Long-chain non-coding RNA n337374 relieves symptoms of respiratory syncytial virus-induced asthma by inhibiting dendritic cell maturation via the CD86 and the ERK pathway

Main Article Content

Shuning Sun
Ming Yao
Lifen Yuan
Jianou Qiao

Keywords

asthma, dendritic cells maturation, lentiviruses, long-chain non-coding RNA, respiratory syncytial virus

Abstract

Background: In this study, we investigated the relationship between long-chain non-coding RNAs (lncRNAs) and respiratory syncytial virus (RSV)-exacerbated asthma.


Methods: Transcriptome microarray was used to detect differentially expressed lncRNAs in dendritic cells (DCs) co-cultured with RSV-infected human airway epithelial cells and DCs infected with RSV. The identified downregulation of lncRNA n337374 was validated using fluorescence RT-qPCR. LncRNA n337374-overexpressing DCs and RSV-exacerbated asthmatic mouse models were established. Airway hyper-reactivity and bronchoalveolar lavage fluid (BALF) were examined, and pathological changes in lung tissues were observed in mice. Surface molecules in DCs were detected by flow cytometry and RT-qPCR and the expression of CD86 and mitogen-activated protein kinases was determined by western blot.


Results: In an RSV-exacerbated asthmatic mouse model, the airway wall was thickened, luminal stenosis was observed, a large number of inflammatory cells were infiltrated in the lung tissue, lung function was impaired, and counts of inflammatory cells in the BALF were increased. The overexpression of lncRNA n337374 ameliorated these pathological changes and improved impaired lung function and inflammation in an asthmatic mouse model. In DCs co-cultured with RSV-infected human airway epithelial cells, CD86 expression was promoted and ERK was markedly phosphorylated. When lncRNA n337374-overexpressing DCs were used in the co-cultures, the expression of CD86 and phosphorylated ERK was decreased.


Conclusion: The results suggest that lncRNA n337374 overexpression may suppress DC maturation by downregulating the CD86 and ERK pathway, subsequently relieving the symptoms of RSV-induced asthma. LncRNA n337374 may be a promising target in the treatment of RSV infection-induced asthma.

Abstract 888 | PDF Downloads 727 HTML Downloads 304 XML Downloads 9

References

1. Becker AB, Abrams EM. Asthma guidelines: the global initiative for asthma in relation to national guidelines. Curr Opin Allergy Clin Immunol. 2017;17:99–103. 10.1097/ACI.0000000000000346

2. Sykes A, Johnston SL. Etiology of asthma exacerbations. J Allergy Clin Immunol. 2008;122:685–8. 10.1016/j.jaci.2008.08.017

3. Jackson DJ, Johnston SL. The role of viruses in acute exacerbations of asthma. J Allergy Clin Immunol. 2010;125:1178–87; quiz 88–9. 10.1016/j.jaci.2010.04.021

4. Bacharier LB, Cohen R, Schweiger T, Yin-DeClue H, Christie C, Zheng J, et al. Determinants of asthma after severe respiratory syncytial virus bronchiolitis. J Allergy Clin Immunol. 2012;130:91–100.e3. 10.1016/j.jaci.2012.02.010

5. Knudson CJ, Varga SM. The relationship between respiratory syncytial virus and asthma. Vet Pathol. 2015;52:97–106. 10.1177/0300985814520639

6. Jalink MB, Langley JM, Dodds L, Andreou P. Severe respiratory syncytial virus infection in preterm infants and later onset of asthma. Pediatr Infect Dis J. 2019;38:1121–5. 10.1097/INF.0000000000002432

7. Hu X, Li X, Hu C, Qin L, He R, Luo L, et al. Respiratory syncytial virus exacerbates OVA-mediated asthma in mice through C5a-C5aR regulating CD4(+)T cells immune responses. Sci Rep. 2017;7:15207. 10.1038/s41598-017-15471-w

8. Bacharier LB, Cohen R, Schweiger T, Yin-DeClue H, Christie C, Zheng J, et al. Determinants of asthma after severe respiratory syncytial virus bronchiolitis. J Allergy Clin Immunol. 2012;130:91–100.e3. 10.1016/j.jaci.2012.02.010

9. Qiu Y-Y, Wu Y, Lin M-J, Bian T, Xiao Y-L, Qin C. LncRNA-MEG3 functions as a competing endogenous RNA to regulate Treg/Th17 balance in patients with asthma by targeting microRNA-17/RORγt. Biomed Pharmacother. 2019;111:386–94. 10.1016/j.biopha.2018.12.080

10. Zhang X-Y, Tang X-Y, Li N, Zhao L-M, Guo Y-L, Li X-S, et al. GAS5 promotes airway smooth muscle cell proliferation in asthma via controlling miR-10a/BDNF signaling pathway. Life Sci. 2018;212:93–101. 10.1016/j.lfs.2018.09.002

11. Awasthi S, Singh B, Welliver RC, Dietert RR. Lung dendritic cell developmental programming, environmental stimuli, and asthma in early periods of life. J Allergy. 2012;2012:176468. 10.1155/2012/176468

12. Chen X, Luo Y, Wang M, Sun L, Huang K, Li Y, et al. Wuhu decoction regulates dendritic cell autophagy in the treatment of respiratory syncytial virus (RSV)-induced mouse asthma by AMPK/ULK1 signaling pathway. Med Sci Monitor Int Med J Exp Clin Res. 2019;25:5389–400. 10.12659/MSM.917692

13. de Graaff PMA, de Jong EC, van Capel TM, van Dijk MEA, Roholl PJM, Boes J, et al. Respiratory syncytial virus infection of monocyte-derived dendritic cells decreases their capacity to activate CD4 T cells. J Immunol. 2005;175:5904–11. 10.4049/jimmunol.175.9.5904

14. Qiao J, Li A, Jin X. TSLP from RSV-stimulated rat airway epithelial cells activates myeloid dendritic cells. Immunol Cell Biol. 2011;89:231–8. 10.1038/icb.2010.85

15. Jin W, Li C, Du T, Hu K, Huang X, Hu Q. DC-SIGN plays a stronger role than DCIR in mediating HIV-1 capture and transfer. Virology. 2014;458–459:83–92. 10.1016/j.virol.2014.04.016

16. Wright GW, Simon RM. A random variance model for detection of differential gene expression in small microarray experiments. Bioinformatics. 2003;19:2448–55. 10.1093/bioinformatics/btg345

17. Blake JA, Chan J, Kishore R, Sternberg PW, Li Y. Gene ontology consortium: going forward. Nucl Acids Res. 2015;43:1049–56. 10.1093/nar/gku1179

18. Du J, Yuan Z, Ma Z, Song J, Xie X, Chen Y. KEGG-PATH: Kyoto encyclopedia of genes and genomes-based pathway analysis using a path analysis model. Mol Biosyst. 2014;10:2441–7. 10.1039/C4MB00287C

19. Haw R, Hermjakob H, D’Eustachio P, Stein L. Reactome pathway analysis to enrich biological discovery in proteomics data sets. Proteomics. 2011;11:3598–613. 10.1002/pmic.201100066

20. Zhou J, Bai W, Liu Q, Cui J, Zhang W. Silencing of ADAM33 restrains proliferation and induces apoptosis of airway smooth muscle cells in ovalbumin-induced asthma model. J Cell Biochem. 2018; 120:1434-1443. 10.1002/jcb.27263

21. Khorasanizadeh M, Eskian M, Gelfand EW, Rezaei N. Mitogen-activated protein kinases as therapeutic targets for asthma. Pharmacol Ther. 2017;174:112–26. 10.1016/j.pharmthera.2017.02.024

22. Nunes C, Pereira AM, Morais-Almeida M. Asthma costs and social impact. Asthma Res Pract. 2017;3:1. 10.1186/s40733-016-0029-3

23. Furuta T, Hasegawa S, Mizutani M, Iwai T, Ohbuchi N, Kawano S, et al. Burden of human metapneumovirus and respiratory syncytial virus infections in asthmatic children. Pediatr Infect Dis J. 2018;37:1107–11. 10.1097/INF.0000000000002038

24. Woodman L, Sutcliffe A, Kaur D, Berry M, Bradding P, Pavord ID, et al. Chemokine concentrations and mast cell chemotactic activity in BAL fluid in patients with eosinophilic bronchitis and asthma, and in normal control subjects. Chest. 2006;130:371–8. 10.1378/chest.130.2.371

25. Peng H, Guerau-de-Arellano M, Mehta VB, Yang Y, Huss DJ, Papenfuss TL, et al. Dimethyl fumarate inhibits dendritic cell maturation via nuclear factor κB (NF-κB) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) and mitogen stress-activated kinase 1 (MSK1) signaling. J Biol Chem. 2012;287:28017–26. 10.1074/jbc.M112.383380

26. O’Sullivan BJ, Thomas R. CD40 ligation conditions dendritic cell antigen-presenting function through sustained activation of NF-kappaB. J Immunol. 2002;168:5491–8. 10.4049/jimmunol.168.11.5491

27. Ma DY, Clark EA. The role of CD40 and CD154/CD40L in dendritic cells. Semin Immunol. 2009;21:265–72. 10.1016/j.smim.2009.05.010

28. Salcedo SP, Marchesini MI, Lelouard H, Fugier E, Jolly G, Balor S, et al. Brucella control of dendritic cell maturation is dependent on the TIR-containing protein Btp1. PLoS Pathog. 2008;4:e21. 10.1371/journal.ppat.0040021

29. Ivanova E, Kyurkchiev D, Altankova I, Dimitrov J, Binakova E, Kyurkchiev S. CD83 monocyte-derived dendritic cells are present in human decidua and progesterone induces their differentiation in vitro. Am J Reprod Immunol. 2005;53:199–205. 10.1111/j.1600-0897.2005.00266.x

30. Suzuki M, Yokota M, Matsumoto T, Ozaki S. Synergic effects of CD40 and CD86 silencing in dendritic cells on the control of allergic diseases. Int Arch Allergy Immunol. 2018;177:87–96. 10.1159/000489862

31. Sun Y, Liu W-Z, Liu T, Feng X, Yang N, Zhou H-F. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduct Res. 2015;35:600–4. 10.3109/10799893.2015.1030412

32. Southworth T, Mason S, Bell A, Ramis I, Calbet M, Domenech A, et al. PI3K, p38 and JAK/STAT signalling in bronchial tissue from patients with asthma following allergen challenge. Biomark Res. 2018;6:14. 10.1186/s40364-018-0128-9

33. Yu F, Sun Y, Yu J, Ding Z, Wang J, Zhang L, et al. ORMDL3 is associated with airway remodeling in asthma via the ERK/MMP-9 pathway. Mol Med Rep. 2017;15:2969–76. 10.3892/mmr.2017.6413

34. Wu H-M, Shen Q-Y, Fang L, Zhang S-H, Shen P-T, Liu Y-J, et al. JNK-TLR9 signal pathway mediates allergic airway inflammation through suppressing melatonin biosynthesis. J Pineal Res. 2016;60:415–23. 10.1111/jpi.12323

35. Jiang L, Huang D, Nie S, Xie M. Polysaccharide isolated from seeds of Plantago asiatica L. induces maturation of dendritic cells through MAPK and NF-κB pathway. Saudi J Biol Sci. 2018;25:1202–7. 10.1016/j.sjbs.2017.09.011

36. Qin T, Ren Z, Huang Y, Song Y, Lin D, Li J, et al. Selenizing Hericium erinaceus polysaccharides induces dendritic cells maturation through MAPK and NF-κB signaling pathways. Int J Biol Macromol. 2017;97:287–98. 10.1016/j.ijbiomac.2017.01.039