Extracellular Vesicles Derived from Adipose Mesenchymal Stem Cells Alleviate PM2.5-Induced Lung Injury and Pulmonary Fibrosis

Yongheng Gao, Jinbo Sun, Chuan Dong, Mingxuan Zhao, Ying Hu, Faguang Jin, Yongheng Gao, Jinbo Sun, Chuan Dong, Mingxuan Zhao, Ying Hu, Faguang Jin

Abstract

BACKGROUND Exposure to PM2.5 (fine particulate matter ≤2.5 μm in aerodynamic diameter) in air increases the risk of lung injury and pulmonary fibrosis (PF). Extracellular vesicles (EVs) derived from adipose mesenchymal stem cells (ADSCs) have been identified as a potential treatment based on the proteins or RNAs delivery and immunomodulatory properties. Here, we assessed the protective effects and mechanisms of ADSCs-EVs on PM2.5-induced lung injury or PF. MATERIAL AND METHODS Rats (male, 6 weeks old) were exposed to PBS or PM2.5 (1.5 mg/kg/day) for 3 days a week for 4 weeks. ADSCs-EVs were extracted by ultracentrifugation. PBS and ADSCs-EVs were administrated through intratracheal instillation. After the end of exposure, the rats were anesthetized and killed. Lung tissues with different treatments were collected for Western blot analysis and HE, IHC, and IF staining analysis. Cells exposed to PM2.5 or "PM2.5+ADSCs-EVs" in vitro were also collected for further Western blotting, qRT-PCR, and IF staining evaluation. RESULTS The results indicated that the initial response of lungs exposed to PM2.5 was lung injury with oxidative stress and inflammation. Long-term PM2.5 exposure resulted in obvious PF in rats. Treatment with ADSCs-EVs decreased PM2.5-induced apoptosis and necrosis in type II alveolar epithelial cells and alleviated lung injury and PF in rats. ADSCs-EVs suppressed reactive oxygen species (ROS) levels and inflammation induced by PM2.5. Furthermore, ADSCs-EVs inhibited TGF-ßRI by transferring let-7d-5p and further mitigated PF. CONCLUSIONS Our results suggest that EVs derived from ADSCs can alleviate PM2.5-induced lung injury and PF.

Conflict of interest statement

Conflict of interest

None.

Figures

Figure 1
Figure 1
PM2.5 exposure induced lung injury and PF. (A) SEM image of PM2.5. (B) Particles size distribution of PM2.5. (C) HE staining of lungs at 6 h after single exposure to PBS (None) or PM2.5. (D) Relative ROS of cells, TGF-β1 and TNF-α concentrations in BALF at different time points (3, 6, 9, 12, and 24 h after PM2.5 single exposure) were detected. (E) TEM of type II alveolar epithelial cells 6 hours after rats were treated with PBS (None) or PM2.5. (F) Masson staining of lung tissues with treatments of PBS or PM2.5 for 4 weeks (Yellow arrows represented PF). (G) IHC staining of TGF-β1 in lungs after treatments of PBS or PM2.5 for 4 weeks. (* p<0.05; ** p<0.01; *** p<0.001).
Figure 2
Figure 2
The identification of ADSCs and AT II cells. (A) Multilineage differentiation of ADSCs (upper left: osteoblast differentiation, upper middle: adipocyte differentiation, upper right: cartilage differentiation) and typical image of ADSCs (lower). (B) SP-C immunofluorescence staining for AT II cells.
Figure 3
Figure 3
ADSCs-EVs alleviated lung injury induced by PM2.5. (A) TEM identification of ADSCs-EVs. (B) NTA analysis of ADSCs-EVs. (C) GM130, Alix, TSG101, and CD63 expressions in ADSCs-EVs and cell lysates were detected by Western blotting. (D) The uptake of ADSCs-EVs in ATII cells was detected by a confocal microscopy. (E) Apoptosis or necrosis of ATII cells with treatments of PBS (None), ADSCs-EVs, PM2.5, or PM2.5+ADSCs-EVs was detected by flow cytometry assay. (F) Six hours after the rats treated with PBS (None), ADSCs-EVs, PM2.5 or PM2.5+ADSCs-EVs, relative ROS level in BALF was measured. (G) Six hours after the rats treated with PBS (None), ADSCs-EVs, PM2.5, or PM2.5+ADSCs-EVs, TNF-α concentration in BALF was evaluated. (H) HE staining of lungs exposed to PBS (None), ADSCs-EVs, PM2.5 or PM2.5+ADSCs-EVs. (* p<0.05; ** p<0.01; *** p<0.001).
Figure 4
Figure 4
ADSCs-EVs alleviated PF induced by PM2.5. Masson staining, IHC staining for TGF-β1, IF staining for TGF-βRI and TGF-βRII in lungs exposed to PBS (None), ADSCs-EVs, PM2.5, or PM2.5+ADSCs-EVs for 4 weeks. (* p

Figure 5

ADSCs-EVs repressed TGF-βRI expression. (…

Figure 5

ADSCs-EVs repressed TGF-βRI expression. ( A ) Western blotting analysis of TGF-β1, TGF-βRI,…

Figure 5
ADSCs-EVs repressed TGF-βRI expression. (A) Western blotting analysis of TGF-β1, TGF-βRI, and TGF-βRII levels in ATII cells under different treatments. (B) The miRNA levels in ATII cells with different treatments were measured by qRT-PCR. (* p<0.05; ** p<0.01; *** p<0.001).

Figure 6

let-7d-5p directly inhibited the expression…

Figure 6

let-7d-5p directly inhibited the expression of TGF-βRI. ( A ) The expression of…

Figure 6
let-7d-5p directly inhibited the expression of TGF-βRI. (A) The expression of TGF-βRI under different treatments was tested by Western blotting. (B) The expression of TGF-βRI mRNA under different treatments was tested by qRT-PCR. (C) Schematic representation of matching sequence between TGF-βRI 3′UTR mRNA and let-7d-5p. (D) Luciferase activity of ATII cells co-transfected with reporter plasmid (“TGF-βRI wt” or “TGF-βRI mt”) and let-7d-5p or miRNA-NC. (* p<0.05; ** p<0.01; *** p<0.001).
Figure 5
Figure 5
ADSCs-EVs repressed TGF-βRI expression. (A) Western blotting analysis of TGF-β1, TGF-βRI, and TGF-βRII levels in ATII cells under different treatments. (B) The miRNA levels in ATII cells with different treatments were measured by qRT-PCR. (* p<0.05; ** p<0.01; *** p<0.001).
Figure 6
Figure 6
let-7d-5p directly inhibited the expression of TGF-βRI. (A) The expression of TGF-βRI under different treatments was tested by Western blotting. (B) The expression of TGF-βRI mRNA under different treatments was tested by qRT-PCR. (C) Schematic representation of matching sequence between TGF-βRI 3′UTR mRNA and let-7d-5p. (D) Luciferase activity of ATII cells co-transfected with reporter plasmid (“TGF-βRI wt” or “TGF-βRI mt”) and let-7d-5p or miRNA-NC. (* p<0.05; ** p<0.01; *** p<0.001).

References

    1. Wuyts WA, Agostini C, Antoniou KM, et al. The pathogenesis of pulmonary fibrosis: A moving target. Eur Respir J. 2013;41:1207–18.
    1. Li N, Hao M, Phalen RF, et al. Particulate air pollutants and asthma. A paradigm for the role of oxidative stress in PM-induced adverse health effects. Clin Immunol. 2003;109:250–65.
    1. Hertz-Picciotto I, Baker RJ, Yap PS, et al. Early childhood lower respiratory illness and air pollution. Environ Health Perspect. 2007;115:1510–18.
    1. Dysart MM, Galvis BR, Russell AG, Barker TH. Environmental particulate (PM2.5) augments stiffness-induced alveolar epithelial cell mechanoactivation of transforming growth factor beta. PLoS One. 2014;9:e106821.
    1. Nair GB, Matela A, Kurbanov D, Raghu G. Newer developments in idiopathic pulmonary fibrosis in the era of anti-fibrotic medications. Expert Rev Respir Med. 2016;10:699–711.
    1. Wang H, Shen X, Tian G, et al. AMPKalpha2 deficiency exacerbates long-term PM2.5 exposure-induced lung injury and cardiac dysfunction. Free Radic Biol Med. 2018;121:202–14.
    1. He M, Ichinose T, Yoshida S, et al. PM2.5-induced lung inflammation in mice: Differences of inflammatory response in macrophages and type II alveolar cells. J Appl Toxicol. 2017;37:1203–18.
    1. Dagher Z, Garcon G, Gosset P, et al. Pro-inflammatory effects of Dunkerque city air pollution particulate matter 2.5 in human epithelial lung cells (L132) in culture. J Appl Toxicol. 2005;25:166–75.
    1. Watterson TL, Sorensen J, Martin R, Coulombe RA., Jr Effects of PM2.5 collected from Cache Valley Utah on genes associated with the inflammatory response in human lung cells. J Toxicol Environ Health A. 2007;70:1731–44.
    1. Li J, Liu K, Liu Y, et al. Exosomes mediate the cell-to-cell transmission of IFN-alpha-induced antiviral activity. Nat Immunol. 2013;14:793–803.
    1. Qin J, Xu Q. Functions and application of exosomes. Acta Pol Pharm. 2014;71:537–43.
    1. Yoon YJ, Kim OY, Gho YS. Extracellular vesicles as emerging intercellular communicasomes. BMB Rep. 2014;47:531–39.
    1. Liu S, Mahairaki V, Bai H, et al. Highly purified human extracellular vesicles produced by stem cells alleviate aging cellular phenotypes of senescent human cells. Stem Cells. 2019;37:779–90.
    1. EL Andaloussi S, Mäger I, Breakefield XO, Wood MJ. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12:347–57.
    1. Thery C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 2009;9:581–93.
    1. Keller S, Sanderson MP, Stoeck A, Altevogt P. Exosomes: From biogenesis and secretion to biological function. Immunol Lett. 2006;107:102–8.
    1. Bhatnagar S, Shinagawa K, Castellino FJ, Schorey JS. Exosomes released from macrophages infected with intracellular pathogens stimulate a proinflammatory response in vitro and in vivo. Blood. 2007;110:3234–44.
    1. Yeo RW, Lai RC, Zhang B, et al. Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Adv Drug Deliv Rev. 2013;65:336–41.
    1. Chang CL, Sung PH, Chen KH, et al. Adipose-derived mesenchymal stem cell-derived exosomes alleviate overwhelming systemic inflammatory reaction and organ damage and improve outcome in rat sepsis syndrome. Am J Transl Res. 2018;10:1053–70.
    1. Fareh M, Almairac F, Turchi L, et al. Cell-based therapy using miR-302-367 expressing cells represses glioblastoma growth. Cell Death Dis. 2017;8:e2713.
    1. Klinge CM. Non-coding RNAs in breast cancer: Intracellular and intercellular communication. Noncoding RNA. 2018;4(4) pii: E40.
    1. Deng X, Zhang F, Rui W, et al. PM2.5-induced oxidative stress triggers autophagy in human lung epithelial A549 cells. Toxicol In Vitro. 2013;27:1762–70.
    1. Billet S, Garcon G, Dagher Z, et al. Ambient particulate matter (PM2.5): Physicochemical characterization and metabolic activation of the organic fraction in human lung epithelial cells (A549) Environ Res. 2007;105:212–23.
    1. Li X, Xie X, Lian W, et al. Exosomes from adipose-derived stem cells overexpressing Nrf2 accelerate cutaneous wound healing by promoting vascularization in a diabetic foot ulcer rat model. Exp Mol Med. 2018;50:29.
    1. Bao L, You B, Shi S, et al. Metastasis-associated miR-23a from nasopharyngeal carcinoma-derived exosomes mediates angiogenesis by repressing a novel target gene TSGA10. Oncogene. 2018;37:2873–89.
    1. Xu X, Qimuge A, Wang H, et al. IRE1alpha/XBP1s branch of UPR links HIF1alpha activation to mediate ANGII-dependent endothelial dysfunction under particulate matter (PM) 2.5 exposure. Sci Rep. 2017;7:13507.
    1. Willis GR, Fernandez-Gonzalez A, Anastas J, et al. Mesenchymal stromal cell exosomes ameliorate experimental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. Am J Respir Crit Care Med. 2018;197:104–16.
    1. Li Y, Li H, Liu S, et al. Pirfenidone ameliorates lipopolysaccharide-induced pulmonary inflammation and fibrosis by blocking NLRP3 inflammasome activation. Mol Immunol. 2018;99:134–44.
    1. Yang L, Lin Z, Wang Y, et al. Nickle(II) ions exacerbate bleomycin-induced pulmonary inflammation and fibrosis by activating the ROS/Akt signaling pathway. Environ Sci Pollut Res Int. 2018;25:4406–18.
    1. Cheng X, He S, Yuan J, et al. Lipoxin A4 attenuates LPS-induced mouse acute lung injury via Nrf2-mediated E-cadherin expression in airway epithelial cells. Free Radi Biol Med. 2016;93:52–66.
    1. Zheng JQ, Zhang GR, Li J, Bi HW. Neutrophil elastase inhibitor suppresses oxidative stress in obese asthmatic rats by activating Keap1/Nrf2 signaling pathway. Eur Rev Med Pharmacol Sci. 2019;23:361–69.
    1. Muthusamy BP, Budi EH, Katsuno Y, et al. ShcA protects against epithelial-mesenchymal transition through compartmentalized inhibition of TGF-beta-induced Smad activation. PLoS Biol. 2015;13:e1002325.
    1. Gurgueira SA, Lawrence J, Coull B, et al. Rapid increases in the steady-state concentration of reactive oxygen species in the lungs and heart after particulate air pollution inhalation. Environ Health Perspect. 2002;110:749–55.
    1. Ghio AJ, Carraway MS, Madden MC. Composition of air pollution particles and oxidative stress in cells, tissues, and living systems. J Toxicol Environ Health B Crit Rev. 2012;15:1–21.
    1. Zhang G, Zou X, Huang Y, et al. Mesenchymal stromal cell-derived extracellular vesicles protect against acute kidney injury through anti-oxidation by enhancing Nrf2/ARE activation in rats. Kidney Blood Press Res. 2016;41:119–28.
    1. Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: Toward cell-free therapeutic applications. Mol Ther. 2015;23:812–23.
    1. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-beta: The master regulator of fibrosis. Nat Rev Nephrol. 2016;12:325–38.
    1. Xu P, Liu J, Derynck R. Post-translational regulation of TGF-beta receptor and Smad signaling. FEBS Lett. 2012;586:1871–84.
    1. Koh W, Sheng CT, Tan B, et al. Analysis of deep sequencing microRNA expression profile from human embryonic stem cells derived mesenchymal stem cells reveals possible role of let-7 microRNA family in downstream targeting of hepatic nuclear factor 4 alpha. BMC Genomics. 2010;11(Suppl 1):S6.
    1. Jiang XC, Gao JQ. Exosomes as novel bio-carriers for gene and drug delivery. Int J Pharm. 2017;521:167–75.
    1. Yousefpour P, Chilkoti A. Co-opting biology to deliver drugs. Biotechnol Bioeng. 2014;111:1699–716.
    1. Corcione A, Benvenuto F, Ferretti E, et al. Human mesenchymal stem cells modulate B-cell functions. Blood. 2006;107:367–72.

Source: PubMed

3
Tilaa