377
Views
21
CrossRef citations to date
0
Altmetric
Review

The regenerative potential of skin and the immune system

, , , , , & show all
Pages 519-532 | Published online: 15 Jul 2019

References

  • Gonzales KAU, Fuchs E. Skin and its regenerative powers: an alliance between stem cells and their niche. Dev Cell. 2017;43(4):387–401. doi:10.1016/j.devcel.2017.10.00129161590
  • Streilein JW. Lymphocyte traffic, T-cell malignancies and the skin. J Invest Dermatol. 1978;71(3):167–171. doi:10.1111/1523-1747.ep1254707129071
  • Cogen AL, Nizet V, Gallo RL. Skin microbiota: a source of disease or defence? Br J Dermatol. 2008;158(3):442–455. doi:10.1111/j.1365-2133.2008.08437.x18275522
  • Grice EA, Segre JA. The skin microbiome. Nat Rev Microbiol. 2011;9(4):244. doi:10.1038/nrmicro253721407241
  • Matejuk MA. Skin Immunity. Arch Immunol Ther Exp (Warsz). 2017;66(1):45–54. doi:10.1007/s00005-017-0477-328623375
  • de Koning HD, Simon A, Zeeuwen PLJM, Schalkwijk J. Pattern recognition receptors in infectious skin diseases. Microbes Infect. 2012;14(11):881–893. doi:10.1016/j.micinf.2012.03.00422516809
  • Janeway CA Jr, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20(1):197–216. doi:10.1146/annurev.immunol.20.083001.08435911861602
  • Friedenstein AJ, Gorskaja JF, Kulagina NN. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol. 1976;4(5):267–274.976387
  • Bautista-Hernández LA, Gómez-Olivares JL, Buentello-Volante B, Bautista-de Lucio VM. Fibroblasts: the unknown sentinels eliciting immune responses against microorganisms. Eur J Microbiol Immunol (Bp). 2017:7(3):151–157. Published 2017 Aug 19. doi:10.1556/1886.2017.00009.29034104
  • Huang Q, Fei J, Yu HJ, Gou YB, Huang XK. Effects of human β-defensin-3 on biofilm formation-regulating genes dltB and icaA in Staphylococcus aureus. Mol Med Resp. 2014;10:825–831.
  • Mahanonda R, SardAIam N, Montreekachon P, et al. IL-8 and IDO expression by human gingival fibroblasts via TLRs. J Immunol. 2007;178(2):1157. doi:10.4049/jimmunol.178.2.1151
  • Kirker KR, James GA, Fleckman P, Orelund JE, Stewart PS. Differential effects of planktonic and biofilm MRSA on human fibroblasts. Wound Repair Regen. 2012;20:253–261. doi:10.1111/j.1524-475X.2012.00769.x22332802
  • Stappers MHT, Thys Y, Oosting M, et al. TLR1, TLR2, and TLR6 gene polymorphisms are associated with increased susceptibility to complicated skin and skin structure infections. J Infect Dis. 2014;210(2):311–318. doi:10.1093/infdis/jiu08024511099
  • Dominici MLBK, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International society for cellular therapy position statement. Cytotherapy. 2006;8(4):315–317. doi:10.1080/1465324060085590516923606
  • Janson D, Rietveld M, Mahé C, Saintigny G, El Ghalbzouri A. Differential effect of extracellular matrix derived from papillary and reticular fibroblasts on epidermal development in vitro. Eur J Dermatol. 2017;27(3):237–246. doi:10.1684/ejd.2017.298428524059
  • Ma T, Wang X, Jiang D. Immune tolerance of mesenchymal stem cells and induction of skin allograft tolerance. Curr Stem Cell Res Ther. 2017;12(5):409–415. doi:10.2174/1574888x1266617030112274428260519
  • Kühbacher A, Henkel H, Stevens P, et al. Dermal fibroblasts play a central role in skin model protection against C. albicans invasion. J Infect Dis. 2017;215. doi:10.1093/infdis/jix153.
  • Wang X, Cheng Q, Li L, et al. Toll-like receptors 2 and 4 mediate the capacity of mesenchymal stromal cells to support the proliferation and differentiation of CD34+ cells. Exp Cell Res. 2012;318(3):196–206. doi:10.1016/j.yexcr.2011.11.00122100911
  • Jackson MV, Morrison TJ, Doherty DF, et al. Mitochondrial transfer via tunneling nanotubes is an important mechanism by which mesenchymal stem cells enhance macrophage phagocytosis in the in vitro and in vivo models of ARDS. Stem Cells. 2016;34(8):2210–2223. doi:10.1002/stem.237227059413
  • Rocher BD, Mencalha AL, Gomes BE, Abdelhay E. Mesenchymal stromal cells impair the differentiation of CD14++ CD16− CD64+ classical monocytes into CD14++ CD16+ CD64++ activate monocytes. Cytotherapy. 2012;14(1):12–25. doi:10.3109/14653249.2011.59479221838603
  • Abumaree MH, Al Jumah MA, Kalionis B, et al. Human placental mesenchymal stem cells (pMSCs) play a role as immune suppressive cells by shifting macrophage differentiation from inflammatory M1 to anti-inflammatory M2 macrophages. Stem Cell Rev Rep. 2013;9(5):620–641. doi:10.1007/s12015-013-9455-2
  • Mei SHJ, Haitsma JJ, Dos Santos CC, et al. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. Am J Respir Crit Care Med. 2010;182(8):1047–1057. PMID: 20558630. doi:10.1164/rccm.201001-0010OC.20558630
  • Burr SP, Dazzi F, Garden OA. Mesenchymal stromal cells and regulatory T cells: the Yin and Yang of peripheral tolerance? Immunol Cell Biol. 2013;91(1):12–18. doi:10.1038/icb.2012.6023146942
  • Asadi M, Farokhi F, Delirezh N, Ganji Bakhsh M, Nejati V, Golami K. Fibroblast and T cells conditioned media induce maturation dendritic cell and promote T helper immune response. Veterinary research forum : an international quarterly journal 2012;3(2):111–118.
  • English K, Barry FP, Mahon BP. Murine mesenchymal stem cells suppress dendritic cell migration, maturation and antigen presentation. Immunol Lett. 2008;115(1):50–58. doi:10.1016/j.imlet.2007.10.00218022251
  • Gay D, Kwon O, Zhang Z, et al Fgf9 from dermal γδ T cells induces hair follicle neogenesis after wounding. Nat Med. 2013;19(7):916–923. doi:10.1038/nm.318123727932
  • Kashem SW, Haniffa M, Kaplan DH. Antigen-presenting cells in the skin. Annu Rev Immunol. 2017;35:469–499. doi:10.1146/annurev-immunol-051116-05221528226228
  • Dos Santos VG, Orfali RL, de Oliveira Titz T, Da Silva Duarte AJ, Sato MN, Aoki V. Evidence of regulatory myeloid dendritic cells and circulating inflammatory epidermal dendritic cells‐like modulated by Toll‐like receptors 2 and 7/8 in adults with atopic dermatitis. Int J Dermatol. 2017;56(6):630–635. doi:10.1111/ijd.1353728083892
  • Kyewski BA, Momburg F, Schirrmacher V. Phenotype of stromal cell‐associated thymocytes in situ is compatible with selection of the T cell repertoire at an “immature” stage of thymic T cell differentiation. Eur J Immunol. 1987;17(7):961–967. doi:10.1002/eji.18301707113111861
  • Khasawneh A, Baráth S, Medgyesi B, et al. Myeloid but not plasmacytoid blood DCs possess Th1 polarizing and Th1/Th17 recruiting capacity in psoriasis. Immunol Lett. 2017;189:109–113. doi:10.1016/j.imlet.2017.04.00528414181
  • Han Z, Chen Y, Zhang Y, et al. MiR‐21/PTEN axis promotes skin wound healing by dendritic cells enhancement. J Cell Biochem. 2017;118(10):3511–3519. doi:10.1002/jcb.2602628374893
  • Gregorio J, Meller S, Conrad C, et al. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J Exp Med. 2010;207(13):2921–2930. doi:10.1084/jem.2010110221115688
  • Jacquemin C, Rambert J, Guillet S, et al. Heat shock protein 70 potentiates interferon alpha production by plasmacytoid dendritic cells: relevance for cutaneous lupus and vitiligo pathogenesis. Br J Dermatol. 2017;177(5):1367–1375. doi:10.1111/bjd.1555028380264
  • Gallucci RM, Sloan DK, Heck JM, Murray AR, O’Dell SJ. Interleukin 6 indirectly induces keratinocyte migration. J Invest Dermatol. 2004;122:764–772. doi:10.1111/j.0022-202X.2004.22323.x15086564
  • Hoeffel G, Wang Y, Greter M, et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J Exp Med. 2012;209(6):1167–1181. doi:10.1084/jem.2012034022565823
  • Minutti CM, Knipper JA, Allen JE, Zaiss DMW. Tissue-specific contribution of macrophages to wound healing. Semin Cell Dev Biol. 2017;61:3–11. ISSN 1084-9521. doi:10.1016/j.semcdb.2016.08.00627521521
  • Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677–686. doi:10.1016/j.it.2004.09.01515530839
  • Gerber JS, Mosser DM. Reversing lipopolysaccharide toxicity by ligating the macrophage Fcγ receptors. J Immunol. 2001;166(11):6861–6868. doi:10.4049/jimmunol.166.11.686111359846
  • Knipper JA, Willenborg S, Brinckmann J, et al. Interleukin-4 receptor α signaling in myeloid cells controls collagen fibril assembly in skin repair. Immunity. 2015;43(4):803–816. doi:10.1016/j.immuni.2015.09.00526474656
  • Lucas T, Waisman A, Ranjan R, et al. Differential roles of macrophages in diverse phases of skin repair. J.Immunol. 2010;184(7):3964–3977. doi:10.4049/jimmunol.090335620176743
  • Willenborg S, Lucas T, vanLoo G, et al. CCR2 recruits an inflammatory macrophage subpopulation critical for angiogenesis in tissue repair. Blood. 2012;120(3):613–625. doi:10.1182/blood-2012-01-40338622577176
  • Wilgus TA, Roy S, McDaniel JC. Neutrophils and wound repair: positive actions and negative reactions. Adv Wound Care. 2013;2(7):379–388. doi:10.1089/wound.2012.0383
  • Gutiérrez-Fernández, Inada M, Balbin M, et al. Increased inflammation delays wound healing in mice deficient in collagenase-2 (MMP-8). FASEB J. 2017;21(10):2580–2591. doi:10.1096/fj.06-7860com
  • Skrzeczynska-Moncznik J, Zabieglo K, Bossowski JP, et al. Eosinophils regulate interferon alpha production in plasmacytoid dendritic cells stimulated with components of neutrophil extracellular traps. Int J Interferon Cytokine Res. 2017;37(3):119–128. doi:10.1089/jir.2016.0036
  • Lamback EB, Resende FSS, Lenzi TCR. Eosinophilic fasciitis. An Bras Dermatol. 2016;91(5):57–59. doi:10.1590/abd1806-4841.2016468328300895
  • Otsuka A, Kabashima K. Contribution of basophils to cutaneous immune reactions and Th2-mediated allergic responses. Front Immunol. 2015;6:393. doi:10.3389/fimmu.2015.0039326284076
  • Larouche J, Sheoran S, Maruyama K, Martino MM. Immune regulation of skin wound healing: mechanisms and novel therapeutic targets. Adv Wound Care. 2018;7(7):209–231. doi:10.1089/wound.2017.0761
  • Schwartz C, Eberle JU, Voehringer D. Basophils in inflammation. Eur J Pharmacol. 2016;778:90–95. doi:10.1016/j.ejphar.2015.04.04925959388
  • Voehringer D. Recent advances in understanding basophil functions in vivo. F1000Res. 2017;6:1464 Published 2017 Aug 15. doi:10.12688/f1000research.11697.128868143
  • Cardamone C, Parente R, Feo GD, Triggiani M. Mast cells as effector cells of innate immunity and regulators of adaptive immunity. Immunol Lett. 2016;178:10–14. doi:10.1016/j.imlet.2016.07.00327393494
  • Igawa S, Nardo AD. Skin microbiome and mast cells. Transl Res. 2017;184:68–76. doi:10.1016/j.trsl.2017.03.00328390799
  • Bonefeld CM, Geisler C. The role of innate lymphoid cells in healthy and inflamed skin. Immunol Lett. 2016;179:25–28. doi:10.1016/j.imlet.2016.01.00526794088
  • Levin C, Bonduelle O, Nuttens C, et al. Critical role for skin-derived migratory DCs and Langerhans cells in TFH and GC responses after intradermal immunization. J Invest Dermatol. 2017;137(9):1905–1913. doi:10.1016/j.jid.2017.04.01628457909
  • Godfrey DI, Pellicci DG, Smyth MJ. The elusive NKT cell antigen–is the search over? Science. 2004;306(5702):1687–1689. doi:10.1126/science.110693215576595
  • Linsen L, Somers V, Stinissen P. Immunoregulation of autoimmunity by natural killer T cells. Hum Immunol. 2005;66(12):1193–1202. doi:10.1016/j.humimm.2006.02.02016690406
  • Koupenova M, Vitseva O, MacKay CR, et al. “Platelet-TLR7 mediates host survival and platelet count during viral infection in the absence of platelet-dependent thrombosis. Blood. 2014; blood-2013. doi:10.1182/blood-2013-11-536003.
  • Zufferey A, Schvartz D, Nolli S, Reny J-L, Sanchez J-C, Fontana P. Characterization of the platelet granule proteome: evidence of the presence of MHC1 in alpha-granules. J Proteomics. 2014;101:130–140. doi:10.1016/j.jprot.2014.02.00824549006
  • Wagner DD, Frenette PS. The vessel wall and its interactions. Blood. 2008;111(11):5271–5281. doi:10.1182/blood-2008-01-07820418502843
  • Hartwig H, Drechsler M, Lievens D, et al. Platelet-derived PF4 reduces neutrophil apoptosis following arterial occlusion. Thromb Haemost. 2014;111(3):562–564. doi:10.1160/TH13-08-0699
  • Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-Canic M. Growth factors and cytokines in wound healing. Wound Repair Regen. 2008;16(5):585–601. doi:10.1111/j.1524-475x.2008.00410.x19128254
  • Bayer A, Lammel J, Lippross S, et al. Platelet-released growth factors induce psoriasin in keratinocytes: implications for the cutaneous barrier. Ann Anat Anz. 2017;213:25–32. doi:10.1016/j.aanat.2017.04.002
  • Chicharro-Alcántara D, Rubio-Zaragoza M, Damiá-Giménez E, et al. Platelet rich plasma: new insights for cutaneous wound healing management. J Funct Biomater. 2018;9(1):10 Published 2018 Jan 18. doi:10.3390/jfb9010010
  • Mann ER, Smith KM, Bernardo D, Al-Hassi HO, Knight SC, Hart AL. Review: Skin and the Immune System. J Clin Exp Dermatol Res 2012;S2:003. doi:10.4172/2155-9554.S2-003
  • Luger TA, Kock A, Kirnbauer R, Schwarz T, Ansel JC. Keratinocyte‐Derived Interleukin 3. Ann N Y Acad Sci. 1988;548(1):253–261. doi:10.1111/j.1749-6632.1988.tb18813.x2470298
  • 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. 2014;22(2):246–255. doi:10.1111/wrr.1215424635175
  • Auger FA, Lacroix D, Germain L. Skin substitutes and wound healing. Skin Pharmacol Physiol. 2009;22:94–102. doi:10.1159/00017886819188757
  • Zanetti M. Cathelicidins, multifunctional peptides of the innate immunity. J Leukoc Biol. 2004;75:39–48. doi:10.1189/jlb.040314712960280
  • Martinon F, Mayor A, Tschopp J. The inflammasomes: guardians of the body. Annu Rev Immunol. 2009;27:229–265. doi:10.1146/annurev.immunol.021908.13271519302040
  • Albanesi C, Scarponi C, Giustizieri M, Girolomoni G. Keratinocytes in inflammatory skin diseases. Curr Drug Targets Inflamm Allergy. 2005;4(3):329–334. doi:10.2174/156801005402203316101542
  • Mc Dermott R, Ziylan U, Spehner D, et al. Birbeck granules are subdomains of endosomal recycling compartment in human epidermal Langerhans cells, which form where Langerin accumulates. Mol Biol Cell. 2002;13(1):317–335. doi:10.1091/mbc.01-06-030011809842
  • Bursch LS, Wang L, Igyarto B, et al. Identification of a novel population of Langerin+ dendritic cells. J Exp Med. 2007;204(13):3147–3156. doi:10.1084/jem.2007196618086865
  • Johnson LA, Banerji S, Lawrance W, et al. Dendritic cells enter lymph vessels by hyaluronan-mediated docking to the endothelial receptor LYVE-1. Nat Immunol. 2017;18(7):762. doi:10.1038/ni.375028504698
  • Fay NS, Larson EC, Jameson JM. Chronic inflammation and γδ T cells. Front Immunol. 2016;7:210. doi:10.3389/fimmu.2016.0021027303404
  • MacLeod AS, Havran WL. Functions of skin-resident γδ T cells. Cell Mol Life Sci. 2011;68(14):2399–2408. doi:10.1007/s00018-011-0702-x21560071
  • Hartwig H, Drechsler M, Lievens D, et al. Platelet-derived PF4 reduces neutrophil apoptosis following arterial occlusion. Thromb Haemost. 2014;111(3):562–564. doi:10.1160/TH13-08-069924258616
  • McMenamin C, Pimm C, McKersey M, Holt PG. Regulation of IgE responses to inhaled antigen in mice by antigen-specific gamma delta T cells. Science. 1994;265(5180):1869–1871. doi:10.1126/science.79164817916481
  • Ramírez-Valle F, Gray EE, Cyster JG. Inflammation induces dermal Vγ4+ γδT17 memory-like cells that travel to distant skin and accelerate secondary IL-17–driven responses. Proc Natl Acad Sci. 2015;112(26):8046–8051. doi:10.1073/pnas.150899011226080440
  • 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. 2005;6(1):73. doi:10.1038/ni115215592472
  • Komori HK, Witherden DA, Kelly R, Sendaydiego K, Jameson JM, Teyton L, Havran WL. Cutting edge: dendritic epidermal γδ T cell ligands are rapidly and locally expressed by keratinocytes following cutaneous wounding. J Immunology 2012;188(7):2972–2976. doi:10.4049/jimmunol.1100887
  • Xu P, Fu X, Xiao N, et al. Involvements of γδT lymphocytes in acute and chronic skin wound repair. Inflammation. 2017;40(4):1416–1427. doi:10.1007/s10753-017-0585-628540539
  • Toulon A, Breton L, Taylor KR, et al. A role for human skin–resident T cells in wound healing. J Exp Med. 2009;206(4):743–750. doi:10.1084/jem.2008178719307328
  • Campbell JJ, O’Connell DJ, Wurbel M-A. Cutting edge: chemokine receptor CCR4 is necessary for antigen-driven cutaneous accumulation of CD4 T cells under physiological conditions. J Immunol. 2007;178(6):3358–3362. doi:10.4049/jimmunol.178.6.335817339428
  • Slominski AT, Manna PR, Tuckey RC. Cutaneous glucocorticosteroidogenesis: securing local homeostasis and the skin integrity. Exp Dermatol. 2014;23(6):369–374. doi:10.1111/exd.1237624888781
  • Fuhlbrigge RC, Kieffer JD, Armerding D, Kupper TS. Cutaneous lymphocyte antigen is a specialized form of PSGL-1 expressed on skin-homing T cells. Nature. 1997;389(6654):978. doi:10.1038/401669353122
  • Richmond JM, Harris JE. Immunology and skin in health and disease. Cold Spring Harb Perspect Med 2014;4(12):a15339. doi:10.1101/cshperspect.a015339
  • Barinova ME, Barinov EF, Sulaieva OM. Functional activity of monocytes and mechanisms of iNOS intracellular regulation during wound process. Fiziol Zh. 2011;57(1):36–44.21516832
  • Gilroy D, De Maeyer R. New insights into the resolution of inflammation. Semin Immunol. 2015:27(3):161–168. Academic Press. doi:10.1016/j.smim.2015.05.003.26037968
  • Cruz MS, Diamond A, Russell A, Jameson JM. Human αβ and γδ T cells in skin immunity and disease. Front Immunol. 2018;9:1304 Published 2018 Jun 6. doi:10.3389/fimmu.2018.0130429928283
  • Supp DM, Wilson-Landy KAILA, Boyce ST. Human dermal microvascular endothelial cells form vascular analogs in cultured skin substitutes after grafting to athymic mice. Faseb J. 2002;16(8):797–804. doi:10.1096/fj.01-0868com12039861
  • Nikolsky IS, Serebrovska TV. Role of hypoxia in stem cell development and functioning. Int J Physiol Pathophysiol Pharmacol. 2010;1(1). doi:10.1615/IntJPhysPathophys.v1.i1.90
  • Serebrovskaya TV, Nikolsky IS, Nikolska VV, Mallet RT, Ishchuk VA. Intermittent hypoxia mobilizes hematopoietic progenitors and augments cellular and humoral elements of innate immunity in adult men. High Alt Med Biol. 2011;12(3):243–252. doi:10.1089/ham.2010.108621962068
  • Grando SA, Pittelkow MR, Schallreuter KU. Adrenergic and cholinergic control in the biology of epidermis: physiological and clinical significance. J Invest Dermatol. 2006;126(9):1948–1965. doi:10.1038/sj.jid.570015116912692
  • Pongratz G, Straub RH. Role of peripheral nerve fibres in acute and chronic inflammation in arthritis. Nat Rev Rheumatol. 2013;9(2):117. doi:10.1038/nrrheum.2012.18123147892
  • Vinik AI, Strotmeyer ES. Diabetic neuropathy. Pathy’s Principles Pract Geriatric Med. 2012;1:751–767. doi:10.1002/9781119952930.ch65