Skip to main content

Skin Wound Healing: Refractory Wounds and Novel Solutions

  • Protocol
  • First Online:
Skin Stem Cells

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1879))

Abstract

This overview of the current state of skin wound healing includes in vitro and in vivo approaches along with some recent clinical trials. From an introduction to wound healing, to tissue engineering as applied to the skin, we cover the basis for the current wound care techniques as well as novel and promising approaches. Special emphasis is given to refractory wounds which include wounds in diabetic patients. Natural compounds have been ever present in wound healing, and so we devote a section to highlighting current attempts to understand their mechanisms and to use them in novel ways.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Pastar I, Stojadinovic O, Yin NC et al (2014) Epithelialization in wound healing: a comprehensive review. Adv Wound Care 3:445–464

    Google Scholar 

  2. Thangapazham RL, Darling TN, Meyerle J (2014) Alteration of skin properties with autologous dermal fibroblasts. Int J Mol Sci 15:8407–8427

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Godwin JW, Rosenthal N (2014) Scar-free wound healing and regeneration in amphibians: immunological influences on regenerative success. Differentiation 87:66–75

    CAS  PubMed  Google Scholar 

  4. Greaves NS, Ashcroft KJ, Baguneid M et al (2013) Current understanding of molecular and cellular mechanisms in fibroplasia and angiogenesis during acute wound healing. J Dermatol Sci 72:206–217

    CAS  PubMed  Google Scholar 

  5. Chadwick SL, Yip C, Ferguson MWJ et al (2013) Repigmentation of cutaneous scars depends on original wound type. J Anat 223:74–82

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Coupland RM (1992) The red cross classification of war wounds: the E.X.C.F.V.M. scoring system. World J Surg 16:910–917

    CAS  PubMed  Google Scholar 

  7. Mangram AJ, Horan TC, Pearson ML et al (1999) Guideline for prevention of surgical site infection, 1999. Infect Control Hosp Epidemiol 20:247–280

    Google Scholar 

  8. Levy SM, Lally KP, Blakely ML et al (2015) Surgical wound misclassification: a multicenter evaluation. J Am Coll Surg 220:323–329

    PubMed  Google Scholar 

  9. Berríos-Torres SI, Umscheid CA, Bratzler DW et al (2017) Centers for disease control and prevention guideline for the prevention of surgical site infection, 2017. JAMA Surg 152:784–791

    PubMed  Google Scholar 

  10. Brem H, Tomic-Canic M (2007) Cellular and molecular basis of wound healing in diabetes. J Clin Invest 117:1219–1222

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Arno AI, Gauglitz GG, Barret JP et al (2014) Up-to-date approach to manage keloids and hypertrophic scars: a useful guide. Burns 40:1255–1266

    PubMed  PubMed Central  Google Scholar 

  12. Beitz JM (2014) Providing quality skin and wound care for the bariatric patient: an overview of clinical challenges. Ostomy Wound Manage 60:12–21

    PubMed  Google Scholar 

  13. Takeo M, Lee W, Ito M (2015) Wound healing and skin regeneration. Cold Spring Harb Perspect Med 5:a023267

    PubMed  PubMed Central  Google Scholar 

  14. Glim JE, van Egmond M, Niessen FB et al (2013) Detrimental dermal wound healing: what can we learn from the oral mucosa? Wound Repair Regen 21:648–660

    PubMed  Google Scholar 

  15. Akita S (2015) Lower extremity wounds in patients with idiopathic thrombocytopenic purpura and systemic lupus erythematosus. Int J Low Extrem Wounds 14:224–230

    CAS  PubMed  Google Scholar 

  16. Fine J-D (2010) Inherited epidermolysis bullosa. Orphanet J Rare Dis 5:12

    PubMed  PubMed Central  Google Scholar 

  17. van der Kooi-Pol MM, Sadaghian Sadabad M, Duipmans JC et al (2013) Topography of distinct Staphylococcus aureus types in chronic wounds of patients with epidermolysis bullosa. PLoS One 8:e67272

    CAS  PubMed  PubMed Central  Google Scholar 

  18. van Koppen CJ, Hartmann RW (2015) Advances in the treatment of chronic wounds: a patent review. Expert Opin Ther Pat 25:931–937

    PubMed  Google Scholar 

  19. Ang GC (2005) History of skin transplantation. Clin Dermatol 23:320–324

    PubMed  Google Scholar 

  20. Janeway CA Jr, Travers P, Walport M et al (2001) Responses to alloantigens and transplant rejection. Garland Science, New York

    Google Scholar 

  21. Ji S, Guvendiren M (2017) Recent advances in bioink design for 3D bioprinting of tissues and organs. Front Bioeng Biotechnol 5:23

    PubMed  PubMed Central  Google Scholar 

  22. Jakus AE, Laronda MM, Rashedi AS et al (2017) “Tissue Papers” from organ-specific decellularized extracellular matrices. Adv Funct Mater 27. https://doi.org/10.1002/adfm.201700992

    PubMed  PubMed Central  Google Scholar 

  23. Patel S, Dhillon MS, Aggarwal S et al (2013) Treatment with platelet-rich plasma is more effective than placebo for knee osteoarthritis: a prospective, double-blind, randomized trial. Am J Sports Med 41:356–364

    PubMed  Google Scholar 

  24. Sampson S, Reed M, Silvers H et al (2010) Injection of platelet-rich plasma in patients with primary and secondary knee osteoarthritis: a pilot study. Am J Phys Med Rehabil 89:961–969

    PubMed  Google Scholar 

  25. Eppley BL, Woodell JE, Higgins J (2004) Platelet quantification and growth factor analysis from platelet-rich plasma: implications for wound healing. Plast Reconstr Surg 114:1502–1508

    PubMed  Google Scholar 

  26. Werner S, Grose R (2003) Regulation of wound healing by growth factors and cytokines. Physiol Rev 83:835–870

    CAS  PubMed  Google Scholar 

  27. Yang HS, Shin J, Bhang SH et al (2011) Enhanced skin wound healing by a sustained release of growth factors contained in platelet-rich plasma. Exp Mol Med 43:622–629

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Law JX, Chowdhury SR, Saim AB et al (2017) Platelet-rich plasma with keratinocytes and fibroblasts enhance healing of full-thickness wounds. J Tissue Viability 26:208–215

    PubMed  Google Scholar 

  29. Suthar M, Gupta S, Bukhari S et al (2017) Treatment of chronic non-healing ulcers using autologous platelet rich plasma: a case series. J Biomed Sci 24:16

    PubMed  PubMed Central  Google Scholar 

  30. Martinez-Zapata MJ, Martí-Carvajal AJ, Solà I et al (2012) Autologous platelet-rich plasma for treating chronic wounds. Cochrane Database Syst Rev 10:CD006899

    PubMed  Google Scholar 

  31. Hu MS, Walmsley GG, Barnes LA et al (2017) Delivery of monocyte lineage cells in a biomimetic scaffold enhances tissue repair. JCI Insight 2. https://doi.org/10.1172/jci.insight.96260

  32. Sinno H, Malholtra M, Lutfy J et al (2013) Topical application of complement C3 in collagen formulation increases early wound healing. J Dermatol Treat 24:141–147

    CAS  Google Scholar 

  33. Tetteh PW, Farin HF, Clevers H (2015) Plasticity within stem cell hierarchies in mammalian epithelia. Trends Cell Biol 25:100–108

    CAS  PubMed  Google Scholar 

  34. Maxson S, Lopez EA, Yoo D et al (2012) Concise review: role of mesenchymal stem cells in wound repair. Stem Cells Transl Med 1:142–149

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Kwon DS, Gao X, Liu YB et al (2008) Treatment with bone marrow-derived stromal cells accelerates wound healing in diabetic rats. Int Wound J 5:453–463

    PubMed  Google Scholar 

  36. Lu D, Chen B, Liang Z et al (2011) Comparison of bone marrow mesenchymal stem cells with bone marrow-derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double-blind, randomized, controlled trial. Diabetes Res Clin Pract 92:26–36

    PubMed  Google Scholar 

  37. Crigler L, Kazhanie A, Yoon T-J et al (2007) Isolation of a mesenchymal cell population from murine dermis that contains progenitors of multiple cell lineages. FASEB J 21:2050–2063

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Kato Y, Iwata T, Washio K et al (2017) Creation and transplantation of an adipose-derived stem cell (ASC) sheet in a diabetic wound-healing model. J Vis Exp. https://doi.org/10.3791/54539

  39. Kim W-S, Park B-S, Sung J-H et al (2007) Wound healing effect of adipose-derived stem cells: a critical role of secretory factors on human dermal fibroblasts. J Dermatol Sci 48:15–24

    CAS  PubMed  Google Scholar 

  40. Hassan WU, Greiser U, Wang W (2014) Role of adipose-derived stem cells in wound healing. Wound Repair Regen 22:313–325

    PubMed  Google Scholar 

  41. Chopinaud M, Labbé D, Creveuil C et al (2017) Autologous adipose tissue graft to treat hypertensive leg ulcer: a pilot study. Dermatology 233:234–241

    CAS  PubMed  Google Scholar 

  42. Paolini G, Amoroso M, Longo B et al (2014) Simplified lipostructure: a technical note. Aesthet Plast Surg 38:78–82

    Google Scholar 

  43. Marks PW, Witten CM, Califf RM (2017) Clarifying stem-cell therapy’s benefits and risks. N Engl J Med 376:1007–1009

    PubMed  Google Scholar 

  44. Morris C, Emsley P, Marland E et al (2009) Use of wound dressings with soft silicone adhesive technology. Paediatr Nurs 21:38–43

    PubMed  Google Scholar 

  45. Kisch T, Weber C, Rapoport DH et al (2015) LPS-stimulated human skin-derived stem cells enhance neo-vascularization during dermal regeneration. PLoS One 10:e0142907

    PubMed  PubMed Central  Google Scholar 

  46. Rnjak J, Wise SG, Mithieux SM et al (2010) Severe burn injuries and the role of elastin in the design of dermal substitutes. Tissue Eng Part B Rev 17:81–91

    Google Scholar 

  47. Vig K, Chaudhari A, Tripathi S et al (2017) Advances in skin regeneration using tissue engineering. Int J Mol Sci 18. https://doi.org/10.3390/ijms18040789

    PubMed Central  Google Scholar 

  48. Laschke MW, Menger MD (2016) Prevascularization in tissue engineering: current concepts and future directions. Biotechnol Adv 34:112–121

    CAS  PubMed  Google Scholar 

  49. Lee J, Kim EH, Shin D et al (2017) Accelerated oral wound healing using a pre-vascularized mucosal cell sheet. Sci Rep 7:10667

    PubMed  PubMed Central  Google Scholar 

  50. Wilson WC, Boland T (2003) Cell and organ printing 1: protein and cell printers. Anat Rec A Discov Mol Cell Evol Biol 272:491–496

    PubMed  Google Scholar 

  51. Ventola CL (2014) Medical applications for 3D printing: current and projected uses. P T 39:704–711

    PubMed  PubMed Central  Google Scholar 

  52. Villar G, Graham AD, Bayley H (2013) A tissue-like printed material. Science 340:48–52

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Graham AD, Olof SN, Burke MJ et al (2017) High-resolution patterned cellular constructs by droplet-based 3D printing. Sci Rep 7:7004

    PubMed  PubMed Central  Google Scholar 

  54. Murphy SV, Atala A (2014) 3D bioprinting of tissues and organs. Nat Biotechnol 32:773–785

    CAS  PubMed  Google Scholar 

  55. Cubo N, Garcia M, Del Cañizo JF et al (2016) 3D bioprinting of functional human skin: production and in vivo analysis. Biofabrication 9:015006

    PubMed  Google Scholar 

  56. Topham J (2002) Why do some cavity wounds treated with honey or sugar paste heal without scarring? J Wound Care 11:53–55

    CAS  PubMed  Google Scholar 

  57. Rogalska T (2016) Healing the Bee’s knees—on honey and wound healing. JAMA Dermatol 152:275

    PubMed  Google Scholar 

  58. Simon A, Traynor K, Santos K et al (2009) Medical honey for wound care—still the ‘latest resort’? Evid Based Complement Alternat Med 6:165–173

    PubMed  Google Scholar 

  59. Can Z, Yildiz O, Sahin H et al (2015) An investigation of Turkish honeys: their physico-chemical properties, antioxidant capacities and phenolic profiles. Food Chem 180:133–141

    CAS  PubMed  Google Scholar 

  60. Wu Z, Chen L, Wu L et al (2015) Classification of Chinese honeys according to their floral origins using elemental and stable isotopic compositions. J Agric Food Chem 63:5388–5394

    CAS  PubMed  Google Scholar 

  61. McLoone P, Warnock M, Fyfe L (2016) Honey: a realistic antimicrobial for disorders of the skin. J Microbiol Immunol Infect 49:161–167

    CAS  PubMed  Google Scholar 

  62. Jenkins R, Cooper R (2012) Improving antibiotic activity against wound pathogens with manuka honey in vitro. PLoS One 7:e45600

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Al-Waili N, Salom K, Al-Ghamdi AA (2011) Honey for wound healing, ulcers, and burns; data supporting its use in clinical practice. ScientificWorldJournal 11:766–787

    PubMed  PubMed Central  Google Scholar 

  64. Cokcetin NN, Pappalardo M, Campbell LT et al (2016) The antibacterial activity of Australian leptospermum honey correlates with methylglyoxal levels. PLoS One 11:e0167780

    PubMed  PubMed Central  Google Scholar 

  65. Majtan J (2014) Honey: an immunomodulator in wound healing. Wound Repair Regen 22:187–192

    PubMed  Google Scholar 

  66. Ranzato E, Martinotti S, Burlando B (2012) Epithelial mesenchymal transition traits in honey-driven keratinocyte wound healing: comparison among different honeys. Wound Repair Regen 20:778–785

    PubMed  Google Scholar 

  67. Kamaratos AV, Tzirogiannis KN, Iraklianou SA et al (2014) Manuka honey-impregnated dressings in the treatment of neuropathic diabetic foot ulcers. Int Wound J 11:259–263

    PubMed  Google Scholar 

  68. Cao X-P, Chen Y-F, Zhang J-L et al (2017) Mechanisms underlying the wound healing potential of propolis based on its in vitro antioxidant activity. Phytomedicine 34:76–84

    CAS  PubMed  Google Scholar 

  69. Sehn E, Hernandes L, Franco SL et al (2009) Dynamics of reepithelialisation and penetration rate of a bee propolis formulation during cutaneous wounds healing. Anal Chim Acta 635:115–120

    CAS  PubMed  Google Scholar 

  70. Henshaw FR, Bolton T, Nube V et al (2014) Topical application of the bee hive protectant propolis is well tolerated and improves human diabetic foot ulcer healing in a prospective feasibility study. J Diabetes Complicat 28:850–857

    PubMed  Google Scholar 

  71. Léguillier T, Lecsö-Bornet M, Lémus C et al (2015) The wound healing and antibacterial activity of five ethnomedical Calophyllum inophyllum oils: an alternative therapeutic strategy to treat infected wounds. PLoS One 10:e0138602

    PubMed  PubMed Central  Google Scholar 

  72. Vergani L, Vecchione G, Baldini F et al (2017) Polyphenolic extract attenuates fatty acid-induced steatosis and oxidative stress in hepatic and endothelial cells. Eur J Nutr. https://doi.org/10.1007/s00394-017-1464-5

    PubMed  Google Scholar 

  73. Dunnill C, Patton T, Brennan J et al (2017) Reactive oxygen species (ROS) and wound healing: the functional role of ROS and emerging ROS-modulating technologies for augmentation of the healing process. Int Wound J 14:89–96

    PubMed  Google Scholar 

  74. Donato-Trancoso A, Monte-Alto-Costa A, Romana-Souza B (2016) Olive oil-induced reduction of oxidative damage and inflammation promotes wound healing of pressure ulcers in mice. J Dermatol Sci 83:60–69

    CAS  PubMed  Google Scholar 

  75. Rosa Ados S, Bandeira LG, Monte-Alto-Costa A et al (2014) Supplementation with olive oil, but not fish oil, improves cutaneous wound healing in stressed mice. Wound Repair Regen 22:537–547

    PubMed  Google Scholar 

  76. Sidgwick GP, McGeorge D, Bayat A (2015) A comprehensive evidence-based review on the role of topicals and dressings in the management of skin scarring. Arch Dermatol Res 307:461–477

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Peppone LJ, Hebl S, Purnell JQ et al (2010) The efficacy of calcitriol therapy in the management of bone loss and fractures: a qualitative review. Osteoporos Int 21:1133–1149

    CAS  PubMed  Google Scholar 

  78. Trujillo V, Marín-Luevano P, González-Curiel I et al (2017) Calcitriol promotes proangiogenic molecules in keratinocytes in a diabetic foot ulcer model. J Steroid Biochem Mol Biol 174:303–311

    CAS  PubMed  Google Scholar 

  79. Hinman CD, Maibach H (1963) Effect of air exposure and occlusion on experimental human skin wounds. Nature 200:377–378

    CAS  PubMed  Google Scholar 

  80. Steed DL (2004) Debridement. Am J Surg 187:71S–74S

    PubMed  Google Scholar 

  81. Fonder MA, Lazarus GS, Cowan DA et al (2008) Treating the chronic wound: a practical approach to the care of nonhealing wounds and wound care dressings. J Am Acad Dermatol 58:185–206

    PubMed  Google Scholar 

  82. Dabiri G, Damstetter E, Phillips T (2016) Choosing a wound dressing based on common wound characteristics. Adv Wound Care 5:32–41

    Google Scholar 

  83. Hussain Z, Thu HE, Shuid AN et al (2018) Recent advances in polymer-based wound dressings for the treatment of diabetic foot ulcer: an overview of state-of-the-art. Curr Drug Targets 19(5):527–550

    CAS  PubMed  Google Scholar 

  84. Kasiewicz LN, Whitehead KA (2017) Recent advances in biomaterials for the treatment of diabetic foot ulcers. Biomater Sci 5:1962–1975

    CAS  PubMed  Google Scholar 

  85. Ramundo J, Gray M (2008) Enzymatic wound debridement. J Wound Ostomy Continence Nurs 35:273–280

    PubMed  Google Scholar 

  86. Cullen B, Watt PW, Lundqvist C et al (2002) The role of oxidised regenerated cellulose/collagen in chronic wound repair and its potential mechanism of action. Int J Biochem Cell Biol 34:1544–1556

    CAS  PubMed  Google Scholar 

  87. Jeschke MG, Sandmann G, Schubert T et al (2005) Effect of oxidized regenerated cellulose/collagen matrix on dermal and epidermal healing and growth factors in an acute wound. Wound Repair Regen 13:324–331

    PubMed  Google Scholar 

  88. Wu S, Applewhite AJ, Niezgoda J et al (2017) Oxidized regenerated cellulose/collagen dressings: review of evidence and recommendations. Adv Skin Wound Care 30:S1–S18

    PubMed  PubMed Central  Google Scholar 

  89. Cuomo R, Grimaldi L, Brandi C et al (2017) Skin graft donor site: a procedure for a faster healing. Acta Biomed 88:310–314

    PubMed  Google Scholar 

  90. Sulaeva I, Henniges U, Rosenau T et al (2015) Bacterial cellulose as a material for wound treatment: properties and modifications. A review. Biotechnol Adv 33:1547–1571

    CAS  PubMed  Google Scholar 

  91. Shen G, Hu X, Guan G et al (2015) Surface modification and characterisation of silk fibroin fabric produced by the layer-by-layer self-assembly of multilayer alginate/regenerated silk fibroin. PLoS One 10:e0124811

    PubMed  PubMed Central  Google Scholar 

  92. Slotta UK, Rammensee S, Gorb S et al (2008) An engineered spider silk protein forms microspheres. Angew Chem Int Ed Engl 47:4592–4594

    CAS  PubMed  Google Scholar 

  93. Wang F, Zhang Y-Q (2015) Bioconjugation of silk fibroin nanoparticles with enzyme and peptide and their characterization. Adv Protein Chem Struct Biol 98:263–291

    CAS  PubMed  Google Scholar 

  94. Bhardwaj N, Rajkhowa R, Wang X et al (2015) Milled non-mulberry silk fibroin microparticles as biomaterial for biomedical applications. Int J Biol Macromol 81:31–40

    CAS  PubMed  Google Scholar 

  95. Spiess K, Lammel A, Scheibel T (2010) Recombinant spider silk proteins for applications in biomaterials. Macromol Biosci 10:998–1007

    CAS  PubMed  Google Scholar 

  96. Petzold J, Aigner TB, Touska F et al (2017) Surface features of recombinant spider silk protein eADF4(κ16)-made materials are well-suited for cardiac tissue engineering. Adv Funct Mater 27:1701427

    Google Scholar 

  97. Shi L, Wang F, Zhu W et al (2017) Self-healing silk fibroin-based hydrogel for bone regeneration: dynamic metal-ligand self-assembly approach. Adv Funct Mater 27:1700591

    Google Scholar 

  98. Karahaliloğlu Z, Ercan B, Denkbaş EB et al (2015) Nanofeatured silk fibroin membranes for dermal wound healing applications. J Biomed Mater Res A 103:135–144

    PubMed  Google Scholar 

  99. Chouhan D, Janani G, Chakraborty B et al (2018) Functionalized PVA-silk blended nanofibrous mats promote diabetic wound healing via regulation of extracellular matrix and tissue remodelling. J Tissue Eng Regen Med 12(3):e1559–e1570

    CAS  PubMed  Google Scholar 

  100. Wang Y, Mithieux SM, Kong Y et al (2015) Tropoelastin incorporation into a dermal regeneration template promotes wound angiogenesis. Adv Healthc Mater 4:577–584

    CAS  PubMed  Google Scholar 

  101. Mithieux SM, Weiss AS (2017) Design of an elastin-layered dermal regeneration template. Acta Biomater 52:33–40

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Rao KM, Rao KSVK, Ramanjaneyulu G et al (2014) Biodegradable sodium alginate-based semi-interpenetrating polymer network hydrogels for antibacterial application. J Biomed Mater Res A 102:3196–3206

    PubMed  Google Scholar 

  103. Kerschenmeyer A, Arlov Ø, Malheiro V et al (2017) Anti-oxidant and immune-modulatory properties of sulfated alginate derivatives on human chondrocytes and macrophages. Biomater Sci 5:1756–1765

    CAS  PubMed  Google Scholar 

  104. Moghadam MN, Pioletti DP (2016) Biodegradable HEMA-based hydrogels with enhanced mechanical properties. J Biomed Mater Res B Appl Biomater 104:1161–1169

    CAS  PubMed  Google Scholar 

  105. Elomaa L, Pan C-C, Shanjani Y et al (2015) Three-dimensional fabrication of cell-laden biodegradable poly(ethylene glycol-co-depsipeptide) hydrogels by visible light stereolithography. J Mater Chem B Mater Biol Med 3:8348–8358

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Gnavi S, Barwig C, Freier T et al (2013) The use of chitosan-based scaffolds to enhance regeneration in the nervous system. Int Rev Neurobiol 109:1–62

    PubMed  Google Scholar 

  107. Mekhail M, Tabrizian M (2014) Injectable chitosan-based scaffolds in regenerative medicine and their clinical translatability. Adv Healthc Mater 3:1529–1545

    CAS  PubMed  Google Scholar 

  108. Thangavel P, Ramachandran B, Chakraborty S et al (2017) Accelerated healing of diabetic wounds treated with L-glutamic acid loaded hydrogels through enhanced collagen deposition and angiogenesis: an in vivo study. Sci Rep 7:10701

    PubMed  PubMed Central  Google Scholar 

  109. Uskoković V, Ghosh S (2016) Carriers for the tunable release of therapeutics: etymological classification and examples. Expert Opin Drug Deliv 13:1729–1741

    PubMed  PubMed Central  Google Scholar 

  110. Zhang L, Ma Y, Pan X et al (2018) A composite hydrogel of chitosan/heparin/poly (γ-glutamic acid) loaded with superoxide dismutase for wound healing. Carbohydr Polym 180:168–174

    CAS  PubMed  Google Scholar 

  111. Abadir P, Hosseini S, Faghih M et al (2017) Topical reformulation of valsartan for treatment of chronic diabetic wounds. J Invest Dermatol 138(2):434–443

    PubMed  Google Scholar 

  112. Zhao L, Niu L, Liang H et al (2017) pH and glucose dual-responsive injectable hydrogels with insulin and fibroblasts as bioactive dressings for diabetic wound healing. ACS Appl Mater Interfaces 9:37563–37574

    CAS  PubMed  Google Scholar 

  113. Banerjee J, Das Ghatak P, Roy S et al (2015) Silver-zinc redox-coupled electroceutical wound dressing disrupts bacterial biofilm. PLoS One 10:e0119531

    PubMed  PubMed Central  Google Scholar 

  114. Singla R, Soni S, Patial V et al (2017) Cytocompatible anti-microbial dressings of Syzygium cumini cellulose nanocrystals decorated with silver nanoparticles accelerate acute and diabetic wound healing. Sci Rep 7:10457

    PubMed  PubMed Central  Google Scholar 

  115. Hwang S-W, Song J-K, Huang X et al (2014) High-performance biodegradable/transient electronics on biodegradable polymers. Adv Mater 26:3905–3911

    CAS  PubMed  Google Scholar 

  116. de Araújo DR, da Silva DC, Barbosa RM et al (2013) Strategies for delivering local anesthetics to the skin: focus on liposomes, solid lipid nanoparticles, hydrogels and patches. Expert Opin Drug Deliv 10:1551–1563

    PubMed  Google Scholar 

  117. Chen G, Li J, Song M et al (2017) A mixed component supramolecular hydrogel to improve mice cardiac function and alleviate ventricular remodeling after acute myocardial infarction. Adv Funct Mater 27:1701–1798

    Google Scholar 

Download references

Acknowledgments

Our undergraduate research (course number SCIR297) is conducted with the support of Montgomery College. We thank our students Nhi Luu and Rachel Tannenbaum for contributing ideas to the project. We also thank Jung Wha Lee and Lauren Kimlin (Virador and Associates) for their support. We appreciate our lively discussions with Jacqueline Muller and her critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Victoria M. Virador .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media New York

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Virador, G.M., de Marcos, L., Virador, V.M. (2018). Skin Wound Healing: Refractory Wounds and Novel Solutions. In: Turksen, K. (eds) Skin Stem Cells. Methods in Molecular Biology, vol 1879. Humana Press, New York, NY. https://doi.org/10.1007/7651_2018_161

Download citation

  • DOI: https://doi.org/10.1007/7651_2018_161

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-8869-3

  • Online ISBN: 978-1-4939-8870-9

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics