Understanding the role of microRNAs in the interaction of mosquitoes with an insect-specific flavivirus Free

Abstract

The genus contains some of the most prevalent vector-borne viruses, such as the dengue, Zika and yellow fever viruses that cause devastating diseases in humans. However, the insect-specific clade of flaviviruses is restricted to mosquito hosts, albeit they have retained the general features of the genus, such as genome structure and replication. The interactions between insect-specific flaviviruses (ISFs) and their mosquito hosts are largely unknown. Pathogenic flaviviruses are known to modulate host-derived microRNAs (miRNAs), a class of non-coding RNAs that are important in controlling gene expression. Alterations in miRNAs may represent changes in host gene expression and promote understanding of virus–host interactions. The role of miRNAs in ISF–mosquito interactions is largely unknown. A recently discovered Australian ISF, Palm Creek virus (PCV), has the ability to suppress medically relevant flaviviruses. Here, we investigated the potential involvement of miRNAs in PCV infection using the model mosquito By combining small-RNA sequencing and bioinformatics analysis, differentially expressed miRNAs were determined. Our results indicated that PCV infection hardly affects host miRNAs. Out of 101 reported miRNAs of , only aae-miR-2940-5p had a significantly altered expression over the course of infection. However, further analysis of aae-miR-2940-5p revealed that this miRNA does not have any direct impact on PCV replication . Thus, overall the results suggest that PCV infection has a limited effect on the mosquito miRNA profile and therefore miRNAs may not play a significant role in the PCV– interaction.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.000832
2017-07-01
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jgv/98/7/1892.html?itemId=/content/journal/jgv/10.1099/jgv.0.000832&mimeType=html&fmt=ahah

References

  1. Blitvich BJ, Firth AE. Insect-specific flaviviruses: a systematic review of their discovery, host range, mode of transmission, superinfection exclusion potential and genomic organization. Viruses 2015; 7:1927–1959 [View Article][PubMed]
    [Google Scholar]
  2. Calzolari M, Ze-Ze L, Vázquez A, Sánchez Seco MP, Amaro F et al. Insect-specific flaviviruses, a worldwide widespread group of viruses only detected in insects. Infect Genet Evol 2016; 40:381–388 [View Article][PubMed]
    [Google Scholar]
  3. Chambers TJ, Hahn CS, Galler R, Rice CM. Flavivirus genome organization, expression, and replication. Annu Rev Microbiol 1990; 44:649–688 [View Article][PubMed]
    [Google Scholar]
  4. McLean BJ, Hobson-Peters J, Webb CE, Watterson D, Prow NA et al. A novel insect-specific flavivirus replicates only in Aedes-derived cells and persists at high prevalence in wild Aedes vigilax populations in Sydney, Australia. Virology 2015; 486:272–283 [View Article][PubMed]
    [Google Scholar]
  5. Tree MO, McKellar DR, Kieft KJ, Watson AM, Ryman KD et al. Insect-specific flavivirus infection is restricted by innate immunity in the vertebrate host. Virology 2016; 497:81–91 [View Article][PubMed]
    [Google Scholar]
  6. Hobson-Peters J, Yam AW, Lu JW, Setoh YX, May FJ et al. A new insect-specific flavivirus from northern Australia suppresses replication of West Nile virus and Murray Valley encephalitis virus in co-infected mosquito cells. PLoS One 2013; 8:e56534 [View Article][PubMed]
    [Google Scholar]
  7. Hall-Mendelin S, McLean BJ, Bielefeldt-Ohmann H, Hobson-Peters J, Hall RA et al. The insect-specific Palm Creek virus modulates West Nile virus infection in and transmission by Australian mosquitoes. Parasit Vectors 2016; 9:414 [View Article][PubMed]
    [Google Scholar]
  8. Donald CL, Kohl A, Schnettler E. New insights into control of arbovirus replication and spread by insect RNA interference pathways. Insects 2012; 3:511–531 [View Article][PubMed]
    [Google Scholar]
  9. Léger P, Lara E, Jagla B, Sismeiro O, Mansuroglu Z et al. Dicer-2- and Piwi-mediated RNA interference in Rift Valley fever virus-infected mosquito cells. J Virol 2013; 87:1631–1648 [View Article][PubMed]
    [Google Scholar]
  10. Schnettler E, Donald CL, Human S, Watson M, Siu RW et al. Knockdown of piRNA pathway proteins results in enhanced Semliki Forest virus production in mosquito cells. J Gen Virol 2013; 94:1680–1689 [View Article][PubMed]
    [Google Scholar]
  11. Schnettler E, Ratinier M, Watson M, Shaw AE, Mcfarlane M et al. RNA interference targets arbovirus replication in Culicoides cells. J Virol 2013; 87:2441–2454 [View Article][PubMed]
    [Google Scholar]
  12. Vodovar N, Bronkhorst AW, van Cleef KW, Miesen P, Blanc H et al. Arbovirus-derived piRNAs exhibit a ping-pong signature in mosquito cells. PLoS One 2012; 7:e30861 [View Article][PubMed]
    [Google Scholar]
  13. Dietrich I, Shi X, Mcfarlane M, Watson M, Blomström AL et al. The antiviral RNAi response in vector and non-vector cells against orthobunyaviruses. PLoS Negl Trop Dis 2017; 11:e0005272 [View Article][PubMed]
    [Google Scholar]
  14. Miesen P, Ivens A, Buck AH, van Rij RP. Small RNA profiling in dengue virus 2-Infected Aedes mosquito cells reveals viral piRNAs and novel host miRNAs. PLoS Negl Trop Dis 2016; 10:e0004452 [View Article][PubMed]
    [Google Scholar]
  15. Miesen P, Joosten J, van Rij RP. PIWIs go viral: arbovirus-derived piRNAs in vector mosquitoes. PLoS Pathog 2016; 12:e1006017 [View Article][PubMed]
    [Google Scholar]
  16. Miesen P, Girardi E, van Rij RP. Distinct sets of PIWI proteins produce arbovirus and transposon-derived piRNAs in Aedes aegypti mosquito cells. Nucleic Acids Res 2015; 43:6545–6556 [View Article][PubMed]
    [Google Scholar]
  17. Keene KM, Foy BD, Sanchez-Vargas I, Beaty BJ, Blair CD et al. RNA interference acts as a natural antiviral response to O'nyong-nyong virus (Alphavirus; Togaviridae) infection of Anopheles gambiae. Proc Natl Acad Sci USA 2004; 101:17240–17245 [View Article][PubMed]
    [Google Scholar]
  18. McFarlane M, Arias-Goeta C, Martin E, O'Hara Z, Lulla A et al. Characterization of Aedes aegypti innate-immune pathways that limit chikungunya virus replication. PLoS Negl Trop Dis 2014; 8:e2994 [View Article][PubMed]
    [Google Scholar]
  19. Sánchez-Vargas I, Scott JC, Poole-Smith BK, Franz AW, Barbosa-Solomieu V et al. Dengue virus type 2 infections of Aedes aegypti are modulated by the mosquito's RNA interference pathway. PLoS Pathog 2009; 5:e1000299 [View Article][PubMed]
    [Google Scholar]
  20. Tavanez JP, Quina AS, Cunha C. Virus and noncoding RNAs: stars in the host–virus interaction game. Future Virol 2014; 9:1077–1087 [View Article]
    [Google Scholar]
  21. Han J, Lee Y, Yeom KH, Kim YK, Jin H et al. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev 2004; 18:3016–3027 [View Article][PubMed]
    [Google Scholar]
  22. Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev 2003; 17:3011–3016 [View Article][PubMed]
    [Google Scholar]
  23. Lee YS, Nakahara K, Pham JW, Kim K, He Z et al. Distinct roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing pathways. Cell 2004; 117:69–81 [View Article][PubMed]
    [Google Scholar]
  24. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell 2003; 115:787–798 [View Article][PubMed]
    [Google Scholar]
  25. Lytle JR, Yario TA, Steitz JA. Target mRNAs are repressed as efficiently by microRNA-binding sites in the 5' UTR as in the 3' UTR. Proc Natl Acad Sci USA 2007; 104:9667–9672 [View Article][PubMed]
    [Google Scholar]
  26. Tay Y, Zhang J, Thomson AM, Lim B, Rigoutsos I. MicroRNAs to Nanog, Oct4 and Sox2 coding regions modulate embryonic stem cell differentiation. Nature 2008; 455:1124–1128 [View Article][PubMed]
    [Google Scholar]
  27. Liu Y, Zhou Y, Wu J, Zheng P, Li Y et al. The expression profile of Aedes albopictus miRNAs is altered by dengue virus serotype-2 infection. Cell Biosci 2015; 5:16 [View Article][PubMed]
    [Google Scholar]
  28. Skalsky RL, Vanlandingham DL, Scholle F, Higgs S, Cullen BR. Identification of microRNAs expressed in two mosquito vectors, Aedes albopictus and Culex quinquefasciatus. BMC Genomics 2010; 11:119 [View Article][PubMed]
    [Google Scholar]
  29. Maharaj PD, Widen SG, Huang J, Wood TG, Thangamani S. Discovery of mosquito saliva microRNAs during CHIKV infection. PLoS Negl Trop Dis 2015; 9:e0003386 [View Article][PubMed]
    [Google Scholar]
  30. Hussain M, Frentiu FD, Moreira LA, O'Neill SL, Asgari S. Wolbachia uses host microRNAs to manipulate host gene expression and facilitate colonization of the dengue vector Aedes aegypti. Proc Natl Acad Sci USA 2011; 108:9250–9255 [View Article][PubMed]
    [Google Scholar]
  31. Zhang G, Hussain M, Asgari S. Regulation of arginine methyltransferase 3 by a Wolbachia-induced microRNA in Aedes aegypti and its effect on Wolbachia and dengue virus replication. Insect Biochem Mol Biol 2014; 53:81–88 [View Article][PubMed]
    [Google Scholar]
  32. Zhang G, Hussain M, O'Neill SL, Asgari S. Wolbachia uses a host microRNA to regulate transcripts of a methyltransferase, contributing to dengue virus inhibition in Aedes aegypti. Proc Natl Acad Sci USA 2013; 110:10276–10281 [View Article][PubMed]
    [Google Scholar]
  33. Slonchak A, Hussain M, Torres S, Asgari S, Khromykh AA. Expression of mosquito microRNA aae-miR-2940-5p is downregulated in response to West Nile virus infection to restrict viral replication. J Virol 2014; 88:8457–8467 [View Article][PubMed]
    [Google Scholar]
  34. Liu YX, Li FX, Liu ZZ, Jia ZR, Zhou YH et al. Integrated analysis of miRNAs and transcriptomes in Aedes albopictus midgut reveals the differential expression profiles of immune-related genes during dengue virus serotype-2 infection. Insect Sci 2016; 23:377–385 [View Article][PubMed]
    [Google Scholar]
  35. Stiburek L, Cesnekova J, Kostkova O, Fornuskova D, Vinsova K et al. YME1L controls the accumulation of respiratory chain subunits and is required for apoptotic resistance, cristae morphogenesis, and cell proliferation. Mol Biol Cell 2012; 23:1010–1023 [View Article][PubMed]
    [Google Scholar]
  36. Dennison NJ, Benmarzouk-Hidalgo OJ, Dimopoulos G. MicroRNA-regulation of Anopheles gambiae immunity to Plasmodium falciparum infection and midgut microbiota. Dev Comp Immunol 2015; 36:170–178 [View Article][PubMed]
    [Google Scholar]
  37. Mead EA, Tu Z. Cloning, characterization, and expression of microRNAs from the Asian malaria mosquito, Anopheles stephensi. BMC Genomics 2008; 9:244 [View Article][PubMed]
    [Google Scholar]
  38. Dusi E, Gougat-Barbera C, Berendonk TU, Kaltz O. Long-term selection experiment produces breakdown of horizontal transmissibility in parasite with mixed transmission mode. Evolution 2015; 69:1069–1076 [View Article][PubMed]
    [Google Scholar]
  39. Aguiar ER, Olmo RP, Marques JT. Virus-derived small RNAs: molecular footprints of host-pathogen interactions. Wiley Interdiscip Rev RNA 2016; 7:824–837 [View Article][PubMed]
    [Google Scholar]
  40. Brackney DE, Scott JC, Sagawa F, Woodward JE, Miller NA et al. C6/36 Aedes albopictus cells have a dysfunctional antiviral RNA interference response. PLoS Negl Trop Dis 2010; 4:e856 [View Article][PubMed]
    [Google Scholar]
  41. Hess AM, Prasad AN, Ptitsyn A, Ebel GD, Olson KE et al. Small RNA profiling of dengue virus-mosquito interactions implicates the PIWI RNA pathway in anti-viral defense. BMC Microbiol 2011; 11:45 [View Article][PubMed]
    [Google Scholar]
  42. Morazzani EM, Wiley MR, Murreddu MG, Adelman ZN, Myles KM. Production of virus-derived ping-pong-dependent piRNA-like small RNAs in the mosquito soma. PLoS Pathog 2012; 8:e1002470 [View Article][PubMed]
    [Google Scholar]
  43. Scott JC, Brackney DE, Campbell CL, Bondu-Hawkins V, Hjelle B et al. Comparison of dengue virus type 2-specific small RNAs from RNA interference-competent and -incompetent mosquito cells. PLoS Negl Trop Dis 2010; 4:e848 [View Article][PubMed]
    [Google Scholar]
  44. Siu RW, Fragkoudis R, Simmonds P, Donald CL, Chase-Topping ME et al. Antiviral RNA interference responses induced by Semliki Forest virus infection of mosquito cells: characterization, origin, and frequency-dependent functions of virus-derived small interfering RNAs. J Virol 2011; 85:2907–2917 [View Article][PubMed]
    [Google Scholar]
  45. Göertz GP, Fros JJ, Miesen P, Vogels CB, van der Bent ML et al. Noncoding subgenomic flavivirus RNA is processed by the mosquito RNA interference machinery and determines West Nile virus transmission by Culex pipiens mosquitoes. J Virol 2016; 90:10145–10159 [View Article][PubMed]
    [Google Scholar]
  46. Goic B, Stapleford KA, Frangeul L, Doucet AJ, Gausson V et al. Virus-derived DNA drives mosquito vector tolerance to arboviral infection. Nat Commun 2016; 7:12410 [View Article][PubMed]
    [Google Scholar]
  47. Zhang G, Etebari K, Asgari S. Wolbachia suppresses cell fusing agent virus in mosquito cells. J Gen Virol 2016; 97:3427–3432 [View Article][PubMed]
    [Google Scholar]
  48. Linsen SE, de Wit E, Janssens G, Heater S, Chapman L et al. Limitations and possibilities of small RNA digital gene expression profiling. Nat Methods 2009; 6:474–476 [View Article][PubMed]
    [Google Scholar]
  49. Clark DC, Lobigs M, Lee E, Howard MJ, Clark K et al. In situ reactions of monoclonal antibodies with a viable mutant of Murray Valley encephalitis virus reveal an absence of dimeric NS1 protein. J Gen Virol 2007; 88:1175–1183 [View Article][PubMed]
    [Google Scholar]
  50. O'Brien CA, Hobson-Peters J, Yam AW, Colmant AM, Mclean BJ et al. Viral RNA intermediates as targets for detection and discovery of novel and emerging mosquito-borne viruses. PLoS Negl Trop Dis 2015; 9:e0003629 [View Article][PubMed]
    [Google Scholar]
  51. Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res 2014; 42:D68–D73 [View Article][PubMed]
    [Google Scholar]
  52. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J Mol Biol 1990; 215:403–410 [View Article][PubMed]
    [Google Scholar]
  53. Antoniewski C. Computing siRNA and piRNA overlap signatures. Methods Mol Biol 2014; 1173:135–146 [View Article][PubMed]
    [Google Scholar]
  54. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat Protoc 2008; 3:1101–1108 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.000832
Loading
/content/journal/jgv/10.1099/jgv.0.000832
Loading

Data & Media loading...

Supplements

Supplementary File 1

PDF

Most cited Most Cited RSS feed