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

The biological processes during wound healing

    Fan Yang

    Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China

    ,
    Xiangjun Bai

    Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China

    ,
    Xiaojing Dai

    MD Anderson Cancer Center, The Advanced Technology Genomics Core, Houston, TX 77030, USA

    &
    Yong Li

    *Author for correspondence:

    E-mail Address: Yong.li@med.wmich.edu

    Department of Orthopedic Surgery & Biomedical Engineering, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI 49008, USA

    Published Online:https://doi.org/10.2217/rme-2020-0066

    Numerous individuals suffer from impaired wound healing, such as chronic ulcers, severe burns and immune disorders, resulting in both public health and economic burdens. Skin is the first line of defense and the largest organ of the human body, however, an incomplete understanding of underlying cellular and molecular mechanisms of dermal repair leads to a lack of effective therapy for healing impaired wounds. There are strong clinical and social needs for improved therapeutic approaches to enhance endogenous tissue repair and regenerative capacity. The purpose of this review is to illuminate the cellular and molecular aspects of the healing process and highlight potential therapeutic strategies to accelerate translational research and the development of clinical therapies in dermal wounds.

    References

    • 1. Raghunathan VK, Thomasy SM, Strom P et al. Tissue and cellular biomechanics during corneal wound injury and repair. Acta Biomater. 58, 291–301 (2017).
    • 2. Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci. Transl. Med. 6(265), 265sr266 (2014).
    • 3. Martin P, Nunan R. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br. J. Dermatol. 173(2), 370–378 (2015).
    • 4. Kurita M, Araoka T, Hishida T et al. In vivo reprogramming of wound-resident cells generates skin epithelial tissue. Nature 561(7722), 243–247 (2018).
    • 5. Garcia-Caballero L, Caneiro J, Gandara M et al. Merkel cells of human oral mucosa express the pluripotent stem cell transcription factor Sox2. Histol. Histopathol. doi:10.14670/HH-18-231 18231 (2020) (Epub ahead of print).
    • 6. Hahn JM, Combs KA, Lloyd CM, Mcfarland KL, Boyce ST, Supp DM. Identification of Merkel cells associated with neurons in engineered skin substitutes after grafting to full thickness wounds. PloS ONE 14(3), e0213325 (2019).
    • 7. Heidari F, Yari A, Rasoolijazi H et al. Bulge hair follicle stem cells accelerate cutaneous wound healing in rats. Wounds 28(4), 132–141 (2016).
    • 8. Lim CH, Sun Q, Ratti K et al. Hedgehog stimulates hair follicle neogenesis by creating inductive dermis during murine skin wound healing. Nat. Commun. 9(1), 4903 (2018).
    • 9. Joost S, Jacob T, Sun X et al. Single-cell transcriptomics of traced epidermal and hair follicle stem cells reveals rapid adaptations during wound healing. Cell Rep. 25(3), 585–597 e587 (2018).
    • 10. Ma T, Fu B, Yang X, Xiao Y, Pan M. Adipose mesenchymal stem cell-derived exosomes promote cell proliferation, migration, and inhibit cell apoptosis via Wnt/beta-catenin signaling in cutaneous wound healing. J. Cell. Biochem. 120(6), 10847–10854 (2019).
    • 11. Malara MM, Blackstone BN, Baumann ME, Bailey JK, Supp DM, Powell HM. Cultured epithelial autograft combined with micropatterned dermal template forms rete ridges in vivo. Tissue Eng. Part A doi:10.1089/ten.TEA.2020.0090 (2020) (Epub ahead of print).
    • 12. Blackstone BN, Malara MM, Baumann ME, Mcfarland KL, Supp DM, Powell HM. Fractional CO2 laser micropatterning of cell-seeded electrospun collagen scaffolds enables rete ridge formation in 3D engineered skin. Acta Biomater. 102, 287–297 (2020).
    • 13. Cangkrama M, Wietecha M, Werner S. Wound repair, scar formation, and cancer: converging on activin. Trends Mol. Med. doi:10.1016/j.molmed.2020.07.009 (2020) (Epub ahead of print).
    • 14. Tottoli EM, Dorati R, Genta I, Chiesa E, Pisani S, Conti B. Skin wound healing process and new emerging technologies for skin wound care and regeneration. Pharmaceutics 12(8), 735–765 (2020).
    • 15. Willenborg S, Eming SA. Cellular networks in wound healing. Science 362(6417), 891–892 (2018).
    • 16. Martino MM, Briquez PS, Guc E et al. Growth factors engineered for super-affinity to the extracellular matrix enhance tissue healing. Science 343(6173), 885–888 (2014).
    • 17. Fui LW, Lok MPW, Govindasamy V, Yong TK, Lek TK, Das AK. Understanding the multifaceted mechanisms of diabetic wound healing and therapeutic application of stem cells conditioned medium in the healing process. J. Tissue Eng. Regen. Med. 13(12), 2218–2233 (2019).
    • 18. Golebiewska EM, Poole AW. Platelet secretion: from haemostasis to wound healing and beyond. Blood Rev. 29(3), 153–162 (2015).
    • 19. Burkhardt MA, Waser J, Milleret V et al. Synergistic interactions of blood-borne immune cells, fibroblasts and extracellular matrix drive repair in an in vitro peri-implant wound healing model. Sci. Rep. 6, 21071 (2016).
    • 20. Jayme TS, Leung G, Wang A et al. Human interleukin-4-treated regulatory macrophages promote epithelial wound healing and reduce colitis in a mouse model. Sci. Adv. 6(23), eaba4376 (2020).
    • 21. Naik S, Larsen SB, Gomez NC et al. Inflammatory memory sensitizes skin epithelial stem cells to tissue damage. Nature 550(7677), 475–480 (2017).
    • 22. Tang D, Zhang J, Yan T et al. FG-4592 accelerates cutaneous wound healing by epidermal stem cell activation via HIF-1alpha stabilization. Cell Physiol. Biochem. 46(6), 2460–2470 (2018).
    • 23. Ko UH, Choi J, Choung J, Moon S, Shin JH. Physicochemically tuned myofibroblasts for wound healing strategy. Sci. Rep. 9(1), 16070 (2019).
    • 24. Duscher D, Maan ZN, Whittam AJ et al. Fibroblast-specific deletion of hypoxia inducible factor-1 critically impairs murine cutaneous neovascularization and wound healing. Plast. Reconstr. Surg. 136(5), 1004–1013 (2015).
    • 25. Tracy LE, Minasian RA, Caterson EJ. Extracellular matrix and dermal fibroblast function in the healing wound. Adv. Wound Care (New Rochelle) 5(3), 119–136 (2016).
    • 26. Kechagia JZ, Ivaska J, Roca-Cusachs P. Integrins as biomechanical sensors of the microenvironment. Nat. Rev. Mol. Cell Biol. 20(8), 457–473 (2019).
    • 27. Bellayr IH, Mu X, Li Y. Biochemical insights into the role of matrix metalloproteinases in regeneration: challenges and recent developments. Future Med. Chem. 1(6), 1095–1111 (2009).
    • 28. Wang J, Wang X, Chen H, Liu J, Tredget EE, Wu Y. Distinctively expressed cytokines by three different inflammation cells and their interaction with keratinocytes in wound healing. Inflammation 40(6), 2151–2162 (2017).
    • 29. Akbari A, Li Y, Kilani RT, Ghahary A. Red blood cell lysate modulates the expression of extracellular matrix proteins in dermal fibroblasts. Mol. Cell. Biochem. doi:10.1007/s11010-012-1400-1 (2012) (Epub ahead of print).
    • 30. Etulain J. Platelets in wound healing and regenerative medicine. Platelets 29(6), 556–568 (2018).
    • 31. Cunnion KM, Krishna NK, Pallera HK et al. Complement activation and STAT4 expression are associated with early inflammation in diabetic wounds. PloS ONE 12(1), e0170500 (2017).
    • 32. Kim H, Wang SY, Kwak G, Yang Y, Kwon IC, Kim SH. Exosome-guided phenotypic switch of M1 to M2 macrophages for cutaneous wound healing. Adv. Sci. (Weinh) 6(20), 1900513 (2019).
    • 33. Fu J, Huang J, Lin M, Xie T, You T. Quercetin promotes diabetic wound healing via switching macrophages from M1 to M2 polarization. J. Surg. Res. 246, 213–223 (2020).
    • 34. Sirbulescu RF, Boehm CK, Soon E et al. Mature B cells accelerate wound healing after acute and chronic diabetic skin lesions. Wound Repair Regen. 25(5), 774–791 (2017).
    • 35. Tanno H, Kawakami K, Ritsu M et al. Contribution of invariant natural killer T cells to skin wound healing. Am. J. Pathol. 185(12), 3248–3257 (2015).
    • 36. Nosbaum A, Prevel N, Truong HA et al. Cutting edge: regulatory t cells facilitate cutaneous wound healing. J. Immunol. 196(5), 2010–2014 (2016).
    • 37. Chen L, Mehta ND, Zhao Y, Dipietro LA. Absence of CD4 or CD8 lymphocytes changes infiltration of inflammatory cells and profiles of cytokine expression in skin wounds, but does not impair healing. Exp. Dermatol. 23(3), 189–194 (2014).
    • 38. Li Y, Wang Y, Zhou L et al. Vgamma4 T cells inhibit the pro-healing functions of dendritic epidermal t cells to delay skin wound closure through IL-17A. Front. Immunol. 9, 240 (2018).
    • 39. Sharp LL, Jameson JM, Cauvi G, Havran WL. Dendritic epidermal T cells regulate skin homeostasis through local production of insulin-like growth factor 1. Nat. Immunol. 6(1), 73–79 (2005).
    • 40. Liu Q, Smith CW, Zhang W, Burns AR, Li Z. NK cells modulate the inflammatory response to corneal epithelial abrasion and thereby support wound healing. Am. J. Pathol. 181(2), 452–462 (2012).
    • 41. Lu D, Insel PA. Cellular mechanisms of tissue fibrosis. 6. Purinergic signaling and response in fibroblasts and tissue fibrosis. Am. J. Physiol. Cell Physiol. 306(9), C779–788 (2014).
    • 42. Vorstandlechner V, Laggner M, Kalinina P et al. Deciphering the functional heterogeneity of skin fibroblasts using single-cell RNA sequencing. FASEB J. 34(3), 3677–3692 (2020).
    • 43. Hu X, Zhang H, Li X, Li Y, Chen Z. Activation of mTORC1 in fibroblasts accelerates wound healing and induces fibrosis in mice. Wound Repair Regen. 28(1), 6–15 (2020).
    • 44. Li Y, Huard J. Differentiation of muscle-derived cells into myofibroblasts in injured skeletal muscle. Am. J. Pathol. 161(3), 895–907 (2002).
    • 45. Park JS, Oh Y, Park YJ et al. Targeting of dermal myofibroblasts through death receptor 5 arrests fibrosis in mouse models of scleroderma. Nat. Commun. 10(1), 1128 (2019).
    • 46. Nakagawa T, Sato W, Kosugi T, Johnson RJ. Uncoupling of VEGF with endothelial NO as a potential mechanism for abnormal angiogenesis in the diabetic nephropathy. J. Diabetes Res. 2013, 184539 (2013).
    • 47. Leoni G, Neumann PA, Sumagin R, Denning TL, Nusrat A. Wound repair: role of immune-epithelial interactions. Mucosal Immunol. 8(5), 959–968 (2015).
    • 48. Wojtowicz AM, Oliveira S, Carlson MW, Zawadzka A, Rousseau CF, Baksh D. The importance of both fibroblasts and keratinocytes in a bilayered living cellular construct used in wound healing. Wound Repair Regen. 22(2), 246–255 (2014).
    • 49. Hu X, Yu W, Sun H, Wang X, Han C. Epidermal cells delivered for cutaneous wound healing. J. Dermatolog. Treat. 23(3), 224–237 (2012).
    • 50. Yang R, Wang J, Chen X, Shi Y, Xie J. Epidermal stem cells in wound healing and regeneration. Stem Cells Int. 2020, 9148310 (2020).
    • 51. Balaji S, King A, Crombleholme TM, Keswani SG. The role of endothelial progenitor cells in postnatal vasculogenesis: implications for therapeutic neovascularization and wound healing. Adv. Wound Care (New Rochelle) 2(6), 283–295 (2013).
    • 52. Hall MA, Robinson H, Chan W, Sevick-Muraca EM. Detection of lymphangiogenesis by near-infrared fluorescence imaging and responses to VEGF-C during healing in a mouse full-dermis thickness wound model. Wound Repair Regen. 21(4), 604–615 (2013).
    • 53. Scherber C, Aranyosi AJ, Kulemann B et al. Epithelial cell guidance by self-generated EGF gradients. Integr. Biol. (Camb) 4(3), 259–269 (2012).
    • 54. Hsu SH, Hsieh PS. Self-assembled adult adipose-derived stem cell spheroids combined with biomaterials promote wound healing in a rat skin repair model. Wound Repair Regen. 23(1), 57–64 (2015).
    • 55. Mascre G, Dekoninck S, Drogat B et al. Distinct contribution of stem and progenitor cells to epidermal maintenance. Nature 489(7415), 257–262 (2012).
    • 56. Baker DJ, Childs BG, Durik M et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 530(7589), 184–189 (2016).
    • 57. Yu T, Gao M, Yang P et al. Insulin promotes macrophage phenotype transition through PI3K/Akt and PPAR-gamma signaling during diabetic wound healing. J. Cell. Physiol. 234(4), 4217–4231 (2019).
    • 58. Patel S, Maheshwari A, Chandra A. Biomarkers for wound healing and their evaluation. J. Wound Care 25(1), 46–55 (2016).
    • 59. 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(3–4), 151–159 (2015).
    • 60. Rafail S, Kourtzelis I, Foukas PG et al. Complement deficiency promotes cutaneous wound healing in mice. J. Immunol. 194(3), 1285–1291 (2015).
    • 61. Korkmaz HI, Ulrich MMW, Van Wieringen WN et al. The local and systemic inflammatory response in a pig burn wound model with a pivotal role for complement. J. Burn Care Res. 38(5), e796–e806 (2017).
    • 62. Denzinger M, Held M, Daigeler A, Krajewski S, Link A. Complement activation at the interface of wound dressings and blood does not influence keratinocyte migration/proliferation in vitro. Wound Repair Regen. 28(4), 573–575 (2020).
    • 63. Mccarty SM, Percival SL. Proteases and delayed wound healing. Adv. Wound Care (New Rochelle) 2(8), 438–447 (2013).
    • 64. Gao M, Nguyen TT, Suckow MA et al. Acceleration of diabetic wound healing using a novel protease-anti-protease combination therapy. Proc. Natl Acad. Sci. USA 112(49), 15226–15231 (2015).
    • 65. Lee P, Chandel NS, Simon MC. Cellular adaptation to hypoxia through hypoxia inducible factors and beyond. Nat. Rev. Mol. Cell Biol. 21(5), 268–283 (2020).
    • 66. Ruthenborg RJ, Ban JJ, Wazir A, Takeda N, Kim JW. Regulation of wound healing and fibrosis by hypoxia and hypoxia-inducible factor-1. Mol. Cells 37(9), 637–643 (2014).
    • 67. Kim KJ, Li B, Winer J et al. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362(6423), 841–844 (1993).
    • 68. Ciarlillo D, Celeste C, Carmeliet P, Boerboom D, Theoret C. A hypoxia response element in the vegfa promoter is required for basal vegfa expression in skin and for optimal granulation tissue formation during wound healing in mice. PloS ONE 12(7), e0180586 (2017).
    • 69. Schmid P, Itin P, Cherry G, Bi C, Cox DA. Enhanced expression of transforming growth factor-beta type I and type II receptors in wound granulation tissue and hypertrophic scar. Am. J. Pathol. 152(2), 485–493 (1998).
    • 70. Kirby GT, Mills SJ, Cowin AJ, Smith LE. Stem cells for cutaneous wound healing. Biomed Res. Int. 2015, 285869 (2015).
    • 71. Joo SY, Lee SY, Cho YS, Seo CH. Clinical utility of extracorporeal shock wave therapy on hypertrophic scars of the hand caused by burn injury: a prospective, randomized, double-blinded study. J. Clin. Med. 9(5), 1376–1387 (2020).
    • 72. Berman B, Maderal A, Raphael B. Keloids and hypertrophic scars: pathophysiology, classification, and treatment. Dermatol. Surg. 43(Suppl. 1), S3–S18 (2017).
    • 73. Escuin-Ordinas H, Li S, Xie MW et al. Cutaneous wound healing through paradoxical MAPK activation by BRAF inhibitors. Nat. Commun. 7, 12348 (2016).
    • 74. Mahmoudi S, Mancini E, Xu L et al. Heterogeneity in old fibroblasts is linked to variability in reprogramming and wound healing. Nature 574(7779), 553–558 (2019).
    • 75. Keyes BE, Liu S, Asare A et al. Impaired epidermal to dendritic t cell signaling slows wound repair in aged skin. Cell 167(5), 1323–1338 e1314 (2016).
    • 76. Pastar I, Wong LL, Egger AN, Tomic-Canic M. Descriptive vs mechanistic scientific approach to study wound healing and its inhibition: is there a value of translational research involving human subjects? Exp. Dermatol. 27(5), 551–562 (2018).
    • 77. Hoang DH, Nguyen TD, Nguyen HP et al. Differential wound healing capacity of mesenchymal stem cell-derived exosomes originated from bone marrow, adipose tissue and umbilical cord under serum- and xeno-free condition. Front. Mol. Biosci. 7, 119 (2020).
    • 78. Stejskalova A, Almquist BD. Using biomaterials to rewire the process of wound repair. Biomater. Sci. 5(8), 1421–1434 (2017).
    • 79. Dimatteo R, Darling NJ, Segura T. In situ forming injectable hydrogels for drug delivery and wound repair. Adv. Drug Deliv. Rev. 127, 167–184 (2018).
    • 80. Hersant B, Sid-Ahmed M, Braud L et al. Platelet-rich plasma improves the wound healing potential of mesenchymal stem cells through paracrine and metabolism alterations. Stem Cells Int. 2019, 1234263 (2019).
    • 81. Zhang L, Zhang B, Liao B et al. Platelet-rich plasma in combination with adipose-derived stem cells promotes skin wound healing through activating Rho GTPase-mediated signaling pathway. Am. J. Transl. Res. 11(7), 4100–4112 (2019).
    • 82. Verma R, Negi G, Kandwal A, Chandra H, Gaur DS, Harsh M. Effect of autologous PRP on wound healing in dental regenerative surgeries and its correlation with PDGF levels. Asian J. Transfus. Sci. 13(1), 47–53 (2019).
    • 83. Jaseem M, Alungal S, Dhiyaneswaran Shamsudeen J. Effectiveness of autologous PRP therapy in chronic nonhealing ulcer: a 2-year retrospective descriptive study. J. Family Med. Prim. Care 9(6), 2818–2822 (2020).
    • 84. Lei X, Yang Y, Shan G, Pan Y, Cheng B. Preparation of ADM/PRP freeze-dried dressing and effect of mice full-thickness skin defect model. Biomed. Mater. 14(3), 035004 (2019).
    • 85. Huang Z, Wu S, Yu T, Hu A. Efficacy of telemedicine for patients with chronic wounds: a meta-analysis of randomized controlled trials. Adv. Wound Care (New Rochelle) doi:10.1089/wound.2020.1169 (2020) (Epub ahead of print).
    • 86. Teot L, Geri C, Lano J, Cabrol M, Linet C, Mercier G. Complex wound healing outcomes for outpatients receiving care via telemedicine, home health, or wound clinic: a randomized controlled trial. Int. J. Low. Extrem. Wounds 19(2), 197–204 (2020).
    • 87. Frear CC, Griffin B, Cuttle L, Mcphail SM, Kimble R. Study of negative pressure wound therapy as an adjunct treatment for acute burns in children (SONATA in C): protocol for a randomised controlled trial. Trials 20(1), 130 (2019).
    • 88. Li Y, Jiang H, Zheng W et al. Bacterial cellulose-hyaluronan nanocomposite biomaterials as wound dressings for severe skin injury repair. J. Mater. Chem. B 3(17), 3498–3507 (2015).
    • 89. Percival SL, Francolini I, Donelli G. Low-level laser therapy as an antimicrobial and antibiofilm technology and its relevance to wound healing. Future Microbiol. 10(2), 255–272 (2015).
    • 90. Spitler R, Ho H, Norpetlian F et al. Combination of low level light therapy and nitrosyl-cobinamide accelerates wound healing. J. Biomed. Opt. 20(5), 051022 (2015).
    • 91. Wang S, Zheng H, Zhou L et al. Injectable redox and light responsive MnO2 hybrid hydrogel for simultaneous melanoma therapy and multidrug-resistant bacteria-infected wound healing. Biomaterials 260, 120314 (2020).
    • 92. Yang N, Zhu M, Xu G, Liu N, Yu C. A near-infrared light-responsive multifunctional nanocomposite hydrogel for efficient and synergistic antibacterial wound therapy and healing promotion. J. Mater. Chem. B 8(17), 3908–3917 (2020).
    • 93. Pan BH, Zhang Q, Lam CH, Yin Yuen H, Kuang S, Zhao X. Petite miracles: insight into the nano-management of scarless wound healing. Drug Discov. Today doi:10.1016/j.drudis.2020.03.013 (2020) (Epub ahead of print).
    • 94. Haalboom M, Blokhuis-Arkes MHE, Beuk RJ et al. Culture results from wound biopsy versus wound swab: does it matter for the assessment of wound infection? Clin. Microbiol. Infect. 25(5), 629e627–629e612 (2019).
    • 95. Wietecha MS, Pensalfini M, Cangkrama M et al. Activin-mediated alterations of the fibroblast transcriptome and matrisome control the biomechanical properties of skin wounds. Nat. Commun. 11(1), 2604 (2020).
    • 96. Guerrero-Juarez CF, Dedhia PH, Jin S et al. Single-cell analysis reveals fibroblast heterogeneity and myeloid-derived adipocyte progenitors in murine skin wounds. Nat. Commun. 10(1), 650 (2019).