Mesenchymal Stromal Cells Are More Effective Than Their Extracellular Vesicles at Reducing Lung Injury Regardless of Acute Respiratory Distress Syndrome Etiology

Johnatas D Silva, Ligia L de Castro, Cassia L Braga, Gisele P Oliveira, Stefano A Trivelin, Carlos M Barbosa-Junior, Marcelo M Morales, Claudia C Dos Santos, Daniel J Weiss, Miquéias Lopes-Pacheco, Fernanda F Cruz, Patricia R M Rocco, Johnatas D Silva, Ligia L de Castro, Cassia L Braga, Gisele P Oliveira, Stefano A Trivelin, Carlos M Barbosa-Junior, Marcelo M Morales, Claudia C Dos Santos, Daniel J Weiss, Miquéias Lopes-Pacheco, Fernanda F Cruz, Patricia R M Rocco

Abstract

Although mesenchymal stromal cells (MSCs) have demonstrated beneficial effects on experimental acute respiratory distress syndrome (ARDS), preconditioning may be required to potentiate their therapeutic effects. Additionally, administration of cell-free products, such as extracellular vesicles (EVs) obtained from MSC-conditioned media, might be as effective as MSCs. In this study, we comparatively evaluated the effects of MSCs, preconditioned or not with serum collected from mice with pulmonary or extrapulmonary ARDS (ARDSp and ARDSexp, respectively), and the EVs derived from these cells on lung inflammation and remodeling in ARDSp and ARDSexp mice. Administration of MSCs (preconditioned or not), but not their EVs, reduced static lung elastance, interstitial edema, and collagen fiber content in both ARDSp and ARDSexp. Although MSCs and EVs reduced alveolar collapse and neutrophil cell counts in lung tissue, therapeutic responses were superior in mice receiving MSCs, regardless of preconditioning. Despite higher total cell, macrophage, and neutrophil counts in bronchoalveolar lavage fluid in ARDSp than ARDSexp, MSCs and EVs (preconditioned or not) led to a similar decrease. In ARDSp, both MSCs and EVs, regardless of preconditioning, reduced levels of tumor necrosis factor- (TNF-) α, interleukin-6, keratinocyte chemoattractant (KC), vascular endothelial growth factor (VEGF), and transforming growth factor- (TGF-) β in lung homogenates. In ARDSexp, TNF-α, interleukin-6, and KC levels were reduced by MSCs and EVs, preconditioned or not; only MSCs reduced VEGF levels, while TGF-β levels were similarly increased in ARDSexp treated either with saline, MSCs, or EVs, regardless of preconditioning. In conclusion, MSCs yielded greater overall improvement in ARDS in comparison to EVs derived from the same number of cells and regardless of the preconditioning status. However, the effects of MSCs and EVs differed according to ARDS etiology.

Conflict of interest statement

The authors declare that there is no conflict of interest regarding the publication of this manuscript.

Figures

Figure 1
Figure 1
Schematic flow chart and timeline of study design. ARDS was induced by administration of Escherichia coli lipopolysaccharide intratracheally (ARDSp) or intraperitoneally (ARDSexp). Control mice (C) received saline solution intratracheally (Cp) or intraperitoneally (Cexp). After 24 h, ARDSp and ARDSexp animals were further randomized to receive saline (50 μL, SAL), bone marrow-derived MSCs (105, 50 μL), or EVs (105, 50 μL), stimulated (MSC serum, EV serum) or not with serum obtained from ARDSp or ARDSexp animals. All data were analyzed on day 2.
Figure 2
Figure 2
Lung histology. Representative photomicrographs of lung parenchyma stained with hematoxylin and eosin from (a) pulmonary ARDS (ARDSp) and (b) extrapulmonary ARDS (ARDSexp) animals. ARDS was induced by administration of Escherichia coli lipopolysaccharide intratracheally (ARDSp) or intraperitoneally (ARDSexp). Control mice (C) received saline solution intratracheally (Cp) or intraperitoneally (Cexp). After 24 h, ARDSp and ARDSexp animals were further randomized to receive saline (50 μL, SAL), bone marrow-derived MSCs (105, 50 μL), or EVs (105, 50 μL), stimulated (MSC serum, EV serum) or not (MSCs, EVs) with serum obtained from ARDSp or ARDSexp animals.
Figure 3
Figure 3
Total and differential cell counts, as well as protein content, in bronchoalveolar lavage fluid in pulmonary ARDS (ARDSp) (a) and extrapulmonary ARDS (ARDSexp) (b) animals. ARDS was induced by administration of Escherichia coli lipopolysaccharide intratracheally (ARDSp) or intraperitoneally (ARDSexp). Control mice (C) received saline solution intratracheally (Cp) or intraperitoneally (Cexp). After 24 h, ARDSp and ARDSexp animals were further randomized to receive saline (50 μL, SAL), bone marrow-derived MSCs (105, 50 μL), or EVs (105, 50 μL), stimulated or not with serum (MSCs, EVs, MSC serum, and EV serum) obtained from ARDSp or ARDSexp animals. Values were expressed as mean ± standard deviation of 6 animals per group. ∗Significantly different from the corresponding C group (p < 0.05). ∗∗Significantly different from the corresponding ARDS group (p < 0.05).
Figure 4
Figure 4
Lung mechanics. Static lung elastance (Est, L) and resistive (ΔP1, L) and viscoelastic (ΔP2, L) pressures in animals with experimental pulmonary ARDS (ARDSp) (a) and extrapulmonary ARDS (ARDSexp) (b). ARDS was induced by administration of Escherichia coli lipopolysaccharide intratracheally (ARDSp) or intraperitoneally (ARDSexp). Control mice (C) received saline solution intratracheally (Cp) or intraperitoneally (Cexp). After 24 h, ARDSp and ARDSexp animals were further randomized to receive saline (50 μL, SAL), bone marrow-derived MSCs (105, 50 μL), or EVs (105, 50 μL), stimulated (MSC serum, EV serum) or not (MSCs, EVs) with serum obtained from ARDSp or ARDSexp animals. Values were expressed as mean + standard deviation of 6 animals per group. ∗Significantly different from the corresponding C group (p < 0.05). ∗∗Significantly different from the corresponding ARDS group (p < 0.05).
Figure 5
Figure 5
Protein levels of mediators in lung tissue. Protein levels of tumor necrosis factor- (TNF-) α, interleukin- (IL-) 6, IL-10, keratinocyte chemoattractant (KC) (a murine IL-8 homolog), vascular endothelial growth factor (VEGF), and transforming growth factor- (TGF-) β in lung tissue homogenate from (a) pulmonary ARDS (ARDSp) and (b) extrapulmonary ARDS (ARDSexp) animals. ARDS was induced by administration of Escherichia coli lipopolysaccharide intratracheally (ARDSp) or intraperitoneally (ARDSexp). Control mice (C) received saline solution intratracheally (Cp) or intraperitoneally (Cexp). After 24 h, ARDSp and ARDSexp animals were further randomized to receive saline (50 μL, SAL), bone marrow-derived MSCs (105, 50 μL), or EVs (105, 50 μL), stimulated (MSC serum, EV serum) or not with serum (MSCs, EVs) obtained from ARDSp or ARDSexp animals. Boxes show the interquartile (P25-P75) range, whiskers denote the range (minimum-maximum), and the horizontal line represents the median of animals per group. ∗Significantly different from the corresponding C group (p < 0.05). ∗∗Significantly different from the corresponding ARDS group (p < 0.05). #Significantly different from the corresponding MSC group (p < 0.05).
Figure 6
Figure 6
Exposure to conditioned media from MSCs or EVs induces a shift in macrophage polarization in vitro to the M2 rather than the M1 phenotype. Alveolar macrophages (105 cells per well) were collected from (a) Cp and ARDSp or (b) Cexp and ARDSexp mice. Cells were cultured in regular conditions (Cp, Cexp, ARDSp-SAL, and ARDSexp-SAL) or with conditioned media obtained from MSCs (105 cells per well) either unstimulated or stimulated with serum (serum) or extracellular vesicles derived from ARDSp or ARDSexp mice for 24 h. Relative gene expression of iNOS, IL-1β, IL-6, IL-10, arginase, and TGF-β was calculated as a ratio of average gene expression compared to expression of the housekeeping gene 36B4 and presented as fold changes relative to the Cp or Cexp group (alveolar macrophages from Cp or Cexp animals cultured with conditioned media from unstimulated MSCs). Results are presented as means + SD of alveolar macrophages pooled from 3 mice/group. All measurements were performed in triplicate. ∗Significantly different from the corresponding C group (p < 0.05). ∗∗Significantly different from the corresponding ARDS group (p < 0.05). #Significantly different from the corresponding MSC group (p < 0.05).

References

    1. Bein T., Grasso S., Moerer O., et al. The standard of care of patients with ARDS: ventilatory settings and rescue therapies for refractory hypoxemia. Intensive Care Medicine. 2016;42(5):699–711. doi: 10.1007/s00134-016-4325-4.
    1. Bellani G., Laffey J. G., Pham T., et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016;315(8):788–800. doi: 10.1001/jama.2016.0291.
    1. Biehl M., Kashyap R., Ahmed A. H., et al. Six-month quality-of-life and functional status of acute respiratory distress syndrome survivors compared to patients at risk: a population-based study. Critical Care. 2015;19(1) doi: 10.1186/s13054-015-1062-y.
    1. Duggal A., Ganapathy A., Ratnapalan M., Adhikari N. K. Pharmacological treatments for acute respiratory distress syndrome: systematic review. Minerva Anestesiologica. 2015;81:567–588.
    1. Cruz F. F., Weiss D. J., Rocco P. R. M. Prospects and progress in cell therapy for acute respiratory distress syndrome. Expert Opinion on Biological Therapy. 2016;16(11):1353–1360. doi: 10.1080/14712598.2016.1218845.
    1. Hayes M., Curley G. F., Masterson C., Devaney J., O’Toole D., Laffey J. G. Mesenchymal stromal cells are more effective than the MSC secretome in diminishing injury and enhancing recovery following ventilator-induced lung injury. Intensive Care Medicine Experimental. 2015;3(1) doi: 10.1186/s40635-015-0065-y.
    1. Antunes M. A., Abreu S. C., Cruz F. F., et al. Effects of different mesenchymal stromal cell sources and delivery routes in experimental emphysema. Respiratory Research. 2014;15(1):p. 118. doi: 10.1186/s12931-014-0118-x.
    1. Curley G. F., Hayes M., Ansari B., et al. Mesenchymal stem cells enhance recovery and repair following ventilator-induced lung injury in the rat. Thorax. 2012;67(6):496–501. doi: 10.1136/thoraxjnl-2011-201059.
    1. Silva J. D., Lopes-Pacheco M., Paz A. H. R., et al. Mesenchymal stem cells from bone marrow, adipose tissue, and lung tissue differentially mitigate lung and distal organ damage in experimental acute respiratory distress syndrome. Critical Care Medicine. 2018;46(2):e132–e140. doi: 10.1097/CCM.0000000000002833.
    1. Mei S. H. J., Haitsma J. J., Dos Santos C. C., et al. Mesenchymal stem cells reduce inflammation while enhancing bacterial clearance and improving survival in sepsis. American Journal of Respiratory and Critical Care Medicine. 2010;182(8):1047–1057. doi: 10.1164/rccm.201001-0010OC.
    1. Maron-Gutierrez T., Silva J. D., Asensi K. D., et al. Effects of mesenchymal stem cell therapy on the time course of pulmonary remodeling depend on the etiology of lung injury in mice. Critical Care Medicine. 2013;41(11):e319–e333. doi: 10.1097/CCM.0b013e31828a663e.
    1. Gupta N., Su X., Popov B., Lee J. W., Serikov V., Matthay M. A. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. Journal of Immunology. 2007;179(3):1855–1863. doi: 10.4049/jimmunol.179.3.1855.
    1. Monsel A., Zhu Y. G., Gudapati V., Lim H., Lee J. W. Mesenchymal stem cell derived secretome and extracellular vesicles for acute lung injury and other inflammatory lung diseases. Expert Opinion on Biological Therapy. 2016;16(7):859–871. doi: 10.1517/14712598.2016.1170804.
    1. Ionescu L., Byrne R. N., van Haaften T., et al. Stem cell conditioned medium improves acute lung injury in mice: in vivo evidence for stem cell paracrine action. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2012;303(11):L967–L977. doi: 10.1152/ajplung.00144.2011.
    1. Zhu Y. G., Feng X. M., Abbott J., et al. Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice. Stem Cells. 2014;32(1):116–125. doi: 10.1002/stem.1504.
    1. Witwer K. W., van Balkom B. W. M., Bruno S., et al. Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications. Journal of Extracellular Vesicles. 2019;8(1) doi: 10.1080/20013078.2019.1609206.
    1. Cruz F. F., Borg Z. D., Goodwin M., 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 Translational Medicine. 2015;4(11):1302–1316. doi: 10.5966/sctm.2014-0280.
    1. Abreu S. C., Lopes-Pacheco M., da Silva A. L., et al. Eicosapentaenoic acid enhances the effects of mesenchymal stromal cell therapy in experimental allergic asthma. Frontiers in Immunology. 2018;9:p. 1147. doi: 10.3389/fimmu.2018.01147.
    1. Saparov A., Ogay V., Nurgozhin T., Jumabay M., Chen W. C. W. Preconditioning of human mesenchymal stem cells to enhance their regulation of the immune response. Stem Cells Int. 2016;2016, article 3924858:1–10. doi: 10.1155/2016/3924858.
    1. Silva L. H. A., Antunes M. A., Dos Santos C. C., Weiss D. J., Cruz F. F., Rocco P. R. M. Strategies to improve the therapeutic effects of mesenchymal stromal cells in respiratory diseases. Stem Cell Research & Therapy. 2018;9(1):p. 45. doi: 10.1186/s13287-018-0802-8.
    1. Abreu S. C., Xisto D. G., de Oliveira T. B., et al. Serum from asthmatic mice potentiates the therapeutic effects of mesenchymal stromal cells in experimental allergic asthma. Stem Cells Translational Medicine. 2019;8(3):301–312. doi: 10.1002/sctm.18-0056.
    1. Pelosi P., D'Onofrio D., Chiumello D., et al. Pulmonary and extrapulmonary acute respiratory distress syndrome are different. European Respiratory Journal. 2003;22(Supplement 42):48s–56s. doi: 10.1183/09031936.03.00420803.
    1. Kilkenny C., Browne W. J., Cuthill I. C., Emerson M., Altman D. G. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biology. 2010;8(6) doi: 10.1371/journal.pbio.1000412.
    1. Bates J. H., Ludwig M. S., Sly P. D., Brown K., Martin J. G., Fredberg J. J. Interrupter resistance elucidated by alveolar pressure measurement in open-chest normal dogs. Journal of Applied Physiology. 1988;65(1):408–414. doi: 10.1152/jappl.1988.65.1.408.
    1. Silva J. D., Paredes B. D., Araújo I. M., et al. Effects of bone marrow-derived mononuclear cells from healthy or acute respiratory distress syndrome donors on recipient lung-injured mice. Critical Care Medicine. 2014;42(7):e510–e524. doi: 10.1097/CCM.0000000000000296.
    1. Hsia C. C., Hyde D. M., Ochs M., Weibel E. R., ATS/ERS Joint Task Force on Quantitative Assessment of Lung Structure An official research policy statement of the American Thoracic Society/European Respiratory Society: standards for quantitative assessment of lung structure. American Journal of Respiratory and Critical Care Medicine. 2010;181(4):394–418. doi: 10.1164/rccm.200809-1522ST.
    1. Araújo I. M., Abreu S. C., Maron-Gutierrez T., et al. Bone marrow-derived mononuclear cell therapy in experimental pulmonary and extrapulmonary acute lung injury. Critical Care Medicine. 2010;38(8):1733–1741. doi: 10.1097/CCM.0b013e3181e796d2.
    1. Hizume D. C., Rivero D. H. R. F., Kasahara D. I., et al. Effects of positive end-expiratory pressure in an experimental model of acute myocardial infarct in Wistar rats. Shock. 2007;27(5):584–589. doi: 10.1097/01.shk.0000248594.79012.d6.
    1. Maron-Gutierrez T., Silva J., Cruz F., et al. Insult-dependent effect of bone marrow cell therapy on inflammatory response in a murine model of extrapulmonary acute respiratory distress syndrome. Stem Cell Research & Therapy. 2013;4(5):p. 123. doi: 10.1186/scrt334.
    1. Zhang X., Goncalves R., Mosser D. M. The isolation and characterization of murine macrophages. Current Protocols in Immunology. 2008;83 doi: 10.1002/0471142735.im1401s83.
    1. Santos F. B., Nagato L. K. S., Boechem N. M., et al. Time course of lung parenchyma remodeling in pulmonary and extrapulmonary acute lung injury. Journal of Applied Physiology. 2006;100(1):98–106. doi: 10.1152/japplphysiol.00395.2005.
    1. Menezes S. L. S., Bozza P. T., Faria Neto H. C. C., et al. Pulmonary and extrapulmonary acute lung injury: inflammatory and ultrastructural analyses. Journal of Applied Physiology. 2005;98(5):1777–1783. doi: 10.1152/japplphysiol.01182.2004.
    1. Xu J., Woods C. R., Mora A. L., et al. Prevention of endotoxin-induced systemic response by bone marrow-derived mesenchymal stem cells in mice. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2007;293(1):L131–L141. doi: 10.1152/ajplung.00431.2006.
    1. Krasnodembskaya A., Samarani G., Song Y., et al. Human mesenchymal stem cells reduce mortality and bacteremia in gram-negative sepsis in mice in part by enhancing the phagocytic activity of blood monocytes. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2012;302(10):L1003–L1013. doi: 10.1152/ajplung.00180.2011.
    1. Li J. W., Wu X. Mesenchymal stem cells ameliorate LPS-induced acute lung injury through KGF promoting alveolar fluid clearance of alveolar type II cells. European Review for Medical and Pharmacological Sciences. 2015;19:2368–2378.
    1. Lee J. W., Fang X., Gupta N., Serikov V., Matthay M. A. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proceedings of the National Academy of Sciences. 2009;106(38):16357–16362. doi: 10.1073/pnas.0907996106.
    1. Luo C. J., Zhang F. J., Zhang L., et al. Mesenchymal stem cells ameliorate sepsis-associated acute kidney injury in mice. Shock. 2014;41(2):123–129. doi: 10.1097/SHK.0000000000000080.
    1. McIntyre L. A., Moher D., Fergusson D. A., et al. Efficacy of mesenchymal stromal cell therapy for acute lung injury in preclinical animal models: a systematic review. PLoS One. 2016;11(1, article e0147170) doi: 10.1371/journal.pone.0147170.
    1. Wilson J. G., Liu K. D., Zhuo H., et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. The Lancet Respiratory Medicine. 2015;3(1):24–32. doi: 10.1016/S2213-2600(14)70291-7.
    1. Matthay M. A., Calfee C. S., Zhuo H., et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. The Lancet Respiratory Medicine. 2019;7(2):154–162. doi: 10.1016/S2213-2600(18)30418-1.
    1. Deng J. C., Standiford T. J. Growth factors and cytokines in acute lung injury. Comprehensive Physiology. 2011;1(1):81–104. doi: 10.1002/cphy.c090011.
    1. Bhatia M., Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. The Journal of Pathology. 2004;202(2):145–156. doi: 10.1002/path.1491.
    1. Lopes-Pacheco M., Bandeira E., Morales M. M. Cell-based therapy for silicosis. Stem Cells Int. 2016;2016, article 5091838:1–9. doi: 10.1155/2016/5091838.
    1. Pittet J. F., Griffiths M. J. D., Geiser T., et al. TGF-beta is a critical mediator of acute lung injury. The Journal of Clinical Investigation. 2001;107(12):1537–1544. doi: 10.1172/JCI11963.
    1. Thickett D. R., Armstrong L., Christie S. J., Millar A. B. Vascular endothelial growth factor may contribute to increased vascular permeability in acute respiratory distress syndrome. American Journal of Respiratory and Critical Care Medicine. 2001;164(9):1601–1605. doi: 10.1164/ajrccm.164.9.2011071.
    1. Lopes-Pacheco M., Ventura T. G., de Oliveira H. D.'. A., et al. Infusion of bone marrow mononuclear cells reduced lung fibrosis but not inflammation in the late stages of murine silicosis. PLoS One. 2014;9(10) doi: 10.1371/journal.pone.0109982.
    1. Chiossone L., Conte R., Spaggiari G. M., et al. Mesenchymal stromal cells induce peculiar alternatively activated macrophages capable of dampening both innate and adaptive immune responses. Stem Cells. 2016;34(7):1909–1921. doi: 10.1002/stem.2369.
    1. Nemeth K., Keane-Myers A., Brown J. M., et al. Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(12):5652–5657. doi: 10.1073/pnas.0910720107.

Source: PubMed

3
Abonner