Comparison of lncRNA and mRNA expression in mouse brains infected by a wild-type and a lab-attenuated Rabies lyssavirus Free

Abstract

Rabies is a lethal disease caused by Rabies lyssavirus, commonly known as rabies virus (RABV), and results in nearly 100 % death once clinical symptoms occur in human and animals. Long non-coding RNAs (lncRNAs) have been reported to be associated with viral infection. But the role of lncRNAs involved in RABV infection is still elusive. In this study, we performed global transcriptome analysis of both of lncRNA and mRNA expression profiles in wild-type (WT) and lab-attenuated RABV-infected mouse brains by using next-generation sequencing. The differentially expressed lncRNAs and mRNAs were analysed by using the edgeR package. We identified 1422 differentially expressed lncRNAs and 4475 differentially expressed mRNAs by comparing WT and lab-attenuated RABV-infected brains. Then we predicted the enriched biological pathways by the Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) database based on the differentially expressed lncRNAs and mRNAs. Our analysis revealed the relationships between lncRNAs and RABV-infection-associated immune response and ion transport-related pathways, which provide a fresh insight into the potential role of lncRNA in immune evasion and neuron injury induced by WT RABV.

Funding
This study was supported by the:
  • National Natural Science Foundation of China (Award 31872451)
    • Principle Award Recipient: LingZhao
  • National Program on Key Research Project of China (Award 2016YFD0500400)
    • Principle Award Recipient: LingZhao
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001538
2020-12-07
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/jgv/102/3/vir001538.html?itemId=/content/journal/jgv/10.1099/jgv.0.001538&mimeType=html&fmt=ahah

References

  1. Fooks AR, Banyard AC, Horton DL, Johnson N, McElhinney LM et al. Current status of rabies and prospects for elimination. The Lancet 2014; 384:1389–1399 [View Article]
    [Google Scholar]
  2. Sokol F, Stanček D, Koprowski H. Structural proteins of rabies virus. J Virol 1971; 7:241249 [View Article]
    [Google Scholar]
  3. Wunner WH, Larson JK, Dietzschold B, Smith CL. The molecular biology of rabies viruses. Clinical Infectious Diseases 1988; 10:S771–S784 [View Article]
    [Google Scholar]
  4. Albertini AA, Ruigrok RW, Blondel D. Rabies virus transcription and replication. Adv Virus Res 2011; 79:1
    [Google Scholar]
  5. Schnell MJ, McGettigan JP, Wirblich C, Papaneri A. The cell biology of rabies virus: using stealth to reach the brain. Nat Rev Microbiol 2010; 8:5161 [View Article]
    [Google Scholar]
  6. Wang ZW, Sarmento L, Wang Y, Li X-qing, Dhingra V et al. Attenuated rabies virus activates, while pathogenic rabies virus evades, the host innate immune responses in the central nervous system. J Virol 2005; 79:12554–12565 [View Article]
    [Google Scholar]
  7. Suja MS, Mahadevan A, Madhusudana SN, Shankar SK. Role of apoptosis in rabies viral encephalitis: a comparative study in mice, canine, and human brain with a review of literature. Patholog Res Int 2011; 2011:37428613 [View Article]
    [Google Scholar]
  8. Nagano T, Fraser P. No-Nonsense functions for long noncoding RNAs. Cell 2011; 145:178–181 [View Article]
    [Google Scholar]
  9. Prensner JR, Chinnaiyan AM. The emergence of lncRNAs in cancer biology. Cancer Discov 2011; 1:391407 [View Article]
    [Google Scholar]
  10. Ouyang J, Hu J, Chen J-L. lncRNAs regulate the innate immune response to viral infection. WIREs RNA 2016; 7:129–143 [View Article]
    [Google Scholar]
  11. Liu S, Wang Z, Chen D, Zhang B, Tian R-R et al. Annotation and cluster analysis of spatiotemporal- and sex-related lncRNA expression in rhesus macaque brain. Genome Res 2017; 27:16081620 [View Article]
    [Google Scholar]
  12. Mercer TR, Dinger ME, Sunkin SM, Mehler MF, Mattick JS. Specific expression of long noncoding RNAs in the mouse brain. Proc Natl Acad Sci U S A 2008; 105:716–721 [View Article]
    [Google Scholar]
  13. Bonasio R, Shiekhattar R. Regulation of transcription by long noncoding RNAs. Annu Rev Genet 2014; 48:433455 [View Article]
    [Google Scholar]
  14. Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet 2009; 10:155–159 [View Article]
    [Google Scholar]
  15. Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Mol Cell 2011; 43:904914 [View Article]
    [Google Scholar]
  16. Batista PJ, Chang HY. Long noncoding RNAs: cellular address codes in development and disease. Cell 2013; 152:1298–1307 [View Article]
    [Google Scholar]
  17. Briggs JA, Wolvetang EJ, Mattick JS, Rinn JL, Barry G. Mechanisms of long non-coding RNAs in mammalian nervous system development, plasticity, disease, and evolution. Neuron 2015; 88:861877 [View Article]
    [Google Scholar]
  18. Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet 2014; 15:7–21 [View Article]
    [Google Scholar]
  19. Sun W, Yang Y, Xu C, Guo J. Regulatory mechanisms of long noncoding RNAs on gene expression in cancers. Cancer Genet 2017; 216-217:105–110 [View Article]
    [Google Scholar]
  20. Wang P. The Opening of Pandora’s Box: An Emerging Role of Long Noncoding RNA in Viral Infections. Front Immunol 2018; 9:3138 [View Article]
    [Google Scholar]
  21. Atianand MK, Caffrey DR, Fitzgerald KA. Immunobiology of long noncoding RNAs. Annu Rev Immunol 2017; 35:177–198 [View Article]
    [Google Scholar]
  22. Fitzgerald KA, Caffrey DR. Long noncoding RNAs in innate and adaptive immunity. Curr Opin Immunol 2014; 26:140–146 [View Article]
    [Google Scholar]
  23. Gomez JA, Wapinski OL, Yang YW, Bureau J-F, Gopinath S et al. The nest long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-γ locus. Cell 2013; 152:743754 [View Article]
    [Google Scholar]
  24. Imamura K, Imamachi N, Akizuki G, Kumakura M, Kawaguchi A et al. Long noncoding RNA NEAT1-Dependent SFPQ relocation from promoter region to paraspeckle mediates IL8 expression upon immune stimuli. Mol Cell 2014; 53:393–406 [View Article]
    [Google Scholar]
  25. Ma H, Han P, Ye W, Chen H, Zheng X et al. The long noncoding RNA NEAT1 exerts Antihantaviral effects by acting as positive feedback for RIG-I signaling. J Virol 2017; 91:JVI.02250-02216 [View Article]
    [Google Scholar]
  26. Quan Z, Chen CY, Yedavalli V, Kuan-Teh J. Neat1 long noncoding RNA and paraspeckle bodies modulate HIV-1 posttranscriptional expression. Mbio 2013; 4:e00596
    [Google Scholar]
  27. Kambara H, Niazi F, Kostadinova L, Moonka DK, Siegel CT et al. Negative regulation of the interferon response by an interferon-induced long non-coding RNA. Nucleic Acids Res 2014; 42:10668–10680 [View Article]
    [Google Scholar]
  28. Sui B, Chen D, Liu W, Wu Q, Tian B et al. A novel antiviral lncRNA, EDAL, shields a T309 O-GlcNAcylation site to promote EZH2 lysosomal degradation. Genome Biol 2020; 21:228 [View Article]
    [Google Scholar]
  29. Yu F, Zhang G, Xiao S, Fang L, Xu G et al. Complete genome sequence of a street rabies virus isolated from a rabid dog in China. J Virol 2012; 86:10890–10891 [View Article]
    [Google Scholar]
  30. Wang ZW, Sarmento L, Wang Y, Li X-qing, Dhingra V et al. Attenuated rabies virus activates, while pathogenic rabies virus evades, the host innate immune responses in the central nervous system. J Virol 2005; 79:12554–12565 [View Article]
    [Google Scholar]
  31. Zhao L, Toriumi H, Kuang Y, Chen H, Fu ZF. The roles of chemokines in rabies virus infection: overexpression may not always be beneficial. J Virol 2009; 83:11808–11818 [View Article][PubMed]
    [Google Scholar]
  32. Trapnell C, Roberts A, Goff L, Pertea G, Kim D et al. Differential gene and transcript expression analysis of RNA-Seq experiments with TopHat and Cufflinks. Nat Protoc 2012; 7:562578 [View Article]
    [Google Scholar]
  33. Kong L, Zhang Y, Ye Z-Q, Liu X-Q, Zhao S-Q et al. Cpc: assess the protein-coding potential of transcripts using sequence features and support vector machine. Nucleic Acids Res 2007; 35:W345W349 [View Article]
    [Google Scholar]
  34. Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-Seq data. Genome Biol 2010; 11:R25–29 [View Article]
    [Google Scholar]
  35. Li J, Witten DM, Johnstone IM, Tibshirani R. Normalization, testing, and false discovery rate estimation for RNA-sequencing data. Biostatistics 2012; 13:523–538 [View Article]
    [Google Scholar]
  36. Tian B, Zhou M, Yang Y, Yu L, Luo Z et al. Lab-Attenuated rabies virus causes abortive infection and induces cytokine expression in astrocytes by activating mitochondrial Antiviral-Signaling protein signaling pathway. Frontiers in immunology 2011; 2017:8
    [Google Scholar]
  37. Charlton FW, Hover S, Fuller J, Hewson R, Fontana J et al. Cellular cholesterol abundance regulates potassium accumulation within endosomes and is an important determinant in bunyavirus entry. Journal of Biological Chemistry 2019; 294:7335–7347 [View Article]
    [Google Scholar]
  38. Hare DN, Collins SE, Mukherjee S, Loo Y-M, Gale M et al. Membrane Perturbation-Associated Ca 2+ Signaling and Incoming Genome Sensing Are Required for the Host Response to Low-Level Enveloped Virus Particle Entry. J Virol 2016; 90:3018–3027 [View Article]
    [Google Scholar]
  39. Chen X, Cao R, Zhong W. Host calcium channels and pumps in viral infections. Cells 2019; 9:94 [View Article]
    [Google Scholar]
  40. Müller M, Slivinski N, Todd EJAA, Khalid H, Li R et al. Chikungunya virus requires cellular chloride channels for efficient genome replication. PLoS Negl Trop Dis 2019; 13:e0007703 [View Article]
    [Google Scholar]
  41. Nakamichi K, Saiki M, Sawada M, Takayama-Ito M, Yamamuro Y et al. Rabies virus-induced activation of mitogen-activated protein kinase and NF-κB signaling pathways regulates expression of CXC and CC chemokine ligands in microglia. J Virol 2005; 79:11801–11812 [View Article]
    [Google Scholar]
  42. Atianand MK, Hu W, Satpathy AT, Shen Y, Ricci EP et al. A long noncoding RNA lincRNA-EPS acts as a transcriptional brake to restrain inflammation. Cell 2016; 165:1672–1685 [View Article]
    [Google Scholar]
  43. Ji S, Zhu M, Zhang J, Cai Y, Zhai X et al. Microarray analysis of lncRNA expression in rabies virus infected human neuroblastoma cells. Infection, Genetics and Evolution 2019; 67:88–100 [View Article]
    [Google Scholar]
  44. Zhao P, Liu S, Zhong Z, Jiang T, Weng R et al. Analysis of expression profiles of long noncoding RNAs and mRNAs in brains of mice infected by rabies virus by RNA sequencing. Sci Rep 2018; 8:11858 [View Article]
    [Google Scholar]
  45. Samuel CE. Antiviral actions of interferons. Clin Microbiol Rev 2001; 14:778–809 [View Article]
    [Google Scholar]
  46. Kao Y-T, Lai MMC, Yu C-Y. How dengue virus circumvents innate immunity. Front Immunol 2018; 9:2860 [View Article][PubMed]
    [Google Scholar]
  47. Lafon M. Evasive strategies in rabies virus infection. Adv Virus Res 2011; 79:33–53
    [Google Scholar]
  48. Scott T, Nel L. Subversion of the immune response by rabies virus. Viruses 2016; 8:231 [View Article]
    [Google Scholar]
  49. Ito N, Moseley GW, Sugiyama M. The importance of immune evasion in the pathogenesis of rabies virus. J Vet Med Sci 2016; 78:1089–1098 [View Article]
    [Google Scholar]
  50. Barkhouse DA, Garcia SA, Bongiorno EK, Lebrun A, Faber M et al. Expression of interferon gamma by a recombinant rabies virus strongly attenuates the pathogenicity of the virus via induction of type I interferon. J Virol 2015; 89:312–322 [View Article]
    [Google Scholar]
  51. Biermer M, Puro R, Schneider RJ. Tumor necrosis factor alpha inhibition of hepatitis B virus replication involves disruption of capsid integrity through activation of NF-κB. J Virol 2003; 77:4033–4042 [View Article]
    [Google Scholar]
  52. Mitchell JP, Carmody RJ. Nf-kappaB and the transcriptional control of inflammation. Int Rev Cell Mol Biol 2018; 335:41–84
    [Google Scholar]
  53. Deng L, Zeng Q, Wang M, Cheng A, Jia R et al. Suppression of NF-κB activity: a viral immune evasion mechanism. Viruses 2018; 10:409 [View Article]
    [Google Scholar]
  54. Bette M, Kaut O, Schäfer MK-H, Weihe E. Constitutive expression of p55TNFR mRNA and mitogen-specific up-regulation of TNFα and p75TNFR mRNA in mouse brain. Journal of Comparative Neurology 2003; 465:417–430 [View Article]
    [Google Scholar]
  55. Faber M, Bette M, Preuss MAR, Pulmanausahakul R, Rehnelt J et al. Overexpression of tumor necrosis factor alpha by a recombinant rabies virus attenuates replication in neurons and prevents lethal infection in mice. J Virol 2005; 79:15405–15416 [View Article]
    [Google Scholar]
  56. Viruses KPG. Apoptosis, and neuroinflammation-a double-edged sword. J Neurovirol 2015; 21:1–7
    [Google Scholar]
  57. Lay S, Prehaud C, Dietzschold B, Lafon M. Glycoprotein of nonpathogenic rabies viruses is a major inducer of apoptosis in human Jurkat T cells. Ann N Y Acad Sci 2003; 1010:577–581 [View Article]
    [Google Scholar]
  58. Thoulouze MI, Lafage M, Yuste VJ, Kroemer G, Susin SA et al. Apoptosis inversely correlates with rabies virus neurotropism. Ann N Y Acad Sci 2003; 1010:598–603 [View Article]
    [Google Scholar]
  59. Sarmento L, Li X-qing, Howerth E, Jackson AC, Fu ZF. Glycoprotein-Mediated induction of apoptosis limits the spread of attenuated rabies viruses in the central nervous system of mice. J Neurovirol 2005; 11:571–581 [View Article][PubMed]
    [Google Scholar]
  60. Hsu K, Seharaseyon J, Dong P, Bour S, Marbán E. Mutual functional destruction of HIV-1 Vpu and host TASK-1 channel. Mol Cell 2004; 14:259–267 [View Article]
    [Google Scholar]
  61. Stauffer S, Feng Y, Nebioglu F, Heilig R, Picotti P et al. Stepwise priming by acidic pH and a high K+ concentration is required for efficient uncoating of influenza A virus cores after penetration. J Virol 2014; 88:13029–13046 [View Article]
    [Google Scholar]
  62. Lazrak A, Iles KE, Liu G, Noah DL, Noah JW et al. Influenza virus M2 protein inhibits epithelial sodium channels by increasing reactive oxygen species. FASEB j. 2009; 23:3829–3842 [View Article]
    [Google Scholar]
  63. H-L J, Song W, Gao Z, X-F S, Nie H-G et al. SARS-CoV proteins decrease levels and activity of human ENaC via activation of distinct PKC isoforms. Am J Physiol Lung Cell Mol Physiol 2009; 296:L372–L383
    [Google Scholar]
  64. ZF F, Jackson AC. Neuronal dysfunction and death in rabies virus infection. J Neurovirol 2005; 11:101–106
    [Google Scholar]
  65. Mitrabhakdi E, Shuangshoti S, Wannakrairot P, Lewis RA, Susuki K et al. Difference in neuropathogenetic mechanisms in human furious and paralytic rabies. J Neurol Sci 2005; 238:3–10 [View Article]
    [Google Scholar]
  66. Iwata M, Komori S, Unno T, Minamoto N, Ohashi H. Modification of membrane currents in mouse neuroblastoma cells following infection with rabies virus. Br J Pharmacol 1999; 126:1691–1698 [View Article]
    [Google Scholar]
  67. Long WF, Burke DC. Interferon production by double-stranded RNA: a comparison of induction by reovirus to that by a synthetic double-stranded polynucleotide. J Gen Virol 1971; 12:1–11 [View Article]
    [Google Scholar]
  68. Vilcek J, Ng MH, Friedman-Kien AE, Krawciw T. Induction of interferon synthesis by synthetic double-stranded polynucleotides. J Virol 1968; 2:648–650 [View Article]
    [Google Scholar]
  69. Son K-N, Liang Z, Lipton HL. Double-Stranded RNA is detected by immunofluorescence analysis in RNA and DNA virus infections, including those by Negative-Stranded RNA viruses. J Virol 2015; 89:9383–9392 [View Article]
    [Google Scholar]
  70. Li J, McGettigan JP, Faber M, Schnell MJ, Dietzschold B. Infection of monocytes or immature dendritic cells (DCs) with an attenuated rabies virus results in DC maturation and a strong activation of the NFκB signaling pathway. Vaccine 2008; 26:419–426 [View Article]
    [Google Scholar]
  71. Yang Y, Huang Y, Gnanadurai CW, Cao S, Liu X et al. The inability of wild-type rabies virus to activate dendritic cells is dependent on the glycoprotein and correlates with its low level of the de novo-Synthesized Leader RNA. J Virol 2015; 89:2157–2169 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001538
Loading
/content/journal/jgv/10.1099/jgv.0.001538
Loading

Data & Media loading...

Supplements

Supplementary material 1

EXCEL

Most cited Most Cited RSS feed