Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis

Jieyuan Zhang, Junjie Guan, Xin Niu, Guowen Hu, Shangchun Guo, Qing Li, Zongping Xie, Changqing Zhang, Yang Wang, Jieyuan Zhang, Junjie Guan, Xin Niu, Guowen Hu, Shangchun Guo, Qing Li, Zongping Xie, Changqing Zhang, Yang Wang

Abstract

Background: Human induced pluripotent stem cell-derived mesenchymal stem cells (hiPSC-MSCs) have emerged as a promising alternative for stem cell transplantation therapy. Exosomes derived from mesenchymal stem cells (MSC-Exos) can play important roles in repairing injured tissues. However, to date, no reports have demonstrated the use of hiPSC-MSC-Exos in cutaneous wound healing, and little is known regarding their underlying mechanisms in tissue repair.

Methods: hiPSC-MSC-Exos were injected subcutaneously around wound sites in a rat model and the efficacy of hiPSC-MSC-Exos was assessed by measuring wound closure areas, by histological and immunofluorescence examinations. We also evaluated the in vitro effects of hiPSC-MSC-Exos on both the proliferation and migration of human dermal fibroblasts and human umbilical vein endothelial cells (HUVECs) by cell-counting and scratch assays, respectively. The effects of exosomes on fibroblast collagen and elastin secretion were studied in enzyme-linked immunosorbent assays and quantitative reverse-transcriptase-polymerase chain reaction (qRT-PCR). In vitro capillary network formation was determined in tube-formation assays.

Results: Transplanting hiPSC-MSC-Exos to wound sites resulted in accelerated re-epithelialization, reduced scar widths, and the promotion of collagen maturity. Moreover, hiPSC-MSC-Exos not only promoted the generation of newly formed vessels, but also accelerated their maturation in wound sites. We found that hiPSC-MSC-Exos stimulated the proliferation and migration of human dermal fibroblasts and HUVECs in a dose-dependent manner in vitro. Similarly, Type I, III collagen and elastin secretion and mRNA expression by fibroblasts and tube formation by HUVECs were also increased with increasing hiPSC-MSC-Exos concentrations.

Conclusions: Our findings suggest that hiPSC-MSC-Exos can facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. These data provide the first evidence for the potential of hiPSC-MSC-Exos in treating cutaneous wounds.

Figures

Figure 1
Figure 1
Characterization of human induced pluripotent stem cell-derived mesenchymal stem cells (hiPSC-MSCs) and hiPSC-MSC-derived exosomes (hiPSC-MSC-Exos). (A) Light microscopy images demonstrating morphological changes occurring during hiPSCs differentiation into fibroblast-like cells. (a) Representative cell morphology of hiPSCs before differentiation. (b) Intermediate phase of differentiating the hiPSCs into MSCs. (c) Typical fibroblast-like morphology of cells. (B) Flow cytometric analysis of the surface markers in hiPSC-MSCs. (C) Assessment of the tri-lineage differentiation capacity of iPSC-MSC-like cells. (a) Alizarin Red staining for osteocytes after 3 weeks in culture with osteogenic medium. (b) Alcian Blue staining for chondrocytes after 4 weeks in culture with chondrogenic medium. (c) Oil Red O staining for adipocytes after 2 weeks in culture with adipogenic medium. The qRT-PCR results for OCN (d), Sox9 (e), and LPL (f) after 7 days in culture with osteo-, chondro-, and adipogenic mediun. (D) Transmission electron microscope images of hiPSC-MSC-Exos morphology. Scale bars = 100 nm and 50 nm, respectively. (E) Detection of CD9, CD63, and CD81 incorporation into hiPSC-MSC-Exos by western blotting.
Figure 2
Figure 2
Rats macroscopic appearances of cutaneous wounds treated with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos. (A) Gross view of wounds treated with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos at 4, 7, and 14 days. (B) The effects of treatment with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos on wound closure at 4, 7, and 14 days. *P < 0.05.
Figure 3
Figure 3
Rats histological analyses of cutaneous wounds treated with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos. (A) H&E staining of wound sections following treatment with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos at 14 days post-wounding. The double-headed arrows indicate the edges of the scar. The effects of PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos on wound re-epithelialization (B) and scar widths (C) at 14 days post-wounding. (D) Evaluation of collagen maturity by Masson’s trichrome staining of wounds following treatment with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos at 14 days post-wounding. Scale bar = 500 μm. *P < 0.05; Ep, Epithelium; F, Follicle.
Figure 4
Figure 4
Immunofluorescence analyses of newly formed vessels and mature vessels. (A) CD31 (red arrows) Immunofluorescence staining of wound sections treated with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos at 7 and 14 days post-wounding. Scale bar = 50 μm. (B) Enumeration of newly formed vessels in wounds after treatment with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos at 7 and 14 days post-wounding. (C) Immunofluorescent triple staining of wound sections treated with PBS, MesenGro hMSC medium or hiPSC-MSC-Exos at 7 and 14 days post-wounding. Endothelial cells (CD31), smooth muscle cells (α-SMA), and cell nuclei (DAPI) fluoresced with red, green, and blue colours, respectively. Mature vessels (green arrows) were dually positive for CD31 and α-SMA. Scale bar = 50 μm. (D) Enumeration of mature vessels in wounds after treatment with PBS, MesenGro hMSC medium, or hiPSC-MSC-Exos at 7 and 14 days post-wounding. *P < 0.05.
Figure 5
Figure 5
The effects of exosomes on the proliferation, migration, and collagen, elastin secretion of human fibroblasts. The light microscopy images (A) and migration rates (B) of human fibroblasts into scratch sites following growth in MesenGro hMSC medium containing 0, 50, or 100 μg/mL hiPSC-MSC-Exos for 12 or 24 h. Scale bar = 250 μm. (C) Fibronectin protein expression of human fibroblasts treated with MesenGro hMSC medium containing 0, 50, or 100 μg/mL hiPSC-MSC-Exos for 24 h. (D) Human fibroblasts proliferation after growth in MesenGro hMSC medium containing 0, 50, or 100 μg/mL hiPSC-MSC-Exos was detected with a CCK-8 kit over 5 days. Secretion of Col I (E), III (F) and elastin (G) by human fibroblasts after growth in MesenGro hMSC medium containing 0, 50, or 100 μg/mL hiPSC-MSC-Exos over 3 days. (G) The Col I (H), III (I) and elastin (J) mRNA expression of human fibroblasts treated with MesenGro hMSC medium containing 0, 50, or 100 μg/mL hiPSC-MSC-Exos over 3 days. *P < 0.05.
Figure 6
Figure 6
The effects of exosomes on HUVECs proliferation and migration. The light microscopy images (A) and migration rates (B) of HUVECs into the scratched area of monolayers following growth in M200 medium containing 0, 50, or 100 μg/mL hiPSC-MSC-Exos for 12 or 24 h. Scale bar = 250 μm. (C) The proliferation of HUVECs grown in M200 containing 0, 50, or 100 μg/mL hiPSC-MSC-Exos was detected over 5 days, using a cell-counting kit.*P < 0.05.
Figure 7
Figure 7
The effects of exosomes on tube formation by HUVECs. (A) HUVECs tube formation was studied by growing cells in Matrigel in M200 medium containing 0, 50, or 100 μg/mL hiPSC-MSC-Exos. Scale bar = 250 μm. Total tube lengths (B) and branch points (C) of HUVECs following growth in M200 medium containing 0, 50, or, 100 μg/mL hiPSC-MSC-Exos for 4, 6, or 18 h. * P < 0.05.

References

    1. 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;34:9393–400. doi: 10.1016/j.biomaterials.2013.08.053.
    1. Lee KB, Choi J, Cho SB, Chung JY, Moon ES, Kim NS, et al. Topical embryonic stem cells enhance wound healing in diabetic rats. J Orthop Res. 2011;29:1554–62. doi: 10.1002/jor.21385.
    1. Biazar E, Keshel SH. The healing effect of stem cells loaded in nanofibrous scaffolds on full thickness skin defects. J Biomed Nanotechnol. 2013;9:1471–82. doi: 10.1166/jbn.2013.1639.
    1. Xin Y, Wang YM, Zhang H, Li J, Wang W, Wei YJ, et al. Aging adversely impacts biological properties of human bone marrow-derived mesenchymal stem cells: implications for tissue engineering heart valve construction. Artif Organs. 2010;34:215–22. doi: 10.1111/j.1525-1594.2009.00824.x.
    1. Zaim M, Karaman S, Cetin G, Isik S. Donor age and long-term culture affect differentiation and proliferation of human bone marrow mesenchymal stem cells. Ann Hematol. 2012;91:1175–86. doi: 10.1007/s00277-012-1438-x.
    1. Park IH, Zhao R, West JA, Yabuuchi A, Huo H, Ince TA, et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature. 2008;451:141–6. doi: 10.1038/nature06534.
    1. Kang L, Wang J, Zhang Y, Kou Z, Gao S. iPS cells can support full-term development of tetraploid blastocyst-complemented embryos. Cell Stem Cell. 2009;5:135–8. doi: 10.1016/j.stem.2009.07.001.
    1. Cartwright P, McLean C, Sheppard A, Rivett D, Jones K, Dalton S. LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism. Development. 2005;132:885–96. doi: 10.1242/dev.01670.
    1. Lian Q, Zhang Y, Zhang J, Zhang HK, Wu X, Zhang Y, et al. Functional mesenchymal stem cells derived from human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation. 2010;121:1113–23. doi: 10.1161/CIRCULATIONAHA.109.898312.
    1. Yen ML, Hou CH, Peng KY, Tseng PC, Jiang SS, Shun CT, et al. Efficient derivation and concise gene expression profiling of human embryonic stem cell-derived mesenchymal progenitors (EMPs) Cell Transplant. 2011;20:1529–45. doi: 10.3727/096368910X564067.
    1. Villa-Diaz LG, Brown SE, Liu Y, Ross AM, Lahann J, Parent JM, et al. Derivation of mesenchymal stem cells from human induced pluripotent stem cells cultured on synthetic substrates. Stem Cells. 2012;30:1174–81. doi: 10.1002/stem.1084.
    1. Himeno T, Kamiya H, Naruse K, Cheng Z, Ito S, Kondo M, et al. Mesenchymal stem cell-like cells derived from mouse induced pluripotent stem cells ameliorate diabetic polyneuropathy in mice. Biomed Res Int. 2013;2013:259187. doi: 10.1155/2013/259187.
    1. Liu J, Chen W, Zhao Z, Xu HH. Reprogramming of mesenchymal stem cells derived from iPSCs seeded on biofunctionalized calcium phosphate scaffold for bone engineering. Biomaterials. 2013;34:7862–72. doi: 10.1016/j.biomaterials.2013.07.029.
    1. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72. doi: 10.1016/j.cell.2007.11.019.
    1. Okano H, Nakamura M, Yoshida K, Okada Y, Tsuji O, Nori S, et al. Steps toward safe cell therapy using induced pluripotent stem cells. Circ Res. 2013;112:523–33. doi: 10.1161/CIRCRESAHA.111.256149.
    1. Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of Mesenchymal Stem cell-based therapy: Current status and perspectives. Cell Transplant. 2014;23(9):1045–59. doi: 10.3727/096368913X667709.
    1. Shen L, Zeng W, Wu YX, Hou CL, Chen W, Yang MC, et al. Neurotrophin-3 accelerates wound healing in diabetic mice by promoting a paracrine response in mesenchymal stem cells. Cell Transplant. 2013;22:1011–21. doi: 10.3727/096368912X657495.
    1. Song M, Heo J, Chun JY, Bae HS, Kang JW, Kang H, et al. The paracrine effects of mesenchymal stem cells stimulate the regeneration capacity of endogenous stem cells in the repair of a bladder-outlet-obstruction-induced overactive bladder. Stem Cells Dev. 2014;23:654–63. doi: 10.1089/scd.2013.0277.
    1. Zhang HC, Liu XB, Huang S, Bi XY, Wang HX, Xie LX, et al. Microvesicles derived from human umbilical cord mesenchymal stem cells stimulated by hypoxia promote angiogenesis both in vitro and in vivo. Stem Cells Dev. 2012;21:3289–97. doi: 10.1089/scd.2012.0095.
    1. Xin H, Li Y, Buller B, Katakowski M, Zhang Y, Wang X, et al. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells. 2012;30:1556–64. doi: 10.1002/stem.1129.
    1. Baglio SR, Pegtel DM, Baldini N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front Physiol. 2012;3:359. doi: 10.3389/fphys.2012.00359.
    1. Sahoo S, Klychko E, Thorne T, Misener S, Schultz KM, Millay M, et al. Exosomes from human CD34(+) stem cells mediate their proangiogenic paracrine activity. Circ Res. 2011;109:724–8. doi: 10.1161/CIRCRESAHA.111.253286.
    1. Muralidharan-Chari V, Clancy JW, Sedgwick A, D'Souza-Schorey C. Microvesicles: mediators of extracellular communication during cancer progression. J Cell Sci. 2010;123:1603–11. doi: 10.1242/jcs.064386.
    1. Pap E, Pallinger E, Pasztoi M, Falus A. Highlights of a new type of intercellular communication: microvesicle-based information transfer. Inflamm Res. 2009;58:1–8. doi: 10.1007/s00011-008-8210-7.
    1. Stoorvogel W, Kleijmeer MJ, Geuze HJ, Raposo G. The biogenesis and functions of exosomes. Traffic. 2002;3:321–30. doi: 10.1034/j.1600-0854.2002.30502.x.
    1. 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;22:845–54. doi: 10.1089/scd.2012.0395.
    1. Roccaro AM, Sacco A, Maiso P, Azab AK, Tai YT, Reagan M, et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest. 2013;123:1542–55. doi: 10.1172/JCI66517.
    1. Zhou Y, Xu H, Xu W, Wang B, Wu H, Tao Y, et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res Ther. 2013;4:34. doi: 10.1186/scrt194.
    1. Buccini S, Haider KH, Ahmed RP, Jiang S, Ashraf M. Cardiac progenitors derived from reprogrammed mesenchymal stem cells contribute to angiomyogenic repair of the infarcted heart. Basic Res Cardiol. 2012;107:301. doi: 10.1007/s00395-012-0301-5.
    1. Liao J, Wu Z, Wang Y, Cheng L, Cui C, Gao Y, et al. Enhanced efficiency of generating induced pluripotent stem (iPS) cells from human somatic cells by a combination of six transcription factors. Cell Res. 2008;18:600–3. doi: 10.1038/cr.2008.51.
    1. Cai J, Li W, Su H, Qin D, Yang J, Zhu F, et al. Generation of human induced pluripotent stem cells from umbilical cord matrix and amniotic membrane mesenchymal cells. J Biol Chem. 2010;285:11227–34. doi: 10.1074/jbc.M109.086389.
    1. Zou L, Luo Y, Chen M, Wang G, Ding M, Petersen CC, et al. A simple method for deriving functional MSCs and applied for osteogenesis in 3D scaffolds. Sci Rep. 2013;3:2243.
    1. Kosaka N, Yoshioka Y, Hagiwara K, Tominaga N, Ochiya T. Functional analysis of exosomal microRNA in cell-cell communication research. Methods Mol Biol. 2013;1024:1–10. doi: 10.1007/978-1-62703-453-1_1.
    1. Montecalvo A, Larregina AT, Morelli AE. Methods of analysis of dendritic cell-derived exosome-shuttle microRNA and its horizontal propagation between dendritic cells. Methods Mol Biol. 2013;1024:19–40. doi: 10.1007/978-1-62703-453-1_3.
    1. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–79.
    1. Lai RC, Arslan F, Lee MM, Sze NS, Choo A, Chen TS, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4:214–22. doi: 10.1016/j.scr.2009.12.003.
    1. Lai RC, Arslan F, Tan SS, Tan B, Choo A, Lee MM, et al. Derivation and characterization of human fetal MSCs: an alternative cell source for large-scale production of cardioprotective microparticles. J Mol Cell Cardiol. 2010;48:1215–24. doi: 10.1016/j.yjmcc.2009.12.021.
    1. Lai RC, Yeo RW, Tan KH, Lim SK. Mesenchymal stem cell exosome ameliorates reperfusion injury through proteomic complementation. Regen Med. 2013;8:197–209. doi: 10.2217/rme.13.4.
    1. Martin P. Wound healing–aiming for perfect skin regeneration. Science. 1997;276:75–81. doi: 10.1126/science.276.5309.75.
    1. Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med. 1999;341:738–46. doi: 10.1056/NEJM199909023411006.
    1. 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:e96161. doi: 10.1371/journal.pone.0096161.
    1. Bian S, Zhang L, Duan L, Wang X, Min Y, Yu H. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J Mol Med (Berl) 2014;92:387–97. doi: 10.1007/s00109-013-1110-5.

Source: PubMed

3
Prenumerera