Cellular and molecular mechanisms of repair in acute and chronic wound healing

P Martin, R Nunan, P Martin, R Nunan

Abstract

A considerable understanding of the fundamental cellular and molecular mechanisms underpinning healthy acute wound healing has been gleaned from studying various animal models, and we are now unravelling the mechanisms that lead to chronic wounds and pathological healing including fibrosis. A small cut will normally heal in days through tight orchestration of cell migration and appropriate levels of inflammation, innervation and angiogenesis. Major surgeries may take several weeks to heal and leave behind a noticeable scar. At the extreme end, chronic wounds - defined as a barrier defect that has not healed in 3 months - have become a major therapeutic challenge throughout the Western world and will only increase as our populations advance in age, and with the increasing incidence of diabetes, obesity and vascular disorders. Here we describe the clinical problems and how, through better dialogue between basic researchers and clinicians, we may extend our current knowledge to enable the development of novel potential therapeutic treatments.

© 2015 British Association of Dermatologists.

Figures

Figure 1
Figure 1
Acute wound healing mechanisms. The healing of an acute wound involves coordinated cellular and molecular responses. (a) Initially immune cells migrate to the wound site and, in addition to clearing invading pathogens, in part they also orchestrate the healing process. (b) Cut epidermal edges upregulate wound-associated genes, thus enabling collective cell migration. (c) Local and blood-borne fibroblasts proliferate and migrate to form the wound granulation tissue, provide structure and signalling cues and deposit new extracellular matrix (ECM). Some fibroblasts differentiate into myofibroblasts to aid wound contraction. (d) The wound bed is perfused with oxygen and nutrients through new blood vessels derived by angiogenesis. (e) Wound healing rates exhibit a positive correlation with innervation, but hyperinnervation after wound closure could contribute to neuropathic pain. EGF, epidermal growth factor; HGF, hepatocyte growth factor; FGF, fibroblast growth factor; KGF, keratinocyte growth factor; MSC, mesenchymal stem cell; nAG, newt anterior gradient protein.
Figure 2
Figure 2
Chronic wound biology. Chronic wounds are often infected and exhibit a persistent aberrant inflammatory profile. Re-epithelialization stalls but wound keratinocytes are hyperproliferative. Granulation tissue is defective and does not nurture healing, in part due to elevated matrix metalloproteases (MMPs) and poor fibroblast infiltration. Neoangiogenesis is poor and fibrin cuffs restrict existing vessels, limiting the diffusion of oxygen through the wound, rendering the wound hypoxic.
Figure 3
Figure 3
Excessive fibrosis. Scars, formed in part as a consequence of inflammatory signals, are comprised of collagen deposited in thick orientated bundles rather than the basket-weave-like fibrils found in normal dermis. Hypertrophic scars have excessive collagen deposition, leading to a raised surface that partially resolves over time. In contrast, keloid scars have thicker collagen bundles, extend beyond the original wound margin and rarely regress. Contractile myofibroblasts are prevalent in hypertrophic scarring but all but are absent in keloid tissue. Keloids can also be characterized by occluded blood vessels. OPN, osteopontin; TGF-β, transforming growth factor-β.

References

    1. Schultz GS, White M, Mitchell R, et al. Epithelial wound healing enhanced by transforming growth factor-alpha and vaccinia growth factor. Science. 1987;235:350–2.
    1. Grose R, Werner S. Wound-healing studies in transgenic and knockout mice. Mol Biotechnol. 2004;28:147–66.
    1. Cooper L, Johnson C, Burslem F, Martin P. Wound healing and inflammation genes revealed by array analysis of ‘macrophageless’ PU.1 null mice. Genome Biol. 2005;6:R5.
    1. Pedersen TX, Leethanakul C, Patel V, et al. Laser capture microdissection-based in vivo genomic profiling of wound keratinocytes identifies similarities and differences to squamous cell carcinoma. Oncogene. 2003;22:3964–76.
    1. Paladini RD, Takahashi K, Bravo NS, Coulombe PA. Onset of re-epithelialization after skin injury correlates with a reorganization of keratin filaments in wound edge keratinocytes: defining a potential role for keratin 16. J Cell Biol. 1996;132:381–97.
    1. Martin P, Nobes CD. An early molecular component of the wound healing response in rat embryos – induction of c-fos protein in cells at the epidermal wound margin. Mech Dev. 1992;38:209–15.
    1. Grose R, Harris BS, Cooper L, et al. Immediate early genes krox-24 and krox-20 are rapidly up-regulated after wounding in the embryonic and adult mouse. Dev Dyn. 2002;223:371–8.
    1. Shaw T, Martin P. Epigenetic reprogramming during wound healing: loss of polycomb-mediated silencing may enable upregulation of repair genes. EMBO Rep. 2009;10:881–6.
    1. Grose R, Hutter C, Bloch W. >et al A crucial role of β1 integrins for keratinocyte migration in vitro and during cutaneous wound repair. Development. 2002;129:2303–15.
    1. Thomason HA, Cooper NH, Ansell DM, et al. Direct evidence that PKCα positively regulates wound re-epithelialization: correlation with changes in desmosomal adhesiveness. J Pathol. 2012;227:346–56.
    1. Gill SE, Parks WC. Metalloproteinases and their inhibitors: regulators of wound healing. Int J Biochem Cell Biol. 2008;40:1334–47.
    1. Chmielowiec J, Borowiak M, Morkel M, et al. c-Met is essential for wound healing in the skin. J Cell Biol. 2007;177:151–62.
    1. Meyer M, Müller AK, Yang J, et al. FGF receptors 1 and 2 are key regulators of keratinocyte migration in vitro and in wounded skin. J Cell Sci. 2012;125:5690–701.
    1. Repertinger SK, Campagnaro E, Fuhrman J, et al. EGFR enhances early healing after cutaneous incisional wounding. J Invest Dermatol. 2004;123:982–9.
    1. Werner S, Smola H, Liao X, et al. The function of KGF in morphogenesis of epithelium and reepithelialization of wounds. Science. 1994;266:819–22.
    1. Ito M, Liu Y, Yang Z, et al. Stem cells in the hair follicle bulge contribute to wound repair but not to homeostasis of the epidermis. Nat Med. 2005;11:1351–4.
    1. Levy V, Lindon C, Zheng Y, et al. Epidermal stem cells arise from the hair follicle after wounding. FASEB J. 2007;21:1358–66.
    1. Ito M, Yang Z, Andl T, et al. Wnt-dependent de novo hair follicle regeneration in adult mouse skin after wounding. Nature. 2007;447:316–20.
    1. Desmouliere A, Geinoz A, Gabbiani F, Gabbiani G. Transforming growth factor-β1 induces α-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J Cell Biol. 1993;122:103–11.
    1. Fathke C, Wilson L, Hutter J, et al. Contribution of bone marrow-derived cells to skin: collagen deposition and wound repair. Stem Cells. 2004;22:812–22.
    1. Ishii G, Sangai T, Sugiyama K, et al. In vivo characterization of bone marrow-derived fibroblasts recruited into fibrotic lesions. Stem Cells. 2005;23:699–706.
    1. Sasaki M, Abe R, Fujita Y, et al. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol. 2008;180:2581–7.
    1. Driskell RR, Lichtenberger BM, Hoste E, et al. Distinct fibroblast lineages determine dermal architecture in skin development and repair. Nature. 2013;504:277–81.
    1. Hopkinson-Woolley J, Hughes D, Gordon S, Martin P. Macrophage recruitment during limb development and wound healing in the embryonic and foetal mouse. J Cell Sci. 1994;107:1159–67.
    1. Adzick NS, Harrison MR, Glick PL, et al. Comparison of fetal, newborn, and adult wound healing by histologic, enzyme-histochemical, and hydroxyproline determinations. J Pediatr Surg. 1985;20:315–19.
    1. Martin P, D'Souza D, Martin J, et al. Wound healing in the PU.1 null mouse – tissue repair is not dependent on inflammatory cells. Curr Biol. 2003;13:1122–8.
    1. Dovi JV, He LK, DiPietro LA. Accelerated wound closure in neutrophil-depleted mice. J Leukoc Biol. 2003;73:448–55.
    1. Lucas T, Waisman A, Ranjan R, et al. Differential roles of macrophages in diverse phases of skin repair. J Immunol. 2010;184:3964–77.
    1. Antsiferova M, Martin C, Huber M, et al. Mast cells are dispensable for normal and activin-promoted wound healing and skin carcinogenesis. J Immunol. 2013;191:6147–55.
    1. Willenborg S, Eckes B, Brinckmann J, et al. Genetic ablation of mast cells redefines the role of mast cells in skin wound healing and bleomycin-induced fibrosis. J Invest Dermatol. 2014;134:2005–15.
    1. Jameson J, Ugarte K, Chen N, et al. A role for skin γδ T cells in wound repair. Science. 2002;296:747–9.
    1. Deppermann C, Cherpokova D, Nurden P, et al. Gray platelet syndrome and defective thrombo-inflammation in Nbeal2-deficient mice. J Clin Invest. 2015 ; doi: .
    1. Bianchi ME, Manfredi AA. Immunology. Dangers in and out. Science. 2009;323:1683–4.
    1. Wong VW, Rustad KC, Akaishi S, et al. Focal adhesion kinase links mechanical force to skin fibrosis via inflammatory signaling. Nat Med. 2012;18:148–52.
    1. Cash JL, Bass MD, Campbell J, et al. Resolution mediator chemerin15 reprograms the wound microenvironment to promote repair and reduce scarring. Curr Biol. 2014;24:1406–14.
    1. Ferguson MW, O'Kane S. Scar-free healing: from embryonic mechanisms to adult therapeutic intervention. Philos Trans R Soc Lond B Biol Sci. 2004;359:839–50.
    1. Mori R, Shaw TJ, Martin P. Molecular mechanisms linking wound inflammation and fibrosis: knockdown of osteopontin leads to rapid repair and reduced scarring. J Exp Med. 2008;205:43–51.
    1. Gerhardt H, Golding M, Fruttiger M, et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J Cell Biol. 2003;161:1163–77.
    1. Rossiter H, Barresi C, Pammer J, et al. Loss of vascular endothelial growth factor A activity in murine epidermal keratinocytes delays wound healing and inhibits tumor formation. Cancer Res. 2004;64:3508–16.
    1. Fantin A, Vieira JM, Gestri G, et al. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood. 2010;116:829–40.
    1. Wallace H. The response of denervated axolotl arms to delayed amputation. J Embryol Exp Morphol. 1984;84:303–7.
    1. Kumar A, Brockes JP. Nerve dependence in tissue, organ, and appendage regeneration. Trends Neurosci. 2012;35:691–9.
    1. Harsum S, Clarke JD, Martin P. A reciprocal relationship between cutaneous nerves and repairing skin wounds in the developing chick embryo. Dev Biol. 2001;238:27–39.
    1. Razzell W, Wood W, Martin P. Swatting flies: modelling wound healing and inflammation in Drosophila. Dis Model Mech. 2011;4:569–74.
    1. Henry KM, Loynes CA, Whyte MK, Renshaw SA. Zebrafish as a model for the study of neutrophil biology. J Leukoc Biol. 2013;94:633–42.
    1. Stramer B, Wood W, Galko MJ, et al. Live imaging of wound inflammation in Drosophila embryos reveals key roles for small GTPases during in vivo cell migration. J Cell Biol. 2005;168:567–73.
    1. Wood W, Jacinto A, Grose R, et al. Wound healing recapitulates morphogenesis in Drosophila embryos. Nat Cell Biol. 2002;4:907–12.
    1. Razzell W, Wood W, Martin P. Recapitulation of morphogenetic cell shape changes enables wound re-epithelialisation. Development. 2014;141:1814–20.
    1. Lesch C, Jo J, Wu Y, et al. A targeted UAS-RNAi screen in Drosophila larvae identifies wound closure genes regulating distinct cellular processes. Genetics. 2010;186:943–57.
    1. Campos I, Geiger JA, Santos AC, et al. Genetic screen in Drosophila melanogaster uncovers a novel set of genes required for embryonic epithelial repair. Genetics. 2010;184:129–40.
    1. Mace KA, Pearson JC, McGinnis W. An epidermal barrier wound repair pathway in Drosophila is mediated by grainy head. Science. 2005;308:381–5.
    1. Ting SB, Caddy J, Hislop N, et al. A homolog of Drosophila grainy head is essential for epidermal integrity in mice. Science. 2005;308:411–13.
    1. Caddy J, Wilanowski T, Darido C, et al. Epidermal wound repair is regulated by the planar cell polarity signaling pathway. Dev Cell. 2010;19:138–47.
    1. Niethammer P, Grabher C, Look AT, Mitchison TJ. A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature. 2009;459:996–9.
    1. Pase L, Layton JE, Wittmann C, et al. Neutrophil-delivered myeloperoxidase dampens the hydrogen peroxide burst after tissue wounding in zebrafish. Curr Biol. 2012;22:1818–24.
    1. Richardson R, Slanchev K, Kraus C, et al. Adult zebrafish as a model system for cutaneous wound-healing research. J Invest Dermatol. 2013;133:1655–65.
    1. Herrick SE, Sloan P, McGurk M, et al. Sequential changes in histologic pattern and extracellular matrix deposition during the healing of chronic venous ulcers. Am J Pathol. 1992;141:1085–95.
    1. Stojadinovic O, Pastar I, Vukelic S, et al. Deregulation of keratinocyte differentiation and activation: a hallmark of venous ulcers. J Cell Mol Med. 2008;12:2675–90.
    1. Brem H, Stojadinovic O, Diegelmann RF, et al. Molecular markers in patients with chronic wounds to guide surgical debridement. Mol Med. 2007;13:30–9.
    1. Pastar I, Stojadinovic O, Krzyzanowska A, et al. Attenuation of the transforming growth factor β-signaling pathway in chronic venous ulcers. Mol Med. 2010;16:92–101.
    1. Trengove NJ, Stacey MC, MacAuley S, et al. Analysis of the acute and chronic wound environments: the role of proteases and their inhibitors. Wound Repair Regen. 1999;7:442–52.
    1. Charles CA, Tomic-Canic M, Vincek V, et al. A gene signature of nonhealing venous ulcers: potential diagnostic markers. J Am Acad Dermatol. 2008;59:758–71.
    1. Loots MA, Lamme EN, Zeegelaar J, et al. Differences in cellular infiltrate and extracellular matrix of chronic diabetic and venous ulcers versus acute wounds. J Invest Dermatol. 1998;111:850–7.
    1. Stojadinovic O, Yin N, Lehmann J, et al. Increased number of Langerhans cells in the epidermis of diabetic foot ulcers correlates with healing outcome. Immunol Res. 2013;57:222–8.
    1. Naghibi M, Smith RP, Baltch AL, et al. The effect of diabetes mellitus on chemotactic and bactericidal activity of human polymorphonuclear leukocytes. Diabetes Res Clin Pract. 1987;4:27–35.
    1. Gottrup F, Jorgensen B. Maggot debridement: an alternative method for debridement. Eplasty. 2011;11:e33.
    1. Smith C, Kavar B. Extensive spinal epidural abscess as a complication of Crohn's disease. J Clin Neurosci. 2010;17:144–6.
    1. Grice EA, Segre JA. Interaction of the microbiome with the innate immune response in chronic wounds. Adv Exp Med Biol. 2012;946:55–68.
    1. Nunan R, Harding KG, Martin P. Clinical challenges of chronic wounds: searching for an optimal animal model to recapitulate their complexity. Dis Model Mech. 2014;7:1205–13.
    1. Lorenz HP, Longaker MT, Perkocha LA, et al. Scarless wound repair: a human fetal skin model. Development. 1992;114:253–9.
    1. Shih B, Bayat A. Genetics of keloid scarring. Arch Dermatol Res. 2010;302:319–39.
    1. Mogili NS, Krishnaswamy VR, Jayaraman M, et al. Altered angiogenic balance in keloids: a key to therapeutic intervention. Transl Res. 2012;159:182–9.
    1. Canady J, Karrer S, Fleck M, Bosserhoff AK. Fibrosing connective tissue disorders of the skin: molecular similarities and distinctions. J Dermatol Sci. 2013;70:151–8.
    1. Reish RG, Eriksson E. Scars: a review of emerging and currently available therapies. Plast Reconstr Surg. 2008;122:1068–78.
    1. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18:1028–40.
    1. Friedman SL, Sheppard D, Duffield JS, Violette S. Therapy for fibrotic diseases: nearing the starting line. Sci Transl Med. 2013;5:167sr1.
    1. Bosanquet DC, Harding KG. Wound duration and healing rates: cause or effect? Wound Repair Regen. 2014;22:143–50.

Source: PubMed

3
Se inscrever