Intradermal injection of human adipose-derived stem cells accelerates skin wound healing in nude mice

Jonathan Rodriguez, Fabien Boucher, Charlotte Lequeux, Audrey Josset-Lamaugarny, Ondine Rouyer, Orianne Ardisson, Héléna Rutschi, Dominique Sigaudo-Roussel, Odile Damour, Ali Mojallal, Jonathan Rodriguez, Fabien Boucher, Charlotte Lequeux, Audrey Josset-Lamaugarny, Ondine Rouyer, Orianne Ardisson, Héléna Rutschi, Dominique Sigaudo-Roussel, Odile Damour, Ali Mojallal

Abstract

Background: The use of stem cells from adipose tissue or adipose-derived stem cells (ASCs) in regenerative medicine could be an interesting alternative to bone marrow stem cells because they are easily accessible and available in large quantities. The aim of this study was to evaluate the potential effect of ASCs on the healing of 12 mm diameter-excisional wounds (around 110 mm(2)) in nude mice.

Methods: Thirty nude mice underwent surgery to create one 12-mm excisional wound per mouse (spontaneous healing, n = 6; Cytocare® 532, n = 12; ASCs, n = 12). The Galiano wound model was chosen to avoid shrinkage and thus slow the spontaneous healing (SH) of mouse skin, making it closer to the physiology of human skin healing. Transparent dressings were used to enable daily healing time measurements to be taken. Immunohistochemistry, histological and blood perfusion analysis were carried out on the healed skin.

Results: The in vivo results showed the effectiveness of using ASCs on reducing the time needed for complete healing to 21.2 days for SH, 17.4 days for vehicle alone (Cytocare® 532) and 14.6 days with the addition of ASCs (p < 0.001). Moreover, cutaneous perfusion of the healed wound was significantly improved in ASC-treated mice compared to SH group, as shown by laser Doppler flowmetry and the quantitation of blood vessels using immunohistochemistry of αsmooth muscle actin.

Conclusions: The tolerance and efficacy of cryopreserved ASCs to accelerate the complete closure of the wound by increasing the maturation of the skin and its blood perfusion, shows their therapeutic benefit in the wound healing context.

Figures

Fig. 1
Fig. 1
Immunophenotyping and trilineage differentiation of ASC. a ASCs were stained with antibodies and analyzed by flow cytometry. ASCs expressed markers CD73, CD90, and HLA ABC and were negative for CD45, CD14, and HLA DR. Representative histograms are shown. b ASCs were cultured in adipogenic, osteogenic or chondrogenic differentiation medium. Cells were fixed and stained with Oil Red O for adipogenesis, Alizarin red for osteogenesis and Alcian blue for chondrogenesis. Representative images are shown. Scale bar = 100 μm. ASCs adipose-derived stem cells, CD cluster of differentiation, HLA human leukocyte antigen
Fig. 2
Fig. 2
Influence of ASCs on wound healing kinetics. a After surgery, nude mice were intradermally injected or not with Cytocare® 532 or ASCs. Representative images are shown for 0, 7, 14, and 21 days post-surgery. Scale bar 5 mm. b Percentage of wound healing was monitored every day until complete wound closure. Results are expressed as means, with error bars indicating SEM. Statistical analysis was performed using the unpaired t-test. * p < 0.05, **p < 0.01 and † p < 0.001 compared to spontaneous healing. c Days to complete wound closure were noted. Results are expressed as means of days needed to reach complete closure and error bars indicate SEM. The Kruskal-Wallis test was followed by Dunn’s multiple comparison tests to estimate the significance of differences for between-group comparisons. *** p < 0.001 compared to spontaneous healing. ASCs adipose-derived stem cells
Fig. 3
Fig. 3
Wound histology after Masson’s trichrome and Picro Sirius red staining. Tissue sections obtained from the wound area at day 27 after cell injection were stained with Masson’s trichrome (left panel) and Picro Sirius red (right panel). Representative micrographs of wound histological images are shown. ASCs adipose-derived stem cells
Fig. 4
Fig. 4
Quantitative and functional aspects of blood perfusion in the healed tissue. a Quantification of perfusion at day 27 after surgery was performed on the healed tissue compared with normal skin using laser Doppler imaging. b Percentage of vasodilation after ACh iontophoresis and c after SNP iontophoresis in SH-, Cytocare® 532- and ASC-treated mice. d One-way ANOVA was followed by a multiple comparison test to estimate the significance of differences for between-group comparisons. Significance was defined at P < 0.05 and ** p < 0.01 vs. control. Tissue sections obtained from the wound area 27 days after cell injection were stained with antibody against alpha smooth muscle actin and blood vessels in the total wound area were quantified. Representative images of the staining are displayed for each group. The Kruskal-Wallis test was followed by Dunn’s multiple comparison test to estimate the significance of differences for between-group comparisons. * p < 0.05 compared to spontaneous healing. All data are expressed as mean ± SEM. Ach acetylcholine, ASCs adipose-derived stem cells, SH spontaneous healing, SNP sodium nitroprusside

References

    1. Mustoe T. Understanding chronic wounds: a unifying hypothesis on their pathogenesis and implications for therapy. Am J Surg. 2004;187:65S–70. doi: 10.1016/S0002-9610(03)00306-4.
    1. Riedel K, Ryssel H, Koellensperger E, Germann G, Kremer T. Pathogenesis of chronic wounds. Chir Z Für Alle Geb Oper Medizen. 2008;79:526–34. German.
    1. Epstein FH, Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med. 1999;341:738–46. doi: 10.1056/NEJM199909023411006.
    1. Sen CK, Gordillo GM, Roy S, Kirsner R, Lambert L, Hunt TK, et al. Human skin wounds: a major and snowballing threat to public health and the economy. Wound Repair Regen. 2009;17:763–71. doi: 10.1111/j.1524-475X.2009.00543.x.
    1. Berlanga-Acosta J. Diabetic lower extremity wounds: the rationale for growth factors-based infiltration treatment. Int Wound J. 2011;8:612–20. doi: 10.1111/j.1742-481X.2011.00840.x.
    1. Hu X, Sun H, Han C, Wang X, Yu W. Topically applied rhGM-CSF for the wound healing: a systematic review. Burns. 2011;37:729–41. doi: 10.1016/j.burns.2010.08.016.
    1. Shankaran V, Brooks M, Mostow E. Advanced therapies for chronic wounds: NPWT, engineered skin, growth factors, extracellular matrices. Dermatol Ther. 2013;26:215–21. doi: 10.1111/dth.12050.
    1. Greaves NS, Iqbal SA, Baguneid M, Bayat A. The role of skin substitutes in the management of chronic cutaneous wounds. Wound Repair Regen. 2013;21:194–210. doi: 10.1111/wrr.12029.
    1. Sibbald RG, Zuker R, Coutts P, Coelho S, Williamson D, Queen D. Using a dermal skin substitute in the treatment of chronic wounds secondary to recessive dystrophic epidermolysis bullosa: a case series. Ostomy Wound Manage. 2005;51:22–46.
    1. Gibbs S, van den Hoogenband HM, Kirtschig G, Richters CD, Spiekstra SW, Breetveld M, et al. Autologous full-thickness skin substitute for healing chronic wounds. Br J Dermatol. 2006;155:267–74. doi: 10.1111/j.1365-2133.2006.07266.x.
    1. Gimble JM, Guilak F. Differentiation potential of adipose derived adult stem (ADAS) cells. Curr Top Dev Biol. 2003;58:137–60. doi: 10.1016/S0070-2153(03)58005-X.
    1. Atalay S, Coruh A, Deniz K. Stromal vascular fraction improves deep partial thickness burn wound healing. Burns. 2014;40:1375–83. doi: 10.1016/j.burns.2014.01.023.
    1. Oedayrajsingh-Varma MJ, van Ham SM, Knippenberg M, Helder MN, Klein-Nulend J, Schouten TE, et al. Adipose tissue-derived mesenchymal stem cell yield and growth characteristics are affected by the tissue-harvesting procedure. Cytotherapy. 2006;8:166–77. doi: 10.1080/14653240600621125.
    1. Bourin P, Bunnell BA, Casteilla L, Dominici M, Katz AJ, March KL, et al. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT) Cytotherapy. 2013;15:641–8. doi: 10.1016/j.jcyt.2013.02.006.
    1. Mitchell JB, McIntosh K, Zvonic S, Garrett S, Floyd ZE, Kloster A, et al. Immunophenotype of human adipose-derived cells: temporal changes in stromal-associated and stem cell-associated markers. Stem Cells. 2006;24:376–85. doi: 10.1634/stemcells.2005-0234.
    1. Maharlooei MK, Bagheri M, Solhjou Z, Jahromi BM, Akrami M, Rohani L, et al. Adipose tissue derived mesenchymal stem cell (AD-MSC) promotes skin wound healing in diabetic rats. Diabetes Res Clin Pract. 2011;93:228–34. doi: 10.1016/j.diabres.2011.04.018.
    1. Huang SP, Hsu CC, Chang SC, Wang CH, Deng SC, Dai NT, et al. Adipose-derived stem cells seeded on acellular dermal matrix grafts enhance wound healing in a murine model of a full-thickness defect. Ann Plast Surg. 2012;69:656–62. doi: 10.1097/SAP.0b013e318273f909.
    1. Ju X, Pan F, Bai S, Tian X, Tong H, Wang J. An experimental study on repairing full-thickness skin wound by human acellular amniotic membrane loaded with adipose-derived stem cells in rats. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 2010;24:143–9. Chinese.
    1. Ebrahimian TG, Pouzoulet F, Squiban C, Buard V, André M, Cousin B, et al. Cell therapy based on adipose tissue-derived stromal cells promotes physiological and pathological wound healing. Arterioscler Thromb Vasc Biol. 2009;29:503–10. doi: 10.1161/ATVBAHA.108.178962.
    1. Lim JS, Yoo G. Effects of adipose-derived stromal cells and of their extract on wound healing in a mouse model. J Korean Med Sci. 2010;25:746–51. doi: 10.3346/jkms.2010.25.5.746.
    1. Lee SH, Lee JH, Cho KH. Effects of human adipose-derived stem cells on cutaneous wound healing in nude mice. Ann Dermatol. 2011;23:150–5. doi: 10.5021/ad.2011.23.2.150.
    1. Lin YC, Grahovac T, Oh SJ, Ieraci M, Rubin JP, Marra KG. Evaluation of a multi-layer adipose-derived stem cell sheet in a full-thickness wound healing model. Acta Biomater. 2013;9:5243–50. doi: 10.1016/j.actbio.2012.09.028.
    1. Amos PJ, Kapur SK, Stapor PC, Shang H, Bekiranov S, Khurgel M, et al. Human adipose-derived stromal cells accelerate diabetic wound healing: impact of cell formulation and delivery. Tissue Eng Part A. 2010;16:1595–606. doi: 10.1089/ten.tea.2009.0616.
    1. Altman AM, Yan Y, Matthias N, Bai X, Rios C, Mathur AB, et al. IFATS collection: human adipose-derived stem cells seeded on a silk fibroin-chitosan scaffold enhance wound repair in a murine soft tissue injury model. Stem Cells. 2009;27:250–8. doi: 10.1634/stemcells.2008-0178.
    1. Nie C, Yang D, Xu J, Si Z, Jin X, Zhang J. Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis. Cell Transplant. 2011;20:205–16. doi: 10.3727/096368910X520065.
    1. Huang SP, Huang CH, Shyu JF, Lee HS, Chen SG, Chan JY, et al. Promotion of wound healing using adipose-derived stem cells in radiation ulcer of a rat model. J Biomed Sci. 2013;20:51. doi: 10.1186/1423-0127-20-51.
    1. Nambu M, Ishihara M, Kishimoto S, Yanagibayashi S, Yamamoto N, Azuma R, et al. Stimulatory effect of autologous adipose tissue-derived stromal cells in an atelocollagen matrix on wound healing in diabetic db/db mice. J Tissue Eng. 2011;2011:158105.
    1. Jiang D, Qi Y, Walker NG, Sindrilaru A, Hainzl A, Wlaschek M, et al. The effect of adipose tissue derived MSCs delivered by a chemically defined carrier on full-thickness cutaneous wound healing. Biomaterials. 2013;34:2501–15. doi: 10.1016/j.biomaterials.2012.12.014.
    1. Yun IS, Jeon YR, Lee WJ, Lee JW, Rah DK, Tark KC, et al. Effect of human adipose derived stem cells on scar formation and remodeling in a pig model: a pilot study. Dermatol Surg. 2012;38:1678–88. doi: 10.1111/j.1524-4725.2012.02495.x.
    1. Hanson SE, Kleinbeck KR, Cantu D, Kim J, Bentz ML, Faucher LD, et al. Local delivery of allogeneic bone marrow and adipose tissue-derived mesenchymal stromal cells for cutaneous wound healing in a porcine model. J Tissue Eng Regen Med. 2013
    1. Forcheron F, Agay D, Scherthan H, Riccobono D, Herodin F, Meineke V, et al. Autologous adipocyte derived stem cells favour healing in a minipig model of cutaneous radiation syndrome. PLoS One. 2012;7(2) doi: 10.1371/journal.pone.0031694.
    1. Hadad I, Johnstone BH, Brabham JG, Blanton MW, Rogers PI, Fellers C, et al. Development of a porcine delayed wound-healing model and its use in testing a novel cell-based therapy. Int J Radiat Oncol. 2010;78:888–96. doi: 10.1016/j.ijrobp.2010.05.002.
    1. Blanton MW, Hadad I, Johnstone BH, Mund JA, Rogers PI, Eppley BL, et al. Adipose stromal cells and platelet-rich plasma therapies synergistically increase revascularization during wound healing. Plast Reconstr Surg. 2009;123(Suppl):56S–64. doi: 10.1097/PRS.0b013e318191be2d.
    1. Pelizzo G, Avanzini MA, Cornaglia AI, Osti M, Romano P, Avolio L, et al. Mesenchymal stromal cells for cutaneous wound healing in a rabbit model: pre-clinical study applicable in the pediatric surgical setting. J Transl Med. 2015;13:219. doi: 10.1186/s12967-015-0580-3.
    1. Hong SJ, Jia SX, 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) doi: 10.1371/journal.pone.0055640.
    1. Galiano RD, Michaels J, 5th, Dobryansky M, Levine JP, Gurtner GC. Quantitative and reproducible murine model of excisional wound healing. Wound Repair Regen. 2004;12:485–92. doi: 10.1111/j.1067-1927.2004.12404.x.
    1. Lequeux C, Rodriguez J, Boucher F, Rouyer O, Damour O, Mojallal A, et al. In vitro and in vivo biocompatibility, bioavailability and tolerance of an injectable vehicle for adipose-derived stem/stromal cells for plastic surgery indications. J Plast Reconstr Aesthet Surg. 2015;68:1491–7. doi: 10.1016/j.bjps.2015.07.022.
    1. Coleman WP, 4th, Hendry SL., 2nd Principles of liposuction. Semin Cutan Med Surg. 2006;25:138–44. doi: 10.1016/j.sder.2006.06.006.
    1. Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 1998;238:265–72. doi: 10.1006/excr.1997.3858.
    1. Sotocinal SG, Sorge RE, Zaloum A, Tuttle AH, Martin LJ, Wieskopf JS, et al. The Rat Grimace Scale: a partially automated method for quantifying pain in the laboratory rat via facial expressions. Mol Pain. 2011;7:55. doi: 10.1186/1744-8069-7-55.
    1. Sigaudo-Roussel D, Demiot C, Fromy B, Koïtka A, Lefthériotis G, Abraham P, et al. Early endothelial dysfunction severely impairs skin blood flow response to local pressure application in streptozotocin-induced diabetic mice. Diabetes. 2004;53:1564–9. doi: 10.2337/diabetes.53.6.1564.
    1. Fromy B, Sigaudo-Roussel D, Gaubert-Dahan ML, Rousseau P, Abraham P, Benzoni D, et al. Aging-associated sensory neuropathy alters pressure-induced vasodilation in humans. J Invest Dermatol. 2009;130:849–55. doi: 10.1038/jid.2009.279.
    1. Chang Q, Li J, Dong Z, Liu L, Lu F. Quantitative volumetric analysis of progressive hemifacial atrophy corrected using stromal vascular fraction-supplemented autologous fat grafts. Dermatol Surg. 2013;39:1465–73.
    1. Gentile P, Orlandi A, Scioli MG, Pasquali CD, Bocchini I, Curcio CB, et al. A comparative translational study: the combined use of enhanced stromal vascular fraction and platelet-rich plasma improves fat grafting maintenance in breast reconstruction. Stem Cells Transl Med. 2012;1:341–51. doi: 10.5966/sctm.2011-0065.
    1. Yoshimura K, Sato K, Aoi N, Kurita M, Hirohi T, Harii K. Cell-assisted lipotransfer for cosmetic breast augmentation: supportive use of adipose-derived stem/stromal cells. Aesthetic Plast Surg. 2008;32:48–55. doi: 10.1007/s00266-007-9019-4.
    1. Mesimäki K, Lindroos B, Törnwall J, Mauno J, Lindqvist C, Kontio R, et al. Novel maxillary reconstruction with ectopic bone formation by GMP adipose stem cells. Int J Oral Maxillofac Surg. 2009;38:201–9. doi: 10.1016/j.ijom.2009.01.001.
    1. Kølle SF, Fischer-Nielsen A, Mathiasen AB, Elberg JJ, Oliveri RS, Glovinski PV, et al. Enrichment of autologous fat grafts with ex-vivo expanded adipose tissue-derived stem cells for graft survival: a randomised placebo-controlled trial. Lancet. 2013;382:1113–20. doi: 10.1016/S0140-6736(13)61410-5.
    1. De la Portilla F, Alba F, García-Olmo D, Herrerías JM, González FX, Galindo A. Expanded allogeneic adipose-derived stem cells (eASCs) for the treatment of complex perianal fistula in Crohn’s disease: results from a multicenter phase I/IIa clinical trial. Int J Colorectal Dis. 2013;28:313–23. doi: 10.1007/s00384-012-1581-9.
    1. Sterodimas A, de Faria J, Nicaretta B, Papadopoulos O, Papalambros E, Illouz YG. Cell-assisted lipotransfer. Aesthet Surg J. 2010;30:78–81. doi: 10.1177/1090820X10362730.
    1. Matsumoto D, Sato K, Gonda K, Takaki Y, Shigeura T, Sato T, et al. Cell-assisted lipotransfer: supportive use of human adipose-derived cells for soft tissue augmentation with lipoinjection. Tissue Eng. 2006;12:3375–82. doi: 10.1089/ten.2006.12.3375.
    1. Le H, Kleinerman R, Lerman OZ, Brown D, Galiano R, Gurtner GC, et al. Hedgehog signaling is essential for normal wound healing. Wound Repair Regen. 2008;16:768–73. doi: 10.1111/j.1524-475X.2008.00430.x.
    1. Nie C, Zhang G, Yang D, Liu T, Liu D, Xu J, et al. Targeted delivery of adipose-derived stem cells via acellular dermal matrix enhances wound repair in diabetic rats. J Tissue Eng Regen Med. 2012;9(3):224–35. doi: 10.1002/term.1622.
    1. Martínez-Santamaría L, Conti CJ, Llames S, García E, Retamosa L, Holguín A, et al. The regenerative potential of fibroblasts in a new diabetes-induced delayed humanised wound healing model. Exp Dermatol. 2013;22:195–201. doi: 10.1111/exd.12097.
    1. Michaels 5th J, Dobryansky M, Galiano RD, Bhatt KA, Ashinoff R, Ceradini DJ, et al. Topical vascular endothelial growth factor reverses delayed wound healing secondary to angiogenesis inhibitor administration. Wound Repair Regen. 2005;13:506–12.
    1. Mojallal A, Lequeux C, Shipkov C, Rifkin L, Rohrich R, Duclos A, et al. Stem cells, mature adipocytes, and extracellular scaffold: what does each contribute to fat graft survival? Aesthetic Plast Surg. 2011;35:1061–72. doi: 10.1007/s00266-011-9734-8.
    1. Meyer K, Palmer JW. The polysaccharide of the vitreous humor. J Biol Chem. 1934;107:629–34.
    1. Gall Y. Hyaluronic acid: structure, metabolism and implication in cicatrisation. Ann Dermatol Vénéréologie. 2010;37 Suppl 1:S30–9. French.
    1. McKee CM, Lowenstein CJ, Horton MR, Wu J, Bao C, Chin BY, et al. Hyaluronan fragments induce nitric-oxide synthase in murine macrophages through a nuclear factor κB-dependent mechanism. J Biol Chem. 1997;272:8013–8. doi: 10.1074/jbc.272.12.8013.
    1. Horton MR, Shapiro S, Bao C, Lowenstein CJ, Noble PW. Induction and regulation of macrophage metalloelastase by hyaluronan fragments in mouse macrophages. J Immunol. 1999;162:4171–6.
    1. Kim YM, Oh SH, Choi JS, Lee S, Ra JC, Lee JH, et al. Adipose-derived stem cell-containing hyaluronic acid/alginate hydrogel improves vocal fold wound healing. Laryngoscope. 2014;124:E64–72. doi: 10.1002/lary.24405.
    1. Junqueira LC, Bignolas G, Brentani RR. Picrosirius staining plus polarization microscopy, a specific method for collagen detection in tissue sections. Histochem J. 1979;11:447–55. doi: 10.1007/BF01002772.
    1. Lc J, Cossermelli W, Brentani R. Differential staining of collagens type I, II and III by Sirius Red and polarization microscopy. Arch Histol Jpn. 1978;41:267–74. doi: 10.1679/aohc1950.41.267.
    1. Metral E, Dos Santos M, Thépot A, Rachidi W, Mojallal A, Auxenfans C, Damour O. Adipose-derived stem cells promote skin homeostasis and prevent its senescence in an in vitro skin model. J Stem Cell Res Ther. 2014;4:194. .

Source: PubMed

3
구독하다