We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Cellular and biological factors involved in healing wounds and burns and treatment options in tissue engineering

    Pegah Moharrami Yeganeh

    School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran

    ,
    Safa Tahmasebi

    Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran

    &
    Abdolreza Esmaeilzadeh

    *Author for correspondence: Tel.: +98 912 141 4281;

    E-mail Address: a46reza@zums.ac.ir

    Department of immunology, School of Medicine, Zanjan University of Medical Science, Zanjan, 4513956111, Iran

    Cancer Gene Therapy Research Center, Zanjan University of Medical Science, Zanjan, Iran

    Published Online:https://doi.org/10.2217/rme-2022-0029

    Severe traumatic wounds and burns have a high chance of mortality and can leave survivors with many functional disabilities and cosmetic problems, including scars. The healing process requires a harmonious interplay of various cells and growth factors. Different structures of the skin house numerous cells, matrix components and growth factors. Any disturbance in the balance between these components can impair the healing process. The function of cells and growth factors can be manipulated and facilitated to aid tissue repair. In the current review, the authors focus on the importance of the skin microenvironment, the pathophysiology of various types of burns, mechanisms and factors involved in skin repair and wound healing and regeneration of the skin using tissue engineering approaches.

    Plain language summary

    Wounds and ulcers, especially burn wounds, are major causes of morbidity and mortality and pose a significant burden for individuals and societies. The skin has numerous structures that play important roles in wound healing via cells and growth factors. Tissue engineering and regenerative medicine represent a rather new field that focuses on manipulating cells and growth factors, aiming to facilitate repair and regeneration of injured tissues and organs. This review focuses on different burn injuries that can result in nonhealing wounds, provides an overview of several cells and growth factors that are involved in the healing process of the skin and introduces various strategies practiced in tissue engineering with regard to cutaneous wound healing.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Dissanaike S. Editorial on trends in burn injury. Ann. Surg. 270(6), 954 (2019).
    • 2. Bhattacharya Sameek. Burns: Etiology and classification. In: Principles and Practice of Burn Care. Sarabahi STVGoel A (Eds). Jaypee Publishers, New Delhi, India, 25–36 (2010).
    • 3. Tiwari V. Burn wound: how it differs from other wounds? Indian J. Plast. Surg. 45(2), 364–373 (2012).
    • 4. Brandão C, Vaz M, Brito IM et al. Electrical burns: a retrospective analysis over a 10-year period. Ann. Burns Fire Disasters 30(4), 268–271 (2017).
    • 5. WHO. Burns. www.who.int/news-room/fact-sheets/detail/burns
    • 6. Kaddoura I, Abu-Sittah G, Ibrahim A, Karamanoukian R, Papazian N. Burn injury: review of pathophysiology and therapeutic modalities in major burns. Ann. Burns Fire Disasters 30(2), 95 (2017).
    • 7. Yousef H, Alhajj M, Sharma S. Anatomy, Skin (Integument), Epidermis. In: StatPearls [Internet]. StatPearls Publishing, Treasure Island, FL, USA (2022). [Updated 2021 Nov 19]. Available from: www.ncbi.nlm.nih.gov/books/NBK470464/
    • 8. Williams AE. Immunology of the Skin. In: Immunology. Williams AE. (Ed.). (2011). https://doi.org/10.1002/9781119998648.ch10
    • 9. Hashmi S, Marinkovich MP. Molecular organization of the basement membrane zone. Clin. Dermatol. 29(4), 398–411 (2011).
    • 10. Walker NJ, King KC. Acute and Chronic Thermal Burn Evaluation and Management. In: StatPearls [Internet]. StatPearls Publishing, Treasure sland, FL, USA (2022). [Updated 2021 Jul 21]. Available from: www.ncbi.nlm.nih.gov/books/NBK430730/
    • 11. Brans T, Dutrieux R, Hoekstra M, Kreis R, Du Pont J. Histopathological evaluation of scalds and contact burns in the pig model. Burns 20, S48–S51 (1994).
    • 12. Button B, Baker RD, Vertrees RA, Allen SE, Brodwick MS, Kramer GC. Quantitative assessment of a circulating depolarizing factor in shock. Shock 15(3), 239–244 (2001).
    • 13. Evans JA, Darlington DN, Gann DS. A circulating factor (s) mediates cell depolarization in hemorrhagic shock. Ann. Surg. 213(6), 549 (1991).
    • 14. Trunkey DD, Illner H, Arango A, Holiday R, Shires GT. Changes in cell membrane function following shock and cross-perfusion. Surgical forum. 25(0), 1–3 (1974).
    • 15. Friedstat J, Brown DA, Levi B. Chemical, electrical, and radiation injuries. Clin. Plast. Surg. 44(3), 657 (2017).
    • 16. Bounds EJ, Khan M, Kok SJ. Electrical Burns. In: StatPearls [Internet]. StatPearls Publishing, FL, USA (2022). [Updated 2021 May 4]. Available from: www.ncbi.nlm.nih.gov/books/NBK519514/
    • 17. Vučenović S, Šetrajčić JP, Vojnović M, Šetrajčić-Tomić AJ, Džambas LD. Physiological processes when an electrical current passes through the tissues and organs. Presented at: Fifth International Conference on radiation and applications in various fields of research. Budva, Montenegro (12.6 - 16/6 2017).
    • 18. Grimnes S. Dielectric breakdown of human skin in vivo. Med. Biol. Eng. Comput. 21(3), 379–381 (1983).
    • 19. Waghmare CM. Radiation burn – from mechanism to management. Burns 39(2), 212–219 (2013).
    • 20. Koenig TR, Wolff D, Mettler FA, Wagner LK. Skin injuries from fluoroscopically guided procedures: part 1, characteristics of radiation injury. AJR Am. J. Roentgenol. 177(1), 3–11 (2001).
    • 21. Palao R, Monge I, Ruiz M, Barret JP. Chemical burns: pathophysiology and treatment. Burns 36(3), 295–304 (2010).
    • 22. Seth R, Chester D, Moiemen N. A review of chemical burns. Trauma 9(2), 81–94 (2007).
    • 23. Robson MC. Plastic Surgery: Principles and Practice. Jurkiewicz EA (Ed.). CV Mosby, MO, USA, 1355–1410 (1990).
    • 24. Williams FN, Lee JO. Chemical burns. In: Total Burn Care (5th Edition). Herndon DN (Ed.). Elsevier, 408–413.e401 (2018). eBook ISBN: 9780323497428
    • 25. Dinis-Oliveira RJ, Carvalho F, Moreira R et al. Clinical and forensic signs related to chemical burns: a mechanistic approach. Burns 41(4), 658–679 (2015).
    • 26. Matshes EW, Taylor KA, Rao VJ. Sulfuric acid injury. Am. J. Forensic Med. Pathol. 29(4), 340–345 (2008).
    • 27. Jelenko C 3rd. Chemicals that ‘burn.’ J. Trauma 14(1), 65–72 (1974).
    • 28. Thorne CH, Gosain AK, Gurtner GC, Mehrara BJ, Rubin JP, Spear Sl. Grabb and Smith's Plastic Surgery (7th Edition). Lippincott Wiliams & Wilkins, London, UK(2014).
    • 29. Scalise A, Bianchi A, Tartaglione C et al. Microenvironment and microbiology of skin wounds: the role of bacterial biofilms and related factors. Semin. Vasc. Surg. 28(3), 151–159 (2015).
    • 30. Li J, Chen J, Kirsner R. Pathophysiology of acute wound healing. Clin. Dermatol. 25(1), 9–18 (2007).
    • 31. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature 453(7193), 314–321 (2008).
    • 32. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ. Res. 92(8), 827–839 (2003).
    • 33. Tizard IR. Immunity at body surfaces. In: Veterinary Immunology. Tizard IR (Ed.). Saunders, PA, USA, 222–234 (2000).
    • 34. Kubo T, Sugimoto K, Kojima T, Sawada N, Sato N, Ichimiya S. Tight junction protein claudin-4 is modulated via Δnp63 in human keratinocytes. Biochem. Biophys. Res. Commun. 455(3–4), 205–211 (2014).
    • 35. Raja SK, Garcia MS, Isseroff RR. Wound re-epithelialization: modulating keratinocyte migration in wound healing. Front. Biosci. 12(3), 2849–2868 (2007).
    • 36. Gasque P, Jaffar-Bandjee MC. The immunology and inflammatory responses of human melanocytes in infectious diseases. J. Infect. 71(4), 413–421 (2015).
    • 37. Plonka PM, Passeron T, Brenner M et al. What are melanocytes really doing all day long…? Exp. Dermatol. 18(9), 799–819 (2009).
    • 38. Rognoni E, Watt FM. Skin cell heterogeneity in development, wound healing, and cancer. Trends Cell Biol. 28(9), 709–722 (2018).
    • 39. Brown TM, Krishnamurthy K. Histology, Dermis. [Updated 2021 Nov 19]. In: StatPearls [Internet]. StatPearls Publishing, FL, USA (2022) Available from: www.ncbi.nlm.nih.gov/books/NBK535346/
    • 40. Stunova A, Vistejnova L. Dermal fibroblasts – a heterogeneous population with regulatory function in wound healing. Cytokine Growth Factor Rev. 39, 137–150 (2018).
    • 41. Darby IA, Zakuan N, Billet F, Desmouliere A. The myofibroblast, a key cell in normal and pathological tissue repair. Cell. Mol. Life Sci. 73(6), 1145–1157 (2016).
    • 42. Darby IA, Hewitson TD. Fibroblast differentiation in wound healing and fibrosis. International review of cytology. International review of cytology 257, 143–179 (2007).
    • 43. Gauglitz GG, Korting HC, Pavicic T, Ruzicka T, Jeschke MG. Hypertrophic scarring and keloids: pathomechanisms and current and emerging treatment strategies. Mol. Med. 17(1), 113–125 (2011).
    • 44. Li Z, Maitz P. Cell therapy for severe burn wound healing. Burns Trauma 6, 13 (2018).
    • 45. Nanba D. Human keratinocyte stem cells: from cell biology to cell therapy. J. Dermatol. Sci. 96(2), 66–72 (2019).
    • 46. Spichkina OG, Kalmykova NV, Kukhareva LV, Voronkina IV, Blinova MI, Pinaev GP. Isolation of human basal keratinocytes by selective adhesion to extracellular matrix proteins. Tsitologiia 48(10), 841–847 (2006).
    • 47. Hasebe Y, Hasegawa S, Hashimoto N et al. Analysis of cell characterization using cell surface markers in the dermis. J. Dermatol. Sci. 62(2), 98–106 (2011).
    • 48. Iwata Y, Akamatsu H, Hasebe Y, Hasegawa S, Sugiura K. Skin-resident stem cells and wound healing. Nihon Rinsho Meneki Gakkai Kaishi 40(1), 1–11 (2017).
    • 49. Iwata Y, Akamatsu H, Hasegawa S et al. The epidermal integrin beta-1 and p75NTR positive cells proliferating and migrating during wound healing produce various growth factors, while the expression of p75NTR is decreased in patients with chronic skin ulcers. J. Dermatol. Sci. 71(2), 122–129 (2013).
    • 50. Hasebe Y, Hasegawa S, Hashimoto N et al. Analysis of cell characterization using cell surface markers in the dermis. J. Dermatol. Sci. 62(2), 98–106 (2011).
    • 51. Iwata Y, Hasebe Y, Hasegawa S et al. Dermal CD271+ cells are closely associated with regeneration of the dermis in the wound healing process. Acta Derm. Venereol. 97(5), 593–600 (2017).
    • 52. Hynds RE, Bonfanti P, Janes SM. Regenerating human epithelia with cultured stem cells: feeder cells, organoids and beyond. EMBO molecular medicine 10(2), 139–150 (2018).
    • 53. Unger C, Gao S, Cohen M et al. Immortalized human skin fibroblast feeder cells support growth and maintenance of both human embryonic and induced pluripotent stem cells. Human reproduction (Oxford, England) 24(10), 2567–2581 (2009).
    • 54. Werner S, Krieg T, Smola H. Keratinocyte–fibroblast interactions in wound healing. J. Invest. Dermatol. 127(5), 998–1008 (2007).
    • 55. Maas-Szabowski N, Shimotoyodome A, Fusenig NE. Keratinocyte growth regulation in fibroblast cocultures via a double paracrine mechanism. J. Cell Sci. 112(12), 1843–1853 (1999).
    • 56. Waelti ER, Inaebnit SP, Rast HP et al. Co-culture of human keratinocytes on post-mitotic human dermal fibroblast feeder cells: production of large amounts of interleukin 6. J. Invest. Dermatol. 98(5), 805–808 (1992).
    • 57. Smola H, Thiekötter G, Fusenig NE. Mutual induction of growth factor gene expression by epidermal–dermal cell interaction. J. Cell Biol. 122(2), 417–429 (1993).
    • 58. Boxman IL, Ruwhof C, Boerman OC, Löwik CW, Ponec M. Role of fibroblasts in the regulation of proinflammatory interleukin IL-1, IL-6 and IL-8 levels induced by keratinocyte-derived IL-1. Arch. Dermatol. Res. 288(7), 391–398 (1996).
    • 59. Karin M, Liu Z, Zandi E. AP-1 function and regulation. Curr. Opin. Cell Biol. 9(2), 240–246 (1997).
    • 60. Szabowski A, Maas-Szabowski N, Andrecht S et al. c-Jun and JunB antagonistically control cytokine-regulated mesenchymal–epidermal interaction in skin. Cell 103(5), 745–755 (2000).
    • 61. Florin L, Maas-Szabowski N, Werner S, Szabowski A, Angel P. Increased keratinocyte proliferation by JUN-dependent expression of PTN and SDF-1 in fibroblasts. J. Cell Sci. 118(9), 1981–1989 (2005).
    • 62. Brazil JC, Quiros M, Nusrat A, Parkos CA. Innate immune cell–epithelial crosstalk during wound repair. J. Clin. Invest. 129(8), 2983–2993 (2019).
    • 63. Brinkmann V, Reichard U, Goosmann C et al. Neutrophil extracellular traps kill bacteria. Science 303(5663), 1532–1535 (2004).
    • 64. Feiken E, Rømer J, Eriksen J, Lund LR. Neutrophils express tumor necrosis factor-alpha during mouse skin wound healing. J. Invest. Dermatol. 105(1), 120–123 (1995).
    • 65. Campbell EL, Bruyninckx WJ, Kelly CJ et al. Transmigrating neutrophils shape the mucosal microenvironment through localized oxygen depletion to influence resolution of inflammation. Immunity 40(1), 66–77 (2014).
    • 66. Chen F, Yang W, Huang X et al. Neutrophils promote amphiregulin production in intestinal epithelial cells through TGF-β and contribute to intestinal homeostasis. J. Immunol. 201(8), 2492–2501 (2018).
    • 67. Ramirez H, Patel SB, Pastar I. The role of TGFβ signaling in wound epithelialization. Adv. Wound Care (New Rochelle) 3(7), 482–491 (2014).
    • 68. Cox DA, Kunz S, Cerletti N, McMaster GK, Burk RR. Wound healing in aged animals – effects of locally applied transforming growth factor beta 2 in different model systems. EXS 61, 287–295 (1992).
    • 69. Gharaee-Kermani M, Phan SH. Role of cytokines and cytokine therapy in wound healing and fibrotic diseases. Curr. Pharm. Des. 7(11), 1083–1103 (2001).
    • 70. Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol. Rev. 83(3), 835–870 (2003).
    • 71. Beer HD, Longaker MT, Werner S. Reduced expression of PDGF and PDGF receptors during impaired wound healing. J. Invest. Dermatol. 109(2), 132–138 (1997).
    • 72. Pierce GF, Tarpley JE, Tseng J et al. Detection of platelet-derived growth factor (PDGF)-AA in actively healing human wounds treated with recombinant PDGF-BB and absence of PDGF in chronic nonhealing wounds. J. Clin. Invest. 96(3), 1336–1350 (1995).
    • 73. Beer HD, Fassler R, Werner S. Glucocorticoid-regulated gene expression during cutaneous wound repair. Vitam. Horm. 59, 217–239 (2000).
    • 74. Niessen FB, Andriessen MP, Schalkwijk J, Visser L, Timens W. Keratinocyte-derived growth factors play a role in the formation of hypertrophic scars. J. Pathol. 194(2), 207–216 (2001).
    • 75. Haisa M, Okochi H, Grotendorst GR. Elevated levels of PDGF alpha receptors in keloid fibroblasts contribute to an enhanced response to PDGF. J. Invest. Dermatol. 103(4), 560–563 (1994).
    • 76. Clark RA. Regulation of fibroplasia in cutaneous wound repair. Am. J. Med. Sci. 306(1), 42–48 (1993).
    • 77. Seppä H, Grotendorst G, Seppä S, Schiffmann E, Martin GR. Platelet-derived growth factor in chemotactic for fibroblasts. J. Cell Biol. 92(2), 584–588 (1982).
    • 78. Deuel TF, Senior RM, Huang J, Griffin G. Chemotaxis of monocytes and neutrophils to platelet-derived growth factor. J. Clin. Invest. 69(4), 1046–1049 (1982).
    • 79. Risau W, Drexler H, Mironov V et al. Platelet-derived growth factor is angiogenic in vivo. Growth Factors 7(4), 261–266 (1992).
    • 80. Nicosia RF, Nicosia S, Smith M. Vascular endothelial growth factor, platelet-derived growth factor, and insulin-like growth factor-1 promote rat aortic angiogenesis in vitro. Am. J. Pathol. 145(5), 1023 (1994).
    • 81. Yun YR, Won JE, Jeon E et al. Fibroblast growth factors: biology, function, and application for tissue regeneration. J. Tissue Eng. 2010, 218142 (2010).
    • 82. Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 103(2), 211–225 (2000).
    • 83. Katoh M, Katoh M. FGF signaling network in the gastrointestinal tract (review). Int. J. Oncol. 29(1), 163–168 (2006).
    • 84. Kolkova K, Novitskaya V, Pedersen N, Berezin V, Bock E. Neural cell adhesion molecule-stimulated neurite outgrowth depends on activation of protein kinase C and the Ras-mitogen-activated protein kinase pathway. J. Neurosci. 20(6), 2238–2246 (2000).
    • 85. Werner S, Breeden M, Hübner G, Greenhalgh DG, Longaker MT. Induction of keratinocyte growth factor expression is reduced and delayed during wound healing in the genetically diabetic mouse. J. Invest. Dermatol. 103(4), 469–473 (1994).
    • 86. Shukla A, Dubey M, Srivastava R, Srivastava BS. Differential expression of proteins during healing of cutaneous wounds in experimental normal and chronic models. Biochem. Biophys. Res. Commun. 244(2), 434–439 (1998).
    • 87. Swift ME, Kleinman HK, Dipietro LA. Impaired wound repair and delayed angiogenesis in aged mice. Lab. Invest. 79(12), 1479–1487 (1999).
    • 88. Broadley K, Aquino A, Woodward S et al. Monospecific antibodies implicate basic fibroblast growth factor in normal wound repair. Lab. Invest. 61(5), 571–575 (1989).
    • 89. Ortega S, Ittmann M, Tsang SH, Ehrlich M, Basilico C. Neuronal defects and delayed wound healing in mice lacking fibroblast growth factor 2. Proc. Natl Acad. Sci. U. S. A. 95(10), 5672–5677 (1998).
    • 90. Werner S, Smola H, Liao X et al. The function of KGF in epithelial morphogenesis and wound reepithelialization. Science 266, 819–822 (1994).
    • 91. Igarashi M, Finch PW, Aaronson SA. Characterization of recombinant human fibroblast growth factor (FGF)-10 reveals functional similarities with keratinocyte growth factor (FGF-7). J. Biol. Chem. 273(21), 13230–13235 (1998).
    • 92. Ornitz DM, Xu J, Colvin JS et al. Receptor specificity of the fibroblast growth factor family. J. Biol. Chem. 271(25), 15292–15297 (1996).
    • 93. Guo L, Degenstein L, Fuchs E. Keratinocyte growth factor is required for hair development but not for wound healing. Genes Dev. 10(2), 165–175 (1996).
    • 94. Schneider MR, Werner S, Paus R, Wolf E. Beyond wavy hairs: the epidermal growth factor receptor and its ligands in skin biology and pathology. Am. J. Pathol. 173(1), 14–24 (2008).
    • 95. Cook PW, Pittelkow MR, Keeble WW, Graves-Deal R, Coffey RJ, Shipley GD. Amphiregulin messenger RNA is elevated in psoriatic epidermis and gastrointestinal carcinomas. Cancer Res. 52(11), 3224–3227 (1992).
    • 96. Piepkorn M. Overexpression of amphiregulin, a major autocrine growth factor for cultured human keratinocytes, in hyperproliferative skin diseases. Am. J. Dermatopathol. 18(2), 165–171 (1996).
    • 97. Rittié L, Kansra S, Stoll SW et al. Differential ErbB1 signaling in squamous cell versus basal cell carcinoma of the skin. Am. J. Pathol. 170(6), 2089–2099 (2007).
    • 98. Dahlhoff M, Schneider MR. Transgenic mouse lines help decipher the roles of EGFR ligands in the skin. Exp. Dermatol. 25(3), 185–186 (2016).
    • 99. Schneider MR, Antsiferova M, Feldmeyer L et al. Betacellulin regulates hair follicle development and hair cycle induction and enhances angiogenesis in wounded skin. J. Invest. Dermatol. 128(5), 1256–1265 (2008).
    • 100. Shirakata Y, Kimura R, Nanba D et al. Heparin-binding EGF-like growth factor accelerates keratinocyte migration and skin wound healing. J. Cell Sci. 118(11), 2363–2370 (2005).
    • 101. Yamazaki S, Iwamoto R, Saeki K et al. Mice with defects in HB-EGF ectodomain shedding show severe developmental abnormalities. J. Cell Biol. 163(3), 469–475 (2003).
    • 102. Su Y, Richmond A. Chemokine regulation of neutrophil infiltration of skin wounds. Adv. Wound Care (New Rochelle) 4(11), 631–640 (2015).
    • 103. Bryant VL, Slade CA. Chemokines, their receptors and human disease: the good, the bad and the itchy. Immunol. Cell Biol. 93(4), 364–371 (2015).
    • 104. Baggiolini M. Chemokines in pathology and medicine. J. Intern. Med. 250(2), 91–104 (2001).
    • 105. Drechsler M, Duchene J, Soehnlein O. Chemokines control mobilization, recruitment, and fate of monocytes in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 35(5), 1050–1055 (2015).
    • 106. Bodnar RJ. Chemokine regulation of angiogenesis during wound healing. Adv. Wound Care (New Rochelle) 4(11), 641–650 (2015).
    • 107. Christopherson K, Hromas R. Chemokine regulation of normal and pathologic immune responses. Stem Cells 19(5), 388–396 (2001).
    • 108. Dipietro LA, Polverini PJ, Rahbe SM, Kovacs EJ. Modulation of JE/MCP-1 expression in dermal wound repair. Am. J. Pathol. 146(4), 868 (1995).
    • 109. Jackman SH, Yoak MB, Keerthy S, Beaver BL. Differential expression of chemokines in a mouse model of wound healing. Ann. Clin. Lab. Sci. 30(2), 201–207 (2000).
    • 110. Wetzler C, Kämpfer H, Stallmeyer B, Pfeilschifter J, Frank S. Large and sustained induction of chemokines during impaired wound healing in the genetically diabetic mouse: prolonged persistence of neutrophils and macrophages during the late phase of repair. J. Invest. Dermatol. 115(2), 245–253 (2000).
    • 111. Engelhardt E, Toksoy A, Goebeler M, Debus S, Bröcker EB, Gillitzer R. Chemokines IL-8, GROα, MCP-1, IP-10, and Mig are sequentially and differentially expressed during phase-specific infiltration of leukocyte subsets in human wound healing. Am. J. Pathol. 153(6), 1849–1860 (1998).
    • 112. Dipietro LA, Burdick M, Low QE, Kunkel SL, Strieter RM. MIP-1alpha as a critical macrophage chemoattractant in murine wound repair. J. Clin. Invest. 101(8), 1693–1698 (1998).
    • 113. Low QE, Drugea IA, Duffner LA et al. Wound healing in MIP-1α-/- and MCP-1-/- mice. Am. J. Pathol. 159(2), 457–463 (2001).
    • 114. Ding J, Tredget EE. The role of chemokines in fibrotic wound healing. Adv. Wound Care (New Rochelle) 4(11), 673–686 (2015).
    • 115. Martins-Green M, Petreaca M, Wang L. Chemokines and their receptors are key players in the orchestra that regulates wound healing. Adv. Wound Care (New Rochelle) 2(7), 327–347 (2013).
    • 116. Rennekampff HO, Hansbrough JF, Woods V Jr, Doré C, Kiessig V, Schröder JM. Role of melanoma growth stimulatory activity (MGSA/gro) on keratinocyte function in wound healing. Arch. Dermatol. Res. 289(4), 204–212 (1997).
    • 117. Jones SA, Wolf M, Qin S, Mackay CR, Baggiolini M. Different functions for the interleukin 8 receptors (IL-8R) of human neutrophil leukocytes: NADPH oxidase and phospholipase D are activated through IL-8R1 but not IL-8R2. Proc. Natl Acad. Sci. U. S. A. 93(13), 6682–6686 (1996).
    • 118. Rennekampff HO, Hansbrough JF, Kiessig V, Doré C, Sticherling M, Schröder JM. Bioactive interleukin-8 is expressed in wounds and enhances wound healing. J. Surg. Res. 93(1), 41–54 (2000).
    • 119. Iocono JA, Colleran KR, Remick DG, Gillespie BW, Ehrlich HP, Garner WL. Interleukin-8 levels and activity in delayed-healing human thermal wounds. Wound Repair Regen. 8(3), 216–225 (2000).
    • 120. Puapatanakul P, Chansritrakul S, Susantitaphong P et al. Interferon-inducible protein 10 and disease activity in systemic lupus erythematosus and lupus nephritis: a systematic review and meta-analysis. Int. J. Mol. Sci. 20(19), 4954 (2019).
    • 121. Luster AD, Cardiff RD, Maclean JA, Crowe K, Granstein RD. Delayed wound healing and disorganized neovascularization in transgenic mice expressing the IP-10 chemokine. Proc. Assoc. Am. Physicians 110(3), 183–196 (1998).
    • 122. Belperio JA, Keane MP, Arenberg DA et al. CXC chemokines in angiogenesis. J. Leukoc. Biol. 68(1), 1–8 (2000).
    • 123. Luster AD, Greenberg SM, Leder P. The IP-10 chemokine binds to a specific cell surface heparan sulfate site shared with platelet factor 4 and inhibits endothelial cell proliferation. J. Exp. Med. 182(1), 219–231 (1995).
    • 124. Singer AJ, Quinn JV, Clark RE, Hollander JE, Group TS. Closure of lacerations and incisions with octylcyanoacrylate: a multicenter randomized controlled trial. Surgery 131(3), 270–276 (2002).
    • 125. Kim H, Kim W, Kang GH et al. Comparison of Leukosan SkinLink with surgical suture for traumatic laceration repair: a randomized controlled trial. Medicine (Baltimore) 97(25), e10918 (2018).
    • 126. Stone RC, Stojadinovic O, Rosa AM et al. A bioengineered living cell construct activates an acute wound healing response in venous leg ulcers. Sci. Transl. Med. 9(371), eaaf8611 (2017).
    • 127. Cogliati B, Vinken M, Silva TC et al. Connexin 43 deficiency accelerates skin wound healing and extracellular matrix remodeling in mice. J. Dermatol. Sci. 79(1), 50–56 (2015).
    • 128. Rhett JM, Jourdan J, Gourdie RG. Connexin 43 connexon to gap junction transition is regulated by zonula occludens-1. Mol. Biol. Cell 22(9), 1516–1528 (2011).
    • 129. Grek CL, Prasad G, Viswanathan V, Armstrong DG, Gourdie RG, Ghatnekar GS. Topical administration of a connexin43-based peptide augments healing of chronic neuropathic diabetic foot ulcers: a multicenter, randomized trial. Wound Repair Regen. 23(2), 203–212 (2015).
    • 130. Ghatnekar GS, Grek CL, Armstrong DG, Desai SC, Gourdie RG. The effect of a connexin43-based peptide on the healing of chronic venous leg ulcers: a multicenter, randomized trial. J. Invest. Dermatol. 135(1), 289–298 (2015).
    • 131. Ghatnekar GS, O'Quinn MP, Jourdan LJ, Gurjarpadhye AA, Draughn RL, Gourdie RG. Connexin43 carboxyl-terminal peptides reduce scar progenitor and promote regenerative healing following skin wounding. Regen. Med. 4(2), 205–223 (2009).
    • 132. Shi A, Li J, Qiu X et al. TGF-β loaded exosome enhances ischemic wound healing in vitro and in vivo. Theranostics 11(13), 6616–6631 (2021). • Introduces a rather novel process and an interesting new product.
    • 133. Atiyeh BS, Costagliola M. Cultured epithelial autograft (CEA) in burn treatment: three decades later. Burns 33(4), 405–413 (2007).
    • 134. Karlsson M, Steinvall I, Olofsson P et al. Sprayed cultured autologous keratinocytes in the treatment of severe burns: a retrospective matched cohort study. Ann. Burns Fire Disasters 33(2), 134 (2020).
    • 135. Yim H, Yang HT, Cho YS et al. Clinical study of cultured epithelial autografts in liquid suspension in severe burn patients. Burns 37(6), 1067–1071 (2011).
    • 136. Schlabe J, Johnen C, Schwartlander R et al. Isolation and culture of different epidermal and dermal cell types from human scalp suitable for the development of a therapeutical cell spray. Burns 34(3), 376–384 (2008).
    • 137. Esteban-Vives R, Young MT, Zhu T et al. Calculations for reproducible autologous skin cell-spray grafting. Burns 42(8), 1756–1765 (2016).
    • 138. Gerlach JC, Johnen C, Ottomann C et al. Method for autologous single skin cell isolation for regenerative cell spray transplantation with non-cultured cells. Int. J. Artif. Organs 34(3), 271–279 (2011).
    • 139. Matsumura H, Gondo M, Imai R, Shibata D, Watanabe K. Chronological histological findings of cultured epidermal autograft over bilayer artificial dermis. Burns 39(4), 705–713 (2013).
    • 140. Jeong JH. Adipose stem cells and skin repair. Curr. Stem Cell Res. Ther. 5(2), 137–140 (2010).
    • 141. Fu X, Fang L, Li X, Cheng B, Sheng Z. Enhanced wound-healing quality with bone marrow mesenchymal stem cells autografting after skin injury. Wound Repair Regen. 14(3), 325–335 (2006).
    • 142. Mao HS, Wang YP, Wang Q et al. Prospective randomized controlled study on clinical effects of autologous skin paste in repairing medium-thickness skin donor site wounds. Zhonghua Shao Shang Za Zhi 37(3), 232–236 (2021).
    • 143. Leung A, Balaji S, Keswani SG. Biology and function of fetal and pediatric skin. Facial Plast. Surg. Clin. North Am. 21(1), 1 (2013).
    • 144. Guo SA, Dipietro LA. Factors affecting wound healing. J. Dent. Res. 89(3), 219–229 (2010).
    • 145. Holmes JHT, Schurr MJ, King BT et al. An open-label, prospective, randomized, controlled, multicenter, phase 1b study of StrataGraft skin tissue versus autografting in patients with deep partial-thickness thermal burns. Burns 45(8), 1749–1758 (2019).
    • 146. Gholian S, Pishgahi A, Shakouri SK et al. Use of autologous conditioned serum dressings in hard-to-heal wounds: a randomised prospective clinical trial. J. Wound Care 31(1), 68–77 (2022). • Examines autologous conditioned serum as a dressing for nonhealing wounds; autologous serum collection and conditioning have been shown to be effective and rather easy to perform.
    • 147. Tissue-Engineered Skin Graft Repair of Autologous Scar Dermal Scaffolds. https://clinicaltrials.gov/show/NCT04389164
    • 148. Autologous Keratinocyte Suspension Versus Adipose-Derived Stem Cell–Keratinocyte Suspension for Post-Burn Raw Area. https://clinicaltrials.gov/show/NCT03686449
    • 149. Study With an Autologous Dermo-epidermal Skin Substitute for the Treatment of Full-Thickness Skin Defects in Adults and Children. https://clinicaltrials.gov/show/NCT03394612
    • 150. Controlled Comparison of a Traditional Dressing Versus a Biologic Dressing Composed of Fetal Fibroblasts and Keratinocytes in Association With a Collagen Matrix on Skin Donor Sites (CICAFAST). https://clinicaltrials.gov/show/NCT03334656
    • 151. Efficacy and Safety of Heberprot-P® in Patients With Advanced Diabetic Foot Ulcer in Dasman Diabetes Institute.. https://clinicaltrials.gov/show/NCT03239457
    • 152. Study With an Autologous Dermo-epidermal Skin Substitute for the Treatment of Burns in Children. https://clinicaltrials.gov/ct2/show/NCT03229564
    • 153. Study With an Autologous Dermo-epidermal Skin Substitute for the Treatment of Burns in Adults. https://clinicaltrials.gov/show/NCT03227146
    • 154. Allogeneic ADSCs and Platelet-Poor Plasma Fibrin Hydrogel to Treat the Patients With Burn Wounds (ADSCs-BWs) (ADSCs-BWs). https://clinicaltrials.gov/show/NCT03113747
    • 155. StrataGraft® Skin Tissue in the Promotion of Autologous Skin Regeneration of Complex Skin Defects Due to Thermal Burns That Contain Intact Dermal Elements. https://clinicaltrials.gov/show/NCT03005106
    • 156. The Activity of Tissue Engineering Skin Substitutes (MSCs). https://clinicaltrials.gov/show/NCT02668042
    • 157. ExpressGraft-C9T1 Skin Tissue as a Treatment of Diabetic Foot Ulcers. https://clinicaltrials.gov/show/NCT02657876
    • 158. EktoTherix Regenerative Tissue Scaffold for Repair of Surgical Excision Wounds. https://clinicaltrials.gov/show/NCT02409628
    • 159. Phase I Study for Autologous Dermal Substitutes and Dermo-epidermal Skin Substitutes for Treatment of Skin Defects. https://clinicaltrials.gov/show/NCT02145130
    • 160. Feasibility Study for Fibroblast Autologous Skin Grafts. https://clinicaltrials.gov/show/NCT01964859
    • 161. A Comparative Efficacy Study: Treatment for Non-healing Diabetic Foot Ulcers. https://clinicaltrials.gov/show/NCT01858545
    • 162. Study Investigating the Safety and Efficacy of HP802-247 in the Treatment of Venous Leg Ulcers >12 cm2 to ≤36 cm2. https://clinicaltrials.gov/show/NCT01737762
    • 163. Study Investigating the Safety and Efficacy of HP802-247 in the Treatment of Venous Leg Ulcers. https://clinicaltrials.gov/show/NCT01656889
    • 164. Effect of Platelet Rich Plasma and Keratinocyte Suspensions on Wound Healing. https://clinicaltrials.gov/show/NCT00856934
    • 165. Application of Cultured Autologous Keratinocytes for Burn Wound Healing (KC). https://clinicaltrials.gov/show/NCT00832156
    • 166. Clinical Application of Autologous Three-Cellular Cultured Skin Substitutes (CSS). https://clinicaltrials.gov/show/NCT00718978
    • 167. Efficacy and Safety Study of DERMAGEN® vs Conventional Treatment to Treat Diabetic Neuropathic Foot Ulcer (DERMAGEN®). https://clinicaltrials.gov/show/NCT00521937
    • 168. Evaluation of Safety and Activity of Celaderm in Healing Venous Leg Ulcers. https://clinicaltrials.gov/show/NCT00399308