Different microRNAs contribute to the protective effect of mesenchymal stem cell-derived microvesicles in LPS induced acute respiratory distress syndrome

Document Type : Original Article

Authors

1 Department of Anesthesiology, Ningbo First Hospital, No. 59 Liuting Street, Haishu District, Ningbo 315010, Zhejiang, China

2 Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong, China

Abstract

Objective(s): The present study aimed to determine whether bone marrow mesenchymal stem cell-derived microvesicles (MSC MVs) were effective in restoring lung tissue structure, and to assess the potential role of miRNAs in the pathogenesis and progression of acute respiratory distress syndrome (ARDS).
Materials and Methods: ARDS was induced by lipopolysaccharide in male C57BL/6 mice. The degree of lung injury was assessed by histological analysis, lung’s wet weight/body weight, and protein levels in the bronchoalveolar lavage fluid (BALF). Sequencing was performed on the BGISEQ-500 platform. Differentially expressed miRNAs (DEMs) were screened with the DEGseq software. The target genes of DEMs were predicted by iRNAhybrid, miRanda, and TargetScan.
Results: Compared with LPS-injured mice, MSC MVs reduced lung water and total protein levels in the BALF, demonstrating a protective effect. 52 miRNAs were differentially expressed following treatment with MSC MVs in ARDS mice. Among them, miR‑532‑5p, miR‑223‑3p, and miR‑744‑5p were significantly regulated. Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed the target genes were mainly located in the cell, organelle, and membrane. Furthermore, KEGG pathways such as ErbB, PI3K-Akt, Ras, MAPK, Toll, and Wnt signaling pathways were the most significant pathways enriched by the target genes.
Conclusion: MSC MVs treatment was involved in alleviating lung injury and promoting lung tissue repair by dysregulated miRNAs.

Keywords


1. Stratton CW, Tang YW. Pathogenesis-directed therapy of 2019 novel coronavirus disease. J Med Virol 2021;93:1320-1342.
2. Ferruelo A, Peñuelas Ó, Lorente JA. MicroRNAs as biomarkers of acute lung injury. Ann Transl Med 2018;6:34.
3. Bosma KJ, Taneja R, Lewis JF. Pharmacotherapy for prevention and treatment of acute respiratory distress syndrome: Current and experimental approaches. Drugs 2010;70:1255-1282.
4. Máca J, Jor O, Holub M, Sklienka P, Burša F, Burda M, et al. Past and present ARDS mortality rates: A systematic review Respir Care 2017;62:113-122.
5. Rubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, et al. Incidence and outcomes of acute lung injury. N Engl J Med 2005;353:1685-1693.
6. Matthay MA, Calfee CS, Zhuo H, Thompson BT, Wilson JG, Levitt JE, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): A randomised phase 2a safety trial. Lancet Respir Med 2019;7:154-162.
7. Tieu A, Lalu MM. An analysis of mesenchymal stem cell-derived extracellular vesicles for preclinical use. ACS Nano 2020;14:9728-9743.
8. Jafarinia M, Alsahebfosoul F. Mesenchymal stem cell-derived extracellular vesicles: A novel cell-free therapy. Immunol Invest 2020;49:758-780.
9. Bandiera S, Pfeffer S, Baumert TF, Zeisel MB. miR-122--a key factor and therapeutic target in liver disease. J Hepatol 2015;62:448-457.
10. Wang JY, Zhang Q, Wang DD, Yan W, Sha HH, Zhao JH, et al. MiR-29a: A potential therapeutic target and promising biomarker in tumors. Biosci Rep 2018;38: 20171265.
11. Bernardo BC, Ooi JY, Lin RC, McMullen JR. miRNA therapeutics: A new class of drugs with potential therapeutic applications in the heart. Future Med Chem 2015;7:1771-1792.
12. Angulo M, Lecuona E, Sznajder JI. Role of microRNAs in lung disease. Arch Bronconeumol 2012;48:325-330.
13. Mokra D, Kosutova P. Biomarkers in acute lung injury. Respir Physiol Neurobiol 2015;209:52-58.
14. Benz F, Roy S, Trautwein C, Roderburg C, Luedde T. Circulating microRNAs as biomarkers for sepsis. Int J Mol Sci 2016;17:78.
15. Hu S, Park J, Liu A, Lee J, Zhang X, Hao Q, et al. Mesenchymal stem cell microvesicles restore protein permeability across primary cultures of injured human lung microvascular endothelial cells. Stem Cells Transl Med 2018;7:615-624.
16. Tang XD, Shi L, Monsel A, Li XY, Zhu HL, Zhu YG, et al. Mesenchymal stem cell microvesicles attenuate acute lung injury in mice partly mediated by Ang-1 mRNA. Stem Cells  2017;35:1849-1859.
17. Wei W, Ma B, Li HY, Jia Y, Lv K, Wang G, et al. Biphasic effects of selective inhibition of transforming growth factor beta1 activin receptor-like kinase on LPS-induced lung injury. Shock 2010;33:218-224.
18. Krüger J, Rehmsmeier M. RNAhybrid: microRNA target prediction easy, fast and flexible. Nucleic Acids Res 2006;34:451-454.
19. John B, Enright AJ, Aravin A, Tuschl T, Sander C, Marks DS. Human MicroRNA targets. PLoS Biol 2004;2:363.
20. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. Elife 2015;4:e05005.
21. Gebert LFR, MacRae IJ. Regulation of microRNA function in animals. Nat Rev Mol Cell Biol 2019;20:21-37.
22. Yang Y, Li L. Depleting microRNA-146a-3p attenuates lipopolysaccharide-induced acute lung injury via up-regulating SIRT1 and mediating NF-κB pathway. J Drug Target 2021;29:420-429.
23. Jiang K, Yang J, Guo S, Zhao G, Wu H, Deng G. Peripheral circulating exosome-mediated delivery of mir-155 as a novel mechanism for acute lung inflammation. Mol Ther 2019;27:1758-1771.
24. Katsha AM, Ohkouchi S, Xin H, Kanehira M, Sun R, Nukiwa T, et al. Paracrine factors of multipotent stromal cells ameliorate lung injury in an elastase-induced emphysema model. Mol Ther 2011;19:196-203.
25. Toma C, Wagner WR, Bowry S, Schwartz A, Villanueva F. Fate of culture-expanded mesenchymal stem cells in the microvasculature: In vivo observations of cell kinetics. Circ Res 2009;104:398-402.
26. Park J, Kim S, Lim H, Liu A, Hu S, Lee J, et al. Therapeutic effects of human mesenchymal stem cell microvesicles in an ex vivo perfused human lung injured with severe Escherichia coli pneumonia. Thorax 2019;74:43-50.
27. Qiu G, Zheng G, Ge M, Wang J, Huang R, Shu Q, et al. Functional proteins of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther 2019;10:359.
28. Phinney DG, Di Giuseppe M, Njah J, Sala E, Shiva S, St Croix CM, et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun 2015;6:8472.
29. Shah T, Qin S, Vashi M, Predescu DN, Jeganathan N, Bardita C, et al. Alk5/Runx1 signaling mediated by extracellular vesicles promotes vascular repair in acute respiratory distress syndrome. Clin Transl Med 2018;7:19.
30. Potter DR, Miyazawa BY, Gibb SL, Deng X, Togaratti PP, Croze RH, et al. Mesenchymal stem cell-derived extracellular vesicles attenuate pulmonary vascular permeability and lung injury induced by hemorrhagic shock and trauma. J Trauma Acute Care Surg 2018;84:245-256.
31. Deny M, Romano M, Denis O, Casimir G, Chamekh M. Progressive control of streptococcus agalactiae-induced innate inflammatory response is associated with time course expression of MicroRNA-223 by neutrophils. Infect Immun 2020;88:563-520.
32. Wang H, Zhang P, Chen W, Feng D, Jia Y, Xie L. Serum microRNA signatures identified by Solexa sequencing predict sepsis patients’ mortality: A prospective observational study. PloS one 2012;7:e38885.
33. Zhao Y, Gan Y, Xu G, Hua K, Liu D. Exosomes from MSCs overexpressing microRNA-223-3p attenuate cerebral ischemia through inhibiting microglial M1 polarization mediated inflammation. Life Sci 2020;260:118403.
34. Yan X, Zeng D, Zhu H, Zhang Y, Shi Y, Wu Y, et al. MiRNA-532-5p regulates CUMS-Induced depression-like behaviors and modulates LPS-Induced proinflammatory cytokine signaling by targeting STAT3. Neuropsychiatr Dis Treat 2020;16:2753-2764.
35. Li J, Panganiban R, Kho AT, McGeachie MJ. Circulating MicroRNAs and treatment response in childhood asthma. Am J Respir Crit Care Med 2020;202:65-72.
36. Qin X, Jiang X, Jiang X, Wang Y, Miao Z, He W, et al. Micheliolide inhibits LPS-induced inflammatory response and protects mice from LPS challenge. Sci Rep 2016;6:23240.
37. Wang X, Quinn PJ, Yan A. Kdo2 -lipid A: Structural diversity and impact on immunopharmacology. Biol Rev Camb Philos Soc 2015;90:408-427.
38. Bode JG, Ehlting C, Häussinger D. The macrophage response towards LPS and its control through the p38(MAPK)-STAT3 axis. Cell Signal 2012;24:1185-1194.
39. Wang D, Lou J, Ouyang C, Chen W, Liu Y, Liu X, et al. Ras-related protein Rab10 facilitates TLR4 signaling by promoting replenishment of TLR4 onto the plasma membrane. Proceedings of the National Academy of Sciences of the United States of America. Proc Natl Acad Sci U S A 2010;107:13806-13811.
40. Villar J, Zhang H, Slutsky AS. Lung repair and regeneration in ARDS: Role of PECAM1 and Wnt signaling. Chest 2019;155:587-594.