12,224
Views
150
CrossRef citations to date
0
Altmetric
Reviews

Recent advances on biomedical applications of scaffolds in wound healing and dermal tissue engineering

, , , , , , , & show all
Pages 691-705 | Received 15 May 2017, Accepted 28 Jun 2017, Published online: 12 Jul 2017

References

  • Jayabalan M. Studies on poly (propylene fumarate-co-caprolactone diol) thermoset composites towards the development of biodegradable bone fixation devices. Int J Biomater. 2009;2009:486710.
  • Do Kim H, Bae EH, Kwon IC, et al. Effect of PEG–PLLA diblock copolymer on macroporous PLLA scaffolds by thermally induced phase separation. Biomaterials. 2004;25:2319–2329.
  • Khan Y, Yaszemski MJ, Mikos AG, et al. Tissue engineering of bone: material and matrix considerations. J Bone Joint Surg. 2008;90:36–42.
  • Dhandayuthapani B, Yoshida Y, Maekawa T, et al. Polymeric scaffolds in tissue engineering application: a review. Int J Polymer Sci. 2011;2011:290602.
  • Baldwin SP, Saltzman WM. Materials for protein delivery in tissue engineering. Adv Drug Deliv Rev. 1998;33:71–86.
  • Nair LS, Laurencin CT. Biodegradable polymers as biomaterials. Progr Polymer Sci. 2007;32:762–798.
  • Sravanthi R. Preparation and characterization of poly (ɛ-caprolactone) PCL scaffolds for tissue engineering applications [Thesis]. Rourkela (India): National Institute of Technology; 2009.
  • Weigel T, Schinkel G, Lendlein A. Design and preparation of polymeric scaffolds for tissue engineering. Expert Rev Med Devices. 2006;3:835–851.
  • Pilehvar-Soltanahmadi Y, Akbarzadeh A, Moazzez-Lalaklo N, et al. An update on clinical applications of electrospun nanofibers for skin bioengineering. Artif Cells Nanomed Biotechnol. 2016;44:1350–1364.
  • Ahmadi-Aghkand F, Gholizadeh-Ghaleh Aziz S, Panahi Y, et al. Recent prospective of nanofiber scaffolds fabrication approaches for skin regeneration. Artif Cells Nanomed Biotechnol. 2016;44:1635–1641.
  • Khang G, Lee SJ, Kim MS, Lee HB. Biomaterials: tissue engineering and scaffolds. In: Webster JG, editor. Encyclopedia of Medical Devices and Instrumentation. New York: Wiley; 2006.
  • Chaignaud BE, Langer R, Vacanti JP. The history of tissue engineering using synthetic biodegradable polymer scaffolds and cells. In: Atala A, Mooney DJ, editors. Synthetic Biodegradable Polymer Scaffolds. Boston (MA): Springer; 1997. p. 1–14.
  • Boyce ST. Cultured skin substitutes: a review. Tissue Eng. 1996;2:255–266.
  • Kneser U, Kaufmann PM, Fiegel HC, et al. Long-term differentiated function of heterotopically transplanted hepatocytes on three-dimensional polymer matrices. J Biomed Mater Res. 1999;47:494–503.
  • Monsour M, Mohammed R, Gorham S, et al. An assessment of a collagen/vicryl composite membrane to repair defects of the urinary bladder in rabbits. Urol Res. 1987;15:235–238.
  • Hadlock T, Sundback C, Hunter D, et al. A polymer foam conduit seeded with Schwann cells promotes guided peripheral nerve regeneration. Tissue Eng. 2000;6:119–127.
  • Hansbrough JF, Morgan JL, Greenleaf GE, et al. Composite grafts of human keratinocytes grown on a polyglactin mesh-cultured fibroblast dermal substitute function as a bilayer skin replacement in full-thickness wounds on athymic mice. J Burn Care Res. 1993;14:485–494.
  • Ishaug-Riley SL, Crane-Kruger GM, Yaszemski MJ, et al. Three-dimensional culture of rat calvarial osteoblasts in porous biodegradable polymers. Biomaterials. 1998;19:1405–1412.
  • Cao Y, Vacanti JP, Paige KT, et al. Transplantation of chondrocytes utilizing a polymer-cell construct to produce tissue-engineered cartilage in the shape of a human ear. Plast Reconstr Surg. 1997;100:297–302.
  • Lin VS, Lee MC, O'Neal S, et al. Ligament tissue engineering using synthetic biodegradable fiber scaffolds. Tissue Eng. 1999;5:443–451.
  • Massia SP, Holecko MM, Ehteshami GR. In vitro assessment of bioactive coatings for neural implant applications. J Biomed Mater Res A. 2004;68:177–186.
  • Broderick E, Infanger S, Turner T, et al. Depressed bone mineralization following high dose TGF-β1 application in an orthopedic implant model. Calcif Tissue Int. 2005;76:379–384.
  • McKinney L, Hollinger JO. A bone regeneration study: transforming growth factor-[beta] 1 and its delivery. J Craniofacial Surg. 1996;7:36–45.
  • Critchlow M, Bland Y, Ashhurst D. The effect of exogenous transforming growth factor-β2 on healing fractures in the rabbit. Bone. 1995;16:521–527.
  • Schmidmaier G, Lucke M, Schwabe P, et al. Collective review: bioactive implants coated with poly (D,L-lactide) and growth factors IGF-I, TGF-β1, or BMP-2 for stimulation of fracture healing. J Long Term Eff Med Implants. 2006;16:61–69.
  • Street J, Bao M, Bunting S, et al. Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc Natl Acad Sci. 2002;99:9656–9661.
  • Becker C, Olde DL, Laeufer T, et al. 'UroMaix' scaffolds: novel collagen matrices for application in tissue engineering of the urinary tract. Int J Artif Organs. 2006;29:764–771.
  • Urist MR. Bone: formation by autoinduction. Science. 1965;150:893–899.
  • Wozney JM, Rosen V, Celeste AJ, et al. Novel regulators of bone formation: molecular clones and activities. Obstet Gynecol Survey. 1989;44:387.
  • Willerth SM, Sakiyama-Elbert SE. Approaches to neural tissue engineering using scaffolds for drug delivery. Adv Drug Deliv Rev. 2007;59:325–338.
  • Huang S, Deng T, Wang Y, et al. Multifunctional implantable particles for skin tissue regeneration: preparation, characterization, in vitro and in vivo studies. Acta Biomaterialia. 2008;4:1057–1066.
  • Danielsson C, Ruault S, Basset-Dardare A, et al. Modified collagen fleece, a scaffold for transplantation of human bladder smooth muscle cells. Biomaterials. 2006;27:1054–1060.
  • Chin CD, Khanna K, Sia SK. A microfabricated porous collagen-based scaffold as prototype for skin substitutes. Biomed Microdevices. 2008;10:459–467.
  • Shields KJ, Beckman MJ, Bowlin GL, et al. Mechanical properties and cellular proliferation of electrospun collagen type II. Tissue Eng. 2004;10:1510–1517.
  • Burke JF, Yannas IV, Quinby WC Jr, et al. Successful use of a physiologically acceptable artificial skin in the treatment of extensive burn injury. Ann Surg. 1981;194:413.
  • Fitton AR, Drew P, Dickson WA. The use of a bilaminate artificial skin substitute (IntegraTM) in acute resurfacing of burns: an early experience. Br J Plastic Surg. 2001;54:208–212.
  • Heimbach DM, Warden GD, Luterman A, et al. Multicenter postapproval clinical trial of Integra® dermal regeneration template for burn treatment. J Burn Care Res. 2003;24:42–48.
  • Aper T, Schmidt A, Duchrow M, et al. Autologous blood vessels engineered from peripheral blood sample. Eur J Vasc Endovasc Surg. 2007;33:33–39.
  • Tamada Y. New process to form a silk fibroin porous 3-D structure. Biomacromolecules. 2005;6:3100–3106.
  • Herbert CB, Bittner GD, Hubbell JA. Effects of fibrinolysis on neurite growth from dorsal root ganglia cultured in two- and three-dimensional fibrin gels. J Comp Neurol. 1996;365:380–391.
  • Sakiyama SE, Schense JC, Hubbell JA. Incorporation of heparin-binding peptides into fibrin gels enhances neurite extension: an example of designer matrices in tissue engineering. FASEB J. 1999;13:2214–2224.
  • Ahmed TA, Dare EV, Hincke M. Fibrin: a versatile scaffold for tissue engineering applications. Tissue Eng Part B Rev. 2008;14:199–215.
  • Boateng JS, Matthews KH, Stevens HN, et al. Wound healing dressings and drug delivery systems: a review. J Pharm Sci. 2008;97:2892–2923.
  • Sofia S, McCarthy MB, Gronowicz G, et al. Functionalized silk-based biomaterials for bone formation. J Biomed Mater Res. 2001;54:139–148.
  • Malafaya PB, Silva GA, Reis RL. Natural–origin polymers as carriers and scaffolds for biomolecules and cell delivery in tissue engineering applications. Adv Drug Deliv Rev. 2007;59:207–233.
  • Li M, Mondrinos MJ, Gandhi MR, et al. Electrospun protein fibers as matrices for tissue engineering. Biomaterials. 2005;26:5999–6008.
  • Daamen WF, Veerkamp J, Van Hest J, et al. Elastin as a biomaterial for tissue engineering. Biomaterials. 2007;28:4378–4398.
  • Sell S, McClure MJ, Barnes CP, et al. Electrospun polydioxanone–elastin blends: potential for bioresorbable vascular grafts disclosure. Several authors have United States and International patents pending concerning technology presented in this article, and this technology has been licensed to NanoMatrix, Inc., in which several authors have a financial interest. Biomed Mater. 2006;1:72.
  • Rnjak-Kovacina J, Wise SG, Li Z, et al. Electrospun synthetic human elastin: collagen composite scaffolds for dermal tissue engineering. Acta Biomaterialia. 2012;8:3714–3722.
  • Kumar MR, Muzzarelli RA, Muzzarelli C, et al. Chitosan chemistry and pharmaceutical perspectives. Chem Rev. 2004;104:6017–6084.
  • Muzzarelli R. Human enzymatic activities related to the therapeutic administration of chitin derivatives. Cell Mol Life Sci. 1997;53:131–140.
  • Okamoto Y, Yano R, Miyatake K, et al. Effects of chitin and chitosan on blood coagulation. Carbohyd Polymers. 2003;53:337–342.
  • Kweon D-K, Song S-B, Park Y-Y. Preparation of water-soluble chitosan/heparin complex and its application as wound healing accelerator. Biomaterials. 2003;24:1595–1601.
  • Ueno H, Mori T, Fujinaga T. Topical formulations and wound healing applications of chitosan. Adv Drug Deliv Rev. 2001;52:105–115.
  • Alemdaroğlu C, Değim Z, Çelebi N, et al. An investigation on burn wound healing in rats with chitosan gel formulation containing epidermal growth factor. Burns. 2006;32:319–327.
  • Mei L, Hu D, Ma J, et al. Preparation, characterization and evaluation of chitosan macroporous for potential application in skin tissue engineering. Int J Biol Macromol. 2012;51:992–997.
  • Choi JS, Yoo HS. Pluronic/chitosan hydrogels containing epidermal growth factor with wound-adhesive and photo-crosslinkable properties. J Biomed Mater Res A. 2010;95:564–573.
  • Chen Y-H, Wang I-J, Young T-H. Formation of keratocyte spheroids on chitosan-coated surface can maintain keratocyte phenotypes. Tissue Eng Part A. 2009;15:2001–2013.
  • Bragulla HH, Homberger DG. Structure and functions of keratin proteins in simple, stratified, keratinized and cornified epithelia. J Anat. 2009;214:516–559.
  • Tachibana A, Furuta Y, Takeshima H, et al. Fabrication of wool keratin sponge scaffolds for long-term cell cultivation. J Biotechnol. 2002;93:165–170.
  • Khalilov RI, Ahmadov IS, Kadirov SG. Two types of kinetics of membrane potential of water plant leaves illuminated by ultraviolet light. Bioelectrochemistry. 2002;58:189–191.
  • Bhardwaj N, Sow WT, Devi D, et al. Silk fibroin–keratin based 3D scaffolds as a dermal substitute for skin tissue engineering. Integr Biol. 2015;7:53–63.
  • Christensen B. Alginates as biomaterials in tissue engineering. In: Rauter AP, Lindhorst TK, editors. Carbohydrate chemistry: chemical and biological approaches. Vol. 37. Cambridge (UK): RSC Publishing; 2011. p. 227–258.
  • Xu H, Ma L, Shi H, et al. Chitosan–hyaluronic acid hybrid film as a novel wound dressing: in vitro and in vivo studies. Polym Adv Technol. 2007;18:869–875.
  • Wounds U. Best practice statement: the use of topical antiseptic/antimicrobial agents in wound management. London (UK): Wounds; 2010.
  • Li X, Chen S, Zhang B, et al. In situ injectable nano-composite hydrogel composed of curcumin, N, O-carboxymethyl chitosan and oxidized alginate for wound healing application. Int J Pharm. 2012;437:(1):110–119.
  • Hooper SJ, Percival SL, Hill KE, et al. The visualisation and speed of kill of wound isolates on a silver alginate dressing. Int Wound J. 2012;9:633–642.
  • Barnett S, Varley S. The effects of calcium alginate on wound healing. Ann R Coll Surg Engl. 1987;69:153.
  • Balakrishnan B, Mohanty M, Umashankar P, et al. Evaluation of an in situ forming hydrogel wound dressing based on oxidized alginate and gelatin. Biomaterials. 2005;26:6335–6342.
  • Sun J, Tan H. Alginate-based biomaterials for regenerative medicine applications. Materials. 2013;6:1285–1309.
  • Straccia MC, d'Ayala GG, Romano I, et al. Alginate hydrogels coated with chitosan for wound dressing. Mar Drugs. 2015;13:2890–2908.
  • Khan F, Ahmad SR. Polysaccharides and their derivatives for versatile tissue engineering application. Macromol Biosci. 2013;13:395–421.
  • Venkatesan J, Pallela R, Kim S-K. Dispersion of single walled carbon nanotubes in marine polysaccharides for bone tissue engineering. J Biomater Tissue Eng. 2014;4:501–505.
  • Yuvarani I, Kumar SS, Venkatesan J, et al. Preparation and characterization of curcumin coated chitosan-alginate blend for wound dressing application. J Biomat Tissue Eng. 2012;2:54–60.
  • Rath G, Johal E, Goyal AK. Development of serratiopeptidase and metronidazole based alginate microspheres for wound healing. Artif Cells Blood Substit Immobil Biotechnol. 2011;39:44–50.
  • Chandika P, Ko S-C, Oh G-W, et al. Fish collagen/alginate/chitooligosaccharides integrated scaffold for skin tissue regeneration application. Int J Biol Macromol. 2015;81:504–513.
  • Venkatesan J, Jayakumar R, Anil S, et al. Development of alginate-chitosan-collagen based hydrogels for tissue engineering. J Biomater Tissue Eng. 2015;5:458–464.
  • Arthanari S, Mani G, Jang JH, et al. Preparation and characterization of gatifloxacin-loaded alginate/poly (vinyl alcohol) electrospun nanofibers. Artif Cells Nanomed Biotechnol. 2016;44:847–852.
  • Chen WJ, Abatangelo G. Functions of hyaluronan in wound repair. Wound Repair Regen. 1999;7:79–89.
  • Gravante G, Delogu D, Giordan N, et al. The use of hyalomatrix PA in the treatment of deep partial-thickness burns. J Burn Care Res. 2007;28:269–274.
  • Madhyastha H, Madhyastha R, Nakajima Y, et al. Regulation of growth factors‐associated cell migration by C‐phycocyanin scaffold in dermal wound healing. Clin Exp Pharmacol Physiol. 2012;39:13–19.
  • Gunatillake P, Mayadunne R, Adhikari R. Recent developments in biodegradable synthetic polymers. Biotechnol Annu Rev. 2006;12:301–347.
  • Moran JM, Pazzano D, Bonassar LJ. Characterization of polylactic acid–polyglycolic acid composites for cartilage tissue engineering. Tissue Eng. 2003;9:63–70.
  • Singhal A, Agrawal C, Athanasiou KA. Salient degradation features of a 50:50 PLA/PGA scaffold for tissue engineering. Tissue Eng. 1996;2:197–207.
  • Khalilov RI, Khomutov GB, Tikhonov AN. Effect of ultraviolet radiation on structural-functional characteristics of the thylakoid membrane. Russ Plant Physiol. 1993;3:338–342.
  • Zhang J, Wu L, Jing D, et al. A comparative study of porous scaffolds with cubic and spherical macropores. Polymer. 2005;46:4979–4985.
  • Gupta V, Sengupta M, Prakash J, Tripathy BC. Tissue engineering and artificial organ. In: Basic and applied aspects of biotechnology. Singapore: Springer; 2017. p. 453–74.
  • Chen G, Ushida T, Tateishi T. Scaffold design for tissue engineering. Macromol Biosci. 2002;2:67–77.
  • Cheng Y-L, Lee M-L. Development of dynamic masking rapid prototyping system for application in tissue engineering. Rapid Prototyp J. 2009;15:29–41.
  • Williams D, Mort E. Enzyme-accelerated hydrolysis of polyglycolic acid. J Bioeng. 1977;1:231–238.
  • Li X, Yang Y, Fan Y, et al. Biocomposites reinforced by fibers or tubes as scaffolds for tissue engineering or regenerative medicine. J Biomed Mater Res A. 2014;102:1580–1594.
  • Slivka MA, Leatherbury NC, Kieswetter K, et al. Porous, resorbable, fiber-reinforced scaffolds tailored for articular cartilage repair. Tissue Eng. 2001;7:767–780.
  • Singh D, Singh MR. Development of antibiotic and debriding enzyme-loaded PLGA microspheres entrapped in PVA-gelatin hydrogel for complete wound management. Artif Cells Blood Subst Biotechnol. 2012;40:345–353.
  • Amirthalingam M, Kasinathan N, Amuthan A, et al. Bioactive PLGA–curcumin microparticle-embedded chitosan scaffold: in vitro and in vivo evaluation. Artif Cells Blood Subst Biotechnol. 2017;45:233–241.
  • Bryant SJ, Anseth KS. Hydrogel properties influence ECM production by chondrocytes photoencapsulated in poly (ethylene glycol) hydrogels. J Biomed Mater Res. 2002;59:63–72.
  • Lee S-H, Moon JJ, Miller JS, et al. Poly (ethylene glycol) hydrogels conjugated with a collagenase-sensitive fluorogenic substrate to visualize collagenase activity during three-dimensional cell migration. Biomaterials. 2007;28:3163–3170.
  • Mann BK, Gobin AS, Tsai AT, et al. Smooth muscle cell growth in photopolymerized hydrogels with cell adhesive and proteolytically degradable domains: synthetic ECM analogs for tissue engineering. Biomaterials. 2001;22:3045–3051.
  • Veronese FM, Pasut G. PEGylation, successful approach to drug delivery. Drug Discov Today. 2005;10:1451–1458.
  • Vahidi M, Frounchi M, Dadbin S. Porous gelatin/poly (ethylene glycol) scaffolds for skin cells. Soft Mater. 2016;15:95–102.
  • Müller HM, Seebach D. Poly (hydroxyalkanoates): a fifth class of physiologically important organic biopolymers? Angew Chem Int Ed Engl. 1993;32:477–502.
  • Philip S, Keshavarz T, Roy I. Polyhydroxyalkanoates: biodegradable polymers with a range of applications. J Chem Technol Biotechnol. 2007;82:233–247.
  • Chen G-Q, Wu Q. The application of polyhydroxyalkanoates as tissue engineering materials. Biomaterials. 2005;26:6565–6578.
  • Ljungberg C, Johansson‐Ruden G, Junemo Boström K, et al. Neuronal survival using a resorbable synthetic conduit as an alternative to primary nerve repair. Microsurgery. 1999;19:259–264.
  • Asran AS, Razghandi K, Aggarwal N, et al. Nanofibers from blends of polyvinyl alcohol and polyhydroxy butyrate as potential scaffold material for tissue engineering of skin. Biomacromolecules. 2010;11:3413–3421.
  • Keshel SH, Biazar E, Rezaei Tavirani M, et al. The healing effect of unrestricted somatic stem cells loaded in collagen-modified nanofibrous PHBV scaffold on full-thickness skin defects. Artif Cells Nanomed Biotechnol. 2014;42:210–216.
  • Allen MJ, Schoonmaker JE, Bauer TW, et al. Preclinical evaluation of a poly (vinyl alcohol) hydrogel implant as a replacement for the nucleus pulposus. Spine. 2004;29:515–523.
  • Chen DH, Leu JC, Huang TC. Transport and hydrolysis of urea in a reactor–separator combining an anion‐exchange membrane and immobilized urease. J Chem Technol Biotechnol. 1994;61:351–357.
  • Zhou Y, Yang D, Nie J. Electrospinning of chitosan/poly (vinyl alcohol)/acrylic acid aqueous solutions. J Appl Polym Sci. 2006;102:5692–5697.
  • Zheng H, Du Y, Yu J, et al. Preparation and characterization of chitosan/poly (vinyl alcohol) blend fibers. J Appl Polym Sci. 2001;80:2558–2565.
  • Zhang Y, Huang X, Duan B, et al. Preparation of electrospun chitosan/poly (vinyl alcohol) membranes. Colloid Polym Sci. 2007;285:855–863.
  • Youssef NA, Gurbanov EM, Haciyeva SR, et al. Antioxidant enzymes, fluctuating asymmetry and morphological changes of urban trees as an ecological indicator of heavy metal stress. Int J Pharm Sci Health Care. 2013;1:1–18.
  • Zeng J, Chen X, Liang Q, et al. Enzymatic degradation of poly (L‐lactide) and poly (ɛ‐caprolactone) electrospun fibers. Macromol Biosci. 2004;4:1118–1125.
  • Valizadeh A, Bakhtiary M, Akbarzadeh A, et al. Preparation and characterization of novel electrospun poly (ε-caprolactone)-based nanofibrous scaffolds. Artif Cells Nanomed Biotechnol. 2016;44:504–509.
  • Yoshimoto H, Shin Y, Terai H, et al. A biodegradable nanofiber scaffold by electrospinning and its potential for bone tissue engineering. Biomaterials. 2003;24:2077–2082.
  • Li W-J, Tuli R, Huang X, et al. Multilineage differentiation of human mesenchymal stem cells in a three-dimensional nanofibrous scaffold. Biomaterials. 2005;26:5158–5166.
  • Bahrami H, Keshel SH, Chari AJ, et al. Human unrestricted somatic stem cells loaded in nanofibrous PCL scaffold and their healing effect on skin defects. Artif Cells Nanomed Biotechnol. 2016;44:1556–1560.
  • Bonvallet PP, Culpepper BK, Bain JL, et al. Microporous dermal-like electrospun scaffolds promote accelerated skin regeneration. Tissue Eng Part A. 2014;20:2434–2445.
  • Bonvallet PP, Schultz MJ, Mitchell EH, et al. Microporous dermal-mimetic electrospun scaffolds pre-seeded with fibroblasts promote tissue regeneration in full-thickness skin wounds. PLoS One. 2015;10:e0122359.
  • Mikos AG, Lyman MD, Freed LE, et al. Wetting of poly(L-lactic acid) and poly(DL-lactic-co-glycolic acid) foams for tissue culture. Biomaterials. 1994;15:55–58.
  • Ebrahimi E, Akbarzadeh A, Abbasi E, et al. Novel drug delivery system based on doxorubicin-encapsulated magnetic nanoparticles modified with PLGA-PEG1000 copolymer. Artif Cells Nanomed Biotechnol. 2016;44:290–297.
  • Yamamoto M, Kato K, Ikada Y. Ultrastructure of the interface between cultured osteoblasts and surface-modified polymer substrates. J Biomed Mater Res. 1997;37:29–36.
  • Zhang R, Ma PX. Porous poly (L-lactic acid)/apatite composites created by biomimetic process. J Biomed Mater Res. 1999;45:285–293.
  • Chen H, Peng Y, Wu S, et al. Electrospun 3D fibrous scaffolds for chronic wound repair. Materials. 2016;9:272.
  • Place ES, George JH, Williams CK, et al. Synthetic polymer scaffolds for tissue engineering. Chem Soc Rev. 2009;38:1139–1151.
  • Kiyotani T, Teramachi M, Takimoto Y, et al. Nerve regeneration across a 25-mm gap bridged by a polyglycolic acid-collagen tube: a histological and electrophysiological evaluation of regenerated nerves. Brain Res. 1996;740:66–74.
  • Miki H, Ando N, Ozawa S, et al. An artificial esophagus constructed of cultured human esophageal epithelial cells, fibroblasts, polyglycolic acid mesh, and collagen. ASAIO J. 1999;45:502–508.
  • Saxena AK, Marler J, Benvenuto M, et al. Skeletal muscle tissue engineering using isolated myoblasts on synthetic biodegradable polymers: preliminary studies. Tissue Eng. 1999;5:525–531.
  • Chen G, Ushida T, Tateishi T. Poly(DL-lactic-co-glycolic acid) sponge hybridized with collagen microsponges and deposited apatite particulates. J Biomed Mater Res. 2001;57:8–14.
  • Khalilov RI, Nasibova AN, Serezhenkov VA, et al. Accumulation of magnetic nanoparticles in plants grown on soils of Apsheron peninsula. Biophysics. 2011;56:316–322.
  • Chen G, Ushida T, Tateishi T. Hybrid biomaterials for tissue engineering: a preparative method for PLA or PLGA–collagen hybrid sponges. Adv Mater. 2000;12:455–457.
  • Chen G, Sato T, Ushida T, et al. The use of a novel PLGA fiber/collagen composite web as a scaffold for engineering of articular cartilage tissue with adjustable thickness. J Biomed Mater Res. 2003;67:1170–1180.
  • Colter K, Shen M, Bell A. Reduction of progesterone release rate through silicone membranes by plasma polymerization. Biomater Med Devices Artif Organs. 1977;5:13–24.
  • Sodhi RN. Application of surface analytical and modification techniques to biomaterial research. J Electron Spectrosc Relat Phenomena. 1996;81:269–284.
  • Brewis D, Briggs D. Adhesion to polyethylene and polypropylene. Polymer. 1981;22:7–16.
  • Reis RL, Neves NM, Mano JF, Gomes ME, Marques AP, Azevedo HS. Natural-based polymers for biomedical applications. Amsterdam (The Netherlands): Elsevier; 2008.
  • Chen G, Ushida T, Tateishi T. A hybrid network of synthetic polymer mesh and collagen sponge. Chem Commun. 2000;1505–1506.
  • Bellincampi LD, Dunn MG. Effect of crosslinking method on collagen fiber-fibroblast interactions. J Appl Polym Sci. 1997;63:1493–1498.
  • Yannas I, Burke JF. Design of an artificial skin. I. Basic design principles. J Biomed Mater Res. 1980;14:65–81.
  • Trasciatti S, Podestà A, Bonaretti S, et al. In vitro effects of different formulations of bovine collagen on cultured human skin. Biomaterials. 1998;19:897–903.
  • Cooper ML, Hansbrough JF, Spielvogel RL, et al. In vivo optimization of a living dermal substitute employing cultured human fibroblasts on a biodegradable polyglycolic acid or polyglactin mesh. Biomaterials. 1991;12:243–248.
  • Yang W-S, Roh H-W, Lee WK, et al. Evaluation of functions and tissue compatibility of poly (D, L-lactic-co-glycolic acid) seeded with human dermal fibroblasts. J Biomater Sci Polymer Ed. 2006;17:151–162.
  • Lu H, Oh HH, Kawazoe N, et al. PLLA–collagen and PLLA–gelatin hybrid scaffolds with funnel-like porous structure for skin tissue engineering. Sci Technol Adv Mater. 2012;13:064210.
  • Sims DC, Butler PE, Casanova R, et al. Injectable cartilage using polyethylene oxide polymer substrates. Plastic Reconstruct Surg. 1996;98:843–850.
  • Gorjikhah F, Azizi Jalalian F, Salehi R, et al. Preparation and characterization of PLGA-β-CD polymeric nanoparticles containing methotrexate and evaluation of their effects on T47D cell line. Artif Cells Nanomed Biotechnol. 2017;45:432–440.
  • Klossner RR, Queen HA, Coughlin AJ, et al. Correlation of chitosan’s rheological properties and its ability to electrospin. Biomacromolecules. 2008;9:2947–2953.
  • Bhattarai N, Edmondson D, Veiseh O, et al. Electrospun chitosan-based nanofibers and their cellular compatibility. Biomaterials. 2005;26:6176–6184.
  • Kriegel C, Kit K, McClements DJ, et al. Electrospinning of chitosan–poly (ethylene oxide) blend nanofibers in the presence of micellar surfactant solutions. Polymer. 2009;50:189–200.
  • Kriegel C, Kit K, McClements DJ, et al. Influence of surfactant type and concentration on electrospinning of chitosan–poly (ethylene oxide) blend nanofibers. Food Biophys. 2009;4:213–228.
  • Zhang J-F, Yang D-Z, Xu F, et al. Electrospun core: shell structure nanofibers from homogeneous solution of poly (ethylene oxide)/chitosan. Macromolecules. 2009;42:5278–5284.
  • Duan B, Dong C, Yuan X, et al. Electrospinning of chitosan solutions in acetic acid with poly(ethylene oxide). J Biomater Sci Polym Ed. 2004;15:797–811.
  • Sun K, Li Z. Preparations, properties and applications of chitosan based nanofibers fabricated by electrospinning. Express Polym Lett. 2011;5:342–361.
  • Zhou Y, Yang D, Chen X, et al. Electrospun water-soluble carboxyethyl chitosan/poly (vinyl alcohol) nanofibrous membrane as potential wound dressing for skin regeneration. Biomacromolecules. 2007;9:349–354.
  • Chen J-P, Chang G-Y, Chen J-K. Electrospun collagen/chitosan nanofibrous membrane as wound dressing. Colloids Surfaces A. 2008;313:183–188.
  • Lu S, Gao W, Gu HY. Construction, application and biosafety of silver nanocrystalline chitosan wound dressing. Burns. 2008;34:623–628.
  • An J, Zhang H, Zhang J, et al. Preparation and antibacterial activity of electrospun chitosan/poly (ethylene oxide) membranes containing silver nanoparticles. Colloid Polym Sci. 2009;287:1425–1434.
  • Hang AT, Tae B, Park JS. Non-woven mats of poly (vinyl alcohol)/chitosan blends containing silver nanoparticles: fabrication and characterization. Carbohyd Polym. 2010;82:472–479.
  • Yoon CS, Ji DS. Effects of In Vitro degradation on the weight loss and tensile properties of PLA/LPCL/HPCL blend fibers. Fibers Polym. 2005;6:13–18.
  • Dai N-T, Williamson M, Khammo N, et al. Composite cell support membranes based on collagen and polycaprolactone for tissue engineering of skin. Biomaterials. 2004;25:4263–4271.
  • Wong VW, Rustad KC, Galvez MG, et al. Engineered pullulan–collagen composite dermal hydrogels improve early cutaneous wound healing. Tissue Eng Part A. 2010;17:631–644.
  • Rnjak J, Wise SG, Mithieux SM, et al. Severe burn injuries and the role of elastin in the design of dermal substitutes. Tissue Eng Part B Rev. 2011;17:81–91.
  • Aberoumandi SM, Mohammadhosseini M, Abasi E, et al. An update on applications of nanostructured drug delivery systems in cancer therapy. Artif Cells Nanomed Biotechnol. 2017;45:1058–1068.
  • Gupta KC, Haider A, Choi Y-r, et al. Nanofibrous scaffolds in biomedical applications. Biomater Res. 2014;18:5.
  • Xie Z, Paras CB, Weng H, et al. Dual growth factor releasing multi-functional nanofibers for wound healing. Acta Biomaterialia. 2013;9:9351–9359.
  • Beachley V, Wen X. Polymer nanofibrous structures: Fabrication, biofunctionalization, and cell interactions. Progr Polym Sci. 2010;35:868–892.
  • Loo Y, Wong Y-C, Cai EZ, et al. Ultrashort peptide nanofibrous hydrogels for the acceleration of healing of burn wounds. Biomaterials. 2014;35:4805–4814.
  • Ninan N, Muthiah M, Yahaya NAB, et al. Antibacterial and wound healing analysis of gelatin/zeolite scaffolds. Colloids Surf B Biointerfaces. 2014;115:244–252.
  • Dreesmann L, Ahlers M, Schlosshauer B. The pro-angiogenic characteristics of a cross-linked gelatin matrix. Biomaterials. 2007;28:5536–5543.
  • Huang W-Y, Yeh C-L, Lin J-H, et al. Development of fibroblast culture in three-dimensional activated carbon fiber-based scaffold for wound healing. J Mater Sci. 2012;23:1465–1478.
  • Matoušková E, Mestak O. The effect of different biologic and biosynthetic wound covers on keratinocyte growth, stratification and differentiation in vitro. J Tissue Eng. 2014;5:2041731414554966.
  • Zajicek R, Mandys V, Mestak O, et al. Human keratinocyte growth and differentiation on acellular porcine dermal matrix in relation to wound healing potential. Sci World J. 2012;2012:727352.
  • Shichu X, Shihui Z, Hengyu L, et al. Feasibility study of composite skin reconstructed by mixing keratinocytes and acellular dermal matrix for wound repair. Swiss Med Week. 2009;139:16.
  • Auger FA, Valle CAL, Guignard R, et al. Skin equivalent produced with human collagen. In Vitro Cell Dev Biol Anim. 1995;31:432–439.
  • El Ghalbzouri A, Hensbergen P, Gibbs S, et al. Fibroblasts facilitate re-epithelialization in wounded human skin equivalents. Lab Invest. 2004;84:102–112.
  • Singh MR, Saraf S, Vyas A, et al. Innovative approaches in wound healing: trajectory and advances. Artif Cells Nanomed Biotechnol. 2013;41:202–212.
  • Falanga V, Margolis D, Alvarez O, et al. Rapid healing of venous ulcers and lack of clinical rejection with an allogeneic cultured human skin equivalent. Human Skin Equivalent Investigators Group. Arch Dermatol. 1998;134:293–300.
  • Brem H, Young J, Tomic-Canic M, et al. Clinical efficacy and mechanism of bilayered living human skin equivalent (HSE) in treatment of diabetic foot ulcers. Surg Technol Int. 2001;11:23–31.
  • Kremer M, Lang E, Berger A. Evaluation of dermal—epidermal skin equivalents (‘composite-skin’) of human keratinocytes in a collagen-glycosaminoglycan matrix (integra™ artificial skin). Br J Plastic Surg. 2000;53:459–465.
  • Supp DM, Boyce ST. Engineered skin substitutes: practices and potentials. Clin Dermatol. 2005;23:403–412.
  • Lam PK, Chan ES, To EW, et al. Development and evaluation of a new composite Laserskin graft. J Trauma. 1999;47:918.
  • Vanscheidt W, Ukat A, Horak V, et al. Treatment of recalcitrant venous leg ulcers with autologous keratinocytes in fibrin sealant: a multinational randomized controlled clinical trial. Wound Repair Regen. 2007;15:308–315.
  • Hansbrough JF, Morgan J, Greenleaf G, et al. Development of a temporary living skin replacement composed of human neonatal fibroblasts cultured in Biobrane, a synthetic dressing material. Surgery. 1994;115:633–644.
  • Dhivya S, Padma VV, Santhini E. Wound dressings: a review. BioMedicine (Taipei). 2015;5:22.
  • Strecker-McGraw MK, Jones TR, Baer DG. Soft tissue wounds and principles of healing. Emergency Med Clin N Am. 2007;25:1–22.
  • Rivera AE, Spencer JM. Clinical aspects of full-thickness wound healing. Clin Dermatol. 2007;25:39–48.
  • Hunt TK, Hopf H, Hussain Z. Physiology of wound healing. Adv Skin Wound Care. 2000;13:6.
  • Stashak TS, Farstvedt E, Othic A. Update on wound dressings: indications and best use. Clin Techn Equine Prac. 2004;3:148–163.
  • Morin RJ, Tomaselli NL. Interactive dressings and topical agents. Clin Plast Surg. 2007;34:643–658.
  • Moshakis V, Fordyce M, Griffiths J, et al. Tegadern versus gauze dressing in breast surgery. Br J Clin Pract. 1984;38:149
  • Buchan I, Andrews J, Lang S, et al. Clinical and laboratory investigation of the composition and properties of human skin wound exudate under semi-permeable dressings. Burns. 1981;7:326–334.
  • Rogozinski WJ. Modifiable, semi-permeable, wound dressing. Google Patents; 1993.
  • Morgan D. Wounds-what should a dressings formulary include? Pharm J. 2009;9:261.
  • Jones V, Grey JE, Harding KG. Wound dressings. BMJ. 2006;332:777.
  • Ashford R, Freear N, Shippen J. An in-vitro study of the pressure-relieving properties of four wound dressings for foot ulcers. J Wound Care. 2001;10:34–38.
  • Razzak MT, Darwis D. Irradiation of polyvinyl alcohol and polyvinyl pyrrolidone blended hydrogel for wound dressing. Radiat Phys Chem. 2001;62:107–113.
  • Thomas DR, Goode PS, LaMaster K, et al. Acemannan hydrogel dressing versus saline dressing for pressure ulcers: a randomized, controlled trial. Adv Skin Wound Care. 1998;11:273–276.
  • Boateng J, Catanzano O. Advanced therapeutic dressings for effective wound healing—a review. J Pharm Sci. 2015;104:3653–3680.
  • Advanced wound dressing. (Srinagarind Med J). 2013;28:18–23.
  • Pudner R. Hydrocolloid dressings. Pract Nurs. 1998;9:17–19.
  • Cullum N. Leg ulcer treatments: a critical review (part 2). Nurs Std. 1994;9:32–36.
  • Skórkowska-Telichowska K, Czemplik M, Kulma A, et al. The local treatment and available dressings designed for chronic wounds. J Am Acad Dermatol. 2013;68:e117–e126.
  • Suzuki Y, Tanihara M, Nishimura Y, et al. In vivo evaluation of a novel alginate dressing. J Biomed Mater Res. 1999;48:522–527.
  • Suzuki Y, Nishimura Y, Tanihara M, et al. Evaluation of a novel alginate gel dressing: Cytotoxicity to fibroblasts in vitro and foreign‐body reaction in pig skin in vivo. J Biomed Mater Res. 1998;39:317–322.
  • Pendry E. The use of alginate dressings in the treatment of diabetic foot ulcers. Diabetic Foot. 2006;9:76–83.
  • Dias A, Braga M, Seabra I, et al. Development of natural-based wound dressings impregnated with bioactive compounds and using supercritical carbon dioxide. Int J Pharm. 2011;408:9–19.
  • Ramshaw JA, Werkmeister JA, Glattauer V. Collagen-based biomaterials. Biotechnol Genet Eng Rev. 1996;13:335–382.
  • Doillon CJ, Silver FH. Collagen-based wound dressing: Effects of hyaluronic acid and firponectin on wound healing. Biomaterials. 1986;7:3–8.
  • Ishihara M, Nakanishi K, Ono K, et al. Photocrosslinkable chitosan as a dressing for wound occlusion and accelerator in healing process. Biomaterials. 2002;23:833–840.
  • Karahaliloglu Z, Kilicay E, Denkbas EB. Antibacterial chitosan/silk sericin 3D porous scaffolds as a wound dressing material. Artif Cells Nanomed Biotechnol. 2016;45:1–14.
  • Wiegand C, Heinze T, Hipler UC. Comparative in vitro study on cytotoxicity, antimicrobial activity, and binding capacity for pathophysiological factors in chronic wounds of alginate and silver‐containing alginate. Wound Repair Regener. 2009;17:511–521.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.