99
Views
6
CrossRef citations to date
0
Altmetric
REVIEW

Multipotent adult progenitor cells: their role in wound healing and the treatment of dermal wounds

, & , MD
Pages 543-550 | Published online: 07 Jul 2009

References

  • Moulin V. Growth factors in the skin wound. Eur J Cell Biol 1995; 68: 1–7
  • Dubay DA, Franz MG. Acute wound healing: the biology of acute wound failure. Surg Clin North Am 2003; 83: 463–81
  • Liechty KW, Crombleholme TM, Cass DL, et al. Diminished interleukin-8 (IL-8) production in the fetal wound healing response. J Surg Res 1998; 77: 80–4
  • Bullard KM, Longaker MT, Lorenz HP. Fetal wound healing: current biology. World J Surg 2003; 27: 54–61
  • Menke NB, Ward KR, Witten TM, et al. Impaired wound healing. Clin Dermatol 2007; 25: 19–25
  • Lazarus GS, Cooper DM, Knighton DR, et al. Definitions and guidelines for assessment of wounds and evaluation of healing. Arch Dermatol 1994; 130: 489–93
  • Brem H, Balledux J, Bloom T, et al. Healing of diabetic foot ulcers and pressure ulcers with human skin equivalents: a new paradigm in wound healing. Arch Surg 2000; 135: 627–34
  • Chen WY, Rogers AA. Recent insights into the causes of chronic leg ulceration in venous diseases and implications on other types of chronic wounds. Wound Repair Regen 2007; 15: 434–49
  • Mekkes JR, Loots MAM, Van Der Wal AC, Bos JD. Causes, investigation and treatment of leg ulceration. Br J Dermatol 2003; 148: 388–401
  • Mustoe YA, O'Shaughnessy K, Kloeters O. Chronic wound pathogenesis and current treatment strategies: a unifying hypothesis. Plast Reconstr Surg 2006; 117(Suppl)35S–41S
  • Cha J, Falanga V. Stem cells in cutaneous wound healing. Clin Dermatol 2007; 25: 73–78
  • Ehrenreich M, Ruszczak Z. Update on tissue-engineered biological dressings. Tissue Eng 2006; 12: 2407–24
  • Griffiths M, Ojeh N, Livingstone R, et al. Survival of Apligraft in acute human wounds. Tissue Eng 2004; 10: 1180–95
  • Evan MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature 1981; 292: 154–6
  • Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science 1998; 282: 1145–7
  • Jiang, Y, Jahagirdar, BN, Reinhardt, RL, , et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature, 418:41–9.
  • Toma JG, Akhavan M, Fernandes KJ, et al. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol 2001; 3: 778–84
  • Ichioka S, Kouraba S, Sekiya N, et al. Bone marrow-impregnated collagen matrix for wound healing: experimental evaluation in a microcirculatory model of angiogenesis, and clinical experience. Br J Plast Surg 2005; 58: 1124–30
  • Badiavas EV, Falanga V. Treatment of chronic wounds with bone marrow-derived cells. Arch Dermatol 2003; 139: 510–16
  • Badillo AT, Redden RA, Zhang L, et al. Treatment of diabetic wounds with fetal murine mesenchymal stromal cells enhances wound closure. Cell Tissue Res 2007; 329: 301–11
  • Javazon EH, Keswani SG, Badillo AT, et al. Enhanced epithelial gap closure and increased angiogenesis in wounds of diabetic mice treated with adult murine bone marrow stromal progenitor cells. Wound Repair Regen 2007; 15: 350–9
  • Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999; 284: 143–7
  • Horwitz E, LeBlanc K, Dominici M, et al. Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy 2005; 7: 393–5
  • Phinney DG, Prockop DJ. Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair current views. Stem Cells 2008; 25: 2896–902
  • Zuk PA, Zhu M, Mizuno H, Huang J, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 2001; 7: 211–28
  • Williams JT, Southerland SS, Souza J, et al. Cells isolated from adult human skeletal muscle capable of differentiating into multiple mesodermal phenotypes. Am Surg 1999; 65: 22–26
  • Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 2000; 109: 235–42
  • In't Anker PS, Scherjon SA, Kleijburg-van der Keur C, et al. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 2003; 102: 1548–9
  • Bieback K, Kluter H. Mesenchymal stromal cells from umbilical cord blood. Curr Stem Cell Res Ther 2007; 2: 310–23
  • O'Donoghue K, Chan J. Human fetal mesenchymal stem cells. Curr Stem Cell Res Ther 2006; 1: 371–86
  • Le Blanc K. Mesenchymal stromal cells: tissue repair and immune modulation. Cytotherapy 2006; 8: 559–61
  • Fox JM, Chamberlain G, Ashton BA, Middleton J. Recent advances into the understanding of mesenchymal stem cell trafficking. Br J Haematol 2007; 137: 491–502
  • Dominici M, 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: 315–7
  • Satoh H, Kishi K, Tanaka T, et al. Transplanted mesenchymal stem cells are effective for skin regeneration in acute cutaneous wounds. Cell Transplant 2004; 13: 405–12
  • Yoshikawa T, Mitsuno H, Nonaka I, et al. Wound therapy by marrow mesenchymal cell transplantation. Plast Reconstr Surg 2008; 121: 860–77
  • Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells 2007; 25: 2648–59
  • Falanga V, Iwamoto S, Chartier M, et al. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng 2007; 13: 1299–312
  • Li H, Fu X, Ouyang Y, et al. Adult bone-marrow-derived mesenchymal stem cells contribute to wound healing of skin appendages. Cell Tissue Res 2006; 326: 725–36
  • Natsu K, Ochi M, Mochizuki Y, et al. Allogeneic bone marrow-derived mesenchymal stromal cells promote the regeneration of injured skeletal muscle without differentiation into myofibers. Tissue Eng 2004; 10: 1093–112
  • Chen L, Tredget EE, Wu PYG, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE 2008; 3: e1886
  • Gnecchi M, He H, Liang OD, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med 2005; 11: 367–8
  • Gnecchi M, He H, Noiseux N, et al. Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J 2006; 20: 661–9
  • Noiseux N, Gnecchi M, Lopez-Ilasaca M, et al. Mesenchymal stem cells overexpressing Akt dramatically repair infracted myocardium and improve cardiac function despite infrequent cellular fusion or differentiation. Mol Ther 2006; 14: 840–50
  • Sobajima, S, Vadala, G, Shimer, A, , et al. Feasibility of a stem cell therapy for intervertebral disc degeneration. Spine J, in press.
  • Stagg J. Immune regulation by mesenchymal stem cells: two sides to the coin. Tissue Antigens 2007; 69: 1–9
  • Le Blanc K, Rasmusson I, Sundberg B, et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004; 363: 1439–41
  • Lee KY, Kong HJ, Mooney DJ. Quantifying interactions between cell receptors and adhesion ligand-modified polymers in solution. Macromol Biosci 2008; 8: 140–5
  • Sands RW, Mooney DJ. Polymers to direct cell fate by controlling the microenvironment. Curr Opin Biotechnol 2007; 18: 448–53
  • Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 2004; 10: 858–64
  • Badillo AT, Chung S, Zhang L, et al. Lentiviral gene transfer of SDF-1α to wounds improved diabetic wound healing. J Surg Research 2007; 143: 35–42
  • Mace KA, Yu DH, Paydar KZ, et al. Sustained expression of HIF-1α in the diabetic environment promotes angiogenesis and cutaneous wound repair. Wound Rep Regen 2007; 15: 636–45
  • Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997; 275: 964–7
  • Asahara T, Takahashi T, Masuda H, et al. VEGF contributes to postnatal neovascularization by mobilizing bone-marrow derived endothelial progenitor cells. EMBO J 1999; 18: 3964–72
  • Hristov M, Weber C. Endothelial progenitor cells: characterization, pathophysiology and possible clinical relevance. J Cell Mol Med 2004; 8: 498–508
  • Gehling UM, Ergün S, Schumacher U, et al. In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood 2000; 95: 3106–12
  • Quirici N, Soligo D, Caneva L, et al. Differentiation and expansion of endothelial cells from human bone marrow CD133+ cells. Br J Haematol 2001; 115: 186–94
  • Garmy-Susini B, Varner JA. Circulating endothelial progenitor cells. Br J Cancer 2005; 93: 855–8
  • Heissig B, Hattori K, Dias S, et al. Recruitment of stem and progenitor cells from bone marrow niches requires MMP-9 mediated release of kit-ligand. Cell 2002; 109: 625–37
  • Fox A, Smythe J, Fisher N, et al. Mobilization of endothelial progenitor cells into the circulation in burned patients. Br J Surg 2008; 95: 244–51
  • Suh W, Kim KL, Kim JM, et al. Transplantation of endothelial progenitor cells accelerates dermal wound healing with increased recruitment of monocytes/macrophages and neovascularization. Stem Cells 2005; 23: 1571–8
  • Kähler C, Weschselberger J, Hilbe W, et al. Peripheral infusion of rat bone marrow derived endothelial progenitor cells leads to homing in acute lung injury. Respir Res 2007; 8: 50
  • Kung EF, Wang F, Schechner JS. In vivo perfusion of human skin substitutes with microvessels formed by adult circulating endothelial progenitor cells. Dermatol Surg 2008; 34: 137–46
  • Rehman J, Li J, Orschell CM, March KL. Peripheral blood ‘endothelial progenitor cells’ are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation 2003; 107: 1164–9
  • Urbich C, Aicher A, Heeschen C, et al. Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells. J Mol Cell Cardiol 2005; 39: 733–42
  • Asai J, Takenaka H, Kusano KF, et al. Topical Sonic Hedgehog gene therapy accelerates wound healing in diabetes by enhancing endothelial progenitor cell-mediated microvascular remodeling. Circulation 2006; 113: 2413–24
  • Zampetaki, A, Kirton, JP, Xu, Q. Vascular repair by endothelial progenitor cells. Cardiovasc Res 2008, in press.
  • Kawamoto A, Losordo DW. Endothelial progenitor cells for cardiovascular regeneration. Trends Cardiovasc Med 2008; 18: 33–7
  • Lam C, Liu YC, Hsu JK, et al. Autologous transplantation of endothelial progenitor cells attenuates acute lung injury in rabbits. Anesthesiology 2008; 108: 392–401
  • Debatin KM, Wei J, Beltinger C. Endothelial progenitor cells for cancer gene therapy. Gene Ther 2008; 15: 780–6
  • Chunmeng ST. Skin: a promising reservoir for adult stem cell populations. Med Hypoth 2004; 62: 683–8
  • Shi C, Zhu Y, Su Y, Cheng T. Stem cells and their applications in skin-cell therapy. Trends Biotech 2006; 24: 48–52
  • Jahoda CA, Whitehouse J, Reynolds AJ, Hole N. Hair follicle dermal cells differentiate into adipogenic and osteogenic lineages. Exp Dermatol 2003; 12: 849–59
  • Toma JG, McKenzie IA, Bagli D, Miller FD. Isolation and characterization of multipotent skin-derived precursors from human skin. Stem Cells 2005; 23: 727–37
  • Morasso MI, Tomic-Canic M. Epidermal stem cells: the cradle of epidermal determination, differentiation and wound healing. Biol Cell 2005; 97: 173–83
  • Ohyama M. Hair follicle bulge: a fascinating reservoir of epithelial stem cells. J Dermatol Sci 2007; 46: 81–9
  • Cotsarelis G. Epithelial stem cells: a folliculocentric view. J Invest Dermatol 2006; 126: 1459–68
  • Paus R, Nickoloff BJ, Ito T. A. ‘hairy’ privilege. Trends Immunol 2005; 26: 32–40
  • Reynolds AJ, Lawrence C, Cserhalmi-Friedman PB, et al. Trans-gender induction of hair follicles. Nature 1999; 402: 33–4
  • Feutz AC, Barrandon Y, Monard D. Control of thrombin signaling through PI3K is a mechanism underlying plasticity between hair follicle dermal sheath and papilla cells. J Cell Sci 2008; 121: 1435–43
  • Bickenbach JR, Stern MM, Grinnell K, et al. Epidermal stem cells have the potential to assist in healing damaged tissues. J Invest Derm Symp Proc 2006; 11: 118–23
  • Hoogduijn MJ, Gorjup E, Genever PG. Comparative characterization of hair follicle dermal stem cells and bone marrow mesenchymal stem cells. Stem Cells Dev 2006; 15: 49–60
  • Miyazaki M, Zuk PA, Zou J, et al. Comparison of human mesenchymal stem cells derived from adipose tissue and bone marrow for ex vivo gene therapy in rat spinal fusion model. Spine 2008; 33: 863–9
  • Keimani AJ, Fathi F, Mowla SJ. Characterization and genetic manipulation of human umbilical cord vein mesenchymal stem cells: potential application in cell based gene therapy. Rejuvenation Res 2008; 11: 379–86
  • Lataillade JJ, Doucet C, Bey E, et al. New approach to radiation burn treatment by dosimetry-guided surgery combined with autologous mesenchymal stem cell therapy. Regen Med 2007; 2: 785–94
  • Badiavas EV, Ford D, Liu P, et al. Long-term bone marrow culture and its clinical potential in chronic wound healing. Wound Repair Regen 2007; 15: 856–65

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.