Current wound healing procedures and potential care (2024)

1. Epstein FH, Singer AJ, Clark RA. Cutaneous wound healing. The New England Journal of Medicine. 1999;341:738–746. [PubMed] [Google Scholar]

2. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453:314–321. [PubMed] [Google Scholar]

3. Korting HC, Schöllmann C, White RJ. Management of minor acute cutaneous wounds: importance of wound healing in a moist environment. J Eur Acad Dermatol Venereol. 2011;25:130–137. [PubMed] [Google Scholar]

4. Sen CK, Gordillo GM, Roy S, Kirsner R, Lambert L, Hunt TK, Gottrup F, Gurtner GC, Longaker MT. Human skin wounds: a major and snowballing threat to public health and the economy. Wound repair and regeneration: official publication of the Wound Healing Society and the European Tissue Repair Society. 2009;17:763–771. [PMC free article] [PubMed] [Google Scholar]

5. Margolis DJ, Hoffstad O, Nafash J, Leonard CE, Freeman CP, Hennessy S, Wiebe DJ. Location, location, location: geographic clustering of lower-extremity amputation among Medicare beneficiaries with diabetes. Diabetes care. 2011;34:2363–2367. [PMC free article] [PubMed] [Google Scholar]

6. Dargaville TR, Farrugia BL, Broadbent JA, Pace S, Upton Z, Voelcker NH. Sensors and imaging for wound healing: a review. Biosensors & bioelectronics. 2013;41:30–42. [PubMed] [Google Scholar]

7. Shahin ES, Dassen T, Halfens RJ. Pressure ulcer prevalence and incidence in intensive care patients: a literature review. Nursing in critical care. 2008;13:71–79. [PubMed] [Google Scholar]

8. Monaco JL, Lawrence WT. Acute wound healing an overview. Clin Plast Surg. 2003;30:1–12. [PubMed] [Google Scholar]

9. Gosain A, DiPietro LA. Aging and wound healing. World J Surg. 2004;28:321–326. [PubMed] [Google Scholar]

10. Greaves NS, Ashcroft KJ, Baguneid M, Bayat A. Current understanding of molecular and cellular mechanisms in fibroplasia and angiogenesis during acute wound healing. J Dermatol Sci. 2013;72:206–217. [PubMed] [Google Scholar]

11. Li J, Chen J, Kirsner R. Pathophysiology of acute wound healing. Clin Dermatol. 2007;25:9–18. [PubMed] [Google Scholar]

12. Enoch S, Price P. Cellular, molecular and biochemical differences in the pathophysiology of healing between acute wounds, chronic wounds and wounds in the aged. World Wide Wounds. 2004 http://www.worldwidewounds.com/2004/august/Enoch/Pathophysiology-Of-Healing.html. [Google Scholar]

13. Menke NB, Ward KR, Witten TM, Bonchev DG, Diegelmann RF. Impaired wound healing. Clin Dermatol. 2007;25:19–25. [PubMed] [Google Scholar]

14. Wilgus TA. Growth Factor-Extracellular Matrix Interactions Regulate Wound Repair. Adv Wound Care (New Rochelle) 2012;1:249–254. [PMC free article] [PubMed] [Google Scholar]

15. Lev-Tov H, Li CS, Dahle S, Isseroff RR. Cellular versus acellular matrix devices in treatment of diabetic foot ulcers: study protocol for a comparative efficacy randomized controlled trial. Trials. 2013;14:8. [PMC free article] [PubMed] [Google Scholar]

16. Stanulis-Praeger BM, Gilchrest BA. Growth factor responsiveness declines during adulthood for human skin-derived cells. Mech Ageing Dev. 1986;34:185–198. [PubMed] [Google Scholar]

17. Ferguson MW, O’Kane S. Scar-free healing: from embryonic mechanisms to adult therapeutic intervention. Philos Trans R Soc B Biol Sci. 2004;359:839–850. [PMC free article] [PubMed] [Google Scholar]

18. Phillips TJ, Kehinde O, Green H, Gilchrest BA. Treatment of skin ulcers with cultured epidermal allografts. Journal of the American Academy of Dermatology. 1989;21:191–199. [PubMed] [Google Scholar]

19. Wainwright DJ. Use of an acellular allograft dermal matrix (AlloDerm) in the management of full-thickness burns. Burns. 1995;21:243–248. [PubMed] [Google Scholar]

20. Leigh IM, Purkis PE. Culture grafted leg ulcers. Clinical and experimental dermatology. 1986;11:650–652. [PubMed] [Google Scholar]

21. Sheridan RL, Tompkins RG. Skin substitutes in burns. Burns: journal of the International Society for Burn Injuries. 1999;25:97–103. [PubMed] [Google Scholar]

22. Harris PA, Leigh IM, Navsaria HA. Pre-confluent keratinocyte grafting: the future for cultured skin replacements? Burns: journal of the International Society for Burn Injuries. 1998;24:591–593. [PubMed] [Google Scholar]

23. Rinnerthaler M, Duschl J, Steinbacher P, Salzmann M, Bischof J, Schuller M, Wimmer H, Peer T, Bauer JW, Richter K. Age-related changes in the composition of the cornified envelope in human skin. Exp Dermatol. 2013;22:329–335. [PubMed] [Google Scholar]

24. Gilchrest BA, Karassik RL, Wilkins LM, Vrabel MA, Maciag T. Autocrine and paracrine growth stimulation of cells derived from human skin. Journal of cellular physiology. 1983;117:235–240. [PubMed] [Google Scholar]

25. Sauder DN, Stanulis-Praeger BM, Gilchrest BA. Autocrine growth stimulation of human keratinocytes by epidermal cell-derived thymocyte-activating factor: implications for skin aging. Arch Dermatol Res. 1988;280:71–76. [PubMed] [Google Scholar]

26. Colwell AS, Faudoa R, Krummel TM, Longaker MT, Lorenz HP. Transforming growth factor-beta, Smad, and collagen expression patterns in fetal and adult keratinocytes. Plast Reconstr Surg. 2007;119:852–857. [PubMed] [Google Scholar]

27. O'Connor NE. Grafting of burns with cultured epithelium prepared from autologous epidermal cells. Lancet. 1981;1:75–78. [PubMed] [Google Scholar]

28. Hefton JM, Caldwell D, Biozes DG, Balin AK, Carter DM. Grafting of skin ulcers with cultured autologous epidermal cells. Journal of the American Academy of Dermatology. 1986;14:399–405. [PubMed] [Google Scholar]

29. Green H, Kehinde O, Thomas J. Growth of cultured human epidermal cells into multiple epithelia suitable for grafting. Proc Natl Acad Sci U S A. 1979;76:5665–5668. [PMC free article] [PubMed] [Google Scholar]

30. Barrandon Y, Green H. Cell migration is essential for sustained growth of keratinocyte colonies: the roles of transforming growth factor-alpha and epidermal growth factor. Cell. 1987;50:1131–1137. [PubMed] [Google Scholar]

31. Lam PK, Chan ES, To EW, Lau CH, Yen SC, King WW. Development and evaluation of a new composite Laserskin graft. J Trauma. 1999;47:918–922. [PubMed] [Google Scholar]

32. Wood F, Martin L, Lewis D, Rawlins J, McWilliams T, Burrows S, Rea S. A prospective randomised clinical pilot study to compare the effectiveness of Biobrane(R) synthetic wound dressing, with or without autologous cell suspension, to the local standard treatment regimen in paediatric scald injuries. Burn: journal of the International Society for Burn Injuries. 2012;38:830–839. [PubMed] [Google Scholar]

33. Balasubramani M, Kumar TR, Babu M. Skin substitutes: a review. Burns. 2001;27:534–544. [PubMed] [Google Scholar]

34. Guerid S, Darwiche SE, Berger MM, Applegate LA, Benathan M, Raffoul W. Autologous keratinocyte suspension in platelet concentrate accelerates and enhances wound healing - a prospective randomized clinical trial on skin graft donor sites: platelet concentrate and keratinocytes on donor sites. Fibrogenesis Tissue Repair. 2013;6:8. [PMC free article] [PubMed] [Google Scholar]

35. Auxenfans C, Menet V, Catherine Z, Shipkov H, Lacroix P, Bertin-Maghit M, Damour O, Braye F. Cultured autologous keratinocytes in the treatment of large and deep burns: A retrospective study over 15 years. Burns. pii: S0305-4179(2014)00200-9. [PubMed] [Google Scholar]

36. Odessey R. Addendum: multicenter experience with cultured epidermal autograft for treatment of burns. The Journal of burn care & rehabilitation. 1992;13:174–180. [PubMed] [Google Scholar]

37. Gallico GG, O'Connor NE, 3rd, Compton CC, Kehinde O, Green H. Permanent coverage of large burn wounds with autologous cultured human epithelium. The New England journal of medicine. 1984;311:448–451. [PubMed] [Google Scholar]

38. Leigh IM, Purkis PE, Navsaria HA, Phillips TJ. Treatment of chronic venous ulcers with sheets of cultured allogenic keratinocytes. The British journal of dermatology. 1987;117:591–597. [PubMed] [Google Scholar]

39. Nanchahal J, Ward CM. New grafts for old? A review of alternatives to autologous skin. Br J Plast Surg. 1992;45:354–363. [PubMed] [Google Scholar]

40. Briggaman RA, Dalldorf FG, Wheeler CE., Jr Formation and origin of basal lamina and anchoring fibrils in adult human skin. The Journal of cell biology. 1971;51:384–395. [PMC free article] [PubMed] [Google Scholar]

41. Madden MR, Finkelstein JL, Staiano-Coico L, Goodwin CW, Shires GT, Nolan EE, Hefton JM. Grafting of cultured allogeneic epidermis on second- and third-degree burn wounds on 26 patients. The Journal of trauma. 1986;26:955–962. [PubMed] [Google Scholar]

42. Woodley DT, Peterson HD, Herzog SR, Stricklin GP, Burgeson RE, Briggaman RA, Cronce DJ, O'Keefe EJ. Burn wounds resurfaced by cultured epidermal autografts show abnormal reconstitution of anchoring fibrils. JAMA: the journal of the American Medical Association. 1988;259:2566–2571. [PubMed] [Google Scholar]

43. Kumagai N, Nishina H, Tanabe H, Hosaka T, Ishida H, Ogino Y. Clinical application of autologous cultured epithelia for the treatment of burn wounds and burn scars. Plastic and reconstructive surgery. 1988;82:99–110. [PubMed] [Google Scholar]

44. Clugston PA, Snelling CF, Macdonald IB, Maledy HL, Boyle JC, Germann E, Courtemanche AD, Wirtz P, Fitzpatrick DJ, Kester DA. Cultured epithelial autografts: three years of clinical experience with eighteen patients. The Journal of burn care & rehabilitation. 1991;12:533–539. [PubMed] [Google Scholar]

45. De Luca M, Albanese E, Bondanza S, Megna M, Ugozzoli L, Molina F, Cancedda R, Santi PL, Bormioli M, Stella M. Multicentre experience in the treatment of burns with autologous and allogenic cultured epithelium, fresh or preserved in a frozen state. Burns: Journal of the International Society for Burn Injuries. 1989;15:303–309. [PubMed] [Google Scholar]

46. Worst PK, Valentine EA, Fusenig NE. Formation of epidermis after reimplantation of pure primary epidermal cell cultures from perinatal mouse skin. Journal of the National Cancer Institute. 1974;53:1061–1064. [PubMed] [Google Scholar]

47. Turley EA, Torrance J. Localization of hyaluronate and hyaluronate-binding protein on motile and non-motile fibroblasts. Experimental cell research. 1985;161:17–28. [PubMed] [Google Scholar]

48. Moczar M, Robert L. Stimulation of cell proliferation by hyaluronidase during in vitro aging of human skin fibroblasts. Experimental gerontology. 1993;28:59–68. [PubMed] [Google Scholar]

49. Kirwan L. Management of difficult wounds with Biobrane. Connecticut medicine. 1995;59:523–529. [PubMed] [Google Scholar]

50. Barret JP, Dziewulski P, Ramzy PI, Wolf SE, Desai MH, Herndon DN. Biobrane versus 1% silver sulfadiazine in second-degree pediatric burns. Plastic and reconstructive surgery. 2000;105:62–65. [PubMed] [Google Scholar]

51. Malmquist JP, Clemens SC, Oien HJ, Wilson SL. Hemostasis of oral surgery wounds with the HemCon Dental Dressing. J Oral Maxillofac Surg. 2008;66:1177–1183. [PubMed] [Google Scholar]

52. Kirichenko AK, Bolshakov IN, Ali-Riza AE, Vlasov AA. Morphological study of burn wound healing with the use of collagen-chitosan wound dressing. Bull Exp Biol Med. 2013;154:692–696. [PubMed] [Google Scholar]

53. Liakos I, Rizzello L, Scurr DJ, Pompa PP, Bayer IS, Athanassiou A. All-natural composite wound dressing films of essential oils encapsulated in sodium alginate with antimicrobial properties. Int J Pharm. 2014;463:137–145. [PubMed] [Google Scholar]

54. Mogoşanu GD, Grumezescu AM. Natural and synthetic polymers for wounds and burns dressing. Int J Pharm. 2013 pii: S0378-5173(13)01083-1. [PubMed] [Google Scholar]

55. Powell HM, Supp DM, Boyce ST. Influence of electrospun collagen on wound contraction of engineered skin substitutes. Biomaterials. 2008;29:834–843. [PubMed] [Google Scholar]

56. Ong SY, Wu J, Moochhala SM, Tan MH, Lu J. Development of a chitosan-based wound dressing with improved hemostatic and antimicrobial properties. Biomaterials. 2008;29:4323. [PubMed] [Google Scholar]

57. Uppal R, Ramaswamy GN, Arnold C, Goodband R, Wang Y. Hyaluronic acid nanofiber wound dressing--production, characterization, and in vivo behavior. J Biomed Mater Res B. 2011;97B:20. [PubMed] [Google Scholar]

58. Dreifke M, Ebraheim NA, Jayasuriya AC. Investigation of Potential Injectable Polymeric Biomaterials for Bone Regeneration. J Biomed Mater Res A. 2013;101:2436–2447. [PMC free article] [PubMed] [Google Scholar]

59. Chen WY, Abatangelo G. Functions of hyaluronan in wound repair. Wound repair and regeneration: official publication of the Wound Healing Society [and] the European Tissue Repair Society. 1999;7:79–89. [PubMed] [Google Scholar]

60. Docherty R, Forrester JV, Lackie JM, Gregory DW. Glycosaminoglycans facilitate the movement of fibroblasts through three-dimensional collagen matrices. Journal of cell science. 1989;92:263–270. [PubMed] [Google Scholar]

61. Weigel PH, Fuller GM, LeBoeuf RD. A model for the role of hyaluronic acid and fibrin in the early events during the inflammatory response and wound healing. Journal of theoretical biology. 1986;11:219–234. [PubMed] [Google Scholar]

62. Feinberg RN, Beebe DC. Hyaluronate in vasculogenesis. Science (New York, N.Y.) 1983;220:1177–1179. [PubMed] [Google Scholar]

63. Xie Y, Upton Z, Richards S, Rizzi SC, Leavesley DI. Hyaluronic acid: evaluation as a potential delivery vehicle for vitronectin: growth factor complexes in wound healing applications. J Control Release. 2011;153:225–232. [PubMed] [Google Scholar]

64. Campo GM, Avenoso A, Campo S, D'Ascola A, Nastasi G, Calatroni A. Molecular size hyaluronan differently modulates toll-like receptor-4 in LPS-induced inflammation in mouse chondrocytes. Biochimie. 2010;92:204–215. [PubMed] [Google Scholar]

65. Ghazi K, Deng-Pichon U, Warnet JM, Rat P. Hyaluronan fragments improve wound healing on in vitro cutaneous model through P2X7 purinoreceptor basal activation: role of molecular weight. PLoS One. 2012;7:48351. [PMC free article] [PubMed] [Google Scholar]

66. Gao F, Liu Y, He Y, Yang C, Wang Y, Shi X, Wei G. Hyaluronan oligosaccharides promote excisional wound healing through enhanced angiogenesis. Matrix Biology. 2010;29:107–116. [PubMed] [Google Scholar]

67. Tolg C, Telmer p, Turley E. Specific sizes of hyaluronan oligosaccharides stimulate fibroblast migration and excisional wound repair. PLoS One. 2014;9:e88479. [PMC free article] [PubMed] [Google Scholar]

68. Lobmann R, Pittasch D, Mühlen I, Lehnert H. J Diabetes Complications. 2003;17:199–204. [PubMed] [Google Scholar]

69. Myers SR, Partha VN, Soranzo C, Price RD, Navsaria HA. Autologous human keratinocytes cultured on membranes composed of benzyl ester of hyaluronic acid for grafting in nonhealing diabetic foot lesions: a pilot study, Hyalomatrix: A Temporary Epidermal Barrier, Hyaluronan Delivery, and Neodermis Induction System for Keratinocyte Stem Cell Therapy. Tissue Engineering. 2007;13:2733–2741. [PubMed] [Google Scholar]

70. Ehrenreich M, Ruszczak Z. Tissue-engineered temporary wound coverings, Important options for the clinician. Acta Dermatovenerol Alp Panonica Adriat. 2006;15:5–13. [PubMed] [Google Scholar]

71. Bradley T, Brown RE, Kucan JO, Smoot EC, Hussmann J., 3rd Toxic epidermal necrolysis: a review and report of the successful use of Biobrane for early wound coverage. Annals of plastic surgery. 1995;35:124–132. [PubMed] [Google Scholar]

72. Egan WC, Clark WR. The toxic shock syndrome in a burn victim. Burns, including thermal injury. 1988;14:135–138. [PubMed] [Google Scholar]

73. Gerding RL, Emerman CL, Effron D, Lukens T, Imbembo AL, Fratianne RB. Outpatient management of partial-thickness burns: Biobrane versus 1% silver sulfadiazine. Annals of emergency medicine. 1990;19:121–124. [PubMed] [Google Scholar]

74. Demling RH. Use of Biobrane in management of scalds. The Journal of burn care & rehabilitation. 1995;16:329–330. [PubMed] [Google Scholar]

75. Phillips LG, Robson MC, Smith DJ, Phillips WA, Gracia WD, McHugh TP, Sullivan WG, Mathoney K, Swartz K, Meltzer T. Uses and abuses of a biosynthetic dressing for partial skin thickness burns. Burns: journal of the International Society for Burn Injuries. 1989;15:254–256. [PubMed] [Google Scholar]

76. Hartford CE, Wang XW, Peterson VM, Rodgers CM, Ketch LL. Healing characteristics of expanded autografts on wounds covered with hom*ograft and Biobrane temporary wound dressing. The Journal of burn care & rehabilitation. 1989;10:476–480. [PubMed] [Google Scholar]

77. Gerding RL, Imbembo AL, Fratianne RB. Biosynthetic skin substitute vs. 1% silver sulfadiazine for treatment of inpatient partial-thickness thermal burns. The Journal of trauma. 1988;28:1265–1269. [PubMed] [Google Scholar]

78. Lal S, Barrow RE, Wolf SE, Chinkes DL, Hart DW, Heggers JP, Herndon DN. Biobrane improves wound healing in burned children without increased risk of infection. Shock (Augusta, Ga.) 2000;14:314–318. discussion 318–319. [PubMed] [Google Scholar]

79. Frew Q, Philp B, Shelley O, Myers S, Navsaria H, Dziewulski P. The use of Biobrane(®) as a delivery method for cultured epithelial autograft in burn patients. Burns. 2013;39:876–880. [PubMed] [Google Scholar]

80. Woodroof EA, Phipps RP, Greenwood JE, Hickerson W, Herndon D. The Search for an Ideal Temporary Skin Substitute: AWBAT Plus, a Combination Product Wound Dressing Medical Device. Eplasty. 2010;10 pii: e60. [PMC free article] [PubMed] [Google Scholar]

81. Wood F, Martin L, Lewis D, Rawlins J, McWilliams T, Burrows S, Rea S. A prospective randomised clinical pilot study to compare the effectiveness of Biobrane® synthetic wound dressing, with or without autologous cell suspension, to the local standard treatment regimen in paediatric scald injuries. Burns. 2012;38:830–839. [PubMed] [Google Scholar]

82. Wessels Q. Engineered alternative skin for partial and full-thickness burns. Bioengineered. 2014;5:161–164. [PMC free article] [PubMed] [Google Scholar]

83. Di Martino A, Sittinger M, Risbud MV. Chitosan: A versatile biopolymer for orthopaedic tissue-engineering. Biomaterials. 2005;26:5983–5990. [PubMed] [Google Scholar]

84. Freier T, Koh HS, Kazazian K, Shoichet MS. Controlling cell adhesion and degradation of CS films by N-acetylation. Biomaterials. 2005;56:5872–5878. [PubMed] [Google Scholar]

85. Jayasuriya AC, Bhat A. Optimization of scaled-up Chitosan Microparticles for Bone Regeneration. Biomedical Materials. 2009;4:55006. [PubMed] [Google Scholar]

86. Jayasuriya AC, Bhat A. Fabrication and characterization of novel hybrid organic/inorganic microparticles to apply in bone regeneration. Journal of Biomedical Materials Research A. 2010;93A:1280–1288. [PubMed] [Google Scholar]

87. Jayasuriya AC, Mauch KJ. In vitro degradation behavior of chitosan based hybrid microparticles. J Biomed Sci Eng. 2011;4:383–390. [PMC free article] [PubMed] [Google Scholar]

88. Moura LI, Dias AM, Leal EC, Carvalho L, de Sousa HC, Carvalho E. Chitosan-based dressings loaded with neurotensin--an efficient strategy to improve early diabetic wound healing. Acta Biomater. 2014;10:843–857. [PubMed] [Google Scholar]

89. Santos TC, Höring B, Reise K, Marques AP, Silva SS, Oliveira JM, Mano JF, Castro AG, Reis RL, van Griensven M. In vivo performance of chitosan/soy-based membranes as wound-dressing devices for acute skin wounds. Tissue Eng Part A. 2013;19:860–869. [PMC free article] [PubMed] [Google Scholar]

90. Akncbay H, Senel S, Ay ZY. Application of chitosan gel in the treatment of chronic periodontitis. J Biomed Mater Res B Appl Biomater. 2007;80:290–296. [PubMed] [Google Scholar]

91. Ong SY, Wu J, Moochhala SM, Tan MH, Lu J. Development of a chitosan-based wound dressing with improved hemostatic and antimicrobial properties. Biomaterials. 2008;29:4323. [PubMed] [Google Scholar]

92. Mi FL, Shyu SS, Wu YB, Lee ST, Shyong JY, Huang RN. Fabrication and characterization of a sponge-like asymmetric chitosan membrane as a wound dressing. Biomaterials. 2001;22:165–173. [PubMed] [Google Scholar]

93. Ribeiro MP, Morgado PI, Miguel SP, Coutinho P, Correia IJ. Dextran-based hydrogel containing chitosan microparticles loaded with growth factors to be used in wound healing. Mater Sci Eng C Mater Biol Appl. 2013;33:2958–2966. [PubMed] [Google Scholar]

94. Park CJ, Clark SG, Lichtensteiger CA, Jamison RD, Johnson AJ. Accelerated wound closure of pressure ulcers in aged mice by chitosan scaffolds with and without bFGF. Acta Biomater. 2009;5:1926–1936. [PubMed] [Google Scholar]

95. Yang HS, Shin J, Bhang SH, Shin JY, Park J, Im GI, Kim CS, Kim BS. Enhanced skin wound healing by a sustained release of growth factors contained in platelet-rich plasma. Exp Mol Med. 2011;43:622–629. [PMC free article] [PubMed] [Google Scholar]

96. Yao C, Yao P, Wu H, Zha Z. Acceleration of wound healing in traumatic ulcers by absorbable collagen sponge containing recombinant basic fibroblast growth factor. Biomed Mater. 2006;1:33–37. [PubMed] [Google Scholar]

97. Yu A, Niiyama H, Kondo S, Yamamoto A, Suzuki R, Kuroyanagi Y. Wound dressing composed of hyaluronic acid and collagen containing EGF or bFGF: comparative culture study. J Biomater Sci Polym Ed. 2013;24:1015–1026. [PubMed] [Google Scholar]

98. Smiell JM. Clinical safety of becaplermin (rhPDGF-BB) gel. Becaplermin Studies Group. Am. J. Surg. 1998;176:68S–73S. [PubMed] [Google Scholar]

99. Steed DL. Clinical evaluation of recombinant human platelet-derived growth factor for the treatment of lower extremity diabetic ulcers, Diabetic Ulcer Study Group. J Vasc Surg. 1995;21 discussion 9–81. [PubMed] [Google Scholar]

100. Singla S, Garg R, Kumar A, Gill C. Efficacy of topical application of beta urogastrone (recombinant human epidermal growth factor) in Wagner's Grade 1 and 2 diabetic foot ulcers: Comparative analysis of 50 patients. J Nat Sci Biol Med. 2014;5:273–277. [PMC free article] [PubMed] [Google Scholar]

101. Mantripragada VP, Jayasuriya AC. IGF-1 release kinetics from chitosan microparticles fabricated using environmentally benign conditions. Mater Sci Eng C Mater Biol Appl. 2014;42:506–516. [PMC free article] [PubMed] [Google Scholar]

102. Mantripragada VP, Jayasuriya AC. Injectable chitosan microparticles incorporating bone morphogenetic protein-7 for bone tissue regeneration. J Biomed Mater Res A. 2014;102:4276–4289. [PMC free article] [PubMed] [Google Scholar]

103. Layliev J, Wilson S, Warren SM, Saadeh PB. Improving Wound Healing with Topical Gene Therapy. Adv Wound Care (New Rochelle) 2012;1:218–223. [PMC free article] [PubMed] [Google Scholar]

104. Nelson CE, Gupta MK, Adolph EJ, Guelcher SA, Duvall CL. siRNA Delivery from an Injectable Scaffold for Wound Therapy. Adv Wound Care (New Rochelle) 2013;2:93–99. [PMC free article] [PubMed] [Google Scholar]

105. Krebs MD, Jeon O, Alsberg E. Localized and sustained delivery of silencing RNA from macroscopic biopolymer hydrogels. J Am Chem Soc. 2009;131:9204. [PubMed] [Google Scholar]

106. Vinas-Castells R, Holladay C, di Luca A, Diaz VM, Pandit A. Snail1 down-regulation using small interfering RNA complexes delivered through collagen scaffolds. Bioconjug Chem. 2009;20:2262. [PubMed] [PubMed] [Google Scholar]

107. Nguyen PD, Tutela JP, Thanik VD, Knobel D, Allen RJ, Chang CC, Levine JP, Warren SM, Saadeh PB. Improved diabetic wound healing through topical silencing of p53 is associated with augmented vasculogenic mediators. Wound Repair Regen. 2010;18:553. [PMC free article] [PubMed] [Google Scholar]

108. Lee JW, Tutela JP, Zoumalan RA, Thanik VD, Nguyen PD, Varjabedian L, Warren SM, Saadeh PB. Inhibition of Smad3 expression in radiation-induced fibrosis using a novel method for topical transcutaneous gene therapy. Arch Otolaryngol. 2010;136:714. [PubMed] [Google Scholar]

109. Madhyastha R, Madhyastha H, Nakajima Y, Omura S, Maruyama M. MicroRNA signature in diabetic wound healing: Promotive role of MIR-21 in fibroblast migration. Int. Wound J. 2012;9:355–361. [PMC free article] [PubMed] [Google Scholar]

110. Van Solingen C, Seghers L, Bijkerk R, Duijs JM, Roeten MK, van Oeveren-Rietdijk AM, Baelde HJ, Monge M, Vos JB, de Boer HC, et al. Antagomir-mediated silencing of endothelial cell specific microRNA-126 impairs ischemia-induced angiogenesis. J. Cell Mol. Med. 2009;13:1577–1585. [PMC free article] [PubMed] [Google Scholar]

111. Pastar I, Khan AA, Stojadinovic O, Lebrun EA, Medina MC, Brem H, Kirsner RS, Jimenez JJ, Leslie C, Tomic-Canic M. Induction of specific microRNAs inhibits cutaneous wound healing. J. Biol. Chem. 2012;287:29324–29335. [PMC free article] [PubMed] [Google Scholar]

112. Wang T, Feng Y, Sun H, Zhang L, Hao L, Shi C, Wang J, Li R, Ran X, Su Y, et al. Mir-21 regulates skin wound healing by targeting multiple aspects of the healing process. Am. J. Pathol. 2012;181:1911–1920. [PubMed] [Google Scholar]

113. Moura J, Børsheim E, Carvalho E. The Role of MicroRNAs in Diabetic Complications-Special Emphasis on Wound Healing. Genes (Basel) 2014;5:926–956. [PMC free article] [PubMed] [Google Scholar]

114. Valls MD, Cronstein BN, Montesinos MC. Adenosine receptor agonists for promotion of dermal wound healing. Biochem Pharmacol. 2009;77:1117–1124. [PMC free article] [PubMed] [Google Scholar]

115. Gu BJ, Wiley JS. Rapid ATP-induced release of matrix metalloproteinase 9 is mediated by the P2X7 receptor. Blood. 2006;107:4946–4953. [PubMed] [Google Scholar]

116. Gendaszewska-Darmach E, Kucharska M. Nucleotide receptors as targets in the pharmacological enhancement of dermal wound healing. Purinergic Signalling. 2011;7:193–206. [PMC free article] [PubMed] [Google Scholar]

117. Jin H, Seo J, Eun SY, Joo YN, Park SW, Lee JH, Chang KC, Kim HJ. P2Y2 R activation by nucleotides promotes skin wound-healing process. Exp Dermatol. 2014;23:480–485. [PubMed] [Google Scholar]

118. Wu Y, Huang S, Enhe J, Ma K, Yang S, Sun T, Fu X. Bone marrow-derived mesenchymal stem cell attenuates skin fibrosis development in mice. Int Wound J. 2013 [PMC free article] [PubMed] [Google Scholar]

119. Carmeliet P, Luttun A. The emerging role of the bone marrow-derived stem cells in [therapeutic] angiogenesis. Haemost. Thromb. 2001;86:289–297. [PubMed] [Google Scholar]

120. Kwon DS, Gao X, Liu YB, Dulchavsky DS, Danyluk AL, Bansal M, Chopp M, McIntosh K, Arbab AS, Dulchavsky SA, Gautam SC. Treatment with bone marrow-derived stromal cells accelerates wound healing in diabetic rats. Int Wound J. 2008;5:453–463. [PMC free article] [PubMed] [Google Scholar]

121. Laverdet B, Micallef L, Lebreton C, Mollard J, Lataillade JJ, Coulomb B, Desmoulière A. Use of mesenchymal stem cells for cutaneous repair and skin substitute elaboration. Pathol Biol (Paris) 2014;62:108–117. [PubMed] [Google Scholar]

122. Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007;100:1249–1260. [PMC free article] [PubMed] [Google Scholar]

123. Yoshimura K, Suga H, Eto H. Adipose-derived stem/progenitor cells: roles in adipose tissue remodeling and potential use for soft tissue augmentation. Regen Med. 2009;4:265–273. [PubMed] [Google Scholar]

124. Iyyanki TS, Dunne LW, Zhang Q, Hubenak J, Turza KC, Butler CE. Adipose-Derived Stem-Cell-Seeded Non-Cross-Linked Porcine Acellular Dermal Matrix Increases Cellular Infiltration, Vascular Infiltration, and Mechanical Strength of Ventral Hernia Repairs. Tissue Eng Part A. 2014 Oct 2; [Epub ahead of print]. [PMC free article] [PubMed] [Google Scholar]

125. Jeong JH. Adipose stem cells and skin repair. Curr Stem Cell Res Ther. 2010 Jun;5(2):137–140. [PubMed] [Google Scholar]

126. Huang SP, Hsu CC, Chang SC, Wang CH, Deng SC, Dai NT, Chen TM, Chan JY, Chen SG, Huang SM. Adipose-derived stem cells seeded on acellular dermal matrix grafts enhance wound healing in a murine model of a full-thickness defect. Ann Plast Surg. 2012;69:656–662. [PubMed] [Google Scholar]

127. Ribeiro J, Pereira T, Amorim I, Caseiro AR, Lopes MA, Lima J, Gartner A, Santos JD, Bártolo PJ, Rodrigues JM, Mauricio AC, Luís AL. Cell therapy with human MSCs isolated from the umbilical cord Wharton jelly associated to a PVA membrane in the treatment of chronic skin wounds. Int J Med Sci. 2014;11:979–987. [PMC free article] [PubMed] [Google Scholar]

128. Shrestha C, Zhao L, Chen K, He H, Mo Z. Enhanced healing of diabetic wounds by subcutaneous administration of human umbilical cord derived stem cells and their conditioned media. Int J Endocrinol. 2013;2013:592454. [PMC free article] [PubMed] [Google Scholar]

129. Azari O, Babaei H, Drakhsahnfar A, Nematollahi-Mahani SN, Poursahebi R, Moshrefi M. Effects of transplanted mesenchymal stem cells isolated from Wharton’s jelly of caprine umbilical cord on cutaneous wound healing: histopathological evaluation. Vet Res Commun. 2011;35:211–222. [PubMed] [Google Scholar]

130. Shohara R, Yamamoto A, Takikawa S, Iwase A, Hibi H, Kikkawa F, Ueda M. Mesenchymal stromal cells of human umbilical cord Wharton’s jelly accelerate wound healing by paracrine mechanisms. Cytotherapy. 2012;14:1171–1181. [PubMed] [Google Scholar]

131. Langer A, Rogowski W. Systematic review of economic evaluations of human cell-derived wound care products for the treatment of venous leg and diabetic foot ulcers. BMC health services research. 2009;9:115. [PMC free article] [PubMed] [Google Scholar]

132. Margolis DJ, Malay DS, Hoffstad OJ, Leonard CE, MaCurdy T, Tan Y, Molina T Y, de Nava KL, Siegel KL. Data Points Publication Series [Internet] Rockville (MD): Agency for Healthcare Research and Quality (US); 2011. Economic Burden of Diabetic Foot Ulcers and Amputations: Data Points #3. -. [PubMed] [Google Scholar]

133. Poon VK, Burd A. In vitro cytotoxity of silver: implication for clinical wound care. Burns. 2004;30:140–147. [PubMed] [Google Scholar]

134. Chan S, Horner SR, Fauchet PM, Miller BL. Identification of Gram negative bacteria using nanoscale silicon microcavities. Journal of the American Chemical Society. 2001;123:11797–11798. [PubMed] [Google Scholar]

135. Bowler PG, Davies BJ, Jones SA. Microbial involvement in chronic wound malodour. Journal of wound care. 1999;8:216–218. [PubMed] [Google Scholar]

136. Di Pietrantonio F, Cannata D, Benetti M, Verona E, Varriale A, Staiano M, D'Auria S. Detection of odorant molecules via surface acoustic wave biosensor array based on odorant-binding proteins. Biosensors & bioelectronics. 2013;41:328–334. [PubMed] [Google Scholar]

137. Nakagami G, Sanada H, Iizaka S, Kadono T, Higashino T, Koyanagi H, Haga N. Predicting delayed pressure ulcer healing using thermography: a prospective cohort study. Journal of wound care. 2010;19:465–466. 468–470. passim. [PubMed] [Google Scholar]

138. Hattori Y Y, Falgout L, Lee W, Jung SY, Poon E E, Lee JW, et al. Multifunctional skin-like electronics for quantitative, clinical monitoring of cutaneous wound healing. Adv Healthc Mater. 2014;3:1597–1607. [PMC free article] [PubMed] [Google Scholar]

139. Atiyeh BS, Ioannovich J, Al-Amm CA, El-Musa KA. Management of acute and chronic open wounds: the importance of moist environment in optimal wound healing. Current pharmaceutical biotechnology. 2002;3:179–195. [PubMed] [Google Scholar]

140. Okan D D, Woo K, Ayello EA, Sibbald G. The role of moisture balance in wound healing. Adv Skin Wound Care. 2007;20:39–53. quiz 53-5. [PubMed] [Google Scholar]

141. Grimstad Ø, Sandanger Ø, Ryan L, Otterdal K, Damaas JK, Pukstad B, Espevik T. Cellular sources and inducers of cytokines present in acute wound fluid. Wound Repair Regen. 2011;3:337–347. [PubMed] [Google Scholar]

142. Katz MH, Alvarez AF, Kirsner RS, Eaglstein WH, Falanga V. Human wound fluid from acute wounds stimulates fibroblast and endothelial cell growth. J Am Acad Dermatol. 1991;25:1054–1058. [PubMed] [Google Scholar]

143. McColl D, Cartlidge B, Connolly P. Real-time monitoring of moisture levels in wound dressings in vitro: an experimental study. International journal of surgery (London, England) 2007;5:316–322. [PubMed] [Google Scholar]

Current wound healing procedures and potential care (2024)

FAQs

What is the current treatment of wound healing? ›

The current wound healing approaches are based on autografts, allografts, and cultured epithelial autografts, and wound dressings based on biocompatible and biodegradable polymers.

What is the current concept in wound healing? ›

It describes the three phases of normal wound healing: inflammatory, proliferative, and remodeling. The inflammatory phase involves hemostasis and inflammation. The proliferative phase mainly involves fibroblast activity and production of new tissue.

What is the 5 step process of wound healing? ›

In adult humans, optimal wound healing involves the following the events: (1) rapid hemostasis; (2) appropriate inflammation; (3) mesenchymal cell differentiation, proliferation, and migration to the wound site; (4) suitable angiogenesis; (5) prompt re-epithelialization (re-growth of epithelial tissue over the wound ...

What is a wound care procedure? ›

Wound care involves every stage of wound management. This includes diagnosing wound type, considering factors that affect wound healing, and the proper treatments for wound management. Once the wound is diagnosed and all factors are considered, the treatment facility can determine the best treatment options.

Which current is used for wound healing? ›

There are three types of electrical current that assist in wound closure and healing: direct current (DC), alternating current, and pulsed current (PC).

What is the best thing for wound healing? ›

Wound healing is most successful in a moist, clean, and warm environment. Some wounds, such as minor cuts and scrapes, can be treated at home. Stop the bleeding with direct pressure, and clean the wound with water. You DO NOT need soap or hydrogen peroxide.

What are new wound healing techniques? ›

Recent developments in advanced wound care technology includes nanotherapeutics, stem cells therapy, bioengineered skin grafts, and 3D bioprinting-based strategies for improving therapeutic outcomes with a focus on skin regeneration with minimal side effects.

What is the process of wound healing? ›

The human adult wound healing process can be divided into 3 or 4 distinct phases. Earlier authors referred to 3 phases—inflammatory, fibroblastic, and maturation, which has also been denoted as inflammatory, proliferation, and remodeling—and this is maintained by some authors.

What is the standard of care wound healing? ›

Wound care should employ comprehensive wound management including appropriate control of complicating factors such as unrelieved pressure, infection, vascular and/or uncontrolled metabolic derangement, and/or nutritional deficiency in addition to appropriate debridement.

What are the 3 basic principles of wound healing? ›

All dermal wounds heal by three basic mechanisms: contraction, connective tissue matrix deposition and epithelialization. Wounds that remain open heal by contraction; the interaction between cells and matrix results in movement of tissue toward the center of the wound.

How do you speed up the healing of a wound? ›

Fresh fruits and vegetables eaten daily will also supply your body with other nutrients essential to wound healing such as vitamin A, copper and zinc. It may help to supplement your diet with extra vitamin C. Keep your wound dressed. Wounds heal faster if they are kept warm.

What is basic wound care? ›

Immediately after the injury, wash thoroughly with clean water and mild soap. Remove any visible dirt or debris from the wound. Apply gentle pressure to stop bleeding. For burn wounds, run cool water over the area or apply a cool, wet cloth. If blisters form, do not pop or drain.

What are the 5 rules of wound care? ›

Wound care is increasingly becoming complex with the introduction of advanced wound technology. However, the entire wound care can be distilled into five basic principles. These five principles include wound assessment, wound cleansing, timely dressing change, selection of appropriate dressings, and antibiotic use.

What is the best medicine for wound healing? ›

A first-aid antibiotic ointment (Bacitracin, Neosporin, Polysporin) can be applied to help prevent infection and keep the wound moist. Continue to care for the wound. Continued care of the wound is also important.

What are the new advances in wound healing? ›

Negative pressure wound therapy

It is a significant, clinically proven advancement in wound care that promotes active wound healing at the cellular level through negative pressure. Pressure used is negative pressure or sub atmospheric pressure (100-125 mm Hg) in a continuous or intermittent manner.

What is the new technology for wound care? ›

Various technologies for wound healing, including targeted and controlled drug delivery45,46, bioelectronics stimulation11, photodynamic therapy47,48, negative pressure wound therapy (NPWT)49, hyperbaric oxygen therapy50 and gene and cell therapy51, have progressed substantially, facilitated by these novel materials.

Which solution is best for wound healing? ›

Normal sterile saline is regarded as the most appropriate and preferred cleansing solution because it is a nontoxic, isotonic solution that does not damage healing tissues. Tap water is commonly used and is therefore of interest as a cleansing solution.

Top Articles
Latest Posts
Article information

Author: Lakeisha Bayer VM

Last Updated:

Views: 6520

Rating: 4.9 / 5 (69 voted)

Reviews: 84% of readers found this page helpful

Author information

Name: Lakeisha Bayer VM

Birthday: 1997-10-17

Address: Suite 835 34136 Adrian Mountains, Floydton, UT 81036

Phone: +3571527672278

Job: Manufacturing Agent

Hobby: Skimboarding, Photography, Roller skating, Knife making, Paintball, Embroidery, Gunsmithing

Introduction: My name is Lakeisha Bayer VM, I am a brainy, kind, enchanting, healthy, lovely, clean, witty person who loves writing and wants to share my knowledge and understanding with you.