309
Views
61
CrossRef citations to date
0
Altmetric
Original Research

CTGF Loaded Electrospun Dual Porous Core-Shell Membrane For Diabetic Wound Healing

, ORCID Icon, ORCID Icon, ORCID Icon &
Pages 8573-8588 | Published online: 31 Oct 2019

References

  • Borys S, Hohendorff J, Frankfurter C, Kiec-Wilk B, Malecki MT. Negative pressure wound therapy use in diabetic foot syndrome—from mechanisms of action to clinical practice. Eur J Clin Invest. 2019;e13067. doi:10.1111/eci.1306730600541
  • Cunha M, Queirós P, Cardoso D, Santos E, Rodrigues M, Apóstolo J. The effectiveness of cleansing solutions for wound treatment: a systematic review. JBI Database System Rev Implement Rep. 2014;12(10):121–151. doi:10.11124/jbisrir-2014-1746
  • Xu F, Zhang C, Graves DT. Abnormal cell responses and role of TNF- α in impaired diabetic wound healing. Biomed Res Int. 2013;2013:1–9. doi:10.1155/2013/754802
  • Bao P, Kodra A, Tomic-Canic M, Golinko MS, Ehrlich HP, Brem H. The role of vascular endothelial growth factor in wound healing. J Surg Res. 2009;153(2):347–358. doi:10.1016/j.jss.2008.04.02319027922
  • Hardwicke J, Schmaljohann D, Boyce D, Thomas D. Epidermal growth factor therapy and wound healing - past, present and future perspectives. Surgeon. 2008;6(3):172–177. doi:10.1016/S1479-666X(08)80114-X18581754
  • Pierce GF, Mustoe TA, Altrock BW, Deuel TF, Thomason A. Role of platelet-derived growth factor in wound healing. J Cell Biochem. 1991;45(4):319–326. doi:10.1002/jcb.240450403
  • Li H, Cao C, Huang A, Man Y. Topically applied connective tissue growth factor/CCN2 Improves diabetic preclinical cutaneous wound healing: potential role for CTGF in human diabetic foot ulcer healing. J Diabetes Res. 2015;2015:1–3. doi:10.1155/2015/512959
  • Kolluru GK, SC B, Kevil CG. Endothelial dysfunction and diabetes: effects on angiogenesis, vascular remodeling, and wound healing. Int J Vasc Med. 2012;2012:1–30. doi:10.1155/2012/918267
  • Brigstock DR. Regulation of angiogenesis and endothelial cell function by connective tissue growth factor (CTGF) and cysteine-rich 61 (CYR61). Angiogenesis. 2002;5(3):153–165. doi:10.1023/A:102382380351012831056
  • Hishikawa K, Nakaki T, Fujii T. Connective tissue growth factor induces apoptosis via caspase 3 in cultured human aortic smooth muscle cells. Eur J Pharmacol. 2000;392(1–2):19–22. doi:10.1016/S0014-2999(00)00115-110748267
  • Shi-Wen X, Leask A, Abraham D. Regulation and function of connective tissue growth factor/CCN2 in tissue repair, scarring and fibrosis. Cytokine Growth Factor Rev. 2008;19(2):133–144. doi:10.1016/j.cytogfr.2008.01.00218358427
  • Frazier K, Williams S, Kothapalli D, Klapper H, Grotendorst GR. Stimulation of fibroblast cell growth, matrix production, and granulation tissue formation by connective tissue growth factor. J Invest Dermatol. 1996;107(3):404–411. doi:10.1111/1523-1747.ep123633898751978
  • Nakerakanti SS, Kapanadze B, Trojanowska M, Ghatnekar A, Markiewicz M. Connective Tissue Growth Factor (CTGF/CCN2) mediates angiogenic effect of S1P in human dermal microvascular endothelial cells. Microcirculation. 2010;18(1):1–11. doi:10.1111/j.1549-8719.2010.00058.x
  • Berlanga-Acosta J, Fernández-Montequín J, Valdés-Pérez C, et al. Diabetic foot ulcers and epidermal growth factor: revisiting the local delivery route for a successful outcome. Biomed Res Int. 2017;2017:1–10. doi:10.1155/2017/2923759
  • Masood N, Ahmed R, Tariq M, et al. Silver nanoparticle impregnated chitosan-PEG hydrogel enhances wound healing in diabetes induced rabbits. Int J Pharm. 2019;559:23–36. doi:10.1016/j.ijpharm.2019.01.01930668991
  • Ahmed R, Tariq M, Ali I, et al. Novel electrospun chitosan/polyvinyl alcohol/zinc oxide nanofibrous mats with antibacterial and antioxidant properties for diabetic wound healing. Int J Biol Macromol. 2018;120:385–393. doi:10.1016/j.ijbiomac.2018.08.05730110603
  • Liu M, Zhang H, Min D, et al. Dual layered wound dressing with simultaneous temperature & antibacterial regulation properties. Mater Sci Eng C. 2019;94:1077–1082. doi:10.1016/j.msec.2018.09.049
  • Zahid AA, Ahmed R, ur Rehman SR, et al. Nitric oxide releasing chitosan-poly (vinyl alcohol) hydrogel promotes angiogenesis in chick embryo model. Int J Biol Macromol 2019;136:901–910. doi:10.1016/j.ijbiomac.2019.06.136
  • Wang J, Cheng Q, Lin L, Jiang L. Synergistic toughening of bioinspired poly(vinyl alcohol)-clay- nanofibrillar cellulose artificial nacre. ACS Nano. 2014;8(3):2739–2745. doi:10.1021/nn406428n24506706
  • Kamoun EA, Kenawy ERS, Chen X. A review on polymeric hydrogel membranes for wound dressing applications: PVA-based hydrogel dressings. J Adv Res. 2017;8(3):217–233. doi:10.1016/j.jare.2017.01.00528239493
  • Hayes JC, Kennedy JE. An evaluation of the biocompatibility properties of a salt-modified polyvinyl alcohol hydrogel for a knee meniscus application. Mater Sci Eng C. 2016;59:894–900. doi:10.1016/j.msec.2015.10.052
  • Hasan A, Soliman S, El Hajj F, Tseng YT, Yalcin HC, Marei HE. Fabrication and in vitro characterization of a tissue engineered PCL-PLLA heart valve. Sci Rep. 2018;8(1):8187. doi:10.1038/s41598-018-26452-y29844329
  • Gandolfi MG, Zamparini F, Degli Esposti M, et al. Polylactic acid-based porous scaffolds doped with calcium silicate and dicalcium phosphate dihydrate designed for biomedical application. Mater Sci Eng C. 2018;82:163–181. doi:10.1016/j.msec.2017.08.040
  • da Silva D, Kaduri M, Poley M, et al. Biocompatibility, biodegradation and excretion of polylactic acid (PLA) in medical implants and theranostic systems. Chem Eng J. 2018;340:9–14. doi:10.1016/J.CEJ.2018.01.01031384170
  • Han X, Wang D, Chen X, Lin H, Qu F. One-pot synthesis of macro-mesoporous bioactive glasses/polylactic acid for bone tissue engineering. Mater Sci Eng C. 2014;43:367–374. doi:10.1016/j.msec.2014.07.042
  • Childs A, Hemraz UD, Castro NJ, Fenniri H, Zhang LG. Novel biologically-inspired rosette nanotube PLLA scaffolds for improving human mesenchymal stem cell chondrogenic differentiation. Biomed Mater. 2013;8(6):065003. doi:10.1088/1748-6041/8/6/06500324225196
  • Hasan A, Memic A, Annabi N, et al. Electrospun scaffolds for tissue engineering of vascular grafts. Acta Biomater. 2014;10(1):11–25. doi:10.1016/j.actbio.2013.08.02223973391
  • Augustine R, Hasan A, Patan NK, et al. Cerium Oxide Nanoparticle Incorporated Electrospun Poly(3-hydroxybutyrate-co-3-hydroxyvalerate) Membranes for Diabetic Wound Healing Applications. ACS Biomater Sci Eng 2019. doi:10.1021/acsbiomaterials.8b01352
  • Tonsomboon K, Butcher AL, Oyen ML. Strong and tough nanofibrous hydrogel composites based on biomimetic principles. Mater Sci Eng C. 2017;72:220–227. doi:10.1016/j.msec.2016.11.025
  • Augustine R, Dan P, Sosnik A, et al. Electrospun poly(vinylidene fluoride-trifluoroethylene)/zinc oxide nanocomposite tissue engineering scaffolds with enhanced cell adhesion and blood vessel formation. Nano Res. 2017;10(10):3358–3376. doi:10.1007/s12274-017-1549-8
  • Augustine R, Sarry F, Kalarikkal N, Thomas S, Badie L, Rouxel D. Surface acoustic wave device with reduced insertion loss by electrospinning P(VDF–trFE)/ZnO nanocomposites. Nanomicro Lett. 2016;8(3):282–290. doi:10.1007/s40820-016-0088-230460288
  • Recek N, Resnik M, Motaln H, et al. Cell adhesion on polycaprolactone modified by plasma treatment. Int J Polym Sci. 2016;2016. doi:10.1155/2016/7354396
  • Augustine R, Kalarikkal N, Thomas S. Clogging-free electrospinning of polycaprolactone using acetic acid/acetone mixture. Polym Plast Technol Eng. 2016;55(5):518–529. doi:10.1080/03602559.2015.1036451
  • Alavarse AC, de Oliveira Silva FW, Colque JT, et al. Tetracycline hydrochloride-loaded electrospun nanofibers mats based on PVA and chitosan for wound dressing. Mater Sci Eng C. 2017;77:271–281. doi:10.1016/j.msec.2017.03.199
  • Butcher AL, Koh CT, Oyen ML. Systematic mechanical evaluation of electrospun gelatin meshes. J Mech Behav Biomed Mater. 2017;69:412–419. doi:10.1016/j.jmbbm.2017.02.00728208112
  • Duque Sánchez L, Brack N, Postma A, Pigram PJ, Meagher L. Surface modification of electrospun fibres for biomedical applications: a focus on radical polymerization methods. Biomaterials. 2016;106:24–45. doi:10.1016/j.biomaterials.2016.08.01127543920
  • Lee S-J, Nowicki M, Harris B, Zhang LG. Fabrication of a highly aligned neural scaffold via a table top stereolithography 3D printing and electrospinning. Tissue Eng Part A. 2016;23(11–12):491–502. doi:10.1089/ten.tea.2016.0353
  • Johnson R, Ding Y, Nagiah N, Monnet E, Tan W. Coaxially-structured fibres with tailored material properties for vascular graft implant. Mater Sci Eng C. 2019;97:1–11. doi:10.1016/j.msec.2018.11.036
  • da Silva TN, Gonçalves RP, Rocha CL, et al. Controlling burst effect with PLA/PVA coaxial electrospun scaffolds loaded with BMP-2 for bone guided regeneration. Mater Sci Eng C. 2019;97:602–612. doi:10.1016/j.msec.2018.12.020
  • Gonçalves RP, da Silva FFF, Picciani PHS, Dias ML. Morphology and thermal properties of core-shell PVA/PLA ultrafine fibers produced by coaxial electrospinning. Mater Sci Appl. 2015;6(02):189–199. doi:10.4236/msa.2015.62022
  • Wang M, Zhou Y, Shi D, et al. Cold atmospheric plasma (CAP)-modified and bioactive protein-loaded core–shell nanofibers for bone tissue engineering applications. Biomater Sci. 2019;7(6):2430–2439. doi:10.1039/c8bm01284a30933194
  • Liu C, Wang C, Zhao Q, et al. Incorporation and release of dual growth factors for nerve tissue engineering using nanofibrous bicomponent scaffolds. Biomed Mater. 2018;13(4):044107. doi:10.1088/1748-605X/aab69329537390
  • Augustine R, Hasan A, Yadu Nath VK, et al. Electrospun polyvinyl alcohol membranes incorporated with green synthesized silver nanoparticles for wound dressing applications. J Mater Sci Mater Med. 2018;29(11):205–212. doi:10.1007/s10856-018-6169-7
  • Abdelwahab MA, Flynn A, Chiou BS, Imam S, Orts W, Chiellini E. Thermal, mechanical and morphological characterization of plasticized PLA-PHB blends. Polym Degrad Stab. 2012;97(9):1822–1828. doi:10.1016/j.polymdegradstab.2012.05.036
  • Chen B-K, Shih -C-C, Chen AF. Ductile PLA nanocomposites with improved thermal stability. Compos Part A Appl Sci Manuf. 2012;43(12):2289–2295. doi:10.1016/J.COMPOSITESA.2012.08.007
  • Hodge RM, Edward GH, Simon GP. Water absorption and states of water in semicrystalline poly(vinyl alcohol) films. Polymer (Guildf). 1996;37(8):1371–1376. doi:10.1016/0032-3861(96)81134-7
  • Firouzi A, Del Gaudio C, Lamastra FR, Montesperelli G, Bianco A. Electrospun polymeric coatings on aluminum alloy as a straightforward approach for corrosion protection. J Appl Polym Sci. 2015;132(2). doi:10.1002/app.41250
  • Golafshan N, Rezahasani R, Tarkesh Esfahani M, Kharaziha M, Khorasani SN. Nanohybrid hydrogels of laponite: PVA-alginate as a potential wound healing material. Carbohydr Polym. 2017;176:392–401. doi:10.1016/j.carbpol.2017.08.07028927623
  • Naseri N, Algan C, Jacobs V, John M, Oksman K, Mathew AP. Electrospun chitosan-based nanocomposite mats reinforced with chitin nanocrystals for wound dressing. Carbohydr Polym. 2014;109:7–15. doi:10.1016/j.carbpol.2014.03.03124815394
  • Augustine R, Kalarikkal N, Thomas S. An in vitro method for the determination of microbial barrier property (MBP) of porous polymeric membranes for skin substitute and wound dressing applications. Tissue Eng Regen Med. 2015;12(1):12–19. doi:10.1007/s13770-014-0032-9
  • Shalumon KT, Anjana J, Mony U, Jayakumar R, Chen J-P. Process study, development and degradation behavior of different size scale electrospun poly(caprolactone) and poly(lactic acid) fibers. J Polym Res. 2018;25(3):82. doi:10.1007/s10965-018-1475-9
  • Kim SJ, Tatman PD, Song DH, Gee AO, Kim DH, Kim SJ. Nanotopographic cues and stiffness control of tendon-derived stem cells from diverse conditions. Int J Nanomedicine. 2018;13:7217–7227. doi:10.2147/IJN.S18174330510414
  • Jiao A, Moerk CT, Penland N, et al. Regulation of skeletal myotube formation and alignment by nanotopographically controlled cell-secreted extracellular matrix. J Biomed Mater Res A. 2018;106(6):1543–1551. doi:10.1002/jbm.a.3635129368451
  • Zhao X, Luo J, Fang C, Xiong J. Investigation of polylactide/poly(ε-caprolactone)/multi-walled carbon nanotubes electrospun nanofibers with surface texture. RSC Adv. 2015;5(120):99179–99187. doi:10.1039/c5ra14301b
  • Sill TJ, von Recum HA. Electrospinning: applications in drug delivery and tissue engineering. Biomaterials. 2008;29(13):1989–2006. doi:10.1016/j.biomaterials.2008.01.01118281090
  • Liu Y, Geever LM, Kennedy JE, Higginbotham CL, Cahill PA, McGuinness GB. Thermal behavior and mechanical properties of physically crosslinked PVA/gelatin hydrogels. J Mech Behav Biomed Mater. 2010;3(2):203–209. doi:10.1016/J.JMBBM.2009.07.00120129419
  • White RJ, Cutting KF. Interventions to avoid maceration of the skin and wound bed. Br J Nurs. 2003;12(20):1186–1201. doi:10.12968/bjon.2003.12.20.1184114685125
  • Moura LIF, Dias AMA, Carvalho E, de Sousa HC. Recent advances on the development of wound dressings for diabetic foot ulcer treatment—A review. Acta Biomater. 2013;9(7):7093–7114. doi:10.1016/J.ACTBIO.2013.03.03323542233
  • Hassan A, Niazi MBK, Hussain A, Farrukh S, Ahmad T. Development of anti-bacterial PVA/starch based hydrogel membrane for wound dressing. J Polym Environ. 2018;26(1):235–243. doi:10.1007/s10924-017-0944-2
  • Porfyris A, Vasilakos S, Zotiadis C, et al. Accelerated ageing and hydrolytic stabilization of poly(lactic acid) (PLA) under humidity and temperature conditioning. Polym Test. 2018;68:315–332. doi:10.1016/J.POLYMERTESTING.2018.04.018
  • Kokabi M, Sirousazar M, Hassan ZM. PVA-clay nanocomposite hydrogels for wound dressing. Eur Polym J. 2007;43(3):773–781. doi:10.1016/j.eurpolymj.2006.11.030
  • Li H, Yang J, Hu X, Liang J, Fan Y, Zhang X. Superabsorbent polysaccharide hydrogels based on pullulan derivate as antibacterial release wound dressing. J Biomed Mater Res A. 2011;98 A(1):31–39. doi:10.1002/jbm.a.3304521523902
  • Dumville JC, Deshpande S, O’Meara S, Speak K. Hydrocolloid dressings for healing diabetic foot ulcers. Cochrane Database Syst Rev. 2013;2013(8):CD009099. doi:10.1002/14651858.CD009099.pub3
  • Wilson CJ, Clegg RE, Leavesley DI, Pearcy MJ. Mediation of biomaterial–cell interactions by adsorbed proteins: a review. Tissue Eng. 2005;11(1–2):1–18. doi:10.1089/ten.2005.11.115738657
  • Miller DR, Hilton JR, Harding KG, Beuker B, Williams DT. Wound dressings in diabetic foot disease. Clin Infect Dis. 2004;39(Supplement_2):S100–S103. doi:10.1086/38327015306987
  • Edwards J, Stapley S. Debridement of diabetic foot ulcers. Cochrane Database Syst Rev. 2010;CD003556. doi:10.1002/14651858.cd003556.pub220091547
  • Ruszczak Z. Effect of collagen matrices on dermal wound healing. Adv Drug Deliv Rev. 2003;55(12):1595–1611. doi:10.1016/J.ADDR.2003.08.00314623403
  • Sakai N, Nakamura M, Lipson KE, et al. Inhibition of CTGF ameliorates peritoneal fibrosis through suppression of fibroblast and myofibroblast accumulation and angiogenesis. Sci Rep. 2017;7(1):5392. doi:10.1038/s41598-017-05624-228710437
  • Wahab N, Cox D, Witherden A, Mason RM. Connective tissue growth factor (CTGF) promotes activated mesangial cell survival via up-regulation of mitogen-activated protein kinase phosphatase-1 (MKP-1). Biochem J. 2007;406(1):131–138. doi:10.1042/bj2006181717489738
  • Fan WH, Pech M, MJ K. Connective tissue growth factor (CTGF) stimulates vascular smooth muscle cell growth and migration in vitro. Eur J Cell Biol. 2000;79(12):915–923. doi:10.1078/0171-9335-0012211152282
  • Crean JKG, Finlay D, Murphy M, et al. The role of p42/44 MAPK and protein kinase B in connective tissue growth factor induced extracellular matrix protein production, cell migration, and actin cytoskeletal rearrangement in human mesangial cells. J Biol Chem. 2002;277(46):44187–44194. doi:10.1074/jbc.M20371520012218048
  • Livshits G, Kobielak A, Fuchs E. Governing epidermal homeostasis by coupling cell-cell adhesion to integrin and growth factor signaling, proliferation, and apoptosis. Proc Natl Acad Sci. 2012;109(13):4886–4891. doi:10.1073/pnas.120212010922411810
  • Nikitorowicz-Buniak J, Denton CP, Abraham D, Stratton R, Feghali-Bostwick C. Partially evoked epithelial-mesenchymal transition (EMT) is associated with increased TGFβ signaling within lesional scleroderma skin. PLoS One. 2015;10(7):e0134092. doi:10.1371/journal.pone.013409226217927
  • Guo S, Dipietro LA. Factors affecting wound healing. J Dent Res. 2010;89(3):219–229. doi:10.1177/002203450935912520139336
  • Shimo T, Nakanishi T, Kimura Y, et al. Inhibition of endogenous expression of connective tissue growth factor by its antisense oligonucleotide and antisense RNA suppresses proliferation and migration of vascular endothelial cells. J Biochem. 1998;124(1):130–140. doi:10.1093/oxfordjournals.jbchem.a0220719644255
  • Nakanishi T, Kuboki T, Tezuka K, et al. Connective tissue growth factor induces the proliferation, migration, and tube formation of vascular endothelial cells in vitro, and angiogenesis in vivo. J Biochem. 2012;126(1):137–145. doi:10.1093/oxfordjournals.jbchem.a022414