Analysis of lncRNA Expression in Patients With Eosinophilic and Neutrophilic Asthma Focusing on LNC_000127

Yujin Zhu, Dan Mao, Wei Gao, Guojing Han, Hong Hu, Yujin Zhu, Dan Mao, Wei Gao, Guojing Han, Hong Hu

Abstract

Long non-coding RNA (lncRNA) is important in many diseases. Some studies have shown that lncRNA affects the pathogenesis of systemic inflammation of asthma. lncRNA regulates gene transcription, protein expression, and epigenetic regulation. However, lncRNAs associated with different airway phenotypes, such as eosinophilic (Eos) and neutrophilic (Neu) asthma have not been identified. The goal of this study was to determine the differences in circulating lncRNA signatures in Eos and Neu samples. Using RNA-sequencing (RNA-seq), lncRNA expression was evaluated in peripheral whole blood samples among Eos patients, Neu patients, and healthy individuals (Control). Bioinformatic analysis was used to predict relevant biological pathways. Quantitative PCR (qPCR) was used to measure gene expression in whole blood samples, Jurkat cells, and human CD4+ T cells. Finally, a novel lncRNA, LNC_000127, was inhibited by transfection of Jurkat cells with a lentiviral vector, and the effect was examined by Human Asthma RT2 Profiler™ PCR Array and western blotting. Compared to control samples, Eos samples contained 190 unique lncRNAs and Neu samples had 166 unique lncRNAs (difference ≥2-fold). KEGG pathway annotation data and GO terms revealed that different lncRNAs are involved in different mechanisms. LNC_000127, was highly expressed in Eos samples before treatment; its expression was increased in Jurkat cells and human CD4+ T cells following stimulation with PMA/CD28. Subsequent analyses revealed that LNC_000127 functions in the Th2 inflammation pathway. The results suggest that lncRNAs are involved in different phenotypes of asthma. Whether the different phenotypes of asthma can be recognized based on these lncRNAs (as biomarkers) requires further analysis. Targeting LNC_000127 may be effective for reducing Th2 inflammation in Eos asthma.

Keywords: RNA-sequencing; eosinophilic asthma; long non-coding RNA; neutrophilic asthma; peripheral whole blood.

Figures

Figure 1
Figure 1
(A) Volcano plot assessment of lncRNA expression between asthma and control groups. Red spots indicate a >2.0-fold change in lncRNA expression between asthma and control groups. Green spots indicate a <0.5-fold change between asthma and control groups. (B) Heat map analysis of differentially expressed lncRNAs between asthma and control group. Blue indicates low lncRNA expression and red indicates high lncRNA expression. (C) Volcano plot assessment of mRNA expression between asthma and control groups. Red spots indicate a >2.0-fold change in mRNA expression between asthma and control groups. Green spots indicate a <0.5-fold change between asthma and control groups. (D) Heat map analysis of differentially expressed mRNAs between asthma and control group. Blue indicates low lncRNA expression and red indicates high lncRNA expression. (E) Venn diagram showing differential expression of mRNAs between asthma and control groups. (F) Venn diagram showing differential expression of lncRNA between asthma and control groups. (G) Gene Ontology (GO) analysis of differentially expressed lncRNAs between asthma and control groups. The most enriched GO terms targeted by dysregulated transcripts were involved in a variety of functions, such as immune response, immune system process, cellular response to stress, and response to stress. (H) Gene Ontology (GO) analysis of differentially expressed lncRNAs between Eos and Neu groups. The enriched GO term targeted by dysregulated transcripts was involved in multiple metabolic processes.
Figure 2
Figure 2
Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of T cell receptor signaling pathway in asthma.
Figure 3
Figure 3
(A) Three selected lncRNA expression levels were validated in vivo. The expression of the selected three lncRNAs was validated by qPCR in three groups in vivo. LNC_000127 was upregulated in Eos samples (p < 0.05). (B) Before and after treatment, the expression of LNC_000127 was validated by qPCR in Eos samples in vivo. LNC_000127 was downregulated after treatment in Eos samples (p < 0.05). (C) lncRNA expression in response to PMA/CD28 stimulation. Jurkat cells were treated without (control) or with 10 ng/mL PMA, and 1 μg/mL CD28 for 8 and 24 h. Levels of selected lncRNAs and GATA3 were determined by qPCR, using the housekeeping gene GAPDH as a reference. Data are shown as the mean ± SD (n = 3) and are representative of one of three independent experiments. p < 0.05 compared to the control group, as analyzed by one-way ANOVA and post-hoc Bonferroni test. (D) Human CD4+ T cells were divided into three groups. One group was treated with 10 ng/mL PMA, and 1 μg/mL CD28 for 8 and 24 h. One group was treated with 10 ng/mL PMA, and 1 μg/mL CD3 for 8 and 24 h. The last group was used as a control. Gene expression results were validated by qPCR. Levels of selected genes (GATA3, LNC_000127, and INF-r) were determined by real-time PCR using the housekeeping gene GAPDH as a reference. Data are shown as the mean ± SD (n = 3) and are representative of one of three independent experiments. A p-value <0.05 compared to the control group, as analyzed by one-way ANOVA and post-hoc Bonferroni test. (E) Expression of LNC_000127 was validated by qPCR. Data are shown as the mean ± SD (n = 3) and are representative of one of three independent experiments. p-Value <0.05 compared to the control group, as analyzed by one-way ANOVA and post-hoc Bonferroni test. (F) Expression of the genes was validated by western blotting. Gene expression in response to PMA/CD28 stimulation. Jurkat cells were treated without (control) or with 10 ng/mL PMA, and 1 μg/mL CD28 for 8 h. β-Actin was used as a reference. The relative abundance of genes was calculated as the ratio of the normalized densitometric values between three samples. Intergroup differences (of the densitometry data) were calculated by the Mann-Whitney U test using SPSS version 11.6 software. p-Value <0.05 was considered to indicate a statistically significant difference.

References

    1. Anderson D. M., Anderson K. M., Chang C. L., Makarewich C. A., Nelson B. R., McAnally J. R., et al. . (2015). A micropeptide encoded by a putative long noncoding RNA regulates muscle performance. Cell 160, 595–606. 10.1016/j.cell.2015.01.009
    1. Austin P. J., Tsitsiou E., Boardman C., Jones S. W., Lindsay M. A., Adcock I. M., et al. . (2017). Transcriptional profiling identifies the long noncoding RNA plasmacytoma variant translocation (PVT1) as a novel regulator of the asthmatic phenotype in human airway smooth muscle. J. Allergy Clin. Immunol. 139, 780–789. 10.1016/j.jaci.2016.06.014
    1. Baines K. J., Simpson J. L., Wood L. G., Scott R. J., Gibson P. G. (2011). Systemic upregulation of neutrophil alpha-defensins and serine proteases in neutrophilic asthma. Thorax 66, 942–947. 10.1136/thx.2010.157719
    1. Beckedorff F. C., Ayupe A. C., Crocci-Souza R., Amaral M. S., Nakaya H. I., Soltys D. T., et al. . (2013). The intronic long noncoding RNA ANRASSF1 recruits PRC2 to the RASSF1A promoter, reducing the expression of RASSF1A and increasing cell proliferation. PLoS Genet. 9:e1003705. 10.1371/journal.pgen.1003705
    1. Chang H. S., Lee T. H., Jun J. A., Baek A. R., Park J. S., Koo S. M., et al. . (2017). Neutrophilic inflammation in asthma: mechanisms and therapeutic considerations. Exp. Rev. Respir. Med. 11, 29–40. 10.1080/17476348.2017.1268919
    1. Chen X., Li X. M., Gu W., Wang D., Chen Y., Guo X. J. (2017). LAT alleviates Th2/Treg imbalance in an OVA-induced allergic asthma mouse model through LAT-PLC-gamma1 interaction. Int. Immunopharmacol. 44, 9–15. 10.1016/j.intimp.2016.12.029
    1. Chung K. F., Wenzel S. E., Brozek J. L., Bush A., Castro M., Sterk P. J., et al. . (2014). International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur. Respir. J. 43, 343–373. 10.1183/09031936.00202013
    1. Du Z., Sun T., Hacisuleyman E., Fei T., Wang X., Brown M., et al. . (2016). Integrative analyses reveal a long noncoding RNA-mediated sponge regulatory network in prostate cancer. Nat. Commun. 7:10982. 10.1038/ncomms10982
    1. Han P., Li W., Lin C. H., Yang J., Shang C., Nuernberg S. T., et al. . (2014). A long noncoding RNA protects the heart from pathological hypertrophy. Nature 514, 102–106. 10.1038/nature13596
    1. Hu G., Tang Q., Sharma S., Yu F., Escobar T. M., Muljo S. A., et al. . (2013). Expression and regulation of intergenic long noncoding RNAs during T cell development and differentiation. Nat. Immunol. 14, 1190–1198. 10.1038/ni.2712
    1. Hu S., Wang X., Shan G. (2016). Insertion of an Alu element in a lncRNA leads to primate-specific modulation of alternative splicing. Nat. Struct. Mol. Biol. 23, 1011–1019. 10.1038/nsmb.3302
    1. Kanduri K., Tripathi S., Larjo A., Mannerström H., Ullah U., Lund R., et al. . (2015). Identification of global regulators of T-helper cell lineage specification. Genome Med. 7:122. 10.1186/s13073-015-0237-0
    1. Keenan C. R., Schuliga M. J., Stewart A. G. (2015). Pro-inflammatory mediators increase levels of the noncoding RNA GAS5 in airway smooth muscle and epithelial cells. Can. J. Physiol. Pharmacol. 93, 203–206. 10.1139/cjpp-2014-0391
    1. Kino T., Hurt D. E., Ichijo T., Nader N., Chrousos G. (2010). Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci. Signal 3:ra8. 10.1126/scisignal.2000568
    1. Koh B. H., Hwang S. S., Kim J. Y., Lee W., Kang M. J., Lee C. G., et al. . (2010). Th2 LCR is essential for regulation of Th2 cytokine genes and for pathogenesis of allergic asthma. Proc. Natl. Acad. Sci. U.S.A. 107, 10614–10619. 10.1073/pnas.1005383107
    1. Kotake Y., Nakagawa T., Kitagawa K., Suzuki S., Liu N., Kitagawa M., et al. . (2011). Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene 30, 1956–1962. 10.1038/onc.2010.568
    1. Li T., Ke Y., Cheng H. (2016). Reasearch progress on the role of neutrophils in asthma. Zhejiang Da Xue Xue Bao Yi Xue Ban 45, 544–549. 10.3785/j.issn.1008-9292.2016.09.15
    1. Matsumoto A., Pasut A., Matsumoto M., Yamashita R., Fung J., Monteleone E., et al. . (2017). mTORC1 and muscle regeneration are regulated by the LINC00961-encoded SPAR polypeptide. Nature 541, 228–232. 10.1038/nature21034
    1. Mercer T. R., Mattick J. S. (2013). Structure and function of long noncoding RNAs in epigenetic regulation. Nat. Struct. Mol. Biol. 20, 300–307. 10.1038/nsmb.2480
    1. Muehling L. M., Lawrence M. G., Woodfolk J. A. (2017). Pathogenic CD4(+) T cells in patients with asthma. J. Allergy Clin. Immunol. 140, 1523–1540. 10.1016/j.jaci.2017.02.025
    1. Nelson B. R., Makarewich C. A., Anderson D. M., Winders B. R., Troupes C. D., Wu F., et al. . (2016). A peptide encoded by a transcript annotated as long noncoding RNA enhances SERCA activity in muscle. Science 351, 271–275. 10.1126/science.aad4076
    1. Perry M. M., Tsitsiou E., Austin P. J., Lindsay M. A., Gibeon D. S., Adcock I. M., et al. . (2014). Role of non-coding RNAs in maintaining primary airway smooth muscle cells. Respir. Res. 15:58. 10.1186/1465-9921-15-58
    1. Persson H., Kwon A. T., Ramilowski J. A., Silberberg G., Söderhäll C., Orsmark-Pietras C., et al. . (2015). Transcriptome analysis of controlled and therapy-resistant childhood asthma reveals distinct gene expression profiles. J. Allergy Clin. Immunol. 136, 638–648. 10.1016/j.jaci.2015.02.026
    1. Qiao Y. Q., Huang M. L., Xu A. T., Zhao D., Ran Z. H., Shen J. (2013). LncRNA DQ786243 affects Treg related CREB and Foxp3 expression in Crohn's disease. J. Biomed. Sci. 20:87. 10.1186/1423-0127-20-87
    1. Quinn J. J., Chang H. Y. (2016). Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 17, 47–62. 10.1038/nrg.2015.10
    1. Reddy M. A., Chen Z., Park J. T., Wang M., Lanting L., Zhang Q., et al. . (2014). Regulation of inflammatory phenotype in macrophages by a diabetes-induced long noncoding RNA. Diabetes 63, 4249–4261. 10.2337/db14-0298
    1. Shan K., Liu C., Liu B. H., Chen X., Dong R., Liu X., et al. . (2017). Circular noncoding RNA HIPK3 mediates retinal vascular dysfunction in diabetes mellitus. Circulation 136, 1629–1642. 10.1161/CIRCULATIONAHA.117.029004
    1. Shinoda K., Hirahara K., Nakayama T. (2017). Maintenance of pathogenic Th2 cells in allergic disorders. Allergol. Int. 66, 369–376. 10.1016/j.alit.2017.03.005
    1. Simpson J. L., Scott R., Boyle M. J., Gibson P. G. (2006). Inflammatory subtypes in asthma: assessment and identification using induced sputum. Respirology 11, 54–61. 10.1111/j.1440-1843.2006.00784.x
    1. Smeets R. L., Fleuren W. W., He X., Vink P. M., Wijnands F., Gorecka M., et al. . (2012). Molecular pathway profiling of T lymphocyte signal transduction pathways; Th1 and Th2 genomic fingerprints are defined by TCR and CD28-mediated signaling. BMC Immunol. 13:12. 10.1186/1471-2172-13-12
    1. Song X., Wang X., Arai S., Kurokawa R. (2012). Promoter-associated noncoding RNA from the CCND1 promoter. Methods Mol. Biol. 809, 609–622. 10.1007/978-1-61779-376-9_39
    1. Spurlock C. F., Tossberg J. T., Guo Y., Collier S. P., Crooke P. S., Aune T. M. (2015). Expression and functions of long noncoding RNAs during human T helper cell differentiation. Nat. Commun. 6:6932. 10.1038/ncomms7932
    1. Tsitsiou E., Williams A. E., Moschos S. A., Patel K., Rossios C., Jiang X., et al. . (2012). Transcriptome analysis shows activation of circulating CD8+ T cells in patients with severe asthma. J. Allergy Clin. Immunol. 129, 95–103. 10.1016/j.jaci.2011.08.011
    1. Villegas V. E., Zaphiropoulos P. G. (2015). Neighboring gene regulation by antisense long non-coding RNAs. Int. J. Mol. Sci. 16, 3251–3266. 10.3390/ijms16023251
    1. Volders P. J., Verheggen K., Menschaert G., Vandepoele K., Martens L., Vandesompele J., et al. (2015). An update on LNCipedia: a database for annotated human lncRNA sequences. Nucl. Acids Res. 43, 4363–4364. 10.1093/nar/gkv295
    1. Wang P., Xue Y., Han Y., Lin L., Wu C., Xu S., et al. . (2014). The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 344, 310–313. 10.1126/science.1251456
    1. Wang Y., Qian C. Y., Li X. P., Zhang Y., He H., Wang J., et al. . (2015). Genome-scale long noncoding RNA expression pattern in squamous cell lung cancer. Sci. Rep. 5:11671. 10.1038/srep11671
    1. Warburton A. J., Boone D. N. (2017). Insights from global analyses of long noncoding RNAs in breast cancer. Curr. Pathobiol. Rep. 5, 23–34. 10.1007/s40139-017-0122-1
    1. Wynn T. A. (2015). Type 2 cytokines: mechanisms and therapeutic strategies. Nat. Rev. Immunol. 15, 271–282. 10.1038/nri3831
    1. Xia F., Dong F., Yang Y., Huang A., Chen S., Sun D., et al. . (2014). Dynamic transcription of long non-coding RNA genes during CD4+ T cell development and activation. PLoS ONE 9:e101588. 10.1371/journal.pone.0101588
    1. Xie C., Yuan J., Li H., Li M., Zhao G., Bu D., et al. . (2014). NONCODEv4: exploring the world of long non-coding RNA genes. Nucl. Acids Res. 42, D98–103. 10.1093/nar/gkt1222
    1. Yu X., Zhe Z., Tang B., Li S., Tang L., Wu Y., et al. . (2017). alpha-Asarone suppresses the proliferation and migration of ASMCs through targeting the lncRNA-PVT1/miR-203a/E2F3 signal pathway in RSV-infected rats. Acta Biochim. Biophys. Sin. 49, 598–608. 10.1093/abbs/gmx048
    1. Zhang X., Lian Z., Padden C., Gerstein M. B., Rozowsky J., Snyder M., et al. . (2009). A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster. Blood 113, 2526–2534. 10.1182/blood-2008-06-162164
    1. Zhang X. Y., Zhang L. X., Tian C. J., Tang X. Y., Zhao L. M., Guo Y. L., et al. . (2016). LncRNAs BCYRN1 promoted the proliferation and migration of rat airway smooth muscle cells in asthma via upregulating the expression of transient receptor potential 1. Am. J. Transl. Res. 8, 3409–3418.

Source: PubMed

3
Abonnere