Skip to main content

Mesenchymal Stem Cells for Cutaneous Wound Healing

  • Chapter
  • First Online:
Concepts and Applications of Stem Cell Biology

Part of the book series: Learning Materials in Biosciences ((LMB))

Abstract

The success of wound healing is impaired in several medical conditions resulting in increased morbidity. The current available therapeutic options frequently fail to promote full tissue regeneration, and stem cell-based therapies have emerged as promising alternatives. Mesenchymal stem/stromal cells (MSCs) have gained relevance within this context not only due to their ability to promote healing through engraftment and further differentiation, but also because of their paracrine effects. Recently, it has been suggested that the secretion of exosomes may be a dominant mechanism by which MSCs exert their healing function, thus granting them a potential new role as active players in cell-free-based therapies. This chapter focuses on the recent advances on MSC-based therapies for the treatment of cutaneous wounds, namely on their mechanisms of action and the strategies adopted to improve their therapeutic efficacy.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 79.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 99.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Heublein H, Bader A, Giri S. Preclinical and clinical evidence for stem cell therapies as treatment for diabetic wounds. Drug Discov Today. 2015;20(6):703–17.

    PubMed  Google Scholar 

  2. Guest JF, Vowden K, Vowden P. The health economic burden that acute and chronic wounds impose on an average clinical commissioning group/health board in the UK. J Wound Care. 2017;26(6):292–303.

    CAS  PubMed  Google Scholar 

  3. Domaszewska-Szostek A, Krzyżanowska M, Siemionow M. Cell-based therapies for chronic wounds tested in clinical studies. Ann Plast Surg. 2019;83(6):e96–e109.

    CAS  PubMed  Google Scholar 

  4. Lau K, Paus R, Tiede S, Day P, Bayat A. Exploring the role of stem cells in cutaneous wound healing. Exp Dermatol. 2009;18(11):921–33.

    CAS  PubMed  Google Scholar 

  5. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453(7193):314–21.

    CAS  PubMed  Google Scholar 

  6. Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med. 1999;341(10):738–46.

    CAS  PubMed  Google Scholar 

  7. Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol. 2008;180(4):2581–7.

    CAS  PubMed  Google Scholar 

  8. Guo S, DiPietro LA. Factors affecting wound healing. J Dent Res. 2010;89(3):219–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Frykberg RG, Banks J. Challenges in the treatment of chronic wounds. Adv Wound Care. 2015;4(9):560–82.

    Google Scholar 

  10. 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(11):1205–13.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Pastar I, Stojadinovic O. Attenuation of the transforming growth Factor β–signaling pathway in chronic venous ulcers. Mol Med. 2010;16(3–4):92–101.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Han G, Ceilley R. Chronic wound healing: a review of current management and treatments. Adv Ther. 2017;34(3):599–610.

    PubMed  PubMed Central  Google Scholar 

  13. Jones V, Grey JE, Harding KG. Wound dressings. BMJ. 2006;332(7544):777–80.

    PubMed  PubMed Central  Google Scholar 

  14. Liu Y, Dulchavsky DS, Gao X, Kwon D, Chopp M, Dulchavsky S, et al. Wound repair by bone marrow stromal cells through growth factor production. J Surg Res. 2006;136(2):336–41.

    CAS  PubMed  Google Scholar 

  15. Weiss ML, Troyer DL. Stem cells in the umbilical cord. Stem Cell Rev. 2006;2(2):155–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Gunawardena TNA, Rahman MT, Abdullah BJJ, Abu Kasim NH. Conditioned media derived from mesenchymal stem cell cultures: the next generation for regenerative medicine. J Tissue Eng Regen Med. 2019;13(4):569–86.

    CAS  PubMed  Google Scholar 

  17. Harrell CR, Fellabaum C, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Molecular mechanisms responsible for therapeutic potential of mesenchymal stem cell-derived Secretome. Cell. 2019;8(5):467.

    CAS  Google Scholar 

  18. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause DS, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–7.

    CAS  Google Scholar 

  19. da Silva Meirelles L. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119(11):2204–13.

    PubMed  Google Scholar 

  20. In ‘t Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GMJS, Claas FHJ, Fibbe WE, et al. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 2004;22(7):1338–1345.

    Google Scholar 

  21. Antonucci I, Stuppia L, Kaneko Y, Yu S, Tajiri N, Bae EC, et al. Amniotic fluid as a rich source of mesenchymal stromal cells for transplantation therapy. Cell Transplant. 2011;20(6):789–96.

    PubMed  Google Scholar 

  22. Campagnoli C. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood. 2001;98(8):2396–402.

    CAS  PubMed  Google Scholar 

  23. Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109(1):235–42.

    CAS  PubMed  Google Scholar 

  24. Miranda JP, Filipe E, Fernandes AS, Almeida JM, Martins JP, De La Fuente A, et al. The human umbilical cord tissue-derived MSC population UCX ® promotes early Motogenic effects on keratinocytes and fibroblasts and G-CSF-mediated mobilization of BM-MSCs when transplanted In vivo. Cell Transplant. 2015;24(5):865–77.

    PubMed  Google Scholar 

  25. Santos JM, Camões SP, Filipe E, Cipriano M, Barcia RN, Filipe M, et al. Three-dimensional spheroid cell culture of umbilical cord tissue-derived mesenchymal stromal cells leads to enhanced paracrine induction of wound healing. Stem Cell Res Ther. 2015;6:90.

    PubMed  PubMed Central  Google Scholar 

  26. Zuk PA. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Santos JM, Bárcia RN, Simões SI, Gaspar MM, Calado S, Água-Doce A, et al. The role of human umbilical cord tissue-derived mesenchymal stromal cells (UCX®) in the treatment of inflammatory arthritis. J Transl Med. 2013;11(1):18.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Rohban R, Pieber TR. Mesenchymal stem and progenitor cells in regeneration: tissue specificity and regenerative potential. Stem Cells Int. 2017;2017:1–16.

    Google Scholar 

  29. Shafiee A, Seyedjafari E, Soleimani M, Ahmadbeigi N, Dinarvand P, Ghaemi N. A comparison between osteogenic differentiation of human unrestricted somatic stem cells and mesenchymal stem cells from bone marrow and adipose tissue. Biotechnol Lett. 2011;33(6):1257–64.

    CAS  PubMed  Google Scholar 

  30. Ardeshirylajimi A, Mossahebi-Mohammadi M, Vakilian S, Langroudi L, Seyedjafari E, Atashi A, et al. Comparison of osteogenic differentiation potential of human adult stem cells loaded on bioceramic-coated electrospun poly (L-lactide) nanofibres. Cell Prolif. 2015;48(1):47–58.

    CAS  PubMed  Google Scholar 

  31. Rohban R, Etchart N, Pieber TR. Vasculogenesis potential of stem cells from various human tissues. BioRxiv. 2016;1–22.

    Google Scholar 

  32. Lin H. The stem-cell niche theory: lessons from flies. Nat Rev Genet. 2002;3(12):931–40.

    CAS  PubMed  Google Scholar 

  33. da Silva Meirelles L, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev. 2009;20(5–6):419–27.

    Google Scholar 

  34. Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell. 2009;4(3):206–16.

    CAS  PubMed  Google Scholar 

  35. He Q, Wan C, Li G. Concise review: multipotent mesenchymal stromal cells in blood. Stem Cells. 2007;25(1):69–77.

    CAS  PubMed  Google Scholar 

  36. Wang C-H, Cherng W-J, Yang N-I, Kuo L-T, Hsu C-M, Yeh H-I, et al. Late-outgrowth endothelial cells attenuate intimal hyperplasia contributed by mesenchymal stem cells after vascular injury. Arterioscler Thromb Vasc Biol. 2007;28(1):54–60.

    PubMed  Google Scholar 

  37. Barminko J, Gray A, Maguire T, Schloss R, Yarmush ML. Mesenchymal stem cell therapy. In: Chase LG, Vemuri MC, editors. Stem cell biology and regenerative medicine. Totowa: Humana Press; 2013. p. 15–39.

    Google Scholar 

  38. Bollag WB, Hill WD. CXCR4 in epidermal keratinocytes: crosstalk within the skin. J Invest Dermatol. 2013;133(11):2505–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Yu J, Li M, Qu Z, Yan D, Li D, Ruan Q. SDF-1/CXCR4-mediated migration of transplanted bone marrow stromal cells towards areas of heart myocardial infarction via activation of PI3K/Akt. J Cardiovasc Pharmacol. 2010;55(5):1.

    Google Scholar 

  40. Wang Y, Deng Y, Zhou G-Q. SDF-1α/CXCR4-mediated migration of systemically transplanted bone marrow stromal cells towards ischemic brain lesion in a rat model. Brain Res. 2008;1195:104–12.

    CAS  PubMed  Google Scholar 

  41. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98(5):1076–84.

    CAS  PubMed  Google Scholar 

  42. El Omar R, Beroud J, Stoltz J-F, Menu P, Velot E, Decot V. Umbilical cord mesenchymal stem cells: the new gold standard for mesenchymal stem cell-based therapies? Tissue Eng Part B Rev. 2014;20(5):523–44.

    PubMed  Google Scholar 

  43. Marfia G, Navone SE, Di Vito C, Ughi N, Tabano S, Miozzo M, et al. Mesenchymal stem cells: potential for therapy and treatment of chronic non-healing skin wounds. Organogenesis. 2015;11(4):183–206.

    PubMed  PubMed Central  Google Scholar 

  44. Miranda JP, Camões SP, Gaspar MM, Rodrigues JS, Carvalheiro M, Bárcia RN, et al. The Secretome derived from 3D-cultured umbilical cord tissue MSCs counteracts manifestations typifying rheumatoid arthritis. Front Immunol. 2019;10:18.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Lai RC, Chen TS, Lim SK. Mesenchymal stem cell exosome: a novel stem cell-based therapy for cardiovascular disease. Regen Med. 2011;6(4):481–92.

    PubMed  Google Scholar 

  46. Ghannam S, Bouffi C, Djouad F, Jorgensen C, Noël D. Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications. Stem Cell Res Ther. 2010;1(1):2.

    PubMed  PubMed Central  Google Scholar 

  47. Chapel A, Bertho JM, Bensidhoum M, Fouillard L, Young RG, Frick J, et al. Mesenchymal stem cells home to injured tissues when co-infused with hematopoietic cells to treat a radiation-induced multi-organ failure syndrome. J Gene Med. 2003;5(12):1028–38.

    PubMed  Google Scholar 

  48. Bárcia RN, Santos JM, Filipe M, Teixeira M, Martins JP, Almeida J, et al. What makes umbilical cord tissue-derived mesenchymal stromal cells superior Immunomodulators when compared to bone marrow derived mesenchymal stromal cells? Stem Cells Int. 2015;2015:583984.

    PubMed  PubMed Central  Google Scholar 

  49. Soleymaninejadian E, Pramanik K, Samadian E. Immunomodulatory properties of mesenchymal stem cells: cytokines and factors. Am J Reprod Immunol. 2012;67(1):1–8.

    CAS  PubMed  Google Scholar 

  50. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105(4):1815–22.

    CAS  PubMed  Google Scholar 

  51. Rafei M, Hsieh J, Fortier S, Li M, Yuan S, Birman E, et al. Mesenchymal stromal cell-derived CCL2 suppresses plasma cell immunoglobulin production via STAT3 inactivation and PAX5 induction. Blood. 2008;112(13):4991–8.

    CAS  PubMed  Google Scholar 

  52. Wang L-T, Ting C-H, Yen M-L, Liu K-J, Sytwu H-K, Wu KK, et al. Human mesenchymal stem cells (MSCs) for treatment towards immune- and inflammation-mediated diseases: review of current clinical trials. J Biomed Sci. 2016;23(1):76.

    PubMed  PubMed Central  Google Scholar 

  53. Levy V, Lindon C, Zheng Y, Harfe BD, Morgan B. a. Epidermal stem cells arise from the hair follicle after wounding. FASEB J. 2007;21(7):1358–66.

    CAS  PubMed  Google Scholar 

  54. Jahoda CA, Reynolds AJ. Hair follicle dermal sheath cells: unsung participants in wound healing. Lancet. 2001;358(9291):1445–8.

    CAS  PubMed  Google Scholar 

  55. Rodriguez-Menocal L, Salgado M, Ford D, Van Badiavas E. Stimulation of skin and wound fibroblast migration by mesenchymal stem cells derived from normal donors and chronic wound patients. Stem Cells Transl Med. 2012;1(3):221–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Liu L, Yu Y, Hou Y, Chai J, Duan H, Chu W, et al. Human umbilical cord mesenchymal stem cells transplantation promotes cutaneous wound healing of severe burned rats. PLoS One. 2014;9(2):e88348.

    PubMed  PubMed Central  Google Scholar 

  57. Basiouny HS, Salama NM, Mohamed Z, Maadawi E, Farag EA. Effect of bone marrow derived mesenchymal stem cells on healing of induced full-thickness skin wounds in albino rat. Int J Stem Cells. 2013;6(1):12–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Luo G, Cheng W, He W, Wang X, Tan J, Fitzgerald M, et al. Promotion of cutaneous wound healing by local application of mesenchymal stem cells derived from human umbilical cord blood. Wound Repair Regen. 2010;18(5):506–13.

    PubMed  Google Scholar 

  59. Kong P, Xie X, Li F, Liu Y, Lu Y. Placenta mesenchymal stem cell accelerates wound healing by enhancing angiogenesis in diabetic Goto-Kakizaki (GK) rats. Biochem Biophys Res Commun. 2013;438(2):410–9.

    CAS  PubMed  Google Scholar 

  60. Hong SJ, Jia S-X, Xie P, Xu W, Leung KP, Mustoe TA, et al. Topically delivered adipose derived stem cells show an activated-fibroblast phenotype and enhance granulation tissue formation in skin wounds. PLoS One. 2013;8(1):e55640.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Shin L, Peterson D. a. Human mesenchymal stem cell grafts enhance normal and impaired wound healing by recruiting existing endogenous tissue stem/progenitor cells. Stem Cells Transl Med. 2013;2(1):33–42.

    CAS  PubMed  Google Scholar 

  62. Yoon BS, Moon J-H, Jun EK, Kim J, Maeng I, Kim JS, et al. Secretory profiles and wound healing effects of human amniotic fluid–derived mesenchymal stem cells. Stem Cells Dev. 2010;19(6):887–902.

    CAS  PubMed  Google Scholar 

  63. Arno AI, Amini-Nik S, Blit PH, Al-Shehab M, Belo C, Herer E, et al. Human Wharton’s jelly mesenchymal stem cells promote skin wound healing through paracrine signaling. Stem Cell Res Ther. 2014;5(1):28.

    PubMed  PubMed Central  Google Scholar 

  64. Mishra PJ. Cell-free derivatives from mesenchymal stem cells are effective in wound therapy. World J Stem Cells. 2012;4(5):35.

    PubMed  PubMed Central  Google Scholar 

  65. Li M, Zhao Y, Hao H, Dai H, Han Q, Tong C, et al. Mesenchymal stem cell–conditioned medium improves the proliferation and migration of keratinocytes in a diabetes-like microenvironment. Int J Low Extrem Wounds. 2015;14(1):73–86.

    CAS  PubMed  Google Scholar 

  66. Lai RC, Arslan F, Lee MM, Sze NSK, Choo A, Chen TS, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4(3):214–22.

    CAS  PubMed  Google Scholar 

  67. Shi Y, Shi H, Nomi A, Lei-lei Z, Zhang B, Qian H. Mesenchymal stem cell–derived extracellular vesicles: a new impetus of promoting angiogenesis in tissue regeneration. Cytotherapy. 2019;21(5):497–508.

    CAS  PubMed  Google Scholar 

  68. Wu P, Zhang B, Shi H, Qian H, Xu W. MSC-exosome: a novel cell-free therapy for cutaneous regeneration. Cytotherapy. 2018;20(3):291–301.

    PubMed  Google Scholar 

  69. Li T, Yan Y, Wang B, Qian H, Zhang X, Shen L, et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013;22(6):845–54.

    CAS  PubMed  Google Scholar 

  70. Marquez-Curtis LA, Gul-Uludag H, Xu P, Chen J, Janowska-Wieczorek A. CXCR4 transfection of cord blood mesenchymal stromal cells with the use of cationic liposome enhances their migration toward stromal cell-derived factor-1. Cytotherapy. 2013;15(7):840–9.

    CAS  PubMed  Google Scholar 

  71. Zhang B, Wang M, Gong A, Zhang X, Wu X, Zhu Y, et al. HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells. 2015;33(7):2158–68.

    CAS  PubMed  Google Scholar 

  72. Zhang J, Guan J, Niu X, Hu G, Guo S, Li Q, et al. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J Transl Med. 2015;13(1):49.

    PubMed  PubMed Central  Google Scholar 

  73. Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Van Badiavas E. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev. 2015;24(14):1635–47.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Kim S, Lee SK, Kim H, Kim TM. Exosomes secreted from induced pluripotent stem cell-derived mesenchymal stem cells accelerate skin cell proliferation. Int J Mol Sci. 2018;19(10):pii: E3119.

    Google Scholar 

  75. Hu L, Wang J, Zhou X, Xiong Z, Zhao J, Yu R, et al. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci Rep. 2016;6(1):32993.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Fang S, Xu C, Zhang Y, Xue C, Yang C, Bi H, et al. Umbilical cord-derived mesenchymal stem cell-derived Exosomal MicroRNAs suppress Myofibroblast differentiation by inhibiting the transforming growth factor-β/SMAD2 pathway during wound healing. Stem Cells Transl Med. 2016;5(10):1425–39.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Noronha Nc NDC, Mizukami A, Caliári-Oliveira C, Cominal JG, Rocha JLM, Covas DT, et al. Priming approaches to improve the efficacy of mesenchymal stromal cell-based therapies. Stem Cell Res Ther. 2019;10(1):1–21.

    Google Scholar 

  78. Chen L, Xu Y, Zhao J, Zhang Z, Yang R, Xie J, et al. Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice. PLoS One. 2014;9(4):e96161.

    PubMed  PubMed Central  Google Scholar 

  79. Jun E, Zhang Q, Yoon B, Moon J-H, Lee G, Park G, et al. Hypoxic conditioned medium from human amniotic fluid-derived mesenchymal stem cells accelerates skin wound healing through TGF-β/SMAD2 and PI3K/Akt pathways. Int J Mol Sci. 2014;15(1):605–28.

    PubMed  PubMed Central  Google Scholar 

  80. Liu G-Y, Liu Y, Lu Y, Qin Y-R, Di G-H, Lei Y-H, et al. Short-term memory of danger signals or environmental stimuli in mesenchymal stem cells: implications for therapeutic potential. Cell Mol Immunol. 2016;13(3):369–78.

    CAS  PubMed  Google Scholar 

  81. Ti D, Hao H, Tong C, Liu J, Dong L, Zheng J, et al. LPS-preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome-shuttled let-7b. J Transl Med. 2015;13(1):308.

    PubMed  PubMed Central  Google Scholar 

  82. Anton D, Burckel H, Josset E, Noel G. Three-dimensional cell culture: a breakthrough in vivo. Int J Mol Sci. 2015;16(3):5517–27.

    Google Scholar 

  83. Hsu S, Hsieh P-S. Self-assembled adult adipose-derived stem cell spheroids combined with biomaterials promote wound healing in a rat skin repair model. Wound Repair Regen. 2015;23(1):57–64.

    PubMed  Google Scholar 

  84. Kwon SH, Bhang SH, Jang H-K, Rhim T, Kim B-S. Conditioned medium of adipose-derived stromal cell culture in three-dimensional bioreactors for enhanced wound healing. J Surg Res. 2015;194(1):8–17.

    CAS  PubMed  Google Scholar 

  85. Xia Z, Zhang C, Zeng Y, Wang T, Ai G. Transplantation of BMSCs expressing hVEGF 165/hBD3 promotes wound healing in rats with combined radiation-wound injury. Int Wound J. 2014;11(3):293–303.

    PubMed  Google Scholar 

  86. Qi Y, Jiang D, Sindrilaru A, Stegemann A, Schatz S, Treiber N, et al. TSG-6 released from intradermally injected mesenchymal stem cells accelerates wound healing and reduces tissue fibrosis in murine full-thickness skin wounds. J Invest Dermatol. 2014;134(2):526–37.

    CAS  PubMed  Google Scholar 

  87. Yang D, Sun S, Wang Z, Zhu P, Yang Z, Zhang B. Stromal cell-derived factor-1 receptor CXCR4-overexpressing bone marrow mesenchymal stem cells accelerate wound healing by migrating into skin injury areas. Cell Reprogram. 2013;15(3):206–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Li Y, Zheng L, Xu X, Song L, Li Y, Li W, et al. Mesenchymal stem cells modified with angiopoietin-1 gene promote wound healing. Stem Cell Res Ther. 2013;4(5):113.

    PubMed  PubMed Central  Google Scholar 

  89. Kortesidis A, Zannettino A, Isenmann S, Shi S, Lapidot T, Gronthos S. Stromal-derived factor-1 promotes the growth, survival, and development of human bone marrow stromal stem cells. Blood. 2005;105(10):3793–801.

    CAS  PubMed  Google Scholar 

  90. Nakamura Y, Ishikawa H, Kawai K, Tabata Y, Suzuki S. Enhanced wound healing by topical administration of mesenchymal stem cells transfected with stromal cell-derived factor-1. Biomaterials. 2013;34(37):9393–400.

    CAS  PubMed  Google Scholar 

  91. Yew T-L, Hung Y-T, Li H-Y, Chen H-W, Chen L-L, Tsai K-S, et al. Enhancement of wound healing by human multipotent stromal cell conditioned medium: the paracrine factors and p38 MAPK activation. Cell Transplant. 2011;20(5):693–706.

    PubMed  Google Scholar 

  92. Shohara R, Yamamoto A, Takikawa S, Iwase A, Hibi H, Kikkawa F, et al. Mesenchymal stromal cells of human umbilical cord Wharton’s jelly accelerate wound healing by paracrine mechanisms. Cytotherapy. 2012;14(10):1171–81.

    CAS  PubMed  Google Scholar 

  93. Fong C-Y, Tam K, Cheyyatraivendran S, Gan S-U, Gauthaman K, Armugam A, et al. Human Wharton’s jelly stem cells and its conditioned medium enhance healing of excisional and diabetic wounds. J Cell Biochem. 2014;115(2):290–302.

    CAS  PubMed  Google Scholar 

  94. Zhang B, Wu X, Zhang X, Sun Y, Yan Y, Shi H, et al. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/β-catenin pathway. Stem Cells Transl Med. 2015;4(5):513–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Wang X, Jiao Y, Pan Y, Zhang L, Gong H, Qi Y, et al. Fetal dermal mesenchymal stem cell-derived exosomes accelerate cutaneous wound healing by activating notch signaling. Stem Cells Int. 2019;2019:1–11.

    CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joana P. Miranda .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2020 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Camões, S.P., Santos, J.M., Carvalho, F., Miranda, J.P. (2020). Mesenchymal Stem Cells for Cutaneous Wound Healing. In: Rodrigues, G., Roelen, B.A.J. (eds) Concepts and Applications of Stem Cell Biology. Learning Materials in Biosciences. Springer, Cham. https://doi.org/10.1007/978-3-030-43939-2_13

Download citation

Publish with us

Policies and ethics