First Characterization of Human Amniotic Fluid Stem Cell Extracellular Vesicles as a Powerful Paracrine Tool Endowed with Regenerative Potential

Carolina Balbi, Martina Piccoli, Lucio Barile, Andrea Papait, Andrea Armirotti, Elisa Principi, Daniele Reverberi, Luisa Pascucci, Pamela Becherini, Luigi Varesio, Massimo Mogni, Domenico Coviello, Tiziano Bandiera, Michela Pozzobon, Ranieri Cancedda, Sveva Bollini, Carolina Balbi, Martina Piccoli, Lucio Barile, Andrea Papait, Andrea Armirotti, Elisa Principi, Daniele Reverberi, Luisa Pascucci, Pamela Becherini, Luigi Varesio, Massimo Mogni, Domenico Coviello, Tiziano Bandiera, Michela Pozzobon, Ranieri Cancedda, Sveva Bollini

Abstract

Human amniotic fluid stem cells (hAFS) have shown a distinct secretory profile and significant regenerative potential in several preclinical models of disease. Nevertheless, little is known about the detailed characterization of their secretome. Herein we show for the first time that hAFS actively release extracellular vesicles (EV) endowed with significant paracrine potential and regenerative effect. c-KIT+ hAFS were isolated from leftover samples of amniotic fluid from prenatal screening and stimulated to enhance EV release (24 hours 20% O2 versus 1% O2 preconditioning). The capacity of the c-KIT+ hAFS-derived EV (hAFS-EV) to induce proliferation, survival, immunomodulation, and angiogenesis were investigated in vitro and in vivo. The hAFS-EV regenerative potential was also assessed in a model of skeletal muscle atrophy (HSA-Cre, SmnF7/F7 mice), in which mouse AFS transplantation was previously shown to enhance muscle strength and survival. hAFS secreted EV ranged from 50 up to 1,000 nm in size. In vitro analysis defined their role as biological mediators of regenerative, paracrine effects while their modulatory role in decreasing skeletal muscle inflammation in vivo was shown for the first time. Hypoxic preconditioning significantly induced the enrichment of exosomes endowed with regenerative microRNAs within the hAFS-EV. In conclusion, this is the first study showing that c-KIT+ hAFS dynamically release EV endowed with remarkable paracrine potential, thus representing an appealing tool for future regenerative therapy. Stem Cells Translational Medicine 2017;6:1340-1355.

Keywords: Apoptosis; Exosomes; Extracellular vesicles; Fetal stem cells; Paracrine communication; Proliferation; Tissue regeneration; miRNA.

© 2017 The Authors Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.

Figures

Figure 1
Figure 1
Hypoxic preconditioning does not significantly affect hAFS stemness and phenotype. (A): Bright field, SSEA4 and NANOG immunostaining images of hAFS under: (a), (a′), and (a′′) 24 hours 20% O2 CTRL conditions; (b), (b′), and (b′′) 24 hours 20% O2 SF conditions; (c), (c′), and (c′′) 24 hours 1% O2 preconditioning in CTRL medium; (d), (d′), and (d′′) 24 hours 1% O2 preconditioning in SF condition; scale bar 50 µm. (B): Western Blot for HIF‐1α expression by hAFS after 24 hours of 20% O2 or 1% O2 preconditioning in complete (CTRL) or SF medium. (C): SSEA4+ hAFS under: (a) 24 hours 20% O2 CTRL condition: 87.8% ± 2.8%; (b) 24 hours 20%O2 SF condition: 89.9% ± 1.5%; (c) 24 hours 1% O2 preconditioning in CTRL medium: 83.0% ± 0.7%; and (d) 24 hours 1% O2 preconditioning in SF condition: 84.1% ± 2.2%; values expressed as mean ± SEM. (D): Real time qRT‐PCR and qualitative RT‐PCR analysis for NANOG and GAPDH expression of hAFS under 24 hours: (a) 20% O2 CTRL and (b) 20% O2 SF conditions, (c) 1% O2 CTRL and (d) 1% O2 SF conditions; +ve: positive control, human embryonic stem cells (****, p < .0001); −ve: negative control: HDF; n.d. (E): hAFS immunophenotype under 24 hours 20% O2 in CTRL (a), or SF conditions (b), or under 24 hours 1% O2 preconditioning, in either CTRL (c), or SF conditions (d). (F): Apoptosis analysis by Annexin V and PI of hAFS under 24 hours: (a) 20% O2 CTRL and (b) 20% O2 SF conditions, (c) 1% O2 CTRL and (d) 1% O2 SF conditions. Abbreviations: CD, cluster of differentiation; hAFS, human amniotic fluid stem cells; HIF‐1α, hypoxia inducible factor‐1 alpha; HLA, human leucocyte antigen; n.d., not determined; PI, propidium iodide; SF, serum‐free.
Figure 2
Figure 2
hAFS secrete EV containing microvesicles and exosomes. (A): Transmission electron microscopy (TEM) analysis of hAFS under 24 hours 20% O2 CTRL (a) or 20% O2 SF conditions (b) and 1% O2 CTRL (c) or 1% O2 SF (d) conditions; scale bars 500 nm. (B): TEM Analysis of 1% O2 hAFS‐EV (hAFS‐EVHypo); scale bar 500 nm in (d′); 200 nm in (d′′); and 100 nm (d′′′ and d′′′′). (C): Nanosight analysis measuring the amount of particles within hAFS‐EVNormo (283.3 ± 64.8 × 106 particle/ml) and hAFS‐EVHypo (283.8 ± 79.9 × 106 particle/ml) released by 106 cells (right panel); values are expressed as mean ± SEM. Representative images of the graphical output are reported in the left panel. (D): BCA assay of the protein concentration of hAFS‐EVHypo (4.2 ± 0.4 μg/106 cells, ***, p < .001, p = .0005) over hAFS‐EVNormo (2.4 ± 0.3 μg/106 cells) released by 106 cells. (E): Western Blot analysis of TSG101, ALIX, GRP94, and βACTIN by hAFS and hAFS‐EV, both under 20% O2 and 1% O2 SF conditions. (F): FACS analysis of hAFS‐EVNormo and hAFS‐EVHypo bound to the ExoCapTM Capture Beads compared to control empty beads (CTRL BEADS) for the exosomal markers CD81 (*, p < .05, p = .049; **, p < .01, p = .005; ***, p < .001, p = .0004), CD9 (*p < .05, p = .047; **, p < .01, p = .0015; ***, p < .001, p = .0002), and CD63 (*, p < .05, p = .02; **, p < .01, p = .004; ***, p < .001, p = .0002). Representative dot plots are illustrated in the left panel, the evaluation of the MFI ratio on the right panels. Abbreviations: CD, cluster of differentiation; EV, extracellular vesicles; hAFS, human amniotic fluid stem cells; Hypo, hypoxic; MFI, mean fluorescent intensity; Normo, normoxic; SF, serum‐free.
Figure 3
Figure 3
hAFS‐EV mediate paracrine effects on target cells. (A): PKH67+ hAFS‐EV uptake by HDF, C2C12, and hPBMC by FACS (values normalized to mode on the y‐axis) and immunostaining (scale bar: 20 µm). PKH67+ cells after treatment with hAFS‐EVNormo and hAFS‐EVHypo were: 65.4% ± 4.4% and 63.5% ± 5.6% HDF (**, p < .01, p = .0033 and p = .0036, respectively); 75.9% ± 3.4% and 83.7% ± 1.1% C2C12 (****, p < .0001); 9.3% ± 2.6% and 12.5% ± 2.5% hPBMC (*, p < .05, p = .017). (B): BrdU Enzyme‐Linked ImmunoSorbent Assay (ELISA) on HDF treated with 1 µg/104, 2 µg/104, and 4 µg/104 cells of hAFS‐EVNormo and hAFS‐EVHypo versus untreated cells (CTRL); ****, p < .0001; #, p referring to hAFS‐EVHypo versus hAFS‐EVNormo: #, p < .05 (p = .033); ##, p < .01 (p = .0077); ###, p < .001 (p = .0002). Values expressed as the fold change of sample absorbance read at 370 nm (reference wavelength approximately 492 nm). (C): MTT assay on C2C12 primed with 1 µg/104, 2 µg/104, and 4 µg/104 cells of hAFS‐EVNormo or hAFS‐EVHypo and exposed to oxidative stress versus untreated cells (CTRL) or damaged cells without hAFS‐EV stimulation (H2O2). ****, p < .0001; #, p referring to hAFS‐EVHypo versus hAFS‐EVNormo: ####, p < .0001. Values expressed as the fold change of the sample absorbance read at 560 and 670 nm. (D): hPBMC proliferation under unstimulated (Naive) and activating (PHA or PWM) conditions with 1 μg/105 cells of hAFS‐EVNormo and hAFS‐EVHypo versus untreated cells (CTRL hPBMC); CPS. (E): FACS analysis of mature CD19+CD27+ B cells treated with 1 μg/105 cells of hAFS‐EVNormo or hAFS‐EVHypo. Naive hPBMC: 3.48% ± 0.86%; CTRL hPBMC + PWM: 8.48% ± 2.63%; hPBMC + PWM + hAFS‐EVNormo: 2.32% ± 0.96%; hPBMC + PWM + hAFS‐EVHypo: 2.36% ± 0.62%. *, p < .05 (p = .045 and p = .047). Abbreviations: CD, cluster of differentiation; CPS, counts per second; DAPI, 4′,6‐diamidino‐2‐phenylindole; EV, extracellular vesicles; hAFS, human amniotic fluid stem cells; HDF, human dermal fibroblast; hPBMC, human peripheral blood mononuclear cell; Hypo, hypoxic; MVB, multivesicular body; Normo, normoxic; PHA, phytohemagglutinin; PWM, pokeweed mitogen; SF, serum‐free.
Figure 4
Figure 4
hAFS‐EV exert a certain degree of angiogenic potential in a mouse Matrigel plug assay. (A): Representative images of Matrigel plugs loaded with 10 μg of hAFS‐EVNormo or hAFS‐EVHypo and recovered after 3 weeks from C57Bl/6J mice; scale bar 5 mm. (B): Evaluation of blood hemoglobin content on the Matrigel plugs loaded with 10 μg of hAFS‐EVNormo or hAFS‐EVHypo versus the PBS control *, p < .05 (p = .04). (C): Real time qRT‐PCR analysis on the recovered plugs for Cdh5, VegfA, and Pecam1 expression as normalized to B2M expression. (D): Representative images of histological evaluation of vessel formation within the Matrigel plug by infiltrating host cells via hematoxylin and eosin and CD31 staining, scale bar: 50 µm. CTRL: vehicle loaded (PBS) Matrigel plug; hAFS‐EVNormo: hAFS‐EVNormo loaded Matrigel plug; hAFS‐EVHypo: hAFS‐EVHypo loaded Matrigel plug. Abbreviations: CD, cluster of differentiation; EV, extracellular vesicles; hAFS, human amniotic fluid stem cells; Hypo, hypoxic; Normo, normoxic.
Figure 5
Figure 5
hAFS‐EV decrease muscle inflammation in a mouse model of muscle atrophy. (A): In vivo procedure. (B): IgG (red), CD68 (red), and LAMININ (green) expression in the Tibialis Anterior muscle of untreated (CTRL) and 1 μg hAFS‐EVNormo‐ and hAFS‐EVHypo‐treated mice at 1 and 7 days post injection (p.i.); scale bar: 100 μm and 50 μm, respectively. (C): IgG‐infiltrated fibers at 1 and 7 days p.i. (CTRL 1 Day: 14.9% ± 3.9%; hAFS‐EVNormo 1 Day: 2.9% ± 1.4%; hAFS‐EVHypo 1 Day: 1.6% ± 0.7%; *, p < .05 p = .031 and p = .016; CTRL 7 Days: 16.1% ± 4.3%; hAFS‐EVNormo 7 Days: 3.8% ± 1.0%; hAFS‐EVHypo 7 Days: 2.0% ± 0.7%; *, p < .05, p = .035) and corresponding mean fluorescent area (CTRL 1 Day: 8.33% ± 3.0%; hAFS‐EVNormo 1 Day: 0.71% ± 0.11%; hAFS‐EVHypo 1 Day: 0.42% ± 0.1%; **, p < .01, p = .002; CTRL 7 Days: 8.90% ± 3.2%; hAFS‐EVNormo 7 Days: 1.64% ± 0.44%; hAFS‐EVHypo 7 Days: 0.80% ± 0.15%; **, p < .01, p = .006 and p = .003). (D): Central nucleated fibers at 1 and 7 days p.i.: CTRL 1 Day: 9.4% ± 1.6%; hAFS‐EVNormo 1 Day: 4.0% ± 1.3%; hAFS‐EVHypo 1 Day: 2.6% ± 1.3%, *, p < .05, p = .015; CTRL 7 Days: 9.8% ± 1.7%; hAFS‐EVNormo 7 Days: 13.1% ± 3.5%; hAFS‐EVHypo 7 Days: 3.7% ± 0.9%; #, p < .05, p = .033, as hAFS‐EVHypo versus hAFS‐EVNormo. (E): Ratio of Nos2 over Arg2 by real time qRT‐PCR; *, p < .05, p = .045 and p = .036 for hAFS‐EVNormo and hAFS‐EVHypo;. A.U. (F): Real time qRT‐PCR for Il‐1α (***, p < .001, p = .0002 and p = .001; *, p < .05, p = .046 and p = .0163, for hAFS‐EVNormo and hAFS‐EVHypo) and Il‐10 (****, p < .0001, **, p < .01, p = .004). Abbreviations: A.U. arbitrary units; CD, cluster of differentiation; DAPI, 4′,6‐diamidino‐2‐phenylindole; EV, extracellular vesicles; hAFS, human amniotic fluid stem cells; Hypo, hypoxic; Normo, normoxic; SF, serum‐free.
Figure 6
Figure 6
hAFS‐EV act as biological carrier of regenerative miRNAs which are significantly increased in the target cells following direct uptake. (A): Analysis of hAFS‐EV small non‐coding RNA content by Agilent technology. (B): Real time qRT‐PCR of hAFS‐EVHypo versus hAFS‐EVNormo showing significant enrichment of the following regenerative miRNAs: miR‐223 (***, p < .001, p = .0001), miR‐146a (**, p < .01, p = .009), miR‐let7c (***, p < .001, p = .0002), miR‐21 (***, p < .001, p = .0003), miR‐199a‐3p (**, p < .01, p = .002), miR‐126, miR‐146b, and miR‐210 (****, p < .0001). (C): Uptake analysis of hAFS‐EV fluorescently labeled RNA by HDF (upper panel) and C2C12 (lower panel) via FACS (on the left representative histogram graphs with corresponding values in the graphs on the right). 88.67% ± 4.21% and 86.73% ± 4.46% of HDF (****, p < .0001) and 91.5% ± 3.38% and 85.97% ± 5.51% of C2C12 cells (****, p < .0001) became fluorescent following incubation with 1 µg/104 cells of SYTO®RNASelect+ hAFS‐EVNormo and hAFS‐EVHypo, showing direct uptake of the marked RNA cargo. (D): Real time qRT‐PCR analysis for the enrichment of specific hAFS‐EV‐delivered miRNAs in the target proliferative HDF: miRNA‐146a, **, p < .01, p = .006; miRNA‐126 and miR‐199a‐3p (*, p < .05, p = .03), miR‐210 (*, p < .05, p = .04). (E): Real time qRT‐PCR analysis for the enrichment of specific hAFS‐EV‐delivered miRNAs in the target C2C12 with or without H2O2 treatment: miR‐let7c (**, p < .01, p = .005 and *, p < .05, p = .04); miR‐21 (**, p < .01, p = .002 and p = .001, respectively); miRNA‐146b (*, p < .05, p = .020 and p = .012); miRNA‐199a‐3p (*, p < .05, p = .013 and **, p < .01 p = .008); and miRNA‐210 (**, p < .01, p = .006 and *, p < .05 p = .02). Gene expression was normalized to human GAPDH and to mouse B2M as internal controls in the HDF and C2C12 cells, respectively. Abbreviations: EV, extracellular vesicles; FITC, fluorescein isothiocyanate; hAFS, human amniotic fluid stem cells; HDF, human dermal fibroblast; Hypo, hypoxic; Normo, normoxic.

References

    1. Gnecchi M, Zhang Z, Ni A et al. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res 2008;103:1204–1219.
    1. Mirotsou M, Jayawardena TM, Schmeckpeper J et al. Paracrine mechanisms of stem cell reparative and regenerative actions in the heart. J Mol Cell Cardiol 2011;50:280–289.
    1. Bollini S, Gentili C, Tasso R et al. The regenerative role of the fetal and adult stem cell secretome. J Clin Med J Clin Med 2013;2:302–327.
    1. Tetta C, Ghigo E, Silengo L et al. Extracellular vesicles as an emerging mechanism of cell‐to‐cell communication. Endocrine 2012;44:11–19.
    1. Lötvall J, Hill AF, Hochberg F et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles 2014;3:26913.
    1. Zhang B, Yeo R, Tan K et al. Focus on extracellular vesicles: Therapeutic potential of stem cell‐derived extracellular vesicles. Int J Mol Sci 2016;17:pii: E174.
    1. Arslan F, Lai RC, Smeets MB et al. Mesenchymal stem cell‐derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res 2013;10:301–312.
    1. Lai RC, Arslan F, Lee MM et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res 2010;4:214–222.
    1. Bian S, Zhang L, Duan L et al. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J Mol Med (Berl) 2014;92:387–397.
    1. Xu W, Qian H, Zhang B et al. HucMSC‐exosome mediated‐Wnt4 signaling is required for cutaneous wound healing. Stem Cells 2015;33:2158–2168.
    1. Kordelas L, Rebmann V, Ludwig A‐K et al. MSC‐derived exosomes: A novel tool to treat therapy‐refractory graft‐versus‐host disease. Leukemia 2014;28:970–973.
    1. Bruno S, Grange C, Collino F et al. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS One 2012;7:e33115.
    1. Tan CY, Lai RC, Wong W et al. Mesenchymal stem cell‐derived exosomes promote hepatic regeneration in drug‐induced liver injury models. Stem Cell Res Ther 2014;5:76.
    1. Xin H, Li Y, Buller B et al. Exosome‐mediated transfer of miR‐133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells 2012;30:1556–1564.
    1. Bobis‐Wozowicz S, Kmiotek K, Sekula M et al. Human induced pluripotent stem cell‐derived microvesicles transmit RNAs and proteins to recipient mature heart cells modulating cell fate and behavior. Stem Cells 2015;33:2748–2761.
    1. De Coppi P, BartschG Jr., Siddiqui MM et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 2007;25:100–106.
    1. Pozzobon M, Piccoli M, Schiavo AA et al. Isolation of c‐Kit+ human amniotic fluid stem cells from second trimester. Methods Mol Biol 2013;1035:191–198.
    1. Schiavo AA, Franzin C, Albiero M et al. Endothelial properties of third‐trimester amniotic fluid stem cells cultured in hypoxia. Stem Cell Res Ther 2015;6:209.
    1. Bollini S, Cheung KK, Riegler J et al. Amniotic fluid stem cells are cardioprotective following acute myocardial infarction. Stem Cells Dev 2011;20:1985–1994.
    1. Smart N, Bollini S, Dubé KN et al. De novo cardiomyocytes from within the activated adult heart after injury 2011. Nature 2011;474:640–644.
    1. Zani A, Cananzi M, Fascetti‐Leon F et al. Amniotic fluid stem cells improve survival and enhance repair of damaged intestine in necrotising enterocolitis via a COX‐2 dependent mechanism. Gut 2014;63:300–309.
    1. Piccoli M, Franzin C, Bertin E et al. Amniotic fluid stem cells restore the muscle cell niche in a HSA‐Cre, SmnF7/F7 mouse model. Stem Cells 2012;30:1675–1684.
    1. Mirabella T, Hartinger J, Lorandi C et al. Proangiogenic soluble factors from amniotic fluid stem cells mediate the recruitment of endothelial progenitors in a model of ischemic fasciocutaneous flap. Stem Cells Dev 2012;21:2179–2188.
    1. Mirabella T, Cilli M, Carlone S et al. Amniotic liquid derived stem cells as reservoir of secreted angiogenic factors capable of stimulating neo‐arteriogenesis in an ischemic model. Biomaterials 2011;32:3689–3699.
    1. Mirabella T, Poggi A, Scaranari M et al. Recruitment of host's progenitor cells to sites of human amniotic fluid stem cells implantation. Biomaterials 2011;32:4218–4227.
    1. Bekeredjian‐Ding I, Foermer S, Kirschning CJ et al. Poke weed mitogen requires Toll‐like receptor ligands for proliferative activity in human and murine B lymphocytes. PLoS One 2012;7:e29806.
    1. Zhang H‐C, Liu X‐B, Huang S 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;21:3289–3297.
    1. Ayers L, Nieuwland R, Kohler M et al. Dynamic microvesicle release and clearance within the cardiovascular system: Triggers and mechanisms. Clin Sci (Lond) 2015;129:915–931.
    1. Gray WD, French KM, Ghosh‐Choudhary S et al. Identification of therapeutic covariant microRNA clusters in hypoxia‐treated cardiac progenitor cell exosomes using systems biology. Circ Res 2015;116:255–263.
    1. Bottai D, Cigognini D, Nicora E et al. Third trimester amniotic fluid cells with the capacity to develop neural phenotypes and with heterogeneity among sub‐populations. Restor Neurol Neurosci 2012;30:55–68.
    1. Chiavegato A, Bollini S, Pozzobon M et al. Human amniotic fluid‐derived stem cells are rejected after transplantation in the myocardium of normal, ischemic, immuno‐suppressed or immuno‐deficient rat 2006 J Mol Cell Cardiol 2007;42:746–759.
    1. Barile L, Lionetti V, Cervio E et al. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovasc Res 2014;103:530–541.
    1. Caradec J, Kharmate G, Hosseini‐Beheshti E et al. Reproducibility and efficiency of serum‐derived exosome extraction methods. Clin Biochem 2014;47:1286–1292.
    1. Connolly KD, Guschina IA, Yeung V et al. Characterisation of adipocyte‐derived extracellular vesicles released pre‐ and post‐adipogenesis. J Extracell Vesicles 2015;24;4:29159.
    1. Grisendi G, Finetti E, Manganaro D et al. Detection of microparticles from human red blood cells by multiparametric flow cytometry. Blood Transfus 2015;13:274–280.
    1. Judge TA, Wu Z, Zheng XG et al. The role of CD80, CD86, and CTLA4 in alloimmune responses and the induction of long‐term allograft survival. J Immunol 1999;162:1947–1951.
    1. Mareschi K, Castiglia S, Sanavio F et al. Immunoregulatory effects on T lymphocytes by human mesenchymal stromal cells isolated from bone marrow, amniotic fluid, and placenta. Exp Hematol 2016;44:138–150.
    1. de Masson A, Le Buanec H, Bouaziz J‐D. Purification and immunophenotypic characterization of human B cells with regulatory functions. Methods Mol Biol 2014;1190:45–52.
    1. Heredia JE, Mukundan L, Chen FM et al. Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell 2013;153:376–388.
    1. Shamim EA, Rider LG, Miller FW. Update on the genetics of the idiopathic inflammatory myopathies. Curr Opin Rheumatol 2000;12:482–491.
    1. Fan J, Kou X, Yang Y et al. MircoRNA‐regulated proinflammatory cytokines in sarcopenia. Mediators Inflamm 2016;2016:1438686.
    1. Deyhle MR, Gier AM, Evans KC et al. Skeletal muscle inflammation following repeated bouts of lengthening contractions in humans. Front Physiol 2016;6:624.
    1. Tidball JG, Villalta SA. Regulatory interactions between muscle and the immune system during muscle regeneration. AJP Regul Integr Comp Physiol 2010;298:R1173–R1187.
    1. Zhang Z, Sun H, Dai H et al. MicroRNA miR‐210 modulates cellular response to hypoxia through the MYC antagonist MNT. Cell Cycle 2009;8:2756–2768.
    1. Won Kim H, Haider HK, Jiang S et al. Ischemic preconditioning augments survival of stem cells via miR‐210 expression by targeting Caspase‐8‐associated protein 2. J Biol Chem 2009;284:33161–33168.
    1. Keerthikumar S, Chisanga D, Ariyaratne D et al. ExoCarta: A web‐based compendium of exosomal cargo. J Mol Biol 2016;428:688–692.
    1. Rani S, Ryan AE, Griffin MD et al. Mesenchymal stem cell‐derived extracellular vesicles: Toward cell‐free therapeutic applications. Mol Ther 2015;23:812–823.
    1. Tetta C, Consiglio AL, Bruno S et al. The role of microvesicles derived from mesenchymal stem cells in tissue regeneration; A dream for tendon repair? Muscles Ligaments Tendons J 2012;2:212–221.
    1. Camussi G, Deregibus MC, Cantaluppi V. Role of stem‐cell‐derived microvesicles in the paracrine action of stem cells. Biochem Soc Trans 2013;41:283–287.
    1. EL Andaloussi S, Mäger I, Breakefield XO et al. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat Rev Drug Discov 2013;12:347–357.
    1. Mohyeldin A, Garzón‐Muvdi T, Quiñones‐Hinojosa A. Oxygen in stem cell biology: A critical component of the stem cell niche. Cell Stem Cell 2010;7:150–161.
    1. Salomon C, Ryan J, Sobrevia L et al. Exosomal signaling during hypoxia mediates microvascular endothelial cell migration and vasculogenesis. PLoS One 2013;8:e68451.
    1. Yu X, Deng L, Wang D et al. Mechanism of TNF‐α autocrine effects in hypoxic cardiomyocytes: Initiated by hypoxia inducible factor 1α, presented by exosomes. J Mol Cell Cardiol 2012;53:848–857.
    1. Jun EK, Zhang Q, Yoon BS et al. Hypoxic conditioned medium from human amniotic fluid‐derived mesenchymal stem cells accelerates skin wound healing through TGF‐β/SMAD2 and PI3K/Akt pathways. Int J Mol Sci 2014;15:605–628.
    1. Kang K, Ma R, Cai W et al. Exosomes secreted from CXCR4 overexpressing mesenchymal stem cells promote cardioprotection via Akt signaling pathway following myocardial infarction. Stem Cells Int 2015;2015:659890.
    1. Kang T, Jones TM, Naddell C et al. Adipose‐derived stem cells induce angiogenesis via microvesicle transport of miRNA‐31. Stem Cells Transl Med 2016;5:440–450.
    1. Di Trapani M, Bassi G, Fontana E et al. Immune regulatory properties of CD117(pos) amniotic fluid stem cells vary according to gestational age. Stem Cells Dev 2015;24:132–143.
    1. Moorefield EC, McKee EE, Solchaga L et al. Cloned, CD117 selected human amniotic fluid stem cells are capable of modulating the immune response. PLoS One 2011;6:e26535.
    1. Gao F, Chiu SM, Motan DAL et al. Mesenchymal stem cells and immunomodulation: Current status and future prospects. Cell Death Dis 2016;7:e2062.
    1. Uccelli A, de Rosbo NK. The immunomodulatory function of mesenchymal stem cells: Mode of action and pathways. Ann NY Acad Sci 2015;1351:114–126.
    1. Mokarizadeh A, Delirezh N, Morshedi A et al. Microvesicles derived from mesenchymal stem cells: Potent organelles for induction of tolerogenic signaling. Immunol Lett 2012;147:47–54.
    1. Blazquez R, Sanchez‐Margallo FM, de la Rosa O et al. Immunomodulatory potential of human adipose mesenchymal stem cells derived exosomes on in vitro stimulated T cells. Front Immunol 2014;5:556.
    1. Zhang B, Yin Y, Lai RC et al. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev 2014;23:1233–1244.
    1. Conforti A, Scarsella M, Starc N et al. Microvescicles derived from mesenchymal stromal cells are not as effective as their cellular counterpart in the ability to modulate immune responses in vitro. Stem Cells Dev 2014;23:2591–2599.
    1. Budoni M, Fierabracci A, Luciano R et al. The immunosuppressive effect of mesenchymal stromal cells on B lymphocytes is mediated by membrane vesicles. Cell Transplant 2013;22:369–379.
    1. Nicole S, Desforges B, Millet G et al. Intact satellite cells lead to remarkable protection against Smn gene defect in differentiated skeletal muscle. J Cell Biol 2003;161:571–582.
    1. Nakamura Y, Miyaki S, Ishitobi H et al. Mesenchymal‐stem‐cell‐derived exosomes accelerate skeletal muscle regeneration. FEBS Lett 2015;589:1257–1265.
    1. Dutt V, Gupta S, Dabur R et al. Skeletal muscle atrophy: Potential therapeutic agents and their mechanisms of action. Pharmacol Res 2015;99:86–100.
    1. Das S, Halushka MK. Extracellular vesicle microRNA transfer in cardiovascular disease. Cardiovasc Pathol 2015;24:199–206.
    1. Zhou Y, Zhou G, Tian C et al. Exosome‐mediated small RNA delivery for gene therapy. Wiley Interdiscip Rev RNA 2016;7:758–771.
    1. Bodempudi V, Hergert P, Smith K et al. miR‐210 promotes IPF fibroblast proliferation in response to hypoxia. Am J Physiol Lung Cell Mol Physiol 2014;307:L283–L294.
    1. Eulalio A, Mano M, Ferro MD et al. Functional screening identifies miRNAs inducing cardiac regeneration. Nature 2012;492:376–381.
    1. Zeng L, He X, Wang Y et al. MicroRNA‐210 overexpression induces angiogenesis and neurogenesis in the normal adult mouse brain. Gene Ther 2014;21:37–43.
    1. Alaiti MA, Ishikawa M, Masuda H et al. Up‐regulation of miR‐210 by vascular endothelial growth factor in ex vivo expanded CD34+ cells enhances cell‐mediated angiogenesis. J Cell Mol Med 2012;16:2413–2421.
    1. Cervio E, Barile L, Moccetti T et al. Exosomes for intramyocardial intercellular communication. Stem Cells Int 2015;2015:482171.
    1. Wang S, Aurora AB, Johnson BA et al. The endothelial‐specific microRNA miR‐126 governs vascular integrity and angiogenesis. Dev Cell 2008;15:261–271.
    1. Wang S, Olson EN. AngiomiRs‐‐Key regulators of angiogenesis. Curr Opin Genet Dev 2009;19:205–211.
    1. Wang B, Yao K, Huuskes BM et al. Mesenchymal stem cells deliver exogenous microRNA let7c via exosomes to attenuate renal fibrosis. Mol Ther 2016; 24:1290–1301.
    1. Haider KH, Idris NM, Kim HW et al. MicroRNA‐21 is a key determinant in IL‐11/Stat3 anti‐apoptotic signalling pathway in preconditioning of skeletal myoblasts. Cardiovasc Res 2010;88:168–178.
    1. Ibrahim AG, Cheng K, Marbán E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Rep 2014;2:606–619.
    1. Recchiuti A, Krishnamoorthy S, Fredman G et al. MicroRNAs in resolution of acute inflammation: Identification of novel resolvin D1‐miRNA circuits. FASEB J 2011;25:544–560.
    1. Sheedy FJ, Palsson‐McDermott E, Hennessy EJ et al. Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR‐21. Nat Immunol 2010;11:141–147.
    1. Asangani IA, Rasheed SAK, Nikolova DA et al. MicroRNA‐21 (miR‐21) post‐transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 2008;27:2128–2136.
    1. Essandoh K, Li Y, Huo J et al. Mirna‐mediated macrophage polarization and its potential role in the regulation of inflammatory response. Shock 2016. 46:122–131.
    1. Ivan M, Huang X. miR‐210: Fine‐tuning the hypoxic response. Adv Exp Med Biol 2014;772:205–227.
    1. Lange‐Consiglio A, Perrini C, Tasquier R et al. Equine amniotic microvesicles and their anti‐inflammatory potential in a tenocyte model in vitro. Stem Cells Dev 2016;25:610–621.
    1. Iglesias DM, El‐Kares R, Taranta A et al. Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro. PLoS One 2012;7.
    1. Zhang G, Zou X, Miao S et al. The anti‐oxidative role of micro‐vesicles derived from human Wharton‐Jelly mesenchymal stromal cells through NOX2/gp91(phox) suppression in alleviating renal ischemia‐reperfusion injury in rats. PLoS One 2014;9:e92129.

Source: PubMed

Подписаться