Please wait a minute...
Frontiers of Materials Science

ISSN 2095-025X

ISSN 2095-0268(Online)

CN 11-5985/TB

Postal Subscription Code 80-974

2018 Impact Factor: 1.701

Front. Mater. Sci.    2021, Vol. 15 Issue (1) : 10-35    https://doi.org/10.1007/s11706-021-0540-1
REVIEW ARTICLE
Electrospinning: An emerging technology to construct polymer-based nanofibrous scaffolds for diabetic wound healing
Atta ur Rehman KHAN1, Yosry MORSI2, Tonghe ZHU3, Aftab AHMAD4, Xianrui XIE1, Fan YU1, Xiumei MO1()
1. State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
2. Faculty of Engineering and Industrial Sciences, Swinburne University of Technology, Boroondara, VIC 3122, Australia
3. Multidisciplinary Center for Advanced Materials of Shanghai University of Engineering Science, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China
4. Department of Bioscience, COMSATS Institute of Information Technology, Pakistan
 Download: PDF(5363 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

A chronic wound in diabetic patients is a major public health concern with socioeconomic and clinical manifestations. The underlying medical condition of diabetic patients deteriorates the wound through physiological, metabolic, molecular, and cellular pathologies. Consequently, a wound enters a vicious pathological inflammatory cycle. Many therapeutic approaches are in practice to manage diabetic wounds hence ensuring the regeneration process. Polymer-based biomaterials have come up with high therapeutic promises. Many efforts have been devoted, over the years, to build an effective wound healing material using polymers. The electrospinning technique, although not new, has turned out to be one of the most effective strategies in building wound healing biomaterials due to the special structural advantages of electrospun nanofibers over the other formulations. In this review, careful integration of all electrospinning approaches has been presented which will not only give an insight into the current updates but also be helpful in the development of new therapeutic material considering pathophysiological conditions of a diabetic wound.

Keywords diabetic wound healing      inflammation      polymers      bioactive substances      hydrogel      electrospinning      nanofibers     
Corresponding Author(s): Xiumei MO   
Online First Date: 11 February 2021    Issue Date: 11 March 2021
 Cite this article:   
Atta ur Rehman KHAN,Yosry MORSI,Tonghe ZHU, et al. Electrospinning: An emerging technology to construct polymer-based nanofibrous scaffolds for diabetic wound healing[J]. Front. Mater. Sci., 2021, 15(1): 10-35.
 URL:  
https://academic.hep.com.cn/foms/EN/10.1007/s11706-021-0540-1
https://academic.hep.com.cn/foms/EN/Y2021/V15/I1/10
Fig.1  Images displaying the events at various phases of wound healing and role of biomaterials during the healing process: (a) Schematic representation of the role of various dressing materials at various phases of wound healing. (b) The description of specific roles played by biomaterials, at various levels, during the wound healing process. Reproduced from Ref. [25] with permission of Elsevier.
Fig.2  Representative image showing the DW environment. Cause and effect are represented by arrows.
Fig.3  The typical electrospinning setup comprises syringe, pump, collector, and power supply.
Fig.4  An overview of various system requirements and parameters of electrospinning affecting the fabrication and characteristics of resultant fibers.
Fig.5  An overview of applications of electrospun nanofibers with a special focus on biomedical applications.
Fig.6  An illustration of various loading techniques for bioactive substances in electrospun nanofibers.
Fig.7  Images displaying the angiogenetic potential through various model: (a) Representative image showing the angiogenetic potential of PHBV/nCeO2-1 material on the CAM model. Reproduced from Ref. [70] with permission of American Chemical Society. (b) Fluorescent image representing tube formation on PC and PCD groups on two-time lines. Reproduced from Ref. [79] with permission of John Wiley and Sons. (c) Image showing immunofluorescence staining of CD31 at 7 and 15 d after surgery. The green color shows a positive area for CD31 depicting blood vessels, nuclei labeled as blue whereas red arrows mark the vascularized area. Reproduced from Ref. [82] with permission of Elsevier.
Growth factor Cells releasing the growth factor Function Ref.
EGF platelets; macrophages re-epithelialization; granulation tissue formation [9,94]
CTGF fibroblasts; endothelial cells; renal cells connective tissue formation; angiogenesis/vascularization; cartilage regeneration [9596]
bFGF keratinocytes; mast cells; fibroblasts; endothelial cells; smooth muscle cells; chondrocytes granulation tissue formation/angiogenesis; re-epithelialization; matrix formation and remodeling [9798]
IGF-1 vascular smooth muscle cells; hepatocytes; hepatic stellate cells; renal fibroblast granulation tissue formation; re-epithelialization [99100]
PDGF platelets; keratinocytes; macrophages; endothelial cells; fibroblasts inflammation; granulation tissue formation/angiogenesis; re-epithelialization; matrix formation and remodeling [101102]
TGF-β platelets; keratinocytes; macrophages; lymphocytes; fibroblasts inflammation; granulation tissue formation; re-epithelialization; matrix formation and remodeling [103104]
TNF-α neutrophils; macrophages collagen expression; re-epithelialization [7,105]
VEGF platelets; neutrophils; macrophages; endothelial cells; smooth muscle cells; fibroblasts granulation tissue formation/angiogenesis/neo-vascularization [8,106107]
Tab.1  Role of various growth factors in the wound healing process [79,94107]
Fig.8  Images displaying the impact of nanofibrous membrane on various in vivo and in vitro studies: (A) Representative images of the size of the wound, blank and treated with the material, recorded at various time points. Reproduced from Ref. [83] with permission of the Royal Society of Chemistry. (B) Images of histocompatibility and bio-absorbability analysis of 10% loaded NAG-PL membrane in vivo: Subcutaneous implantation of 10 NAG-PL scaffolds in mice for 7, 14 and 28 d with green arrows pointed toward the location of scaffolds (a); Masson’s trichrome staining of the scaffolds at 7, 14 and 28 d with green arrows pointed to the collagen infiltration (original magnification ×100) (b). Reproduced from Ref. [92] with permission of Elsevier. (C) Effect of MMP-9 content noted on day 14: DAPI-labeled nuclei (blue) (d–f); Cy3-conjugated secondary antibody (orange) (g–i); RhPDGF-BB-eluting PLGA-collagen hybrid scaffold up-regulated MMP-9 in the dermis (double arrow). Auto-fluorescence (a–c). Reproduced from Ref. [112] with permission of the Royal Society of Chemistry. (D) SEM images showing random, aligned, and crossed orientation of fibers (i); Fluorescent images of fibroblast cultured on various nanofibers F-actin (green) stained with phalloidin-FITC and nuclei (blue) with DAPI (ii). Reproduced from Ref. [118] with permission of the Royal Society of Chemistry.
1 I D Federation. Diabetes Facts and Figures. Ninth edition, 2019
2 L Yazdanpanah, M Nasiri, S Adarvishi. Literature review on the management of diabetic foot ulcer. World Journal of Diabetes, 2015, 6(1): 37–53
https://doi.org/10.4239/wjd.v6.i1.37 pmid: 25685277
3 M S Miller. Use of topical recombinant human platelet-derived growth factor-BB (becaplermin) in healing of chronic mixed arteriovenous lower extremity diabetic ulcers. The Journal of Foot and Ankle Surgery, 1999, 38(3): 227–231
https://doi.org/10.1016/S1067-2516(99)80058-1 pmid: 10384364
4 S Baker, A R Iorio. Application of Apligraf skin graft substitute along with autologous platelet derived growth factors in the treatment of diabetic foot ulcer. Foot, 2008, 18(4): 181–182
https://doi.org/10.1016/j.foot.2008.11.004
5 A A Omar, A I D Mavor, A M Jones, et al.. Treatment of venous leg ulcers with Dermagraft. European Journal of Vascular and Endovascular Surgery, 2004, 27(6): 666–672
https://doi.org/10.1016/j.ejvs.2004.03.001 pmid: 15121121
6 U Freudenberg, A Zieris, K Chwalek, et al.. Heparin desulfation modulates VEGF release and angiogenesis in diabetic wounds. Journal of Controlled Release, 2015, 220(Pt A): 79–88
https://doi.org/10.1016/j.jconrel.2015.10.028 pmid: 26478015
7 R L Brown, M P Breeden, D G Greenhalgh. PDGF and TGF-α act synergistically to improve wound healing in the genetically diabetic mouse. The Journal of Surgical Research, 1994, 56(6): 562–570
https://doi.org/10.1006/jsre.1994.1090 pmid: 8015312
8 P Losi, E Briganti, C Errico, et al.. Fibrin-based scaffold incorporating VEGF- and bFGF-loaded nanoparticles stimulates wound healing in diabetic mice. Acta Biomaterialia, 2013, 9(8): 7814–7821
https://doi.org/10.1016/j.actbio.2013.04.019 pmid: 23603001
9 S M Choi, K M Lee, H J Kim, et al.. Effects of structurally stabilized EGF and bFGF on wound healing in type I and type II diabetic mice. Acta Biomaterialia, 2018, 66: 325–334
https://doi.org/10.1016/j.actbio.2017.11.045 pmid: 29203426
10 Z Lei, G Singh, Z Min, et al.. Bone marrow-derived mesenchymal stem cells laden novel thermo-sensitive hydrogel for the management of severe skin wound healing. Materials Science & Engineering C, 2018, 90: 159–167
https://doi.org/10.1016/j.msec.2018.04.045 pmid: 29853078
11 L Kaisang, W Siyu, F Lijun, et al.. Adipose-derived stem cells seeded in Pluronic F-127 hydrogel promotes diabetic wound healing. The Journal of Surgical Research, 2017, 217: 63–74
https://doi.org/10.1016/j.jss.2017.04.032 pmid: 28595815
12 K B Lee, J Choi, S B Cho, et al.. Topical embryonic stem cells enhance wound healing in diabetic rats. Journal of Orthopaedic Research, 2011, 29(10): 1554–1562
https://doi.org/10.1002/jor.21385 pmid: 21469178
13 K Kaushik, A Das. Endothelial progenitor cell therapy for chronic wound tissue regeneration. Cytotherapy, 2019, 21(11): 1137–1150
https://doi.org/10.1016/j.jcyt.2019.09.002 pmid: 31668487
14 S G Keswani, A B Katz, F Y Lim, et al.. Adenoviral mediated gene transfer of PDGF-B enhances wound healing in type I and type II diabetic wounds. Wound Repair and Regeneration, 2004, 12(5): 497–504
https://doi.org/10.1111/j.1067-1927.2004.12501.x pmid: 15453831
15 A T Badillo, S Chung, L Zhang, et al.. Lentiviral gene transfer of SDF-1α to wounds improves diabetic wound healing. The Journal of Surgical Research, 2007, 143(1): 35–42
https://doi.org/10.1016/j.jss.2007.03.051 pmid: 17950070
16 K A Belek, A A Dunn, L W T Alkureishi, et al.. Attenuation of the abnormal inflammatory response in diabetic wounds with Hox gene therapy. Journal of the American College of Surgeons, 2009, 209(3S): S71
https://doi.org/10.1016/j.jamcollsurg.2009.06.172
17 V Kant, D Kumar, R Prasad, et al.. Combined effect of substance P and curcumin on cutaneous wound healing in diabetic rats. The Journal of Surgical Research, 2017, 212: 130–145
https://doi.org/10.1016/j.jss.2017.01.011 pmid: 28550899
18 S Hamed, Y Ullmann, D Egozi, et al.. Fibronectin potentiates topical erythropoietin-induced wound repair in diabetic mice. The Journal of Investigative Dermatology, 2011, 131(6): 1365–1374
https://doi.org/10.1038/jid.2011.15 pmid: 21326299
19 C H Lee, K C Hung, M J Hsieh, et al.. Core–shell insulin-loaded nanofibrous scaffolds for repairing diabetic wounds. Nanomedicine: Nanotechnology, Biology, and Medicine, 2020, 24: 102123
https://doi.org/10.1016/j.nano.2019.102123 pmid: 31711999
20 B Y Yang, C H Hu, W C Huang, et al.. Effects of bilayer nanofibrous scaffolds containing curcumin/lithospermi radix extract on wound healing in streptozotocin-induced diabetic rats. Polymers, 2019, 11(11): 1745
https://doi.org/10.3390/polym11111745 pmid: 31653001
21 M A Abu-Al-Basal. Healing potential of Rosmarinus officinalis L. on full-thickness excision cutaneous wounds in alloxan-induced-diabetic BALB/c mice. Journal of Ethnopharmacology, 2010, 131(2): 443–450
https://doi.org/10.1016/j.jep.2010.07.007 pmid: 20633625
22 T W Lau, F F Y Lam, K M Lau, et al.. Pharmacological investigation on the wound healing effects of Radix Rehmanniae in an animal model of diabetic foot ulcer. Journal of Ethnopharmacology, 2009, 123(1): 155–162
https://doi.org/10.1016/j.jep.2009.02.010 pmid: 19429355
23 S Mazumdar, A K Ghosh, M Dinda, et al.. Evaluation of wound healing activity of ethanol extract of Annona reticulata L. leaf both in vitro and in diabetic mice model. Journal of Traditional and Complementary Medicine, 2019, doi:10.1016/j.jtcme.2019.12.001
https://doi.org/10.1016/j.jtcme.2019.12.001
24 A C N Marchianti, E N Sakinah, U Elfiah, et al.. Gel formulations of Merremia mammosa (Lour.) accelerated wound healing of the wound in diabetic rats. Journal of Traditional and Complementary Medicine, 2019, doi:10.1016/j.jtcme.2019.12.002
https://doi.org/10.1016/j.jtcme.2019.12.002
25 D Chouhan, N Dey, N Bhardwaj, et al.. Emerging and innovative approaches for wound healing and skin regeneration: Current status and advances. Biomaterials, 2019, 216: 119267
https://doi.org/10.1016/j.biomaterials.2019.119267 pmid: 31247480
26 T C Alba-Loureiro, C D Munhoz, J O Martins, et al.. Neutrophil function and metabolism in individuals with diabetes mellitus. Brazilian Journal of Medical and Biological Research, 2007, 40(8): 1037–1044
https://doi.org/10.1590/S0100-879X2006005000143 pmid: 17665039
27 J A Smith. Regulation of cytokine production by the unfolded protein response; Implications for infection and autoimmunity. Frontiers in Immunology, 2018, 9: 422
https://doi.org/10.3389/fimmu.2018.00422 pmid: 29556237
28 S Dangwal, B Stratmann, C Bang, et al.. Impairment of wound healing in patients with type 2 diabetes mellitus influences circulating microRNA patterns via inflammatory cytokines. Arteriosclerosis, Thrombosis, and Vascular Biology, 2015, 35(6): 1480–1488
https://doi.org/10.1161/ATVBAHA.114.305048 pmid: 25814674
29 S Biswas, S Roy, J Banerjee, et al.. Hypoxia inducible microRNA 210 attenuates keratinocyte proliferation and impairs closure in a murine model of ischemic wounds. Proceedings of the National Academy of Sciences of the United States of America, 2010, 107(15): 6976–6981
https://doi.org/10.1073/pnas.1001653107 pmid: 20308562
30 M Sinha, S Ghatak, S Roy, et al.. MicroRNA-200b as a switch for inducible adult angiogenesis. Antioxidants & Redox Signaling, 2015, 22(14): 1257–1272
https://doi.org/10.1089/ars.2014.6065 pmid: 25761972
31 S Jhamb, V N Vangaveti, U H Malabu. Genetic and molecular basis of diabetic foot ulcers: Clinical review. Journal of Tissue Viability, 2016, 25(4): 229–236
https://doi.org/10.1016/j.jtv.2016.06.005 pmid: 27372176
32 U Förstermann, T Münzel. Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation, 2006, 113(13): 1708–1714
https://doi.org/10.1161/CIRCULATIONAHA.105.602532 pmid: 16585403
33 K Clement, P Boutin, P Froguel. Genetics of obesity. American Journal of Pharmacogenomics, 2002, 2(3): 177–187
https://doi.org/10.2165/00129785-200202030-00003 pmid: 12383024
34 D El-Lebedy, M Kafoury, D Abd-El Haleem, et al.. Paraoxonase-1 gene Q192R and L55M polymorphisms and risk of cardiovascular disease in Egyptian patients with type 2 diabetes mellitus. Journal of Diabetes and Metabolic Disorders, 2014, 13(1): 124
https://doi.org/10.1186/s40200-014-0125-y pmid: 25551104
35 S Deakin, I Leviev, M Gomaraschi, et al.. Enzymatically active paraoxonase-1 is located at the external membrane of producing cells and released by a high affinity, saturable, desorption mechanism. The Journal of Biological Chemistry, 2002, 277(6): 4301–4308
https://doi.org/10.1074/jbc.M107440200 pmid: 11726658
36 S Shenhar-Tsarfaty, N Waiskopf, K Ofek, et al.. Atherosclerosis and arteriosclerosis parameters in stroke patients associate with paraoxonase polymorphism and esterase activities. European Journal of Neurology, 2013, 20(6): 891–898
https://doi.org/10.1111/ene.12074 pmid: 23305304
37 L S Angelo, R Kurzrock. Vascular endothelial growth factor and its relationship to inflammatory mediators. Clinical Cancer Research, 2007, 13(10): 2825–2830
https://doi.org/10.1158/1078-0432.CCR-06-2416 pmid: 17504979
38 B Bruhn-Olszewska, A Korzon-Burakowska, M Gabig-Cimińska, et al.. Molecular factors involved in the development of diabetic foot syndrome. Acta Biochimica Polonica, 2012, 59(4): 507–513
https://doi.org/10.18388/abp.2012_2085 pmid: 23251910
39 D G Armstrong, E B Jude. The role of matrix metalloproteinases in wound healing. Journal of the American Podiatric Medical Association, 2002, 92(1): 12–18
https://doi.org/10.7547/87507315-92-1-12 pmid: 11796794
40 R G Sibbald, K Y Woo. The biology of chronic foot ulcers in persons with diabetes. Diabetes-Metabolism Research and Reviews, 2008, 24(S1): S25–S30
https://doi.org/10.1002/dmrr.847 pmid: 18442179
41 A Formhals. Process and apparatus for preparing artificial threads. US Patent, 1 975 504, 1934
42 J Zeleny. The electrical discharge from liquid points, and a hydrostatic method of measuring the electric intensity at their surfaces. Physical Review, 1914, 3(2): 69–91
https://doi.org/10.1103/PhysRev.3.69
43 X Xie, Y Chen, X Wang, et al.. Electrospinning nanofiber scaffolds for soft and hard tissue regeneration. Journal of Materials Science & Technology, 2020, 59: 243–261
https://doi.org/10.1016/j.jmst.2020.04.037
44 L Vysloužilová, M Buzgo, P Pokorný, et al.. Needleless coaxial electrospinning: A novel approach to mass production of coaxial nanofibers. International Journal of Pharmaceutics, 2017, 516(1–2): 293–300
https://doi.org/10.1016/j.ijpharm.2016.11.034 pmid: 27851978
45 A U R Khan, K Huang, Z Jinzhong, et al.. PLCL/silk fibroin based antibacterial nano wound dressing encapsulating oregano essential oil: Fabrication, characterization and biological evaluation. Colloids and Surfaces B: Biointerfaces, 2020, 196: 111352
https://doi.org/10.1016/j.colsurfb.2020.111352 pmid: 32919244
46 A R Khan, M Nadeem, M A Bhutto, et al.. Physico-chemical and biological evaluation of PLCL/SF nanofibers loaded with oregano essential oil. Pharmaceutics, 2019, 11(8): 386
https://doi.org/10.3390/pharmaceutics11080386
47 C Maderuelo, A Zarzuelo, J M Lanao. Critical factors in the release of drugs from sustained release hydrophilic matrices. Journal of Controlled Release, 2011, 154(1): 2–19
https://doi.org/10.1016/j.jconrel.2011.04.002 pmid: 21497624
48 S Iqbal, M H Rashid, A S Arbab, et al.. Encapsulation of anticancer drugs (5-fluorouracil and paclitaxel) into polycaprolactone (PCL) nanofibers and in vitro testing for sustained and targeted therapy. Journal of Biomedical Nanotechnology, 2017, 13(4): 355–366
https://doi.org/10.1166/jbn.2017.2353 pmid: 28845137
49 T Tadros. Principles of emulsion stabilization with special reference to polymeric surfactants. Journal of Cosmetic Science, 2006, 57(2): 153–169
pmid: 16688378
50 A Camerlo, A M Bühlmann-Popa, C Vebert-Nardin, et al.. Environmentally controlled emulsion electrospinning for the encapsulation of temperature-sensitive compounds. Journal of Materials Science, 2014, 49(23): 8154–8162
https://doi.org/10.1007/s10853-014-8524-5
51 X B Xiong, H Uludağ, A Lavasanifar. Engineering of amphiphilic block copolymers for drug and gene delivery. In: M M de Villiers, P Aramwit, G S Kwon, eds. Nanotechnology in Drug Delivery. New York, NY: Springer New York, 2009, 385–422
52 Y N Jiang, H Y Mo, D G Yu. Electrospun drug-loaded core–sheath PVP/zein nanofibers for biphasic drug release. International Journal of Pharmaceutics, 2012, 438(1–2): 232–239
https://doi.org/10.1016/j.ijpharm.2012.08.053 pmid: 22981688
53 M Yoshida, R Langer, A Lendlein, et al.. From advanced biomedical coatings to multi-functionalized biomaterials. Polymer Reviews, 2006, 46(4): 347–375
54 H S Yoo, T G Kim, T G Park. Surface-functionalized electrospun nanofibers for tissue engineering and drug delivery. Advanced Drug Delivery Reviews, 2009, 61(12): 1033–1042
https://doi.org/10.1016/j.addr.2009.07.007 pmid: 19643152
55 B S Kim, S W Park, P T Hammond. Hydrogen-bonding layer-by-layer-assembled biodegradable polymeric micelles as drug delivery vehicles from surfaces. ACS Nano, 2008, 2(2): 386–392
https://doi.org/10.1021/nn700408z pmid: 19206641
56 B Thierry, P Kujawa, C Tkaczyk, et al.. Delivery platform for hydrophobic drugs: Prodrug approach combined with self-assembled multilayers. Journal of the American Chemical Society, 2005, 127(6): 1626–1627
https://doi.org/10.1021/ja045077s pmid: 15700982
57 T G Kim, T G Park. Biomimicking extracellular matrix: cell adhesive RGD peptide modified electrospun poly(D,L-lactic-co-glycolic acid) nanofiber mesh. Tissue Engineering, 2006, 12(2): 221–233
https://doi.org/10.1089/ten.2006.12.221 pmid: 16548681
58 J S Choi, K W Leong, H S Yoo. In vivo wound healing of diabetic ulcers using electrospun nanofibers immobilized with human epidermal growth factor (EGF). Biomaterials, 2008, 29(5): 587–596
https://doi.org/10.1016/j.biomaterials.2007.10.012 pmid: 17997153
59 S Cui, X Sun, K Li, et al.. Polylactide nanofibers delivering doxycycline for chronic wound treatment. Materials Science & Engineering C, 2019, 104: 109745
https://doi.org/10.1016/j.msec.2019.109745 pmid: 31499963
60 S Tort, F Acartürk, A Beşikci. Evaluation of three-layered doxycycline-collagen loaded nanofiber wound dressing. International Journal of Pharmaceutics, 2017, 529(1–2): 642–653
https://doi.org/10.1016/j.ijpharm.2017.07.027 pmid: 28705624
61 S Tort, F T Demiroz, S C Cevher, et al.. The effect of a new wound dressing on wound healing: Biochemical and histopathological evaluation. Burns, 2020, 46(1): 143–155
https://doi.org/10.1016/j.burns.2019.02.013 pmid: 31862280
62 M Ranjbar-Mohammadi, S Rabbani, S H Bahrami, et al.. Antibacterial performance and in vivo diabetic wound healing of curcumin loaded gum tragacanth/poly(ε-caprolactone) electrospun nanofibers. Materials Science & Engineering C, 2016, 69: 1183–1191
https://doi.org/10.1016/j.msec.2016.08.032 pmid: 27612816
63 C Dwivedi, I Pandey, H Pandey, et al.. In vivo diabetic wound healing with nanofibrous scaffolds modified with gentamicin and recombinant human epidermal growth factor. Journal of Biomedical Materials Research Part A, 2018, 106(3): 641–651
https://doi.org/10.1002/jbm.a.36268 pmid: 28986947
64 R Ahmed, M Tariq, I Ali, et al.. Novel electrospun chitosan/polyvinyl alcohol/zinc oxide nanofibrous mats with antibacterial and antioxidant properties for diabetic wound healing. International Journal of Biological Macromolecules, 2018, 120(Pt A): 385–393
https://doi.org/10.1016/j.ijbiomac.2018.08.057 pmid: 30110603
65 P H Lin, M Sermersheim, H Li, et al.. Zinc in wound healing modulation. Nutrients, 2017, 10(1): 16
https://doi.org/10.3390/nu10010016 pmid: 29295546
66 G Cicco, F Giorgino, S Cicco. Wound healing in diabetes: hemorheological and microcirculatory aspects. Oxygen Transport to Tissue XXXII. Springer, 2011, 263–269
67 F Giacco, M Brownlee. Oxidative stress and diabetic complications. Circulation Research, 2010, 107(9): 1058–1070
https://doi.org/10.1161/CIRCRESAHA.110.223545 pmid: 21030723
68 A Burns, W T Self. Antioxidant inorganic nanoparticles and their potential applications in biomedicine. In: Smart Nanoparticles for Biomedicine. Elsevier, 2018, 159–169
https://doi.org/10.1016/B978-0-12-814156-4.00011-2
69 S S Lee, W Song, M Cho, et al.. Antioxidant properties of cerium oxide nanocrystals as a function of nanocrystal diameter and surface coating. ACS Nano, 2013, 7(11): 9693–9703
https://doi.org/10.1021/nn4026806 pmid: 24079896
70 R Augustine, A Hasan, N K Patan, et al.. Cerium oxide nanoparticle incorporated electrospun poly(3-hydroxybutyrate-co-3-hydroxyvalerate) membranes for diabetic wound healing applications. ACS Biomaterials Science & Engineering, 2020, 6(1): 58–70
https://doi.org/10.1021/acsbiomaterials.8b01352
71 F Liu, X Li, L Wang, et al.. Sesamol incorporated cellulose acetate–zein composite nanofiber membrane: An efficient strategy to accelerate diabetic wound healing. International Journal of Biological Macromolecules, 2020, 149: 627–638
https://doi.org/10.1016/j.ijbiomac.2020.01.277 pmid: 32004602
72 A D Pinzón-García, P Cassini-Vieira, C C Ribeiro, et al.. Efficient cutaneous wound healing using bixin-loaded PCL nanofibers in diabetic mice. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 2017, 105(7): 1938–1949
https://doi.org/10.1002/jbm.b.33724 pmid: 27292445
73 S Jagtap, K Meganathan, V Wagh, et al.. Chemoprotective mechanism of the natural compounds, epigallocatechin-3-O-gallate, quercetin and curcumin against cancer and cardiovascular diseases. Current Medicinal Chemistry, 2009, 16(12): 1451–1462
https://doi.org/10.2174/092986709787909578 pmid: 19355899
74 S Das, J Tanwar, S Hameed, et al.. Antimicrobial potential of epigallocatechin-3-gallate (EGCG): a green tea polyphenol. Journal of Biochemical and Pharmacological Research, 2014, 2(3): 167–174
75 Y C Shin, D M Shin, E J Lee, et al.. Hyaluronic acid/PLGA core/shell fiber matrices loaded with EGCG beneficial to diabetic wound healing. Advanced Healthcare Materials, 2016, 5(23): 3035–3045
https://doi.org/10.1002/adhm.201600658 pmid: 27805803
76 R C Riddle, R Khatri, E Schipani, et al.. Role of hypoxia-inducible factor-1α in angiogenic-osteogenic coupling. Journal of Molecular Medicine, 2009, 87(6): 583–590
https://doi.org/10.1007/s00109-009-0477-9 pmid: 19415227
77 B L Krock, N Skuli, M C Simon. Hypoxia-induced angiogenesis: good and evil. Genes & Cancer, 2011, 2(12): 1117–1133
https://doi.org/10.1177/1947601911423654 pmid: 22866203
78 K J Woo, T J Lee, J W Park, et al.. Desferrioxamine, an iron chelator, enhances HIF-1 accumulation via cyclooxygenase-2 signaling pathway. Biochemical and Biophysical Research Communications, 2006, 343(1): 8–14
https://doi.org/10.1016/j.bbrc.2006.02.116 pmid: 16527254
79 H Chen, P Jia, H Kang, et al.. Upregulating Hif-1α by hydrogel nanofibrous scaffolds for rapidly recruiting angiogenesis relative cells in diabetic wound. Advanced Healthcare Materials, 2016, 5(8): 907–918
https://doi.org/10.1002/adhm.201501018 pmid: 26891197
80 M Hägg, S Wennström. Activation of hypoxia-induced transcription in normoxia. Experimental Cell Research, 2005, 306(1): 180–191
https://doi.org/10.1016/j.yexcr.2005.01.017 pmid: 15878343
81 Q Zhang, J H Oh, C H Park, et al.. Effects of dimethyloxalylglycine-embedded poly(ε-caprolactone) fiber meshes on wound healing in diabetic rats. ACS Applied Materials & Interfaces, 2017, 9(9): 7950–7963
https://doi.org/10.1021/acsami.6b15815 pmid: 28211272
82 X Ren, Y Han, J Wang, et al.. An aligned porous electrospun fibrous membrane with controlled drug delivery — An efficient strategy to accelerate diabetic wound healing with improved angiogenesis. Acta Biomaterialia, 2018, 70: 140–153
https://doi.org/10.1016/j.actbio.2018.02.010 pmid: 29454159
83 W Gao, L Sun, X Fu, et al.. Enhanced diabetic wound healing by electrospun core-sheath fibers loaded with dimethyloxalylglycine. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2018, 6(2): 277–288
https://doi.org/10.1039/C7TB02342A pmid: 32254170
84 S Bhakta, P E Faira, L A Salata, et al.. Determination of relative in vivo osteoconductivity of modified potassium fluorrichterite glass-ceramics compared with 45S5 bioglass. Journal of Materials Science: Materials in Medicine, 2012, 23(10): 2521–2529
https://doi.org/10.1007/s10856-012-4707-2 pmid: 22752884
85 F Tan, M Naciri, M Al-Rubeai. Osteoconductivity and growth factor production by MG63 osteoblastic cells on bioglass-coated orthopedic implants. Biotechnology and Bioengineering, 2011, 108(2): 454–464
https://doi.org/10.1002/bit.22955 pmid: 20872820
86 X Yan, B Chen, Y Lin, et al.. Acceleration of diabetic wound healing by collagen-binding vascular endothelial growth factor in diabetic rat model. Diabetes Research and Clinical Practice, 2010, 90(1): 66–72
https://doi.org/10.1016/j.diabres.2010.07.001 pmid: 20667614
87 H S Kim, H S Yoo. In vitro and in vivo epidermal growth factor gene therapy for diabetic ulcers with electrospun fibrous meshes. Acta Biomaterialia, 2013, 9(7): 7371–7380
https://doi.org/10.1016/j.actbio.2013.03.018 pmid: 23528498
88 I Allan, H Newman, M Wilson. Antibacterial activity of particulate Bioglass(R) against supra- and subgingival bacteria. Biomaterials, 2001, 22(12): 1683–1687
https://doi.org/10.1016/S0142-9612(00)00330-6 pmid: 11374470
89 Q Chen, J Wu, Y Liu, et al.. Electrospun chitosan/PVA/bioglass nanofibrous membrane with spatially designed structure for accelerating chronic wound healing. Materials Science & Engineering C, 2019, 105: 110083
https://doi.org/10.1016/j.msec.2019.110083 pmid: 31546466
90 W Gao, W Jin, Y Li, et al.. A highly bioactive bone extracellular matrix-biomimetic nanofibrous system with rapid angiogenesis promotes diabetic wound healing. Journal of Materials Chemis-try B: Materials for Biology and Medicine, 2017, 5(35): 7285–7296
https://doi.org/10.1039/C7TB01484H pmid: 32264178
91 J Li, F Lv, H Xu, et al.. A patterned nanocomposite membrane for high-efficiency healing of diabetic wound. Journal of Materials Chemistry B: Materials for Biology and Medicine, 2017, 5(10): 1926–1934
https://doi.org/10.1039/C7TB00124J pmid: 32263946
92 F Lv, J Wang, P Xu, et al.. A conducive bioceramic/polymer composite biomaterial for diabetic wound healing. Acta Biomaterialia, 2017, 60: 128–143
https://doi.org/10.1016/j.actbio.2017.07.020 pmid: 28713016
93 F Zafari, S Shirian, M Sadeghi, et al.. CD93 hematopoietic stem cells improve diabetic wound healing by VEGF activation and downregulation of DAPK-1. Journal of Cellular Physiology, 2020, 235(3): 2366–2376
https://doi.org/10.1002/jcp.29142 pmid: 31549396
94 S Thönes, S Rother, T Wippold, et al.. Hyaluronan/collagen hydrogels containing sulfated hyaluronan improve wound healing by sustained release of heparin-binding EGF-like growth factor. Acta Biomaterialia, 2019, 86: 135–147
https://doi.org/10.1016/j.actbio.2019.01.029 pmid: 30660005
95 T Shimo, T Nakanishi, T Nishida, et al.. Connective tissue growth factor induces the proliferation, migration, and tube formation of vascular endothelial cells in vitro, and angiogenesis in vivo. Journal of Biochemistry, 1999, 126(1): 137–145
https://doi.org/10.1093/oxfordjournals.jbchem.a022414 pmid: 10393331
96 M K Das, S Basak, M S Ahmed, et al.. Connective tissue growth factor induces tube formation and IL-8 production in first trimester human placental trophoblast cells. European Journal of Obstetrics, Gynecology, and Reproductive Biology, 2014, 181: 183–188
https://doi.org/10.1016/j.ejogrb.2014.07.045 pmid: 25150958
97 J Zhan, H Xu, Y Zhong, et al.. Surface modification of patterned electrospun nanofibrous films via the adhesion of DOPA-bFGF and DOPA-ponericin G1 for skin wound healing. Materials & Design, 2020, 188: 108432
https://doi.org/10.1016/j.matdes.2019.108432
98 A Shamloo, M Sarmadi, Z Aghababaie, et al.. Accelerated full-thickness wound healing via sustained bFGF delivery based on a PVA/chitosan/gelatin hydrogel incorporating PCL microspheres. International Journal of Pharmaceutics, 2018, 537(1–2): 278–289
https://doi.org/10.1016/j.ijpharm.2017.12.045 pmid: 29288809
99 E Emmerson, L Campbell, F C Davies, et al.. Insulin-like growth factor-1 promotes wound healing in estrogen-deprived mice: New insights into cutaneous IGF-1R/ERα cross talk. The Journal of Investigative Dermatology, 2012, 132(12): 2838–2848
https://doi.org/10.1038/jid.2012.228 pmid: 22810305
100 M R Rajasekaran, S R Lee, K H Kim, et al.. Role of GIV/Girdin and integrin signaling pathways in urethral stricture fibrogenesis. Journal of Urology, 2018, 199(4S): e470–e471
https://doi.org/10.1016/j.juro.2018.02.1163
101 K Xu, N An, H Zhang, et al.. Sustained-release of PDGF from PLGA microsphere embedded thermo-sensitive hydrogel promoting wound healing by inhibiting autophagy. Journal of Drug Delivery Science and Technology, 2020, 55: 101405
https://doi.org/10.1016/j.jddst.2019.101405
102 R Judith, M Nithya, C Rose, et al.. Application of a PDGF-containing novel gel for cutaneous wound healing. Life Sciences, 2010, 87(1–2): 1–8
https://doi.org/10.1016/j.lfs.2010.05.003 pmid: 20470785
103 D Mokoena, S S Dhilip Kumar, N N Houreld, et al.. Role of photobiomodulation on the activation of the Smad pathway via TGF-β in wound healing. Journal of Photochemistry and Photobiology B: Biology, 2018, 189: 138–144
https://doi.org/10.1016/j.jphotobiol.2018.10.011 pmid: 30343208
104 S Y Choi, B H Kim, B K Huh, et al.. Tranilast-delivery surgical sutures to ameliorate wound healing by reducing scar formation through regulation of TGF-β expression and fibroblast recruitment. Journal of Industrial and Engineering Chemistry, 2018, 67: 469–477
https://doi.org/10.1016/j.jiec.2018.07.021
105 L P Pereira, M R L Mota, L A C Brizeno, et al.. Modulator effect of a polysaccharide-rich extract from Caesalpinia ferrea stem barks in rat cutaneous wound healing: Role of TNF-α, IL-1β, NO, TGF-β. Journal of Ethnopharmacology, 2016, 187: 213–223
https://doi.org/10.1016/j.jep.2016.04.043 pmid: 27125588
106 G Liu, X Wang, X Sun, et al.. The effect of urine-derived stem cells expressing VEGF loaded in collagen hydrogels on myogenesis and innervation following after subcutaneous implantation in nude mice. Biomaterials, 2013, 34(34): 8617–8629
https://doi.org/10.1016/j.biomaterials.2013.07.077 pmid: 23932297
107 P Bao, A Kodra, M Tomic-Canic, et al.. The role of vascular endothelial growth factor in wound healing. The Journal of Surgical Research, 2009, 153(2): 347–358
https://doi.org/10.1016/j.jss.2008.04.023 pmid: 19027922
108 M C Robson, L G Phillips, W T Lawrence, et al.. The safety and effect of topically applied recombinant basic fibroblast growth factor on the healing of chronic pressure sores. Annals of Surgery, 1992, 216(4): 401–408
https://doi.org/10.1097/00000658-199210000-00002 pmid: 1417189
109 R Augustine, A A Zahid, A Hasan, et al.. CTGF loaded electrospun dual porous core–shell membrane for diabetic wound healing. International Journal of Nanomedicine, 2019, 14: 8573–8588
https://doi.org/10.2147/IJN.S224047 pmid: 31802870
110 D Chouhan, G Janani, B Chakraborty, et al.. Functionalized PVA–silk blended nanofibrous mats promote diabetic wound healing via regulation of extracellular matrix and tissue remodelling. Journal of Tissue Engineering and Regenerative Medicine, 2018, 12(3): e1559–e1570
https://doi.org/10.1002/term.2581 pmid: 28987032
111 I Garcia-Orue, G Gainza, F B Gutierrez, et al.. Novel nanofibrous dressings containing rhEGF and Aloe vera for wound healing applications. International Journal of Pharmaceutics, 2017, 523(2): 556–566
https://doi.org/10.1016/j.ijpharm.2016.11.006 pmid: 27825864
112 C H Lee, Y K Chao, S H Chang, et al.. Nanofibrous rhPDGF-eluting PLGA–collagen hybrid scaffolds enhance healing of diabetic wounds. RSC Advances, 2016, 6(8): 6276–6284
https://doi.org/10.1039/C5RA21693A
113 M H Lima, A M Caricilli, L L de Abreu, et al.. Topical insulin accelerates wound healing in diabetes by enhancing the AKT and ERK pathways: A double-blind placebo-controlled clinical trial. PLoS One, 2012, 7(5): e36974
https://doi.org/10.1371/journal.pone.0036974 pmid: 22662132
114 S E Greenway, L E Filler, F L Greenway. Topical insulin in wound healing: a randomised, double-blind, placebo-controlled trial. Journal of Wound Care, 1999, 8(10): 526–528
https://doi.org/10.12968/jowc.1999.8.10.26217 pmid: 10827659
115 O Rezvani, E Shabbak, A Aslani, et al.. A randomized, double-blind, placebo-controlled trial to determine the effects of topical insulin on wound healing. Ostomy/Wound Management, 2009, 55(8): 22–28
pmid: 19717853
116 C H Lee, S H Chang, W J Chen, et al.. Augmentation of diabetic wound healing and enhancement of collagen content using nanofibrous glucophage-loaded collagen/PLGA scaffold membranes. Journal of Colloid and Interface Science, 2015, 439: 88–97
https://doi.org/10.1016/j.jcis.2014.10.028 pmid: 25463179
117 S A Majd, M R Khorasgani, S J Moshtaghian, et al.. Application of chitosan/PVA nano fiber as a potential wound dressing for streptozotocin-induced diabetic rats. International Journal of Biological Macromolecules, 2016, 92: 1162–1168
https://doi.org/10.1016/j.ijbiomac.2016.06.035 pmid: 27492559
118 L Sun, W Gao, X Fu, et al.. Enhanced wound healing in diabetic rats by nanofibrous scaffolds mimicking the basketweave pattern of collagen fibrils in native skin. Biomaterials Science, 2018, 6(2): 340–349
https://doi.org/10.1039/C7BM00545H pmid: 29265119
[1] Yuxuan MAO, Mingming PAN, Huilin YANG, Xiao LIN, Lei YANG. Injectable hydrogel wound dressing based on strontium ion cross-linked starch[J]. Front. Mater. Sci., 2020, 14(2): 232-241.
[2] Zhicun WANG, Xiaoman HAN, Yixi WANG, Kenan MEN, Lin CUI, Jianning WU, Guihua MENG, Zhiyong LIU, Xuhong GUO. Facile preparation of low swelling, high strength, self-healing and pH-responsive hydrogels based on the triple-network structure[J]. Front. Mater. Sci., 2019, 13(1): 54-63.
[3] Xin PAN, Binbin SUN, Xiumei MO. Electrospun polypyrrole-coated polycaprolactone nanoyarn nerve guidance conduits for nerve tissue engineering[J]. Front. Mater. Sci., 2018, 12(4): 438-446.
[4] Liu YUAN, Wenshuai FAN, Linyingjun HAN, Changan GUO, Zuoqin YAN, Meifang ZHU, Xiumei MO. Evaluation of hydrogels for soft tissue adhesives in vitro and in vivo analyses[J]. Front. Mater. Sci., 2018, 12(1): 95-104.
[5] Wanyu ZHAO, Jian LI, Bingbing FAN, Gang SHAO, Hailong WANG, Bozhen SONG, Shengnan WEI, Rui ZHANG. Microwave synthesis of chain-like zircona nanofibers through carbon-induced self-assembly growth[J]. Front. Mater. Sci., 2017, 11(4): 353-357.
[6] Shenglian YAO, Xujia FENG, Wenhao LI, Lu-Ning WANG, Xiumei WANG. Regulation of RAW 264.7 macrophages behavior on anodic TiO2 nanotubular arrays[J]. Front. Mater. Sci., 2017, 11(4): 318-327.
[7] Lulu WEI, Beibei LU, Lei LI, Jianning WU, Zhiyong LIU, Xuhong GUO. One-step synthesis and self-assembly behavior of thermo-responsive star-shaped β-cyclodextrin--(P(MEO 2MA-co-PEGMA))21 copolymers[J]. Front. Mater. Sci., 2017, 11(3): 223-232.
[8] Juan WANG,Binbin SUN,Muhammad Aqeel BHUTTO,Tonghe ZHU,Kui YU,Jiayu BAO,Yosry MORSI,Hany EL-HAMSHARY,Mohamed EL-NEWEHY,Xiumei MO. Fabrication and characterization of Antheraea pernyi silk fibroin-blended P(LLA-CL) nanofibrous scaffolds for peripheral nerve tissue engineering[J]. Front. Mater. Sci., 2017, 11(1): 22-32.
[9] Fengyi GUAN,Jiaju LU,Xiumei WANG. A novel honeycomb cell assay kit designed for evaluating horizontal cell migration in response to functionalized self-assembling peptide hydrogels[J]. Front. Mater. Sci., 2017, 11(1): 13-21.
[10] Xuran GUO,Kaile ZHANG,Mohamed EL-AASSAR,Nanping WANG,Hany EL-HAMSHARY,Mohamed EL-NEWEHY,Qiang FU,Xiumei MO. The comparison of the Wnt signaling pathway inhibitor delivered electrospun nanoyarn fabricated with two methods for the application of urethroplasty[J]. Front. Mater. Sci., 2016, 10(4): 346-357.
[11] Li-Da HOU,Zhen LI,Yu PAN,MuhammadIqbal SABIR,Yu-Feng ZHENG,Li LI. A review on biodegradable materials for cardiovascular stent application[J]. Front. Mater. Sci., 2016, 10(3): 238-259.
[12] Junfeng ZHOU,Liang CHENG,Xiaodan SUN,Xiumei WANG,Shouhong JIN,Junxiang LI,Qiong WU. Neurogenic differentiation of human umbilical cord mesenchymal stem cells on aligned electrospun polypyrrole/polylactide composite nanofibers with electrical stimulation[J]. Front. Mater. Sci., 2016, 10(3): 260-269.
[13] Qilin WEI,Feiyang XU,Xingjian XU,Xue GENG,Lin YE,Aiying ZHANG,Zengguo FENG. The multifunctional wound dressing with core–shell structured fibers prepared by coaxial electrospinning[J]. Front. Mater. Sci., 2016, 10(2): 113-121.
[14] Jianchao ZHAN,Yosry MORSI,Hany EI-HAMSHARY,Salem S. AL-DEYAB,Xiumei MO. In vitro evaluation of electrospun gelatin–glutaraldehyde nanofibers[J]. Front. Mater. Sci., 2016, 10(1): 90-100.
[15] Xi LIU,Bin PI,Hui WANG,Xiu-Mei WANG. Self-assembling peptide nanofiber hydrogels for central nervous system regeneration[J]. Front. Mater. Sci., 2015, 9(1): 1-13.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed