Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice

Lei Chen, Yingbin Xu, Jingling Zhao, Zhaoqiang Zhang, Ronghua Yang, Julin Xie, Xusheng Liu, Shaohai Qi, Lei Chen, Yingbin Xu, Jingling Zhao, Zhaoqiang Zhang, Ronghua Yang, Julin Xie, Xusheng Liu, Shaohai Qi

Abstract

Growing evidence indicates that bone marrow-derived mesenchymal stem cells (BM-MSCs) enhance wound repair via paracrine. Because the extent of environmental oxygenation affects the innate characteristics of BM-MSCs, including their stemness and migration capacity, the current study set out to elucidate and compare the impact of normoxic and hypoxic cell-culture conditions on the expression and secretion of BM-MSC-derived paracrine molecules (e.g., cytokines, growth factors and chemokines) that hypothetically contribute to cutaneous wound healing in vivo. Semi-quantitative reverse transcriptase polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) analyses of normoxic and hypoxic BM-MSCs and their conditioned medium fractions showed that the stem cells expressed and secreted significantly higher amounts of basic fibroblast growth factor (bFGF),vascular endothelial growth factor A (VEGF-A) interleukin 6 (IL-6) and interleukin 8 (IL-8) under hypoxic conditions. Moreover, hypoxic BM-MSC-derived conditioned medium (hypoCM) vs. normoxic BM-MSC-derived conditioned medium (norCM) or vehicle control medium significantly enhanced the proliferation of keratinocytes, fibroblasts and endothelial cells, the migration of keratinocytes, fibroblasts, endothelial cells and monocytes, and the formation of tubular structures by endothelial cells cultured on Matrigel matrix. Consistent with these in vitro results, skin wound contraction was significantly accelerated in Balb/c nude mice treated with topical hypoCM relative to norCM or the vehicle control. Notably increased in vivo cell proliferation, neovascularization as well as recruitment of inflammatory macrophages and evidently decreased collagen I, and collagen III were also found in the hypoCM-treated group. These findings suggest that BM-MSCs promote murine skin wound healing via hypoxia-enhanced paracrine.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Proliferation of BM-MSCs in serum-containing…
Figure 1. Proliferation of BM-MSCs in serum-containing complete culture medium (A) and serum-free α-MEM (B).
Cell proliferation were examined under conditions of hypoxia or normoxia. Data are given as the means± the SEM; *p< 0.05 compared with the norCM group at indicated time points. #p< 0.05 compared with indicated earlier time points.
Figure 2. Relative mRNA and secreted protein…
Figure 2. Relative mRNA and secreted protein expression levels of wound-healing-related growth factors, cytokines and chemokines in normoxic and hypoxic BM-MSCs or conditioned medium.
(A) RT-PCR assays were performed to measure mRNA levels in BM-MSCs. (B, C, D and E) ELISA assays were performed to measure secreted protein levels in BM-MSC norCM and hypoCM. Data are given as the means± the SEM; *p< 0.05 compared with the expression level of each factor under normoxic culture conditions.
Figure 3. Effects of BM-MSC-derived conditioned medium…
Figure 3. Effects of BM-MSC-derived conditioned medium samples on paracrine cell proliferation and migration.
Equal numbers of keratinocytes, fibroblasts and HUVECs were incubated with vehicle control medium, norCM or hypoCM. Cell proliferation was evaluated at indicated time points (A, C and E). Data are given as the means±the SEM; *p< 0.05 compared with the vehicle control or the norCM group. #p< 0.05 the vehicle control compared with the norCM group. Equal numbers of keratinocytes, fibroblasts, HUVECs and CD14+ monocytes were added to the upper chambers of 24-well transwell plates, with the indicated medium added to the lower chambers (n = 4 wells per treatment). Cells that migrated to the bottom of the filter were stained and evaluated (B, D, F and G). Data are given as the means± the SEM;*p< 0.05 compared with the vehicle control or the norCM group. #p< 0.05 the vehicle control compared with the norCM group.
Figure 4. Increased capillary-like tube formation stimulated…
Figure 4. Increased capillary-like tube formation stimulated by BM-MSC hypoCM.
(A) HUVECs were seeded onto a Matrigel matrix and incubated with vehicle control medium, hypoCM or norCM for 12 h. (B)Tube formation was quantified. Scale bar, 400 µm for images in (A) (200×). Results are given as the means ± the SEM; *p< 0.05 compared with the vehicle control or the norCM group.
Figure 5. Effects of BM-MSC hypoCM on…
Figure 5. Effects of BM-MSC hypoCM on murine skin wound healing.
Representative macroscopic views of cutaneous wounds are shown on day 11 after treatment with vehicle control medium (A), norCM (B) or hypoCM (C). (D) The fraction of the wound area at each indicated time point in comparison to the original wound area was quantified as described in Material and methods, and plotted. Values are givenas the means± the SEM;*p<0.01 compared with the vehicle control or the norCM group.
Figure 6. IHC evaluation of wounded mouse…
Figure 6. IHC evaluation of wounded mouse skin.
Wound sections were evaluated on day 11 by staining with anti-Ki67 and anti-F4/80 antibodies. The numbers of Ki67+ proliferating cells (A, C) and recruited F4/80+ macrophages (B, D) in each of 4 randomly chosen high-power fields in the dermis were counted. Scale bar, 100 µm (400×). Data are expressed as the mean±the SEM;*p<0.05 compared with the vehicle control or the norCM group.
Figure 7. Angiogenesis in the murine skin…
Figure 7. Angiogenesis in the murine skin after wounding.
Wound sections were evaluated on day 11 by staining with anti-CD31 antibody. Representative CD31+ vessels are shown (A). The extent of vascularization was determined by assessing the number of CD31+ vessels in each of 4 randomly chosen high-power fields within the injury site (B). Scale bar, 100 µm for images in (A) (400×). Results are given as the means ± the SEM; *p< 0.05 compared with the vehicle control or the norCM group.
Figure 8. Collagen I and collagen III…
Figure 8. Collagen I and collagen III synthesis analysis.
(A). Evaluation of relative collagen I, collagen III mRNA expression. (B) Staining of collagen I and collagen III in scars treated with vehicle control, norCM or BM-MSC hypoCM. Scale bar, 100 µm (400×). Data are expressed as the mean±the SEM; *p<0.05 compared with day 20. #p< 0.05 compared with the vehicle control or the norCM group at day 40.

References

    1. Gurtner GC, Werner S, Barrandon Y, Longaker MT (2008) Wound repair and regeneration. Nature 453: 314–321.
    1. Sherman RA (2009) Maggot therapy takes us back to the future of wound care: new and improved maggot therapy for the 21st century. J Diabetes Sci Technol 3: 336–344.
    1. Otto WR, Wright NA (2011) Mesenchymal stem cells: from experiment to clinic. Fibrogenesis Tissue Repair 4: 20.
    1. Newman RE, Yoo D, LeRoux MA, Danilkovitch-Miagkova A (2009) Treatment of inflammatory diseases with mesenchymal stem cells. Inflamm Allergy Drug Targets 8: 110–123.
    1. Rosario CM, Yandava BD, Kosaras B, Zurakowski D, Sidman RL, et al. (1997) Differentiation of engrafted multipotent neural progenitors towards replacement of missing granule neurons in meander tail cerebellum may help determine the locus of mutant gene action. Development 124: 4213–4224.
    1. Li TS, Takahashi M, Ohshima M, Qin SL, Kubo M, et al. (2008) Myocardial repair achieved by the intramyocardial implantation of adult cardiomyocytes in combination with bone marrow cells. Cell Transplant 17: 695–703.
    1. Hocking AM, Gibran NS (2010) Mesenchymal stem cells: paracrine signaling and differentiation during cutaneous wound repair. Exp Cell Res 316: 2213–2219.
    1. Das R, Jahr H, van Osch GJ, Farrell E (2010) The role of hypoxia in bone marrow-derived mesenchymal stem cells: considerations for regenerative medicine approaches. Tissue Eng Part B Rev 16: 159–168.
    1. Meng F, He A, Zhang Z, Zhang Z, Lin Z, et al. (2013) Chondrogenic differentiation of ATDC5 and hMSCs could be induced by a novel scaffold-tricalcium phosphate-collagen-hyaluronan without any exogenous growth factors in vitro. J Biomed Mater Res A.
    1. Park IW, Wang JF, Groopman JE (2001) HIV-1 Tat promotes monocyte chemoattractant protein-1 secretion followed by transmigration of monocytes. Blood 97: 352–358.
    1. Caldelari R, Suter MM, Baumann D, De Bruin A, Muller E (2000) Long-term culture of murine epidermal keratinocytes. J Invest Dermatol 114: 1064–1065.
    1. Kim MH, Kim HB, Yoon SP, Lim SC, Cha MJ, et al. (2013) Colon cancer progression is driven by APEX1-mediated upregulation of Jagged. J Clin Invest.
    1. Zudaire E, Gambardella L, Kurcz C, Vermeren S (2011) A computational tool for quantitative analysis of vascular networks. PLoS One 6: e27385.
    1. Kyriakides TR, Wulsin D, Skokos EA, Fleckman P, Pirrone A, et al. (2009) Mice that lack matrix metalloproteinase-9 display delayed wound healing associated with delayed reepithelization and disordered collagen fibrillogenesis. Matrix Biol 28: 65–73.
    1. Javazon EH, Keswani SG, Badillo AT, Crombleholme TM, Zoltick PW, et al. (2007) Enhanced epithelial gap closure and increased angiogenesis in wounds of diabetic mice treated with adult murine bone marrow stromal progenitor cells. Wound Repair Regen 15: 350–359.
    1. Di-Poi N, Ng CY, Tan NS, Yang Z, Hemmings BA, et al. (2005) Epithelium-mesenchyme interactions control the activity of peroxisome proliferator-activated receptor beta/delta during hair follicle development. Mol Cell Biol 25: 1696–1712.
    1. Martin P (1997) Wound healing—aiming for perfect skin regeneration. Science 276: 75–81.
    1. Singer AJ, Clark RA (1999) Cutaneous wound healing. N Engl J Med 341: 738–746.
    1. Galeano M, Altavilla D, Cucinotta D, Russo GT, Calo M, et al. (2004) Recombinant human erythropoietin stimulates angiogenesis and wound healing in the genetically diabetic mouse. Diabetes 53: 2509–2517.
    1. Kinnaird T, Stabile E, Burnett MS, Shou M, Lee CW, et al. (2004) Local delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation 109: 1543–1549.
    1. Mayer H, Bertram H, Lindenmaier W, Korff T, Weber H, et al. (2005) Vascular endothelial growth factor (VEGF-A) expression in human mesenchymal stem cells: autocrine and paracrine role on osteoblastic and endothelial differentiation. J Cell Biochem 95: 827–839.
    1. Lee EY, Xia Y, Kim WS, Kim MH, Kim TH, et al. (2009) Hypoxia-enhanced wound-healing function of adipose-derived stem cells: increase in stem cell proliferation and up-regulation of VEGF and bFGF. Wound Repair Regen 17: 540–547.
    1. Chen L, Tredget EE, Wu PY, Wu Y (2008) Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One 3: e1886.
    1. Smith AN, Willis E, Chan VT, Muffley LA, Isik FF, et al. (2010) Mesenchymal stem cells induce dermal fibroblast responses to injury. Exp Cell Res 316: 48–54.
    1. Kim WS, Park BS, Sung JH, Yang JM, Park SB, et al. (2007) Wound healing effect of adipose-derived stem cells: a critical role of secretory factors on human dermal fibroblasts. J Dermatol Sci 48: 15–24.
    1. Grayson WL, Zhao F, Bunnell B, Ma T (2007) Hypoxia enhances proliferation and tissue formation of human mesenchymal stem cells. Biochem Biophys Res Commun 358: 948–953.
    1. Potier E, Ferreira E, Andriamanalijaona R, Pujol JP, Oudina K, et al. (2007) Hypoxia affects mesenchymal stromal cell osteogenic differentiation and angiogenic factor expression. Bone 40: 1078–1087.
    1. Badolato R, Oppenheim JJ (1996) Role of cytokines, acute-phase proteins, and chemokines in the progression of rheumatoid arthritis. Semin Arthritis Rheum 26: 526–538.
    1. Djouad F, Charbonnier LM, Bouffi C, Louis-Plence P, Bony C, et al. (2007) Mesenchymal stem cells inhibit the differentiation of dendritic cells through an interleukin-6-dependent mechanism. Stem Cells 25: 2025–2032.
    1. Verhaegen PD, Marle JV, Kuehne A, Schouten HJ, Gaffney EA, et al. (2012) Collagen bundle morphometry in skin and scar tissue: a novel distance mapping method provides superior measurements compared to Fourier analysis. J Microsc 245: 82–89.
    1. Verhaegen PD, Schouten HJ, Tigchelaar-Gutter W, van Marle J, van Noorden CJ, et al. (2012) Adaptation of the dermal collagen structure of human skin and scar tissue in response to stretch: an experimental study. Wound Repair Regen 20: 658–666.
    1. Xie JL, Bian HN, Qi SH, Chen HD, Li HD, et al. (2008) Basic fibroblast growth factor (bFGF) alleviates the scar of the rabbit ear model in wound healing. Wound Repair Regen 16: 576–581.

Source: PubMed

Подписаться