Concise review: bone marrow-derived stem/progenitor cells in cutaneous repair and regeneration

Yaojiong Wu, Robert C H Zhao, Edward E Tredget, Yaojiong Wu, Robert C H Zhao, Edward E Tredget

Abstract

Our understanding of the role of bone marrow (BM)-derived cells in cutaneous homeostasis and wound healing had long been limited to the contribution of inflammatory cells. Recent studies, however, suggest that the BM contributes a significant proportion of noninflammatory cells to the skin, which are present primarily in the dermis in fibroblast-like morphology and in the epidermis in a keratinocyte phenotype; and the number of these BM-derived cells increases markedly after wounding. More recently, several studies indicate that mesenchymal stem cells derived from the BM could significantly impact wound healing in diabetic and nondiabetic animals, through cell differentiation and the release of paracrine factors, implying a profound therapeutic potential. This review discusses the most recent understanding of the contribution of BM-derived noninflammatory cells to cutaneous homeostasis and wound healing.

Figures

Figure 1
Figure 1
Bone marrow-derived mesenchymal stem cells (BM-MSCs) in cutaneous regeneration. (A, B): Keratinocytes loaded on a collagen gel containing BM-MSCs formed rete ridge-like structure, whereas keratinocytes loaded on a collagen gel containing dermal fibroblasts did not. Images adapted from Aoki et al. [58]. Image courtesy of Molecular Biology of the Cell. (C): BM-MSCs (green) from GFP-expressing mice were injected around the excisional wound and applied on the wound bed in Matrigel in Balb/C mice. At day 7, some mesenchymal stem cells (yellow) expressed keratinocyte marker cytokeratins (red) and formed structures similar to those observed in the study of Aoki et al. (A). Abbreviations: BMSC, bone morrow-derived mesenchymal stem cells; H&E, hematoxylin and eosin stain.
Figure 2
Figure 2
Paracrine effect of Bone marrow-derived mesenchymal stem cells (BM-MSCs) in wound healing. BM-MSCs (green) in a cutaneous wound release growth factors such as IGF-1, EGF, and KGF to promote the proliferation of keratinocytes, release proangiogenic cytokines such as VEGF-a and Ang-1 to enhance angiogenesis, release chemokines such as MIP-1 to recruit monocytes into the wound, and release cytokines such as SDF-1 and G-CSF to recruit EPCs into the wound. Abbreviations: ang-1, angiopoietin-1; EGF, epidermal growth factor; EPC, endothelial progenitor cell; FB, fibroblast; G-CSF, granulocyte colony-stimulating factor; IGF, insulin-like growth factor; KGF, keratinocyte growth factor; MIP-1, macrophage inflammatory protein-1; MSC, mesenchymal stem cell; SDF1, Stromal cell-derived factor-1; VEGF, vascular endothelial growth factor.

References

    1. Fathke C, Wilson L, Hutter J, et al. Contribution of bone marrow-derived cells to skCollagen deposition and wound repair. Stem Cells. 2004;22:812–822.
    1. Deng W, Han Q, Liao L, et al. Engrafted bone marrow-derived flk-(1+) mesenchymal stem cells regenerate skin tissue. Tissue Eng. 2005;11:110–119.
    1. Brittan M, Braun KM, Reynolds LE, et al. Bone marrow cells engraft within the epidermis and proliferate in vivo with no evidence of cell fusion. J Pathol. 2005;205:1–13.
    1. Devine SM, Cobbs C, Jennings M, et al. Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood. 2003;101:2999–3001.
    1. Kawada H, Fujita J, Kinjo K, et al. Nonhematopoietic mesenchymal stem cells can be mobilized and differentiate into cardiomyocytes after myocardial infarction. Blood. 2004;104:3581–3587.
    1. Wu Y, Chen L, Scott PG, et al. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells. 2007;25:2648–2659.
    1. 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;13:1299–1312.
    1. Phillips RJ, Burdick MD, Hong K, et al. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J Clin Invest. 2004;114:438–446.
    1. Bellini A, Mattoli S. The role of the fibrocyte, a bone marrow-derived mesenchymal progenitor, in reactive and reparative fibroses. Lab Invest. 2007;87:858–870.
    1. Urbich C, Dimmeler S. Endothelial progenitor cells: Characterization and role in vascular biology. Circ Res. 2004;95:343–353.
    1. Hirschi KK, Ingram DA, Yoder MC. Assessing identity, phenotype, and fate of endothelial progenitor cells. Arterioscler Thromb Vasc Biol. 2008;28:1584–1595.
    1. Lagasse E, Connors H, Al Dhalimy M, et al. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med. 2000;6:1229–1234.
    1. Orlic D, Kajstura J, Chimenti S, et al. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;410:701–705.
    1. Nygren JM, Jovinge S, Breitbach M, et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat Med. 2004;10:494–501.
    1. Balsam LB, Wagers AJ, Christensen JL, et al. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature. 2004;428:668–673.
    1. Horwitz EM. MSC: A coming of age in regenerative medicine. Cytotherapy. 2006;8:194–195.
    1. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147.
    1. Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276:71–74.
    1. McNiece I. Delivering cellular therapies: Lessons learned from ex vivo culture and clinical applications of hematopoietic cells. Semin Cell Dev Biol. 2007;18:839–845.
    1. Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res. 2004;95:9–20.
    1. Sutherland FWH, Perry TE, Yu Y, et al. From stem cells to viable autologous semilunar heart valve. Circulation. 2005;111:2783–2791.
    1. Noel D, Djouad F, Jorgense C. Regenerative medicine through mesenchymal stem cells for bone and cartilage repair. Curr Opin Investig Drugs. 2002;3:1000–1004.
    1. Awad HA, Butler DL, Boivin GP, et al. Autologous mesenchymal stem cell-mediated repair of tendon. Tissue Eng. 1999;5:267–277.
    1. Awad HA, Butler DL, Harris MT, et al. In vitro characterization of mesenchymal stem cell-seeded collagen scaffolds for tendon repair: Effects of initial seeding density on contraction kinetics. J Biomed Mater Res. 2000;51:233–240.
    1. Wakitani S, Yamamoto T. Response of the donor and recipient cells in mesenchymal cell transplantation to cartilage defect. Microsc Res Tech. 2002;58:14–18.
    1. Murphy JM, Fink DJ, Hunziker EB, et al. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum. 2003;48:3464–3474.
    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. Castro-Malaspina H, Gay RE, Resnick G, et al. Characterization of human bone marrow fibroblast colony-forming cells (CFU-F) and their progeny. Blood. 1980;56:289–301.
    1. Chen L, Tredget EE, Wu PY, et al. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One. 2008;3:e1886.
    1. Vaage J, Lindblad WJ. Production of collagen type I by mouse peritoneal macrophages. J Leukoc Biol. 1990;48:274–280.
    1. Martin P. Wound healing–Aiming for perfect skin regeneration. Science. 1997;276:75–81.
    1. Armour A, Scott PG, Tredget EE. Cellular and molecular pathology of HTS: Basis for treatment. Wound Repair Regen. 2007;15(Suppl 1):S6–S17.
    1. Schnoor M, Cullen P, Lorkowski J, et al. Production of type VI collagen by human macrophages: A new dimension in macrophage functional heterogeneity. J Immunol. 2008;180:5707–5719.
    1. Roufosse CA, Direkze NC, Otto WR, et al. Circulating mesenchymal stem cells. Int J Biochem Cell Biol. 2004;36:585–597.
    1. Shih DT, Lee DC, Chen SC, et al. Isolation and characterization of neurogenic mesenchymal stem cells in human scalp tissue. Stem Cells. 2005;23:1012–1020.
    1. 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.
    1. Nuttall ME, Patton AJ, Olivera DL, et al. Human trabecular bone cells are able to express both osteoblastic and adipocytic phenotype: Implications for osteopenic disorders. J Bone Miner Res. 1998;13:371–382.
    1. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: Implications for cell-based therapies. Tissue Eng. 2001;7:211–228.
    1. Lee RH, Kim B, Choi I, et al. Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem. 2004;14:311–324.
    1. Jeong JA, Hong SH, Gang EJ, et al. Differential gene expression profiling of human umbilical cord blood-derived mesenchymal stem cells by DNA microarray. Stem Cells. 2005;23:584–593.
    1. Bieback K, Kern S, Kluter H, et al. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells. 2004;22:625–634.
    1. Silva WA, Jr, Covas DT, Panepucci RA, et al. The profile of gene expression of human marrow mesenchymal stem cells. Stem Cells. 2003;21:661–669.
    1. Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: Candidate MSC-like cells from umbilical cord. Stem Cells. 2003;21:105–110.
    1. Tondreau T, Meuleman N, Delforge A, et al. Mesenchymal stem cells derive from CD133 positive cells in mobilized peripheral blood and cord blood: Proliferation, Oct-4 expression and plasticity. Stem Cells. 2005;23:1105–1112.
    1. Otsuru S, Tamai K, Yamazaki T, et al. Circulating bone marrow-derived osteoblast progenitor cells are recruited to the bone-forming site by the CXCR4/stromal cell-derived factor-1 pathway. Stem Cells. 2008;26:223–234.
    1. Ripa RS, Haack-Sorensen M, Wang Y, et al. Bone marrow derived mesenchymal cell mobilization by granulocyte-colony stimulating factor after acute myocardial infarction: Results from the Stem Cells in Myocardial Infarction (STEMMI) trial. Circulation. 2007;116(11) Suppl:I24–I30.
    1. Kuznetsov SA, Mankani MH, Gronthos S, et al. Circulating skeletal stem cells. J Cell Biol. 2001;153:1133–1140.
    1. Fernandez M, Simon V, Herrera G, et al. Detection of stromal cells in peripheral blood progenitor cell collections from breast cancer patients. Bone Marrow Transplant. 1997;20:265–271.
    1. Wexler SA, Donaldson C, Denning-Kendall P, et al. Adult bone marrow is a rich source of human mesenchymal ‘stem’ cells but umbilical cord and mobilized adult blood are not. Br J Haematol. 2003;121:368–374.
    1. Lazarus HM, Haynesworth SE, Gerson SL, et al. Human bone marrow-derived mesenchymal (stromal) progenitor cells (MPCs) cannot be recovered from peripheral blood progenitor cell collections. J Hematother. 1997;6:447–455.
    1. Kassis I, Zangi L, Rivkin R, et al. Isolation of mesenchymal stem cells from G-CSF-mobilized human peripheral blood using fibrin microbeads. Bone Marrow Transplant. 2006;37:967–976.
    1. Rochefort GY, Delorme B, Lopez A, et al. Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells. 2006;24:2202–2208.
    1. Badiavas EV, Abedi M, Butmarc J, et al. Participation of bone marrow derived cells in cutaneous wound healing. J Cell Physiol. 2003;196:245–250.
    1. Borue X, Lee S, Grove J, et al. Bone marrow-derived cells contribute to epithelial engraftment during wound healing. Am J Pathol. 2004;165:1767–1772.
    1. Baxter MA, Wynn RF, Jowitt SN, et al. Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion. Stem Cells. 2004;22:675–682.
    1. Harris RG, Herzog EL, Bruscia EM, et al. Lack of a fusion requirement for development of bone marrow-derived epithelia. Science. 2004;305:90–93.
    1. Kataoka K, Medina RJ, Kageyama T, et al. Participation of adult mouse bone marrow cells in reconstitution of skin. Am J Pathol. 2003;163:1227–1231.
    1. Aoki S, Toda S, Ando T, et al. Bone marrow stromal cells, preadipocytes, and dermal fibroblasts promote epidermal regeneration in their distinctive fashions. Mol Biol Cell. 2004;15:4647–4657.
    1. Duffield JS, Bonventre JV. Kidney tubular epithelium is restored without replacement with bone marrow-derived cells during repair after ischemic injury. Kidney Int. 2005;68:1956–1961.
    1. Ma Y, Xu Y, Xiao Z, et al. Reconstruction of chemically burned rat corneal surface by bone marrow-derived human mesenchymal stem cells. Stem Cells. 2006;24:315–321.
    1. Phinney DG, Prockop DJ. Concise review: Mesenchymal stem/multipotent stromal cells: The state of transdifferentiation and modes of tissue repair—current views. Stem Cells. 2007;25:2896–2902.
    1. Krampera M, Pasini A, Rigo A, et al. HB-EGF/HER-1 signaling in bone marrow mesenchymal stem cells: Inducing cell expansion and reversibly preventing multilineage differentiation. Blood. 2005;106:59–66.
    1. Crisostomo PR, Wang M, Wairiuko GM, et al. High passage number of stem cells adversely affects stem cell activation and myocardial protection. Shock. 2006;26:575–580.
    1. Fu X, Fang L, Li X, et al. Enhanced wound-healing quality with bone marrow mesenchymal stem cells autografting after skin injury. Wound Repair Regen. 2006;14:325–335.
    1. McFarlin K, Gao X, Liu YB, et al. Bone marrow-derived mesenchymal stromal cells accelerate wound healing in the rat. Wound Repair Regen. 2006;14:471–478.
    1. Wu Y, Wang J, Scott PG, et al. Bone marrow-derived stem cells in wound healing: A review. Wound Repair Regen. 2007;15(Suppl 1):S18–S26.
    1. Kwon DS, Gao X, Liu YB, et al. Treatment with bone marrow-derived stromal cells accelerates wound healing in diabetic rats. Int Wound J. 2008;5:453–463.
    1. Galiano RD, Michaels J, Dobryansky M, et al. Quantitative and reproducible murine model of excisional wound healing. Wound Repair Regen. 2004;12:485–492.
    1. Carlson MA, Thompson JS. Wound matrix attachment regulates actin content and organization in cells of the granulation tissue. Wound Repair Regen. 2005;13:84–92.
    1. Chen L, Tredget EE, Liu C, et al. Analysis of allogenicity of mesenchymal stem cells in engraftment and wound healing in mice. PLoS One. 2009;4:e7119.
    1. Zhou S, Greenberger JS, Epperly MW, et al. Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts. Aging Cell. 2008;7:335–343.
    1. Caplan AI. Why are MSCs therapeutic? New data: New insight. J Pathol. 2009;217:318–324.
    1. Carbonare LD, Valenti MT, Zanatta M, et al. Circulating mesenchymal stem cells with abnormal osteogenic differentiation in patients with osteoporosis. Arthritis Rheum. 2009;60:3356–3365.
    1. Murphy JM, Dixon K, Beck S, et al. Reduced chondrogenic and adipogenic activity of mesenchymal stem cells from patients with advanced osteoarthritis. Arthritis Rheum. 2002;46:704–713.
    1. Heeschen C, Lehmann R, Honold J, et al. Profoundly reduced neovascularization capacity of bone marrow mononuclear cells derived from patients with chronic ischemic heart disease. Circulation. 2004;109:1615–1622.
    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;180:2581–2587.
    1. Kinnaird T, Stabile E, Burnett MS, et al. Local delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation. 2004;109:1543–1549.
    1. Mayer H, Bertram H, Lindenmaier W, et al. Vascular endothelial growth factor (VEGF-A) expression in human mesenchymal stem cells: Autocrine and paracrine role on osteoblastic and endothelial differentiation. J Cell Biochem. 2005;95:827–839.
    1. Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med. 1999;341:738–746.
    1. Galeano M, Altavilla D, Cucinotta D, et al. Recombinant human erythropoietin stimulates angiogenesis and wound healing in the genetically diabetic mouse. Diabetes. 2004;53:2509–2517.
    1. Desbois-Mouthon C, Wendum D, Cadoret A, et al. Hepatocyte proliferation during liver regeneration is impaired in mice with liver-specific IGF-1R knockout. FASEB J. 2006;20:773–775.
    1. Welch S, Plank D, Witt S, et al. Cardiac-specific IGF-1 expression attenuates dilated cardiomyopathy in tropomodulin-overexpressing transgenic mice. Circ Res. 2002;90:641–648.
    1. Keller ET, Wanagat J, Ershler WB. Molecular and cellular biology of interleukin-6 and its receptor. Front Biosci. 1996;1:d340–d357.
    1. Xing L, Schwarz EM, Boyce BF. Osteoclast precursors, RANKL/RANK, and immunology. Immunol Rev. 2005;208:19–29.
    1. Falanga V. Wound healing and its impairment in the diabetic foot. Lancet. 2005;366:1736–1743.
    1. Goova MT, Li J, Kislinger T, et al. Blockade of receptor for advanced glycation end-products restores effective wound healing in diabetic mice. Am J Pathol. 2001;159:513–525.
    1. Martin P, Leibovich SJ. Inflammatory cells during wound repair: The good, the bad and the ugly. Trends Cell Biol. 2005;15:599–607.
    1. Amado LC, Saliaris AP, Schuleri KH, et al. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci USA. 2005;102:11474–11479.
    1. DiPietro LA, Burdick M, Low QE, et al. MIP-1alpha as a critical macrophage chemoattractant in murine wound repair. J Clin Invest. 1998;101:1693–1698.
    1. Dewald O, Zymek P, Winkelmann K, et al. CCL2/monocyte chemoattractant protein-1 regulates inflammatory responses critical to healing myocardial infarcts. Circ Res. 2005;96:881–889.
    1. Pierce GF, Mustoe TA, Lingelbach J, et al. Transforming growth factor {beta} reverses the glucocorticoid-induced wound-healing deficit in rats: Possible regulation in macrophages by platelet-derived growth factor. Proc Natl Acad Sci USA. 1989;86:2229–2233.
    1. Danon D, Kowatch MA, Roth GS. Promotion of wound repair in old mice by local injection of macrophages. Proc Natl Acad Sci USA. 1989;86:2018–2020.
    1. Arnold F, West DC. Angiogenesis in wound healing. Pharmacol Ther. 1991;52:407–422.
    1. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964–967.
    1. Shi Q, Rafii S, Wu MH-D, et al. Evidence for circulating bone marrow-derived endothelial cells. Blood. 1998;92:362–367.
    1. Asahara T, Masuda H, Takahashi T, et al. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 1999;85:221–228.
    1. Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med. 2003;9:702–712.
    1. Mendez-Ferrer S, Lucas D, Battista M, et al. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452:442–447.
    1. Lucas D, Battista M, Shi PA, et al. Mobilized hematopoietic stem cell yield depends on species-specific circadian timing. Cell Stem Cell. 2008;3:364–366.
    1. Busik JV, Tikhonenko M, Bhatwadekar A, et al. Diabetic retinopathy is associated with bone marrow neuropathy and a depressed peripheral clock. J Exp Med. 2009;206:2897–2906.
    1. Schmidt-Lucke C, Rossig L, Fichtlscherer S, et al. Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: Proof of concept for the clinical importance of endogenous vascular repair. Circulation. 2005;111:2981–2987.
    1. Werner N, Kosiol S, Schiegl T, et al. Circulating endothelial progenitor cells and cardiovascular outcomes. New Engl J Med. 2005;353:999–1007.
    1. Grisar J, Aletaha D, Steiner CW, et al. Depletion of endothelial progenitor cells in the peripheral blood of patients with rheumatoid arthritis. Circulation. 2005;111:204–211.
    1. Hoetzer GL, van Guilder GP, Irmiger HM, et al. Aging, exercise, and endothelial progenitor cell clonogenic and migratory capacity in men. J Appl Physiol. 2007;102:847–852.
    1. Lee SH, Wolf PL, Escudero R, et al. Early expression of angiogenesis factors in acute myocardial ischemia and infarction. N Engl J Med. 2000;342:626–633.
    1. Pillarisetti K, Gupta SK. Cloning and relative expression analysis of rat stromal cell derived factor-1 (SDF-1)1: SDF-1 alpha mRNA is selectively induced in rat model of myocardial infarction. Inflammation. 2001;25:293–300.
    1. Gill M, Dias S, Hattori K, et al. Vascular trauma induces rapid but transient mobilization of VEGFR2(+)AC133(+) endothelial precursor cells. Circ Res. 2001;88:167–174.
    1. 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;23:1571–1578.
    1. Rehman J, Li J, Orschell CM, et al. Peripheral blood “endothelial progenitor cells” are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation. 2003;107:1164–1169.
    1. Edelberg JM, Tang L, Hattori K, et al. Young adult bone marrow-derived endothelial precursor cells restore aging-impaired cardiac angiogenic function. Circ Res. 2002;90:E89–E93.
    1. Barcelos LS, Duplaa C, Krankel N, et al. Human CD133+ progenitor cells promote the healing of diabetic ischemic ulcers by paracrine stimulation of angiogenesis and activation of Wnt signaling. Circ Res. 2009;104:1095–1102.
    1. Loomans CJ, de Koning EJ, Staal FJ, et al. Endothelial progenitor cell dysfunction: A novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes. 2004;53:195–199.
    1. Tepper OM, Galiano RD, Capla JM, et al. Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation. 2002;106:2781–2786.
    1. Stepanovic V, Awad O, Jiao C, et al. Leprdb diabetic mouse bone marrow cells inhibit skin wound vascularization but promote wound healing. Circ Res. 2003;92:1247–1253.
    1. Gallagher KA, Liu ZJ, Xiao M, et al. Diabetic impairments in NO-mediated endothelial progenitor cell mobilization and homing are reversed by hyperoxia and SDF-1 alpha. J Clin Invest. 2007;117:1249–1259.
    1. Chesney J, Bacher M, Bender A, et al. The peripheral blood fibrocyte is a potent antigen-presenting cell capable of priming naive T cells in situ. Proc Natl Acad Sci USA. 1997;94:6307–6312.
    1. Quan TE, Cowper S, Wu SP, et al. Circulating fibrocytes: Collagen-secreting cells of the peripheral blood. Int J Biochem Cell Biol. 2004;36:598–606.
    1. Bucala R, Spiegel LA, Chesney J, et al. Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair. Mol Med. 1994;1:71–81.
    1. Okada H, Kalluri R. Cellular and molecular pathways that lead to progression and regression of renal fibrogenesis. Curr Mol Med. 2005;5:467–474.
    1. Mori L, Bellini A, Stacey MA, et al. Fibrocytes contribute to the myofibroblast population in wounded skin and originate from the bone marrow. Exp Cell Res. 2005;304:81–90.
    1. Yang L, Scott PG, Dodd C, et al. Identification of fibrocytes in postburn hypertrophic scar. Wound Repair Regen. 2005;13:398–404.
    1. Wang J, Dodd C, Shankowsky HA, et al. Deep dermal fibroblasts contribute to hypertrophic scarring. Lab Invest. 2008;88:1278–1290.
    1. Abe R, Donnelly SC, Peng T, et al. Peripheral blood fibrocytes: Differentiation pathway and migration to wound sites. J Immunol. 2001;166:7556–7562.
    1. Ortiz LA, Gambelli F, McBride C, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci USA. 2003;100:8407–8411.

Source: PubMed

3
Abonnere