Human adipose tissue mesenchymal stromal cells and their extracellular vesicles act differentially on lung mechanics and inflammation in experimental allergic asthma

Ligia Lins de Castro, Debora Gonçalves Xisto, Jamil Zola Kitoko, Fernanda Ferreira Cruz, Priscilla Christina Olsen, Patricia Albuquerque Garcia Redondo, Tatiana Paula Teixeira Ferreira, Daniel Jay Weiss, Marco Aurélio Martins, Marcelo Marcos Morales, Patricia Rieken Macedo Rocco, Ligia Lins de Castro, Debora Gonçalves Xisto, Jamil Zola Kitoko, Fernanda Ferreira Cruz, Priscilla Christina Olsen, Patricia Albuquerque Garcia Redondo, Tatiana Paula Teixeira Ferreira, Daniel Jay Weiss, Marco Aurélio Martins, Marcelo Marcos Morales, Patricia Rieken Macedo Rocco

Abstract

Background: Asthma is a chronic inflammatory disease that can be difficult to treat due to its complex pathophysiology. Most current drugs focus on controlling the inflammatory process, but are unable to revert the changes of tissue remodeling. Human mesenchymal stromal cells (MSCs) are effective at reducing inflammation and tissue remodeling; nevertheless, no study has evaluated the therapeutic effects of extracellular vesicles (EVs) obtained from human adipose tissue-derived MSCs (AD-MSC) on established airway remodeling in experimental allergic asthma.

Methods: C57BL/6 female mice were sensitized and challenged with ovalbumin (OVA). Control (CTRL) animals received saline solution using the same protocol. One day after the last challenge, each group received saline, 105 human AD-MSCs, or EVs (released by 105 AD-MSCs). Seven days after treatment, animals were anesthetized for lung function assessment and subsequently euthanized. Bronchoalveolar lavage fluid (BALF), lungs, thymus, and mediastinal lymph nodes were harvested for analysis of inflammation. Collagen fiber content of airways and lung parenchyma were also evaluated.

Results: In OVA animals, AD-MSCs and EVs acted differently on static lung elastance and on BALF regulatory T cells, CD3+CD4+ T cells, and pro-inflammatory mediators (interleukin [IL]-4, IL-5, IL-13, and eotaxin), but similarly reduced eosinophils in lung tissue, collagen fiber content in airways and lung parenchyma, levels of transforming growth factor-β in lung tissue, and CD3+CD4+ T cell counts in the thymus. No significant changes were observed in total cell count or percentage of CD3+CD4+ T cells in the mediastinal lymph nodes.

Conclusions: In this immunocompetent mouse model of allergic asthma, human AD-MSCs and EVs effectively reduced eosinophil counts in lung tissue and BALF and modulated airway remodeling, but their effects on T cells differed in lung and thymus. EVs may hold promise for asthma; however, further studies are required to elucidate the different mechanisms of action of AD-MSCs versus their EVs.

Keywords: Asthma; Extracellular vesicles; Inflammation; Mesenchymal stromal cells; Remodeling.

Figures

Fig. 1
Fig. 1
Characterization of EVs. a Representative graph of the intensity and hydrodynamic diameter of extracellular vesicle samples analyzed using the dynamic light scattering technique. Graph showing two populations of extracellular vesicles obtained from human adipose tissue-derived mesenchymal stromal cells, one of lower intensity and medium size, characteristic of exosomes, and another with greater intensity and average size, characteristic of microvesicles. b, c, and d Scanning electron microscopy of mesenchymal stromal cells. b Mesenchymal stromal cells before induction of cellular stress (fetal bovine serum deprivation), showing the presence of few extracellular vesicles. c Image obtained 12 hours after cellular stress induction, showing increase in the number of vesicles in the cell surface. d Higher magnification of image C, showing extracellular vesicles with some indicative sizes
Fig. 2
Fig. 2
Experimental design. C57BL/6 female mice were divided into two groups: CTRL (sensitized and challenged with saline) and OVA (sensitized and challenged with ovalbumin). Seven intraperitoneal (i.p.) sensitizations were performed. On days 40, 43, and 46 after first sensitization, an intratracheal challenge (i.t.) was performed. Treatment was administered intravenously (i.v.); 1 day after the last challenge and 7 days after this treatment, the animals were euthanized for data acquisition. The treatment consisted of saline (SAL), human adipose-derived mesenchymal stromal cells (AD-MSC), or extracellular vesicles (EV) at a dose equivalent to 105 AD-MSC (37 μg of total protein)
Fig. 3
Fig. 3
Lung mechanics. a Static lung elastance (Est,L). b Lung resistive (ΔP1, white bar) and viscoelastic (ΔP2, gray bar) pressures. Data are presented as means ± SD of seven animals/group. CTRL mice sensitized and challenged with saline, OVA mice sensitized and challenged with ovalbumin, OVA-SAL OVA mice treated with saline, OVA-AD-MSC OVA mice treated with mesenchymal stromal cells derived from adipose tissue (AD-MSCs), OVA-EV OVA mice treated with extracellular vesicles derived from AD-MSCs. CTRL versus OVA-SAL (Est,L p = 0.04, ΔP1, p = 0.003, ΔP2, p = 0.005)
Fig. 4
Fig. 4
Representative photomicrographs of lung parenchyma (upper panels) and airway (lower panels) stained with Sirius Red under polarization. Percentage of collagen fiber content in lung parenchyma and airway. Levels of transforming growth factor beta (TGF-β) in lung tissue. Data are presented as means + SD of seven animals/group. CTRL mice sensitized and challenged with saline, OVA mice sensitized and challenged with ovalbumin, OVA-SAL OVA mice treated with saline, OVA-AD-MSC OVA mice treated with mesenchymal stromal cells derived from adipose tissue (AD-MSCs), OVA-EV OVA mice treated with extracellular vesicles derived from AD-MSCs. CTRL versus OVA-SAL (parenchyma p < 0.0001, airway p = 0.0002, TGF-β p = 0.0007)
Fig. 5
Fig. 5
Representative photomicrographs of lung parenchyma stained with Sirius Red. Squares indicate peribronchial area. Lower panel: number of eosinophils in the peribronchial area. Data are presented as means + SD of seven animals/group. aCTRL mice sensitized and challenged with saline, bOVA-SALOVA mice sensitized and challenged with ovalbumin and then treated with saline, cOVA-AD-MSC OVA mice treated with mesenchymal stromal cells derived from adipose tissue (AD-MSCs), dOVA-EV OVA mice treated with extracellular vesicles derived from AD-MSCs, e Number of eosinophils in lung parenchyma. CTRL versus OVA-SAL (p < 0.0001)
Fig. 6
Fig. 6
Total cell count and percentage of T cells (CD3+CD4+), Treg cells (CD4+CD25+Foxp3+), and eosinophils (Siglec-F+) in bronchoalveolar lavage fluid (BALF). Data are presented as means + SD of ten animals/group. CTRL mice sensitized and challenged with saline, OVA mice sensitized and challenged with ovalbumin, OVA-SAL OVA mice treated with saline, OVA-AD-MSC OVA mice treated with mesenchymal stromal cells derived from adipose tissue (AD-MSCs), OVA-EV OVA mice treated with extracellular vesicles derived from AD-MSCs. CTRL versus OVA-SAL (total cell count p < 0.0001, eosinophils p = 0.0006, Treg cells p < 0.0001, T cells p = 0.034)
Fig. 7
Fig. 7
Levels of interleukin (IL)-4, IL-5, IL-13, eotaxin, IL-10, and interferon (IFN)-γ. Data are presented as median and interquartile range of seven animals/group. CTRL mice sensitized and challenged with saline, OVA mice sensitized and challenged with ovalbumin, OVA-SAL OVA mice treated with saline, OVA-AD-MSC OVA mice treated with mesenchymal stromal cells derived from adipose tissue (AD-MSCs), OVA-EV OVA mice treated with extracellular vesicles derived from AD-MSCs. CTRL versus OVA-SAL (IL-13 p = 0.004, eotaxin p = 0.0011)
Fig. 8
Fig. 8
Total cell count and percentage of T cells (CD3+CD4+) counts in thymus and mediastinal lymph nodes. Data are presented as means + SD of ten animals/group. CTRL mice sensitized and challenged with saline, OVA mice sensitized and challenged with ovalbumin, OVA-SAL OVA mice treated with saline, OVA-AD-MSC OVA mice treated with mesenchymal stromal cells derived from adipose tissue (AD-MSCs), OVA-EV OVA mice treated with extracellular vesicles derived from AD-MSCs. CTRL versus OVA-SAL (thymus p = 0.0002)

References

    1. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention. 2016. Available from . Accessed 13 June 2017.
    1. Barnes PJ. New therapies for asthma: is there any progress? Trends Pharmacol Sci. 2010;31(7):335–43. doi: 10.1016/j.tips.2010.04.009.
    1. Papierniak ES, Lowenthal DT, Harman E. Novel therapies in asthma: leukotriene antagonists, biologic agents, and beyond. Am J Ther. 2013;20(1):79–103. doi: 10.1097/MJT.0b013e31826915c2.
    1. Singh D, Petavy F, Macdonald AJ, Lazaar AL, O’Connor BJ. The inhaled phosphodiesterase 4 inhibitor GSK256066 reduces allergen challenge responses in asthma. Respir Res. 2010;11:26. doi: 10.1186/1465-9921-11-26.
    1. Yazid S, Sinniah A, Solito E, Calder V, Flower RJ. Anti-allergic cromones inhibit histamine and eicosanoid release from activated human and murine mast cells by releasing Annexin A1. PLoS One. 2013;8(3):e58963. doi: 10.1371/journal.pone.0058963.
    1. Cho KS, Park MK, Kang SA, Park HY, Hong SL, Park HK, Yu HS, Roh HJ. Adipose-derived stem cells ameliorate allergic airway inflammation by inducing regulatory T cells in a mouse model of asthma. Mediat Inflamm. 2014;2014:436476. doi: 10.1155/2014/436476.
    1. Park HK, Cho KS, Park HY, Shin DH, Kim YK, Jung JS, Park SK, Roh HJ. Adipose-derived stromal cells inhibit allergic airway inflammation in mice. Stem Cells Dev. 2010;19(11):1811–8. doi: 10.1089/scd.2009.0513.
    1. Marinas-Pardo L, Mirones I, Amor-Carro O, Fraga-Iriso R, Lema-Costa B, Cubillo I, Rodriguez Milla MA, Garcia-Castro J, Ramos-Barbon D. Mesenchymal stem cells regulate airway contractile tissue remodeling in murine experimental asthma. Allergy. 2014;69(6):730–40. doi: 10.1111/all.12392.
    1. Cho KS, Lee JH, Park MK, Park HK, Yu HS, Roh HJ. Prostaglandin E2 and transforming growth factor-beta play a critical role in suppression of allergic airway inflammation by adipose-derived stem cells. PLoS One. 2015;10(7):e0131813. doi: 10.1371/journal.pone.0131813.
    1. Martinez-Gonzalez I, Cruz MJ, Moreno R, Morell F, Munoz X, Aran JM. Human mesenchymal stem cells resolve airway inflammation, hyperreactivity, and histopathology in a mouse model of occupational asthma. Stem Cells Dev. 2014;23(19):2352–63. doi: 10.1089/scd.2013.0616.
    1. Cruz FF, Borg ZD, Goodwin M, Sokocevic D, Wagner DE, Coffey A, Antunes M, Robinson KL, Mitsialis SA, Kourembanas S, et al. Systemic administration of human bone marrow-derived mesenchymal stromal cell extracellular vesicles ameliorates aspergillus hyphal extract-induced allergic airway inflammation in immunocompetent mice. Stem Cells Transl Med. 2015;4(11):1302–16. doi: 10.5966/sctm.2014-0280.
    1. Bourin P, Bunnell BA, Casteilla L, Dominici M, Katz AJ, March KL, Redl H, Rubin JP, Yoshimura K, Gimble JM. 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(6):641–8. doi: 10.1016/j.jcyt.2013.02.006.
    1. Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S, Collino F, Morando L, Busca A, Falda M, Bussolati B, et al. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J Am Soc Nephrol. 2009;20(5):1053–67. doi: 10.1681/ASN.2008070798.
    1. Silva PL, Passaro CP, Cagido VR, Bozza M, Dolhnikoff M, Negri EM, Morales MM, Capelozzi VL, Zin WA, Rocco PR. Impact of lung remodelling on respiratory mechanics in a model of severe allergic inflammation. Respir Physiol Neurobiol. 2008;160(3):239–48. doi: 10.1016/j.resp.2007.10.009.
    1. Abreu SC, Antunes MA, de Castro JC, de Oliveira MV, Bandeira E, Ornellas DS, Diaz BL, Morales MM, Xisto DG, Rocco PR. Bone marrow-derived mononuclear cells vs. mesenchymal stromal cells in experimental allergic asthma. Respir Physiol Neurobiol. 2013;187(2):190–8. doi: 10.1016/j.resp.2013.03.014.
    1. Xisto DG, Farias LL, Ferreira HC, Picanco MR, Amitrano D, Lapa ESJR, Negri EM, Mauad T, Carnielli D, Silva LF, et al. Lung parenchyma remodeling in a murine model of chronic allergic inflammation. Am J Respir Crit Care Med. 2005;171(8):829–37. doi: 10.1164/rccm.200408-997OC.
    1. Burburan SM, Xisto DG, Ferreira HC, Riva Ddos R, Carvalho GM, Zin WA, Rocco PR. Lung mechanics and histology during sevoflurane anesthesia in a model of chronic allergic asthma. Anesth Analg. 2007;104(3):631–7. doi: 10.1213/01.ane.0000255073.96001.cb.
    1. Bates JH, Ludwig MS, Sly PD, Brown K, Martin JG, Fredberg JJ. Interrupter resistance elucidated by alveolar pressure measurement in open-chest normal dogs. J Appl Physiol (1985) 1988;65(1):408–14.
    1. Saldiva PH, Zin WA, Santos RL, Eidelman DH, Milic-Emili J. Alveolar pressure measurement in open-chest rats. J Appl Physiol (1985) 1992;72(1):302–6.
    1. Ferreira TP, Mariano LL, Ghilosso-Bortolini R, de Arantes AC, Fernandes AJ, Berni M, Cecchinato V, Uguccioni M, Maj R, Barberis A, et al. Potential of PEGylated Toll-like receptor 7 ligands for controlling inflammation and functional changes in mouse models of asthma and silicosis. Front Immunol. 2016;7:95. doi: 10.3389/fimmu.2016.00095.
    1. Lowe AP, Broadley KJ, Nials AT, Ford WR, Kidd EJ. Adjustment of sensitisation and challenge protocols restores functional and inflammatory responses to ovalbumin in guinea-pigs. J Pharmacol Toxicol Methods. 2015;72:85–93. doi: 10.1016/j.vascn.2014.10.007.
    1. Meyerholz DK, Griffin MA, Castilow EM, Varga SM. Comparison of histochemical methods for murine eosinophil detection in an RSV vaccine-enhanced inflammation model. Toxicol Pathol. 2009;37(2):249–55. doi: 10.1177/0192623308329342.
    1. Abreu SC, Antunes MA, Mendonca L, Branco VC, de Melo EB, Olsen PC, Diaz BL, Weiss DJ, Paredes BD, Xisto DG, et al. Effects of bone marrow mononuclear cells from healthy or ovalbumin-induced lung inflammation donors on recipient allergic asthma mice. Stem Cell Res Ther. 2014;5(5):108. doi: 10.1186/scrt496.
    1. Abreu SC, Antunes MA, Xisto DG, Cruz FF, Branco VC, Bandeira E, Zola Kitoko J, de Araujo AF, Dellatorre-Texeira L, Olsen PC, et al. Bone marrow, adipose, and lung tissue-derived murine mesenchymal stromal cells release different mediators and differentially affect airway and lung parenchyma in experimental asthma. Stem Cells Transl Med. 2017;6(6):1557–67. doi: 10.1002/sctm.16-0398.
    1. da Silva AL, Magalhaes RF, Branco VC, Silva JD, Cruz FF, Marques PS, Ferreira TP, Morales MM, Martins MA, Olsen PC, et al. The tyrosine kinase inhibitor dasatinib reduces lung inflammation and remodelling in experimental allergic asthma. Br J Pharmacol. 2016;173(7):1236–47. doi: 10.1111/bph.13430.
    1. Antunes MA, Abreu SC, Silva AL, Parra-Cuentas ER, Ab’Saber AM, Capelozzi VL, Ferreira TP, Martins MA, Silva PM, Rocco PR. Sex-specific lung remodeling and inflammation changes in experimental allergic asthma. J Appl Physiol (1985) 2010;109(3):855–63. doi: 10.1152/japplphysiol.00333.2010.
    1. Zhu Y, Liu T, Song K, Fan X, Ma X, Cui Z. Adipose-derived stem cell: a better stem cell than BMSC. Cell Biochem Funct. 2008;26(6):664–75. doi: 10.1002/cbf.1488.
    1. Lee JK, Park SR, Jung BK, Jeon YK, Lee YS, Kim MK, Kim YG, Jang JY, Kim CW. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 2013;8(12):e84256. doi: 10.1371/journal.pone.0084256.
    1. Monsel A, Zhu YG, Gennai S, Hao Q, Hu S, Rouby JJ, Rosenzwajg M, Matthay MA, Lee JW. Therapeutic effects of human mesenchymal stem cell-derived microvesicles in severe pneumonia in mice. Am J Respir Crit Care Med. 2015;192(3):324–36. doi: 10.1164/rccm.201410-1765OC.
    1. Zhu YG, Feng XM, Abbott J, Fang XH, Hao Q, Monsel A, Qu JM, Matthay MA, Lee JW. Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice. Stem Cells. 2014;32(1):116–25. doi: 10.1002/stem.1504.
    1. Hamid Q, Tulic M. Immunobiology of asthma. Annu Rev Physiol. 2009;71:489–507. doi: 10.1146/annurev.physiol.010908.163200.
    1. Borowski A, Kuepper M, Horn U, Knupfer U, Zissel G, Hohne K, Luttmann W, Krause S, Virchow JC, Jr, Friedrich K. Interleukin-13 acts as an apoptotic effector on lung epithelial cells and induces pro-fibrotic gene expression in lung fibroblasts. Clin Exp Allergy. 2008;38(4):619–28. doi: 10.1111/j.1365-2222.2008.02944.x.
    1. Bellini A, Marini MA, Bianchetti L, Barczyk M, Schmidt M, Mattoli S. Interleukin (IL)-4, IL-13, and IL-17A differentially affect the profibrotic and proinflammatory functions of fibrocytes from asthmatic patients. Mucosal Immunol. 2012;5(2):140–9. doi: 10.1038/mi.2011.60.
    1. Rankin SM, Conroy DM, Williams TJ. Eotaxin and eosinophil recruitment: implications for human disease. Mol Med Today. 2000;6(1):20–7. doi: 10.1016/S1357-4310(99)01635-4.
    1. Kovacs EJ. Fibrogenic cytokines: the role of immune mediators in the development of scar tissue. Immunol Today. 1991;12(1):17–23. doi: 10.1016/0167-5699(91)90107-5.
    1. Halwani R, Al-Muhsen S, Al-Jahdali H, Hamid Q. Role of transforming growth factor-beta in airway remodeling in asthma. Am J Respir Cell Mol Biol. 2011;44(2):127–33. doi: 10.1165/rcmb.2010-0027TR.
    1. Makinde T, Murphy RF, Agrawal DK. The regulatory role of TGF-beta in airway remodeling in asthma. Immunol Cell Biol. 2007;85(5):348–56. doi: 10.1038/sj.icb.7100044.
    1. Raundhal M, Morse C, Khare A, Oriss TB, Milosevic J, Trudeau J, Huff R, Pilewski J, Holguin F, Kolls J, et al. High IFN-gamma and low SLPI mark severe asthma in mice and humans. J Clin Invest. 2015;125(8):3037–50. doi: 10.1172/JCI80911.
    1. Moore KW, O’Garra A, de Waal MR, Vieira P, Mosmann TR. Interleukin-10. Annu Rev Immunol. 1993;11:165–90. doi: 10.1146/annurev.iy.11.040193.001121.
    1. da Silva AL, Martini SV, Abreu SC, Samary Cdos S, Diaz BL, Fernezlian S, de Sa VK, Capelozzi VL, Boylan NJ, Goya RG, et al. DNA nanoparticle-mediated thymulin gene therapy prevents airway remodeling in experimental allergic asthma. J Control Release. 2014;180:125–33. doi: 10.1016/j.jconrel.2014.02.010.
    1. Matsumoto K, Inoue H, Tsuda M, Honda Y, Kibe A, Machida K, Yoshiura Y, Nakanishi Y. Different roles of interleukin-10 in onset and resolution of asthmatic responses in allergen-challenged mice. Respirology. 2005;10(1):18–26. doi: 10.1111/j.1440-1843.2005.00647.x.
    1. Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99(10):3838–43. doi: 10.1182/blood.V99.10.3838.
    1. Krampera M, Glennie S, Dyson J, Scott D, Laylor R, Simpson E, Dazzi F. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood. 2003;101(9):3722–9. doi: 10.1182/blood-2002-07-2104.
    1. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105(4):1815–22. doi: 10.1182/blood-2004-04-1559.
    1. Luz-Crawford P, Kurte M, Bravo-Alegria J, Contreras R, Nova-Lamperti E, Tejedor G, Noel D, Jorgensen C, Figueroa F, Djouad F, et al. Mesenchymal stem cells generate a CD4 + CD25 + Foxp3+ regulatory T cell population during the differentiation process of Th1 and Th17 cells. Stem Cell Res Ther. 2013;4(3):65. doi: 10.1186/scrt216.
    1. Lathrop MJ, Brooks EM, Bonenfant NR, Sokocevic D, Borg ZD, Goodwin M, Loi R, Cruz F, Dunaway CW, Steele C, et al. Mesenchymal stromal cells mediate Aspergillus hyphal extract-induced allergic airway inflammation by inhibition of the Th17 signaling pathway. Stem Cells Transl Med. 2014;3(2):194–205. doi: 10.5966/sctm.2013-0061.

Source: PubMed

3
Subscribe