
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX; The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX.
| |
Collapse
|
11
|
Wang F, Ye Y, Zhang Z, Teng W, Sun H, Chai X, Zhou X, Chen J, Mou H, Eloy Y, Jin X, Chen L, Shao Z, Wu Y, Shen Y, Liu A, Lin P, Wang J, Yu X, Ye Z. PDGFR in PDGF-BB/PDGFR Signaling Pathway Does Orchestrates Osteogenesis in a Temporal Manner. RESEARCH (WASHINGTON, D.C.) 2023; 6:0086. [PMID: 37223474 PMCID: PMC10202377 DOI: 10.34133/research.0086] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/13/2023] [Indexed: 12/01/2023]
Abstract
Platelet-derived growth factor-BB (PDGF-BB)/platelet-derived growth factor receptor-β (PDGFR-β) pathway is conventionally considered as an important pathway to promote osteogenesis; however, recent study suggested its role during osteogenesis to be controversial. Regarding the differential functions of this pathway during 3 stages of bone healing, we hypothesized that temporal inhibition of PDGF-BB/PDGFR-β pathway could shift the proliferation/differentiation balance of skeletal stem and progenitor cells, toward osteogenic lineage, which leads to improved bone regeneration. We first validated that inhibition of PDGFR-β at late stage of osteogenic induction effectively enhanced differentiation toward osteoblasts. This effect was also replicated in vivo by showing accelerated bone formation when block PDGFR-β pathway at late stage of critical bone defect healing mediated using biomaterials. Further, we found that such PDGFR-β inhibitor-initiated bone healing was also effective in the absence of scaffold implantation when administrated intraperitoneally. Mechanistically, timely inhibition of PDGFR-β blocked extracellular regulated protein kinase 1/2 pathway, which shift proliferation/differentiation balance of skeletal stem and progenitor cell to osteogenic lineage by upregulating osteogenesis-related products of Smad to induce osteogenesis. This study offered updated understanding of the use of PDGFR-β pathway and provides new insight routes of action and novel therapeutic methods in the field of bone repair.
Collapse
Affiliation(s)
- Fangqian Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Yuxiao Ye
- School of Material Science and Engineering, University of New South Wales, Sydney 2052, Australia
| | - Zengjie Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Wangsiyuan Teng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Hangxiang Sun
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xupeng Chai
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xingzhi Zhou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jiayu Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Haochen Mou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Yinwang Eloy
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xiaoqiang Jin
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Liang Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Zhenxuan Shao
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Yan Wu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Yue Shen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - An Liu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Peng Lin
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jianwei Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xiaohua Yu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Zhaoming Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| |
Collapse
|
12
|
Dzierżyńska M, Sawicka J, Deptuła M, Sosnowski P, Sass P, Peplińska B, Pietralik-Molińska Z, Fularczyk M, Kasprzykowski F, Zieliński J, Kozak M, Sachadyn P, Pikuła M, Rodziewicz-Motowidło S. Release systems based on self-assembling RADA16-I hydrogels with a signal sequence which improves wound healing processes. Sci Rep 2023; 13:6273. [PMID: 37072464 PMCID: PMC10113214 DOI: 10.1038/s41598-023-33464-w] [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: 01/06/2023] [Accepted: 04/13/2023] [Indexed: 05/03/2023] Open
Abstract
Self-assembling peptides can be used for the regeneration of severely damaged skin. They can act as scaffolds for skin cells and as a reservoir of active compounds, to accelerate scarless wound healing. To overcome repeated administration of peptides which accelerate healing, we report development of three new peptide biomaterials based on the RADA16-I hydrogel functionalized with a sequence (AAPV) cleaved by human neutrophil elastase and short biologically active peptide motifs, namely GHK, KGHK and RDKVYR. The peptide hybrids were investigated for their structural aspects using circular dichroism, thioflavin T assay, transmission electron microscopy, and atomic force microscopy, as well as their rheological properties and stability in different fluids such as water or plasma, and their susceptibility to digestion by enzymes present in the wound environment. In addition, the morphology of the RADA-peptide hydrogels was examined with a unique technique called scanning electron cryomicroscopy. These experiments enabled us to verify if the designed peptides increased the bioactivity of the gel without disturbing its gelling processes. We demonstrate that the physicochemical properties of the designed hybrids were similar to those of the original RADA16-I. The materials behaved as expected, leaving the active motif free when treated with elastase. XTT and LDH tests on fibroblasts and keratinocytes were performed to assess the cytotoxicity of the RADA16-I hybrids, while the viability of cells treated with RADA16-I hybrids was evaluated in a model of human dermal fibroblasts. The hybrid peptides revealed no cytotoxicity; the cells grew and proliferated better than after treatment with RADA16-I alone. Improved wound healing following topical delivery of RADA-GHK and RADA-KGHK was demonstrated using a model of dorsal skin injury in mice and histological analyses. The presented results indicate further research is warranted into the engineered peptides as scaffolds for wound healing and tissue engineering.
Collapse
Affiliation(s)
- Maria Dzierżyńska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Justyna Sawicka
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdańsk, Gdańsk, Poland
| | - Paweł Sosnowski
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Piotr Sass
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | | | | | - Martyna Fularczyk
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | | | - Jacek Zieliński
- Department of Surgical Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Maciej Kozak
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Poznań, Poland
| | - Paweł Sachadyn
- Laboratory for Regenerative Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
13
|
Massidda MW, Im B, Lee J, Baker AB. Mechanical conditioning of human mesenchymal stem cells for enhancing vascular regeneration. STAR Protoc 2023; 4:102103. [PMID: 36853695 PMCID: PMC9929623 DOI: 10.1016/j.xpro.2023.102103] [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: 10/11/2022] [Revised: 12/14/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are an appealing cell type for therapeutic applications but remain limited by poor efficacy in clinical trials. Here, we describe a conditioning technique that enhances the vascular regenerative properties of hMSCs and increases their expression of endothelial cell and pericyte markers. We also describe an alginate gel encapsulation protocol for delivering the conditioned cells. For complete details on the use and execution of this protocol, please refer to Lee et al. (2021).1.
Collapse
Affiliation(s)
- Miles W Massidda
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, USA
| | - ByungGee Im
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, USA
| | - Jason Lee
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, USA
| | - Aaron B Baker
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA; The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
14
|
Wishart TFL, Lovicu FJ. Heparan sulfate proteoglycans (HSPGs) of the ocular lens. Prog Retin Eye Res 2023; 93:101118. [PMID: 36068128 DOI: 10.1016/j.preteyeres.2022.101118] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) reside in most cells; on their surface, in the pericellular milieu and/or extracellular matrix. In the eye, HSPGs can orchestrate the activity of key signalling molecules found in the ocular environment that promote its development and homeostasis. To date, our understanding of the specific roles played by individual HSPG family members, and the heterogeneity of their associated sulfated HS chains, is in its infancy. The crystalline lens is a relatively simple and well characterised ocular tissue that provides an ideal stage to showcase and model the expression and unique roles of individual HSPGs. Individual HSPG core proteins are differentially localised to eye tissues in a temporal and spatial developmental- and cell-type specific manner, and their loss or functional disruption results in unique phenotypic outcomes for the lens, and other ocular tissues. More recent work has found that different HS sulfation enzymes are also presented in a cell- and tissue-specific manner, and that disruption of these different sulfation patterns affects specific HS-protein interactions. Not surprisingly, these sulfated HS chains have also been reported to be required for lens and eye development, with dysregulation of HS chain structure and function leading to pathogenesis and eye-related phenotypes. In the lens, HSPGs undergo significant and specific changes in expression and function that can drive pathology, or in some cases, promote tissue repair. As master signalling regulators, HSPGs may one day serve as valuable biomarkers, and even as putative targets for the development of novel therapeutics, not only for the eye but for many other systemic pathologies.
Collapse
Affiliation(s)
- Tayler F L Wishart
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, NSW, Australia.
| | - Frank J Lovicu
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, NSW, Australia; Save Sight Institute, The University of Sydney, NSW, Australia.
| |
Collapse
|
15
|
Tang X, He T, Li X, Liu Y, Wu Y, You G, Li J, Yun Y, Wu L, Li L, Kang J. Clinical features and independent predictors of postoperative refractory trauma to anal fistula combined with T2DM: A propensity score-matched analysis-retrospective cohort study. Front Surg 2023; 10:1119113. [PMID: 36911620 PMCID: PMC9998506 DOI: 10.3389/fsurg.2023.1119113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 02/26/2023] Open
Abstract
Background Refractory wound is a common postoperative complication in anal fistula surgery, when combined with type 2 diabetes mellitus (T2DM) it presents a slower recovery time and more complex wound physiology. The study aims to investigate factors associated with wound healing in patients with T2DM. Materials and methods 365 T2DM patients who underwent anal fistula surgery at our institution were recruited from June 2017 to May 2022. Through propensity score-matched (PSM) analysis, multivariate logistic regression analysis was applied to determine independent risk factors affecting wound healing. Results 122 pairs of patients with no significant differences were successfully established in matched variables. Multivariate logistic regression analysis revealed that uric acid (OR: 1.008, 95% CI: 1.002-1.015, p = 0.012), maximal fasting blood glucose (FBG) (OR: 1.489, 95% CI: 1.028-2.157, p = 0.035) and random intravenous blood glucose (OR: 1.130, 95% CI: 1.008-1.267, p = 0.037) elevation and the incision at 5 o'clock under the lithotomy position (OR: 3.510, 95% CI: 1.214-10.146, p = 0.020) were independent risk factors for impeding wound healing. However, neutrophil percentage fluctuating within the normal range can be considered as an independent protective factor (OR: 0.906, 95% CI: 0.856-0.958, p = 0.001). After executing the receiver operating characteristic (ROC) curve analysis, it was found that the maximum FBG expressed the largest under curve area (AUC), glycosylated hemoglobin (HbA1c) showed the strongest sensitivity at the critical value and maximum postprandial blood glucose (PBG) had the highest specificity at the critical value. To promote high-quality healing of anal wounds in diabetic patients, clinicians should not only pay attention to surgical procedures but also take above-mentioned indicators into consideration.
Collapse
Affiliation(s)
- Xiao Tang
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Taohong He
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyi Li
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ya Liu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuqi Wu
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gehang You
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Li
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Yun
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lei Wu
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Li
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jian Kang
- Department of Proctology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Tong S, Li Q, Liu Q, Song B, Wu J. Recent advances of the nanocomposite hydrogel as a local drug delivery for diabetic ulcers. Front Bioeng Biotechnol 2022; 10:1039495. [PMID: 36267448 PMCID: PMC9577098 DOI: 10.3389/fbioe.2022.1039495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic ulcer is a serious complication of diabetes. Compared with that of healthy people, the skin of patients with a diabetic ulcer is more easily damaged and difficult to heal. Without early intervention, the disease will become increasingly serious, often leading to amputation or even death. Most current treatment methods cannot achieve a good wound healing effect. Numerous studies have shown that a nanocomposite hydrogel serves as an ideal drug delivery method to promote the healing of a diabetic ulcer because of its better drug loading capacity and stability. Nanocomposite hydrogels can be loaded with one or more drugs for application to chronic ulcer wounds to promote rapid wound healing. Therefore, this paper reviews the latest progress of delivery systems based on nanocomposite hydrogels in promoting diabetic ulcer healing. Through a review of the recent literature, we put forward the shortcomings and improvement strategies of nanocomposite hydrogels in the treatment of diabetic ulcers.
Collapse
Affiliation(s)
- Sen Tong
- School of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qingyu Li
- School of Medicine, Jianghan University, Wuhan, China
| | - Qiaoyan Liu
- School of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Bo Song
- School of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- *Correspondence: Bo Song, ; Junzi Wu,
| | - Junzi Wu
- School of Basic Medical, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- *Correspondence: Bo Song, ; Junzi Wu,
| |
Collapse
|
17
|
Takematsu E, Massidda M, Auster J, Chen PC, Im B, Srinath S, Canga S, Singh A, Majid M, Sherman M, Dunn A, Graham A, Martin P, Baker AB. Transmembrane stem cell factor protein therapeutics enhance revascularization in ischemia without mast cell activation. Nat Commun 2022; 13:2497. [PMID: 35523773 PMCID: PMC9076913 DOI: 10.1038/s41467-022-30103-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 04/08/2022] [Indexed: 11/30/2022] Open
Abstract
Stem cell factor (SCF) is a cytokine that regulates hematopoiesis and other biological processes. While clinical treatments using SCF would be highly beneficial, these have been limited by toxicity related to mast cell activation. Transmembrane SCF (tmSCF) has differential activity from soluble SCF and has not been explored as a therapeutic agent. We created novel therapeutics using tmSCF embedded in proteoliposomes or lipid nanodiscs. Mouse models of anaphylaxis and ischemia revealed the tmSCF-based therapies did not activate mast cells and improved the revascularization in the ischemic hind limb. Proteoliposomal tmSCF preferentially acted on endothelial cells to induce angiogenesis while tmSCF nanodiscs had greater activity in inducing stem cell mobilization and recruitment to the site of injury. The type of lipid nanocarrier used altered the relative cellular uptake pathways and signaling in a cell type dependent manner. Overall, we found that tmSCF-based therapies can provide therapeutic benefits without off target effects.
Collapse
Affiliation(s)
- Eri Takematsu
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Miles Massidda
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Jeff Auster
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Po-Chih Chen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - ByungGee Im
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Sanjana Srinath
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Sophia Canga
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Aditya Singh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Marjan Majid
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Michael Sherman
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Andrew Dunn
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Annette Graham
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, G4 0BA, Scotland, UK
| | - Patricia Martin
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, G4 0BA, Scotland, UK
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA.
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA.
- The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, USA.
- Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
18
|
Ahmed R, Augustine R, Chaudhry M, Akhtar UA, Zahid AA, Tariq M, Falahati M, Ahmad IS, Hasan A. Nitric oxide-releasing biomaterials for promoting wound healing in impaired diabetic wounds: State of the art and recent trends. Pharmacotherapy 2022; 149:112707. [PMID: 35303565 DOI: 10.1016/j.biopha.2022.112707] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022]
Abstract
Impaired diabetic wounds are serious pathophysiological complications associated with persistent microbial infections including failure in the closure of wounds, and the cause of a high frequency of lower limb amputations. The healing of diabetic wounds is attenuated due to the lack of secretion of growth factors, prolonged inflammation, and/or inhibition of angiogenic activity. Diabetic wound healing can be enhanced by supplying nitric oxide (NO) endogenously or exogenously. NO produced inside the cells by endothelial nitric oxide synthase (eNOS) naturally aids wound healing through its beneficial vasculogenic effects. However, during hyperglycemia, the activity of eNOS is affected, and thus there becomes an utmost need for the topical supply of NO from exogenous sources. Thus, NO-donors that can release NO are loaded into wound healing patches or wound coverage matrices to treat diabetic wounds. The burst release of NO from its donors is prevented by encapsulating them in polymeric hydrogels or nanoparticles for supplying NO for an extended duration of time to the diabetic wounds. In this article, we review the etiology of diabetic wounds, wound healing strategies, and the role of NO in the wound healing process. We further discuss the challenges faced in translating NO-donors as a clinically viable nanomedicine strategy for the treatment of diabetic wounds with a focus on the use of biomaterials for the encapsulation and in vivo controlled delivery of NO-donors.
Collapse
Affiliation(s)
- Rashid Ahmed
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar; Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, Mirpur 10250, AJK, Pakistan; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA
| | - Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar
| | - Maryam Chaudhry
- Department of Continuing Education, University of Oxford, OX1 2JD Oxford, United Kingdom
| | - Usman A Akhtar
- Department of Chemical Engineering, College of Engineering, Qatar University, Doha 2713, Qatar
| | - Alap Ali Zahid
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar
| | - Muhammad Tariq
- Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, Mirpur 10250, AJK, Pakistan
| | - Mojtaba Falahati
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Irfan S Ahmad
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA; Department of Agricultural and Biological Engineering, University of Illinois at Urbana Champaign, IL, USA; Carle Illinois College of Medicine, University of Illinois at Urbana Champaign, IL, USA
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| |
Collapse
|
19
|
Kandregula B, Narisepalli S, Chitkara D, Mittal A. Exploration of Lipid-Based Nanocarriers as Drug Delivery Systems in Diabetic Foot Ulcer. Mol Pharm 2022; 19:1977-1998. [PMID: 35481377 DOI: 10.1021/acs.molpharmaceut.1c00970] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a chronic manifestation characterized by high levels of glucose in the blood resulting in several complications including diabetic wounds and ulcers, which predominantly require a longer duration of treatment and adversely affect the quality of life of the patients. Nanotechnology-based therapeutics (both intrinsic and extrinsic types) have emerged as a promising treatment in diabetic foot ulcer/chronic wounds owing to their unique characteristics and specific functional properties. In this review, we have focused on the significance of the use of lipids in the healing of diabetic ulcers, their interaction with the injured skin, and recent trends in lipid-based nanocarriers for the healing of diabetic wounds. Lipid nanocarriers are also being investigated for gene therapy in diabetic wound healing to encapsulate nucleic acids such as siRNA and miRNA, which could silence the expression of inflammatory cytokines overexpressed in chronic wounds. Additionally, these are also being explored for encapsulating proteins, peptides, growth factors, and other biological genetic material as therapeutic agents. Lipid-based nanocarriers encompassing a wide variety of carriers such as liposomes, niosomes, ethosomes, solid lipid nanoparticles, and lipidoid nanoparticles that are explored for the treatment of foot ulcers supplemented with relevant research studies have been discussed in the present review. Lipid-based nanodrug delivery systems have demonstrated promising wound healing potential, particularly in diabetic conditions due to the enhanced efficacy of the entrapped active molecules.
Collapse
Affiliation(s)
- Bhaskar Kandregula
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India
| | - Saibhargav Narisepalli
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India.,Department of Cellular and Molecular Biology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
20
|
PDGF-BB-derived supramolecular hydrogel for promoting skin wound healing. J Nanobiotechnology 2022; 20:201. [PMID: 35473604 PMCID: PMC9044828 DOI: 10.1186/s12951-022-01390-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/20/2022] [Indexed: 12/24/2022] Open
Abstract
Chronic wounds represent a major challenge to the present healthcare system. In recent decades, many topical therapies have been investigated for the treatment of chronic wounds, including different types of wound dressings, antimicrobial agents, and cell therapy. Platelet-derived growth factor (PDGF) plays an important role in wound healing and has been approved for treatment of wounds related to diabetes mellitus. However, the high cost and short retention time of PDGF protein have limited its wide application. To overcome this challenge, we designed a PDGF-mimicking peptide by connecting PDGF epitope VRKIEIVRKK and self-assembling motif derived from β-amyloid peptide. The resultant peptide can self-assemble into a fibril-rich network and leads to supramolecular hydrogelation with good stability. The hydrophilic epitope can be exposed on the surface of nanofibrils, which might contribute to the binding and activation of PDGF receptors. The forming hydrogel is able to induce the growth and migration of vascular endothelial cells and promote the formation of vascular branches. In the full-thickness skin wounds of healthy mice, after the application of the hydrogel, the density of neovascularization marked by CD31 was greater than that in the control group on Day 3. Larger collagen deposition and a thicker epidermis were observed on Day 12. These results demonstrate that the hydrogel can stimulate collagen deposition and angiogenesis, enhance skin regeneration, and show an excellent therapeutic effect. Taken together, this work not only provides new insight into the design of bioactive peptides but also offers a promising biomaterial for wound healing.
Collapse
|
21
|
Pérez LA, Leyton L, Valdivia A. Thy-1 (CD90), Integrins and Syndecan 4 are Key Regulators of Skin Wound Healing. Front Cell Dev Biol 2022; 10:810474. [PMID: 35186924 PMCID: PMC8851320 DOI: 10.3389/fcell.2022.810474] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Acute skin wound healing is a multistage process consisting of a plethora of tightly regulated signaling events in specialized cells. The Thy-1 (CD90) glycoprotein interacts with integrins and the heparan sulfate proteoglycan syndecan 4, generating a trimolecular complex that triggers bi-directional signaling to regulate diverse aspects of the wound healing process. These proteins can act either as ligands or receptors, and they are critical for the successful progression of wound healing. The expression of Thy-1, integrins, and syndecan 4 is controlled during the healing process, and the lack of expression of any of these proteins results in delayed wound healing. Here, we review and discuss the roles and regulatory events along the stages of wound healing that support the relevance of Thy-1, integrins, and syndecan 4 as crucial regulators of skin wound healing.
Collapse
Affiliation(s)
- Leonardo A. Pérez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| | - Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| |
Collapse
|
22
|
Hu Y, Gu J, Lin J, Wang Y, Yang F, Yin J, Yu Z, Wu S, Lv H, Ji X, Wang S. (-)-Epigallocatechin-3-gallate (EGCG) modulates polarized macrophages to suppress M1 phenotype and promote M2 polarization in vitro and in vivo. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
23
|
Bender EC, Kraynak CA, Huang W, Suggs LJ. Cell-Inspired Biomaterials for Modulating Inflammation. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:279-294. [PMID: 33528306 DOI: 10.1089/ten.teb.2020.0276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Inflammation is a crucial part of wound healing and pathogen clearance. However, it can also play a role in exacerbating chronic diseases and cancer progression when not regulated properly. A subset of current innate immune engineering research is focused on how molecules such as lipids, proteins, and nucleic acids native to a healthy inflammatory response can be harnessed in the context of biomaterial design to promote healing, decrease disease severity, and prolong survival. The engineered biomaterials in this review inhibit inflammation by releasing anti-inflammatory cytokines, sequestering proinflammatory cytokines, and promoting phenotype switching of macrophages in chronic inflammatory disease models. Conversely, other biomaterials discussed here promote inflammation by mimicking pathogen invasion to inhibit tumor growth in cancer models. The form that these biomaterials take spans a spectrum from nanoparticles to large-scale hydrogels to surface coatings on medical devices. Cell-inspired molecules have been incorporated in a variety of creative ways, including loaded into or onto the surface of biomaterials or used as the biomaterials themselves.
Collapse
Affiliation(s)
- Elizabeth C Bender
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| | - Chelsea A Kraynak
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| | - Wenbai Huang
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA.,Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas, USA
| | - Laura J Suggs
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
24
|
Chu C, Zhao X, Rung S, Xiao W, Liu L, Qu Y, Man Y. Application of biomaterials in periodontal tissue repair and reconstruction in the presence of inflammation under periodontitis through the foreign body response: Recent progress and perspectives. J Biomed Mater Res B Appl Biomater 2021; 110:7-17. [PMID: 34142745 DOI: 10.1002/jbm.b.34891] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Periodontitis would cause dental tissue damage locally. Biomaterials substantially affect the surrounding immune microenvironment through treatment-oriented local inflammatory remodeling in dental periodontitis. This remodeling process is conducive to wound healing and periodontal tissue regeneration. Recent progress in understanding the foreign body response (FBR) and immune regulation, including cell heterogeneity, and cell-cell and cell-material interactions, has provided new insights into the design criteria for biomaterials applied in treatment of periodontitis. This review discusses recent progress and perspectives in the immune regulation effects of biomaterials to augment or reconstruct soft and hard tissue in an inflammatory microenvironment of periodontitis.
Collapse
Affiliation(s)
- Chenyu Chu
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiwen Zhao
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shengan Rung
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenlan Xiao
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Liu
- State Key Laboratory of Biotherapy and Laboratory, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Yili Qu
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Man
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Xu Z, Liang B, Tian J, Wu J. Anti-inflammation biomaterial platforms for chronic wound healing. Biomater Sci 2021; 9:4388-4409. [PMID: 34013915 DOI: 10.1039/d1bm00637a] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nowadays, there has been an increase in the number of people with chronic wounds, which has resulted in serious health problems worldwide. The rate-limiting stage of chronic wound healing has been found to be the inflammation stage, and strategies for shortening the prolonged inflammatory response have proven to be effective for increasing the healing rate. Recently, various anti-inflammatory strategies (such as anti-inflammatory drugs, antioxidant, NO regulation, antibacterial, immune regulation and angiogenesis) have attracted attention as potential therapeutic pathways. Moreover, various biomaterial platforms based on anti-inflammation therapy strategies have also emerged in the spotlight as potential therapies to accelerate the repair of chronic wounds. In this review, we systematically investigated the advances of various biomaterial platforms based on anti-inflammation strategies for chronic wound healing, to provide valuable guidance for future breakthroughs in chronic wound treatment.
Collapse
Affiliation(s)
- Zejun Xu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, P. R. China.
| | - Biao Liang
- Center of Digestive Endoscopy, Guangdong Second Provincial general Hospital, No. 466, Xingang Middle Road, Guangzhou 510317, Haizhu District, China.
| | - Junzhang Tian
- Center of Digestive Endoscopy, Guangdong Second Provincial general Hospital, No. 466, Xingang Middle Road, Guangzhou 510317, Haizhu District, China.
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, P. R. China.
| |
Collapse
|
26
|
Applications of Nanosized-Lipid-Based Drug Delivery Systems in Wound Care. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11114915] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Impaired wound healing is an encumbering public health issue that increases the demand for developing new therapies in order to minimize health costs and enhance treatment efficacy. Available conventional therapies are still unable to maximize their potential in penetrating the skin at the target site and accelerating the healing process. Nanotechnology exhibits an excellent opportunity to enrich currently available medical treatments, enhance standard care and manage wounds. It is a promising approach, able to address issues such as the permeability and bioavailability of drugs with reduced stability or low water solubility. This paper focuses on nanosized-lipid-based drug delivery systems, describing their numerous applications in managing skin wounds. We also highlight the relationship between the physicochemical characteristics of nanosized, lipid-based drug delivery systems and their impact on the wound-healing process. Different types of nanosized-lipid-based drug delivery systems, such as vesicular systems and lipid nanoparticles, demonstrated better applicability and enhanced skin penetration in wound healing therapy compared with conventional treatments. Moreover, an improved chemically and physically stable drug delivery system, with increased drug loading capacity and enhanced bioavailability, has been shown in drugs encapsulated in lipid nanoparticles. Their applications in wound care show potential for overcoming impediments, such as the inadequate bioavailability of active agents with low solubility. Future research in nanosized-lipid-based drug delivery systems will allow the achievement of increased bioavailability and better control of drug release, providing the clinician with more effective therapies for wound care.
Collapse
|
27
|
Li Z, Bratlie KM. The Influence of Polysaccharides-Based Material on Macrophage Phenotypes. Macromol Biosci 2021; 21:e2100031. [PMID: 33969643 DOI: 10.1002/mabi.202100031] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 02/03/2023]
Abstract
Macrophage polarization is a key factor in determining the success of implanted tissue engineering scaffolds. Polysaccharides (derived from plants, animals, and microorganisms) are known to modulate macrophage phenotypes by recognizing cell membrane receptors. Numerous studies have developed polysaccharide-based materials into functional biomaterial substrates for tissue regeneration and pharmaceutical application due to their immunostimulatory activities and anti-inflammatory response. They are used as hydrogel substrates, surface coatings, and drug delivery carriers. In addition to their innate immunological functions, the newly endowed physical and chemical properties, including substrate modulus, pore size/porosity, surface binding chemistry, and the mole ratio of polysaccharides in hybrid materials may regulate macrophage phenotypes more precisely. Growing evidence indicates that the sulfation pattern of glycosaminoglycans and proteoglycans expressed on polarized macrophages leads to the changes in protein binding, which may alter macrophage phenotype and influence the immune response. A comprehensive understanding of how different types of polysaccharide-based materials alter macrophage phenotypic changes can be beneficial to predict transplantation/implantation outcomes. This review focuses on recent advances in promoting wound healing and balancing macrophage phenotypes using polysaccharide-based substrates/coatings and new directions to address the limitations in the current understanding of macrophage responses to polysaccharides.
Collapse
Affiliation(s)
- Zhuqing Li
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, 50011, USA.,Department of Chemical & Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| |
Collapse
|
28
|
Blanco-Fernandez B, Castaño O, Mateos-Timoneda MÁ, Engel E, Pérez-Amodio S. Nanotechnology Approaches in Chronic Wound Healing. Adv Wound Care (New Rochelle) 2021; 10:234-256. [PMID: 32320364 PMCID: PMC8035922 DOI: 10.1089/wound.2019.1094] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/04/2020] [Indexed: 12/28/2022] Open
Abstract
Significance: The incidence of chronic wounds is increasing due to our aging population and the augment of people afflicted with diabetes. With the extended knowledge on the biological mechanisms underlying these diseases, there is a novel influx of medical technologies into the conventional wound care market. Recent Advances: Several nanotechnologies have been developed demonstrating unique characteristics that address specific problems related to wound repair mechanisms. In this review, we focus on the most recently developed nanotechnology-based therapeutic agents and evaluate the efficacy of each treatment in in vivo diabetic models of chronic wound healing. Critical Issues: Despite the development of potential biomaterials and nanotechnology-based applications for wound healing, this scientific knowledge is not translated into an increase of commercially available wound healing products containing nanomaterials. Future Directions: Further studies are critical to provide insights into how scientific evidences from nanotechnology-based therapies can be applied in the clinical setting.
Collapse
Affiliation(s)
- Barbara Blanco-Fernandez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Oscar Castaño
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Electronics and Biomedical Engineering, Universitat de Barcelona (UB), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Bioelectronics Unit and Nanobioengineering Lab, Institute for Nanoscience and Nanotechnology of the University of Barcelona (IN2UB), Barcelona, Spain
| | - Miguel Ángel Mateos-Timoneda
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Materials Science and Metallurgical Engineering, Polytechnic University of Catalonia (UPC), Barcelona, Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Materials Science and Metallurgical Engineering, Polytechnic University of Catalonia (UPC), Barcelona, Spain
| | - Soledad Pérez-Amodio
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Materials Science and Metallurgical Engineering, Polytechnic University of Catalonia (UPC), Barcelona, Spain
| |
Collapse
|
29
|
Raina N, Rani R, Gupta M. Angiogenesis: Aspects in wound healing. ENDOTHELIAL SIGNALING IN VASCULAR DYSFUNCTION AND DISEASE 2021:77-90. [DOI: 10.1016/b978-0-12-816196-8.00010-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
30
|
ADAMTS proteases and the tumor immune microenvironment: Lessons from substrates and pathologies. Matrix Biol Plus 2020; 9:100054. [PMID: 33718860 PMCID: PMC7930849 DOI: 10.1016/j.mbplus.2020.100054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
The relationship of ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) proteases with inflammatory processes was anticipated since their discovery. Although knowledge of these extracellular proteases in different contexts continues to grow, many questions remain unanswered. In this review, we summarize the most important studies of ADAMTSs and their substrates in inflammation and in the immune system of non-oncological disorders. In addition, we update the findings on cancer and highlight their emerging role in the tumor immune microenvironment. Although the overall functions of extracellular molecules are known to be modulated by proteolysis, specific activities attributed to intact proteins and cleaved fragments in the context of inflammation are still subject to debate. A better understanding of ADAMTS activities will help to elucidate their contribution to the immune phenotype and to open up new therapeutic and diagnostic possibilities.
Collapse
|
31
|
Deptuła M, Karpowicz P, Wardowska A, Sass P, Sosnowski P, Mieczkowska A, Filipowicz N, Dzierżyńska M, Sawicka J, Nowicka E, Langa P, Schumacher A, Cichorek M, Zieliński J, Kondej K, Kasprzykowski F, Czupryn A, Janus Ł, Mucha P, Skowron P, Piotrowski A, Sachadyn P, Rodziewicz-Motowidło S, Pikuła M. Development of a Peptide Derived from Platelet-Derived Growth Factor (PDGF-BB) into a Potential Drug Candidate for the Treatment of Wounds. Adv Wound Care (New Rochelle) 2020; 9:657-675. [PMID: 33124966 PMCID: PMC7698658 DOI: 10.1089/wound.2019.1051] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/13/2019] [Indexed: 12/16/2022] Open
Abstract
Objective: This study evaluated the use of novel peptides derived from platelet-derived growth factor (PDGF-BB) as potential wound healing stimulants. One of the compounds (named PDGF2) was subjected for further research after cytotoxicity and proliferation assays on human skin cells. Further investigation included evaluation of: migration and chemotaxis of skin cells, immunological and allergic safety, the transcriptional analyses of adipose-derived stem cells (ASCs) and dermal fibroblasts stimulated with PDGF2, and the use of dorsal skin wound injury model to evaluate the effect of wound healing in mice. Approach: Colorimetric lactate dehydrogenase and tetrazolium assays were used to evaluate the cytotoxicity and the effect on proliferation. PDGF2 effect on migration and chemotaxis was also checked. Immunological safety and allergic potential were evaluated with a lymphocyte activation and basophil activation test. Transcriptional profiles of ASCs and primary fibroblasts were assessed after stimulation with PDGF2. Eight-week-old BALB/c female mice were used for dorsal skin wound injury model. Results: PDGF2 showed low cytotoxicity, pro-proliferative effects on human skin cells, high immunological safety, and accelerated wound healing in mouse model. Furthermore, transcriptomic analysis of ASCs and fibroblasts revealed the activation of processes involved in wound healing and indicated its safety. Innovation: A novel peptide derived from PDGF-BB was proved to be safe drug candidate in wound healing. We also present a multifaceted in vitro model for the initial screening of new compounds that may be potentially useful in wound healing stimulation. Conclusion: The results show that peptide derived from PDGF-BB is a promising drug candidate for wound treatment.
Collapse
Affiliation(s)
- Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Gdansk, Poland
| | - Przemysław Karpowicz
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Anna Wardowska
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Gdansk, Poland
- Department of Clinical Immunology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Sass
- Laboratory for Regenerative Biotechnology, Gdansk University of Technology, Gdansk, Poland
| | - Paweł Sosnowski
- Laboratory for Regenerative Biotechnology, Gdansk University of Technology, Gdansk, Poland
| | | | | | - Maria Dzierżyńska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Justyna Sawicka
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Ewa Nowicka
- Department of Clinical Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Paulina Langa
- Department of Clinical Immunology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| | | | | | - Jacek Zieliński
- Department of Surgical Oncology, and Medical University of Gdansk, Gdansk, Poland
| | - Karolina Kondej
- Department of Plastic Surgery, Medical University of Gdansk, Gdansk, Poland
| | | | - Artur Czupryn
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | | | - Piotr Mucha
- Department of Biochemistry, and Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Piotr Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | | | - Paweł Sachadyn
- Laboratory for Regenerative Biotechnology, Gdansk University of Technology, Gdansk, Poland
| | | | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Gdansk, Poland
- Department of Clinical Immunology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
32
|
Characterization of Alginate from Sargassum duplicatum and the Antioxidant Effect of Alginate–Okra Fruit Extracts Combination for Wound Healing on Diabetic Mice. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10176082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Diabetes mellitus is a group of metabolic disorders characterized by high blood-glucose levels over a prolonged period that causes complications when an open wound is present. Alginate is an antioxidant and a good absorbent of exudates. Okra fruit contains flavonoids that can act as antioxidants. The antioxidant properties of extracts combination reduce blood-glucose levels significantly to accelerate the activities of wound-healing processes on diabetic mice. Alginate was characterized by Size Exclusion Chromatography-Multiple Angle Laser Light Scattering (SEC-MALLS), thermal stability and Proton Nuclear Magnetic Resonance (1H-NMR). The evaluation of wound healing on 36 male mice were divided into 12 groups including normal control (NC), diabetics control (DC), alginate (DA) and alginate–okra (DAO) groups in three different times by histopathology test on skin tissue. The results of SEC-MALLS analysis showed that alginate as single and homogeneous polysaccharide. The 1H-NMR spectrum showed that the mannuronate/guluronate ratio of the used alginate was 0.91. Alginate, okra fruit extract and their combination were classified as moderate and strong antioxidants. The numbers of fibrocytes, fibroblasts, collagen densities had significantly increased from three to seven days. In contrast, wound width, neutrophil, macrophages had significantly decreased at 14 days. The administration of extracts combination increased the re-epithelization of the wound area and wound-healing process on diabetic mice.
Collapse
|
33
|
Docosahexaenoic Acid Improves Diabetic Wound Healing in a Rat Model by Restoring Impaired Plasticity of Macrophage Progenitor Cells. Plast Reconstr Surg 2020; 145:942e-950e. [PMID: 32332536 DOI: 10.1097/prs.0000000000006739] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Chronic inflammation associated with delayed diabetic wound healing is induced by disturbed polarization of macrophages derived mainly from predisposed progenitor cells in bone marrow. Docosahexaenoic acid plays a critical role in regulating the function of macrophage progenitor cells. The authors evaluated whether docosahexaenoic acid accelerates diabetic wound healing in rats. METHODS Streptozotocin-induced diabetic rats divided into control and docosahexaenoic acid-treated groups (n = 10) were subjected to paired dorsal skin wounds. Docosahexaenoic acid (100 mg/kg per day) was orally supplemented 2 weeks before wounding until termination. The wound healing process was recorded 0, 7, and 14 days after wounding. At day 7, blood perfusion was measured by laser Doppler perfusion imaging; angiogenesis was compared using immunofluorescent CD31 and α-smooth muscle actin staining; macrophage polarization was detected using immunofluorescence for CD68, CD206, and inducible nitric oxide synthase. Hematoxylin and eosin staining was used to examine wound healing at day 14. Activation status of macrophages derived from bone marrow cells in normal, diabetic, and docosahexaenoic acid-treated diabetic rats was determined in vitro using Western blotting and enzyme-linked immunosorbent assay. RESULTS Docosahexaenoic acid significantly accelerated wound healing 7 and 14 days (p < 0.01) after wounding. Increased vessel densities (1.96-fold; p < 0.001) and blood perfusion (2.56-fold; p < 0.001) were observed in docosahexaenoic acid-treated wounds. Immunofluorescence revealed more CD206 and fewer inducible nitric oxide synthase-positive macrophages (p < 0.001) in treated wounds. Furthermore, macrophages derived from diabetic rats expressed higher levels of inducible nitric oxide synthase and tumor necrosis factor-α and lower arginase-1 and interleukin-10 (p < 0.05). CONCLUSION Docosahexaenoic acid accelerates diabetic wound healing at least in part by restoring impaired plasticity of macrophage progenitor cells.
Collapse
|
34
|
Ghasemi Y, Ghoshoon MB, Taheri M, Negahdaripour M, Nouri F. Cloning, expression and purification of human PDGF-BB gene in Escherichia coli: New approach in PDGF-BB protein production. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
Walimbe T, Panitch A. Proteoglycans in Biomedicine: Resurgence of an Underexploited Class of ECM Molecules. Front Pharmacol 2020; 10:1661. [PMID: 32082161 PMCID: PMC7000921 DOI: 10.3389/fphar.2019.01661] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/18/2019] [Indexed: 01/02/2023] Open
Abstract
Proteoglycans have emerged as biomacromolecules with important roles in matrix remodeling, homeostasis, and signaling in the past two decades. Due to their negatively charged glycosaminoglycan chains as well as distinct core protein structures, they interact with a variety of molecules, including matrix proteins, growth factors, cytokines and chemokines, pathogens, and enzymes. This led to the dawn of glycan therapies in the 20th century, but this research was quickly overshadowed by readily available DNA and protein-based therapies. The recent development of recombinant technology and advances in our understanding of proteoglycan function have led to a resurgence of these molecules as potential therapeutics. This review focuses on the recent preclinical efforts that are bringing proteoglycan research and therapies back to the forefront. Examples of studies using proteoglycan cores and mimetics have also been included to give the readers a perspective on the wide-ranging and extensive applications of these versatile molecules. Collectively, these advances are opening new avenues for targeting diseases at a molecular level, and providing avenues for the development of new and exciting treatments in regenerative medicine.
Collapse
Affiliation(s)
- Tanaya Walimbe
- Laboratory of Engineered Therapeutics, Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Alyssa Panitch
- Laboratory of Engineered Therapeutics, Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
36
|
Growth factors with enhanced syndecan binding generate tonic signalling and promote tissue healing. Nat Biomed Eng 2019; 4:463-475. [PMID: 31685999 DOI: 10.1038/s41551-019-0469-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 09/20/2019] [Indexed: 12/31/2022]
Abstract
Growth factors can stimulate tissue regeneration, but the side effects and low effectiveness associated with suboptimal delivery systems have impeded their use in translational regenerative medicine. Physiologically, growth factor interactions with the extracellular matrix control their bioavailability and spatiotemporal cellular signalling. Growth factor signalling is also controlled at the cell surface level via binding to heparan sulfate proteoglycans, such as syndecans. Here we show that vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor-BB (PDGF-BB) that were engineered to have a syndecan-binding sequence trigger sustained low-intensity signalling (tonic signalling) and reduce the desensitization of growth factor receptors. We also show in mouse models that tonic signalling leads to superior morphogenetic activity, with syndecan-binding growth factors inducing greater bone regeneration and wound repair than wild-type growth factors, as well as reduced tumour growth (associated with PDGF-BB delivery) and vascular permeability (triggered by VEGF-A). Tonic signalling via syndecan binding may also enhance the regenerative capacity of other growth factors.
Collapse
|
37
|
Syndecan-4 Inhibits the Development of Pulmonary Fibrosis by Attenuating TGF-β Signaling. Int J Mol Sci 2019; 20:ijms20204989. [PMID: 31600983 PMCID: PMC6834137 DOI: 10.3390/ijms20204989] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022] Open
Abstract
Syndecan-4 is a transmembrane heparan sulfate proteoglycan expressed in a variety of cells, and its heparan sulfate glycosaminoglycan side chains bind to several proteins exhibiting various biological roles. The authors have previously demonstrated syndecan-4′s critical roles in pulmonary inflammation. In the current study, however, its role in pulmonary fibrosis was evaluated. Wild-type and syndecan-4-deficient mice were injected with bleomycin, and several parameters of inflammation and fibrosis were analyzed. The mRNA expression of collagen and α-smooth muscle action (α-SMA) in lung tissues, as well as the histopathological lung fibrosis score and collagen content in lung tissues, were significantly higher in the syndecan-4-deficient mice. However, the total cell count and cell differentiation in bronchoalveolar lavage fluid were equivalent between the wild-type and syndecan-4-deficient mice. Although there was no difference in the TGF-β expression in lung tissues between the wild-type and syndecan-4-deficient mice, significantly more activation of Smad3 in lung tissues was observed in the syndecan-4-deficient mice compared to the wild-type mice. Furthermore, in the in vitro experiments using lung fibroblasts, the co-incubation of syndecan-4 significantly inhibited TGF-β-induced Smad3 activation, collagen and α-SMA upregulation. Moreover, syndecan-4 knock-down by siRNA increased TGF-β-induced Smad3 activation and upregulated collagen and α-SMA expression. These findings showed that syndecan-4 inhibits the development of pulmonary fibrosis, at least in part, through attenuating TGF-β signaling.
Collapse
|
38
|
Veith AP, Henderson K, Spencer A, Sligar AD, Baker AB. Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev 2019; 146:97-125. [PMID: 30267742 DOI: 10.1016/j.addr.2018.09.010] [Citation(s) in RCA: 492] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 09/15/2018] [Accepted: 09/24/2018] [Indexed: 12/19/2022]
Abstract
The enhancement of wound healing has been a goal of medical practitioners for thousands of years. The development of chronic, non-healing wounds is a persistent medical problem that drives patient morbidity and increases healthcare costs. A key aspect of many non-healing wounds is the reduced presence of vessel growth through the process of angiogenesis. This review surveys the creation of new treatments for healing cutaneous wounds through therapeutic angiogenesis. In particular, we discuss the challenges and advancement that have been made in delivering biologic, pharmaceutical and cell-based therapies as enhancers of wound vascularity and healing.
Collapse
|
39
|
Suppression of PDGF induces neuronal apoptosis after neonatal cerebral hypoxia and ischemia by inhibiting P-PI3K and P-AKT signaling pathways. Brain Res 2019; 1719:77-88. [PMID: 31082354 DOI: 10.1016/j.brainres.2019.05.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 02/05/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) always results in severe neurologic dysfunction, nevertheless effective treatments are limited and the underlying mechanism also remains unclear. In this study, we firstly established the neonatal HIE model in the postnatal day 7 SD rats, Zea-Longa score and TTC staining were employed to assess the neurological behavior and infarct volume of the brain after cerebral hypoxia-ischemia (HI). Afterwards, protein chip was adopted to detect the differential proteins in the right cortex, hippocampus and lung, ultimately, PDGF was noticed. Then, immunohistochemistry, immunofluorescence double staining of NeuN/PDGF, and western blot were used to validate the expression level of PDGF in the cortex and hippocampus at 6 hours (h), 12 h and 24 h after HI. To determine the role of PDGF, the primary cortical neurons were prepared and performed PDGF shRNA administration. The results showed that HIE induced a severe behavioral dysfunction and brain infarction in neonatal rats, and the expression of PDGF in right cortex and hippocampus was remarkably increased after HI. Whereas, suppressing PDGF resulted in a significant loss of neurons and inhibition of neurite growth. Moreover, the protein level of P-PI3K and P-AKT signaling pathways were largely decreased following PDGF-shRNA application in the cortical neurons. In conclusion, PDGF suppression aggravated neuronal dysfunction, and the underlying mechanism is associated with inhibiting the phosphorylation of P-PI3K and P-AKT. Together, PDGF regulating PI3K and AKT may be an important panel in HIE events and therefore may provide possible strategy for the treatment of HIE in future clinic trail.
Collapse
|
40
|
Paige JT, Kremer M, Landry J, Hatfield SA, Wathieu D, Brug A, Lightell DJ, Spiller KL, Woods TC. Modulation of inflammation in wounds of diabetic patients treated with porcine urinary bladder matrix. Regen Med 2019; 14:269-277. [PMID: 31020913 PMCID: PMC6886567 DOI: 10.2217/rme-2019-0009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Aim: To determine if porcine urinary bladder matrix (UBM) treatment is associated with modulation of wound inflammation in diabetic patients. Patients & methods: mRNA associated with M1 and M2 macrophages were measured in wounds of diabetic and nondiabetic patients pre- and post-treatment with UBM and an M1:M2 score was calculated. Results: Wound tissue from diabetic subjects exhibited elevated M1:M2 scores compared with nondiabetic patients, suggesting a greater pro-inflammatory state prior to treatment. Post-treatment, there was significantly greater reduction in the magnitude of the individual M1:M2 scores in the diabetic patients resulting in similar levels in both groups of patients. Conclusions: UBM may assist in diabetic wound healing by restoring an inflammatory state similar to that of nondiabetic patients.
Collapse
Affiliation(s)
- John T Paige
- Department of Surgery, LSU Health New Orleans, School of Medicine, New Orleans, LA 70112, USA
| | - Michael Kremer
- Departments of Physiology & Medicine, Tulane School of Medicine, New Orleans, LA 70112, USA
| | - Jace Landry
- Department of Surgery, LSU Health New Orleans, School of Medicine, New Orleans, LA 70112, USA
| | - Samuel A Hatfield
- Departments of Physiology & Medicine, Tulane School of Medicine, New Orleans, LA 70112, USA
| | - Donald Wathieu
- Departments of Physiology & Medicine, Tulane School of Medicine, New Orleans, LA 70112, USA
| | - Aaron Brug
- Departments of Physiology & Medicine, Tulane School of Medicine, New Orleans, LA 70112, USA
| | - Daniel J Lightell
- Departments of Physiology & Medicine, Tulane School of Medicine, New Orleans, LA 70112, USA
| | - Kara L Spiller
- School of Biomedical Engineering Science & Health Systems, Drexel University, Philadelphia, 19104 PA, USA
| | - T Cooper Woods
- Departments of Physiology & Medicine, Tulane School of Medicine, New Orleans, LA 70112, USA,*Author for correspondence: Tel.: +1 504 988 2588;
| |
Collapse
|
41
|
Liu Y, Zhou S, Gao Y, Zhai Y. Electrospun nanofibers as a wound dressing for treating diabetic foot ulcer. Asian J Pharm Sci 2019; 14:130-143. [PMID: 32104445 PMCID: PMC7032134 DOI: 10.1016/j.ajps.2018.04.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/25/2018] [Accepted: 04/19/2018] [Indexed: 01/14/2023] Open
Abstract
Diabetes is one of the most prevalent diseases in the world with high-mortality and complex complications including diabetic foot ulcer (DFU). It has been reported that the difficulties in repairing the wound related to DFU has much relationship with the wound infection, change of inflammatory responses, lack of extracellular matrix (ECM), and the failure of angiogenesis. Following the development of medical materials and pharmaceutical technology, nanofibers has been developed by electrospinning with huge porosity, excellent humidity absorption, a better oxygen exchange rate, and some antibacterial activities. That is to say, as a potential material, nanofibers must be a wonderful candidate for the DFU treatment with so many benefits. Careful selection of polymers from natural resource and synthetic resource can widen the nanofibrous application. Popular methods applied for the nanofibrous fabrication consist of uniaxial electrospinning and coaxial electrospinning. Furthermore, nanofibers loading chemical, biochemical active pharmaceutical ingredient (API) or even stem cells can be wonderful dosage forms for the treatment of DFU. This review summarizes the present techniques applied in the fabrication of nanofibrous dressing (ND) that utilizes a variety of materials and active agents to offer a better health care for the patients suffering from DFU.
Collapse
Affiliation(s)
- Yan Liu
- Shenyang Pharmaceutical University, No.103, Shenyang 110016, China
| | - Shiya Zhou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanlin Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinglei Zhai
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
42
|
Wang L, Wang F, Zhao L, Yang W, Wan X, Yue C, Mo Z. Mesenchymal Stem Cells Coated by the Extracellular Matrix Promote Wound Healing in Diabetic Rats. Stem Cells Int 2019; 2019:9564869. [PMID: 30833970 PMCID: PMC6369500 DOI: 10.1155/2019/9564869] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/20/2018] [Accepted: 11/11/2018] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To investigate the effects of mesenchymal stem cells (MSCs) coated by the extracellular matrix (ECM) on wound healing in diabetic rats. METHODS Mesenchymal stem cells were cocultured with ECM. Cell viabilities were evaluated using MTT assay. The diabetes model was established using both STZ and high-glucose/fat methods in SD rats. A wound area was made on the middle of the rats' back. MSCs or ECM-MSCs were used to treat the rats. HE staining and CD31 immunohistochemistry were used to detect the skin thickness and angiogenesis. Western blotting and qRT-PCR were conducted to determine the level of VEGF-α, PDGF, and EGF. RESULTS It was observed that treatment of ECM had no significant effects on the cell viability of ECM-MSCs. Wound area assay showed that both MSCs and ECM-MSCs could enhance the wound healing of diabetic rats and ECM-MSCs could further promote the effects. Both MSCs and ECM-MSCs could enhance angiogenesis and epithelialization of the wounds, as well as the expression of VEGF-α, PDGF, and EGF in wound tissues, while ECM-MSC treatment showed more obvious effects. CONCLUSION Mesenchymal stem cells coated by the extracellular matrix could promote wound healing in diabetic rats. Our study may offer a novel therapeutic method for impaired diabetic wound healing.
Collapse
Affiliation(s)
- Linhao Wang
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Fang Wang
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Liling Zhao
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Wenjun Yang
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Xinxing Wan
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Chun Yue
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| | - Zhaohui Mo
- Department of Endocrinology and Metabolism, Third Xiangya Hospital of Central South University, China
| |
Collapse
|
43
|
Optimization and characterization of electrospun polycaprolactone coated with gelatin-silver nanoparticles for wound healing application. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 91:318-329. [DOI: 10.1016/j.msec.2018.05.039] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 04/30/2018] [Accepted: 05/10/2018] [Indexed: 02/07/2023]
|
44
|
Monteforte A, Lam B, Sherman MB, Henderson K, Sligar AD, Spencer A, Tang B, Dunn AK, Baker AB. * Glioblastoma Exosomes for Therapeutic Angiogenesis in Peripheral Ischemia. Tissue Eng Part A 2018; 23:1251-1261. [PMID: 28699397 DOI: 10.1089/ten.tea.2016.0508] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Peripheral ischemia as a result of occlusive vascular disease is a widespread problem in patients older than the age of 65. Angiogenic therapies that can induce microvascular growth have great potential for providing a long-lasting solution for patients with ischemia and would provide an appealing alternative to surgical and percutaneous interventions. However, many angiogenic therapies have seen poor efficacy in clinical trials, suggesting that patients with long-term peripheral ischemia have considerable therapeutic resistance to angiogenic stimuli. Glioblastoma is one of the most angiogenic tumor types, inducing robust vessel growth in the area surrounding the tumor. One major angiogenic mechanism used by the tumor cells to induce blood vessel growth is the production of exosomes and other extracellular vesicles that can carry pro-angiogenic and immunomodulatory signals. Here, we explored whether the pro-angiogenic aspects of glioblastoma-derived exosomes could be harnessed to promote angiogenesis and healing in the context of peripheral ischemic disease. We demonstrate that the exosomes derived from glioblastoma markedly enhance endothelial cell proliferation and increase endothelial tubule formation in vitro. An analysis of the microRNA expression using next generation sequencing identified that exosomes contained a high concentration of miR-221. In addition, we found that glioblastoma exosomes contained significant amounts of the proteoglycans glypican-1 and syndecan-4, which can serve as co-receptors for angiogenic factors, including fibroblast growth factor-2 (FGF-2). In a hindlimb ischemia model in mice, we found that the exosomes promoted enhanced revascularization in comparison to control alginate gels and FGF-2 treatment alone. Taken together, our results support the fact that glioblastoma-derived exosomes have powerful effects in increasing revascularization in the context of peripheral ischemia.
Collapse
Affiliation(s)
- Anthony Monteforte
- 1 Department of Biomedical Engineering, University of Texas at Austin , Texas
| | - Brian Lam
- 1 Department of Biomedical Engineering, University of Texas at Austin , Texas
| | - Michael B Sherman
- 2 Department of Biochemistry and Molecular Biology, University of Texas Medical Branch , Galveston, Texas
| | - Kayla Henderson
- 1 Department of Biomedical Engineering, University of Texas at Austin , Texas
| | - Andrew D Sligar
- 1 Department of Biomedical Engineering, University of Texas at Austin , Texas
| | - Adrianne Spencer
- 1 Department of Biomedical Engineering, University of Texas at Austin , Texas
| | - Brian Tang
- 1 Department of Biomedical Engineering, University of Texas at Austin , Texas
| | - Andrew K Dunn
- 1 Department of Biomedical Engineering, University of Texas at Austin , Texas
| | - Aaron B Baker
- 1 Department of Biomedical Engineering, University of Texas at Austin , Texas.,3 Institute for Cellular and Molecular Biology, University of Texas at Austin , Austin, Texas.,4 Institute for Computational Engineering and Sciences, University of Texas at Austin , Austin, Texas.,5 Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin , Austin, Texas
| |
Collapse
|
45
|
Choi JU, Lee SW, Pangeni R, Byun Y, Yoon IS, Park JW. Preparation and in vivo evaluation of cationic elastic liposomes comprising highly skin-permeable growth factors combined with hyaluronic acid for enhanced diabetic wound-healing therapy. Acta Biomater 2017; 57:197-215. [PMID: 28476587 DOI: 10.1016/j.actbio.2017.04.034] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/24/2017] [Accepted: 04/28/2017] [Indexed: 11/16/2022]
Abstract
To enhance the therapeutic effects of exogenous administration of growth factors (GFs) in the treatment of chronic wounds, we constructed GF combinations of highly skin-permeable epidermal growth factor (EGF), insulin-like growth factor-I (IGF-I), and platelet-derived growth factor-A (PDGF-A). We genetically conjugated a low-molecular-weight protamine (LMWP) to the N-termini of these GFs to form LMWP-EGF, LMWP-IGF-I, and LMWP-PDGF-A. Subsequently, these molecules were complexed with hyaluronic acid (HA). Combinations of native or LMWP-fused GFs significantly promoted fibroblast proliferation and the synthesis of procollagen, with a magnification of these results observed after the GFs were complexed with HA. The optimal proportions of LMWP-EGF, LMWP-IGF-I, LMWP-PDGF-A, and HA were 1, 1, 0.02, and 200, respectively. After confirming the presence of a synergistic effect, we incorporated the LMWP-fused GFs-HA complex into cationic elastic liposomes (ELs) of 107±0.757nm in diameter and a zeta potential of 56.5±1.13mV. The LMWP-fused GFs had significantly improved skin permeation compared with native GFs. The in vitro wound recovery rate of the LMWP-fused GFs-HA complex was 23% higher than that of cationic ELs composed of LMWP-fused GFs alone. Moreover, the cationic ELs containing the LMWP-fused GFs-HA complex significantly accelerated the wound closure rate in a diabetic mouse model and the wound size was maximally decreased by 65% and 58% compared to cationic ELs loaded with vehicle or native GFs-HA complex, respectively. Thus, topical treatment with cationic ELs loaded with the LMWP-fused GFs-HA complex synergistically enhanced the healing of chronic wounds, exerting both rapid and prolonged effects. STATEMENT OF SIGNIFICANCE We believe that our study makes a significant contribution to the literature, because it demonstrated the potential application of cationic elastic liposomes as topical delivery systems for growth factors (GFs) that have certain limitations in their therapeutic effects (e.g., low percutaneous absorption of GFs at the lesion site and the requirement for various GFs at different healing stages). Topical treatment with cationic elastic liposomes loaded with highly skin-permeable low-molecular-weight protamine (LMWP)-fused GFs-hyaluronic acid (HA) complex synergistically enhanced the healing of diabetic wounds, exerting both rapid and prolonged effects.
Collapse
Affiliation(s)
- Jeong Uk Choi
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seong Wook Lee
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Rudra Pangeni
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam 58554, Republic of Korea
| | - Youngro Byun
- Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - In-Soo Yoon
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.
| | - Jin Woo Park
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam 58554, Republic of Korea.
| |
Collapse
|
46
|
Qin Y, Zhu Y, Luo F, Chen C, Chen X, Wu M. Killing two birds with one stone: dual blockade of integrin and FGF signaling through targeting syndecan-4 in postoperative capsular opacification. Cell Death Dis 2017; 8:e2920. [PMID: 28703800 PMCID: PMC5550862 DOI: 10.1038/cddis.2017.315] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/03/2017] [Accepted: 06/05/2017] [Indexed: 12/12/2022]
Abstract
The most common complication after cataract surgery is postoperative capsular opacification, which includes anterior capsular opacification (ACO) and posterior capsular opacification (PCO). Increased adhesion of lens epithelial cells (LECs) to the intraocular lens material surface promotes ACO formation, whereas proliferation and migration of LECs to the posterior capsule lead to the development of PCO. Cell adhesion is mainly mediated by the binding of integrin to extracellular matrix proteins, while cell proliferation and migration are regulated by fibroblast growth factor (FGF). Syndecan-4 (SDC-4) is a co-receptor for both integrin and FGF signaling pathways. Therefore, SDC-4 may be an ideal therapeutic target for the prevention and treatment of postoperative capsular opacification. However, how SDC-4 contributes to FGF-mediated proliferation, migration, and integrin-mediated adhesion of LECs is unclear. Here, we found that downregulation of SDC-4 inhibited FGF signaling through the blockade of ERK1/2 and PI3K/Akt/mTOR activation, thus suppressing cell proliferation and migration. In addition, downregulation of SDC-4 suppressed integrin-mediated cell adhesion through inhibiting focal adhesion kinase (FAK) phosphorylation. Moreover, SDC-4 knockout mice exhibited normal lens morphology, but had significantly reduced capsular opacification after injury. Finally, SDC-4 expression level was increased in the anterior capsule LECs of age-related cataract patients. Taken together, we for the first time characterized the key regulatory role of SDC-4 in FGF and integrin signaling in human LECs, and provided the basis for future pharmacological interventions of capsular opacification.
Collapse
Affiliation(s)
- Yingyan Qin
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yi Zhu
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Furong Luo
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Chuan Chen
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xiaoyun Chen
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Mingxing Wu
- State key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
47
|
Ishii Y, Hamashima T, Yamamoto S, Sasahara M. Pathogenetic significance and possibility as a therapeutic target of platelet derived growth factor. Pathol Int 2017; 67:235-246. [DOI: 10.1111/pin.12530] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/27/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Yoko Ishii
- Department of Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama 930-0194 Japan
| | - Takeru Hamashima
- Department of Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama 930-0194 Japan
| | - Seiji Yamamoto
- Department of Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama 930-0194 Japan
| | - Masakiyo Sasahara
- Department of Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama 930-0194 Japan
| |
Collapse
|
48
|
Das S, Baker AB. Biomaterials and Nanotherapeutics for Enhancing Skin Wound Healing. Front Bioeng Biotechnol 2016; 4:82. [PMID: 27843895 PMCID: PMC5087310 DOI: 10.3389/fbioe.2016.00082] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023] Open
Abstract
Wound healing is an intricate process that requires complex coordination between many cell types and an appropriate extracellular microenvironment. Chronic wounds often suffer from high protease activity, persistent infection, excess inflammation, and hypoxia. While there has been intense investigation to find new methods to improve cutaneous wound care, the management of chronic wounds, burns, and skin wound infection remain challenging clinical problems. Ideally, advanced wound dressings can provide enhanced healing and bridge the gaps in the healing processes that prevent chronic wounds from healing. These technologies have great potential for improving outcomes in patients with poorly healing wounds but face significant barriers in addressing the heterogeneity and clinical complexity of chronic or severe wounds. Active wound dressings aim to enhance the natural healing process and work to counter many aspects that plague poorly healing wounds, including excessive inflammation, ischemia, scarring, and wound infection. This review paper discusses recent advances in the development of biomaterials and nanoparticle therapeutics to enhance wound healing. In particular, this review focuses on the novel cutaneous wound treatments that have undergone significant preclinical development or are currently used in clinical practice.
Collapse
Affiliation(s)
- Subhamoy Das
- Department of Biomedical Engineering, University of Texas at Austin , Austin, TX , USA
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA; Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|