1887

Abstract

Mosquito-borne flaviviruses are responsible for viral infections and represent a considerable public health burden. is the principal vector of dengue virus (DENV), therefore understanding the intrinsic virus–host interactions is vital, particularly in the presence of the endosymbiont which blocks virus replication in mosquitoes. Here, we examined the transcriptional response of -transinfected Aag2 cells to DENV infection. We identified differentially expressed immune genes that play a key role in the activation of anti-viral defence such as the Toll and immune deficiency pathways. Further, genes encoding cytosine and N-adenosine methyltransferases and SUMOylation, involved in post-transcriptional modifications, an antioxidant enzyme, and heat-shock response were up-regulated at the early stages of DENV infection and are reported here for the first time. Additionally, several long non-coding RNAs were among the differentially regulated genes. Our results provide insight into -transinfected ’s initial virus recognition and transcriptional response to DENV infection.

Funding
This study was supported by the:
  • Australian Research Council (Award DP190102048)
    • Principle Award Recipient: SassanAsgari
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001694
2022-01-10
2024-04-18
Loading full text...

Full text loading...

/deliver/fulltext/jgv/103/1/jgv001694.html?itemId=/content/journal/jgv/10.1099/jgv.0.001694&mimeType=html&fmt=ahah

References

  1. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW et al. The global distribution and burden of dengue. Nature 2013; 496:504–507 [View Article] [PubMed]
    [Google Scholar]
  2. Guzman MG, Halstead SB, Artsob H, Buchy P, Farrar J et al. Dengue: a continuing global threat. Nat Rev Microbiol 2010; 8:S7–S16 [View Article]
    [Google Scholar]
  3. Liu-Helmersson J, Brännström Å, Sewe MO, Semenza JC, Rocklöv J. Estimating past, present, and future trends in the global distribution and abundance of the arbovirus vector Aedes aegypti under climate change scenarios. Front Public Health 2019; 7:148 [View Article] [PubMed]
    [Google Scholar]
  4. Kraemer MUG, Sinka ME, Duda KA, Mylne A, Shearer FM et al. The global compendium of Aedes aegypti and Ae. albopictus occurrence. Sci Data 2015; 2:150035 [View Article] [PubMed]
    [Google Scholar]
  5. 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]
  6. Kumar A, Srivastava P, Sirisena P, Dubey SK, Kumar R et al. Mosquito innate immunity. Insects 2018; 9:E95 [View Article] [PubMed]
    [Google Scholar]
  7. Bartholomay LC, Michel K. Mosquito Immunobiology: The intersection of vector health and vector competence. Annu Rev Entomol 2018; 63:145–167 [View Article] [PubMed]
    [Google Scholar]
  8. Sánchez-Vargas I, Scott JC, Poole-Smith BK, Franz AWE, 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]
  9. Messina JP, Brady OJ, Scott TW, Zou C, Pigott DM et al. Global spread of dengue virus types: mapping the 70 year history. Trends Microbiol 2014; 22:138–146 [View Article] [PubMed]
    [Google Scholar]
  10. Castro MC, Wilson ME, Bloom DE. Disease and economic burdens of dengue. Lancet Infect Dis 2017; 17:e70–e78 [View Article]
    [Google Scholar]
  11. Bian G, Xu Y, Lu P, Xie Y, Xi Z. The endosymbiotic bacterium Wolbachia induces resistance to dengue virus in Aedes aegypti . PLoS Pathog 2010; 6:e1000833 [View Article] [PubMed]
    [Google Scholar]
  12. Walker T, Johnson PH, Moreira LA, Iturbe-Ormaetxe I, Frentiu FD et al. The wMel Wolbachia strain blocks dengue and invades caged Aedes aegypti populations. Nature 2011; 476:450–453 [View Article] [PubMed]
    [Google Scholar]
  13. Indriani C, Tantowijoyo W, Rancès E, Andari B, Prabowo E et al. Reduced dengue incidence following deployments of Wolbachia-infected Aedes aegypti in Yogyakarta, Indonesia: a quasi-experimental trial using controlled interrupted time series analysis. Gates Open Res 2020; 4:50 [View Article] [PubMed]
    [Google Scholar]
  14. Hoffmann AA, Montgomery BL, Popovici J, Iturbe-Ormaetxe I, Johnson PH et al. Successful establishment of Wolbachia in Aedes populations to suppress dengue transmission. Nature 2011; 476:454–457 [View Article] [PubMed]
    [Google Scholar]
  15. Moreira LA, Iturbe-Ormaetxe I, Jeffery JA, Lu G, Pyke AT et al. A Wolbachia symbiont in Aedes aegypti limits infection with dengue, chikungunya, and Plasmodium . Cell 2009; 139:1268–1278 [View Article] [PubMed]
    [Google Scholar]
  16. Bonizzoni M, Dunn WA, Campbell CL, Olson KE, Marinotti O et al. Complex modulation of the Aedes aegypti transcriptome in response to dengue virus infection. PLoS One 2012; 7:e50512 [View Article] [PubMed]
    [Google Scholar]
  17. Colpitts TM, Cox J, Vanlandingham DL, Feitosa FM, Cheng G et al. Alterations in the Aedes aegypti transcriptome during infection with West Nile, Dengue and Yellow fever viruses. PLoS Pathog 2011; 7:e1002189 [View Article] [PubMed]
    [Google Scholar]
  18. Koh C, Allen SL, Herbert RI, McGraw EA, Chenoweth SF. The transcriptional response of Aedes aegypti with variable extrinsic incubation periods for dengue virus. Genome Biol Evol 2018; 10:3141–3151 [View Article]
    [Google Scholar]
  19. Etebari K, Hegde S, Saldaña MA, Widen SG, Wood TG et al. Global transcriptome analysis of Aedes aegypti mosquitoes in response to Zika virus infection. mSphere 2017; 2:e00456-17 [View Article]
    [Google Scholar]
  20. Li M-J, Lan C-J, Gao H-T, Xing D, Gu Z-Y et al. Transcriptome analysis of Aedes aegypti Aag2 cells in response to dengue virus-2 infection. Parasit Vectors 2020; 13:421 [View Article] [PubMed]
    [Google Scholar]
  21. Hitakarun A, Ramphan S, Wikan N, Smith DR. Analysis of the virus propagation profile of 14 dengue virus isolates in Aedes albopictus C6/36 cells. BMC Res Notes 2020; 13:481 [View Article] [PubMed]
    [Google Scholar]
  22. Bhattacharya T, Newton ILG, Hardy RW. Viral RNA is a target for Wolbachia-mediated pathogen blocking. PLoS Pathog 2020; 16:e1008513 [View Article] [PubMed]
    [Google Scholar]
  23. Rainey SM, Martinez J, McFarlane M, Juneja P, Sarkies P et al. Wolbachia blocks viral genome replication early in infection without a transcriptional response by the endosymbiont or host small RNA pathways. PLoS Pathog 2016; 12:e1005536 [View Article] [PubMed]
    [Google Scholar]
  24. Leitner M, Bishop C, Asgari S. Transcriptional response of Wolbachia to dengue virus infection in cells of the mosquito Aedes aegypti . mSphere 2021e0043321 [View Article] [PubMed]
    [Google Scholar]
  25. Bishop C, Parry R, Asgari S. Effect of Wolbachia wAlbB on a positive-sense RNA negev-like virus: a novel virus persistently infecting Aedes albopictus mosquitoes and cells. J Gen Virol 2020; 101:216–225 [View Article] [PubMed]
    [Google Scholar]
  26. Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol 2010; 11:R25 [View Article] [PubMed]
    [Google Scholar]
  27. McCarthy DJ, Chen Y, Smyth GK. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res 2012; 40:4288–4297 [View Article] [PubMed]
    [Google Scholar]
  28. Ye J, Coulouris G, Zaretskaya I, Cutcutache I, Rozen S et al. Primer-BLAST: A tool to design target-specific primers for polymerase chain reaction. BMC Bioinformatics 2012; 13:134
    [Google Scholar]
  29. Pfaffl MW, Horgan GW, Dempfle L. Relative expression software tool (REST) for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res 2002; 30:e36 [View Article] [PubMed]
    [Google Scholar]
  30. Götz S, García-Gómez JM, Terol J, Williams TD, Nagaraj SH et al. High-throughput functional annotation and data mining with the Blast2GO suite. Nucleic Acids Res 2008; 36:3420–3435 [View Article] [PubMed]
    [Google Scholar]
  31. Jones P, Binns D, Chang H-Y, Fraser M, Li W et al. InterProScan 5: genome-scale protein function classification. Bioinformatics 2014; 30:1236–1240 [View Article] [PubMed]
    [Google Scholar]
  32. Huerta-Cepas J, Szklarczyk D, Heller D, Hernández-Plaza A, Forslund SK et al. eggNOG 5.0: a hierarchical, functionally and phylogenetically annotated orthology resource based on 5090 organisms and 2502 viruses. Nucleic Acids Res 2019; 47:D309–D314 [View Article] [PubMed]
    [Google Scholar]
  33. Peterson TML, Luckhart S. A mosquito 2-Cys peroxiredoxin protects against nitrosative and oxidative stresses associated with malaria parasite infection. Free Radic Biol Med 2006; 40:1067–1082 [View Article] [PubMed]
    [Google Scholar]
  34. Rhee SG, Woo HA, Kil IS, Bae SH. Peroxiredoxin functions as a peroxidase and a regulator and sensor of local peroxides. J Biol Chem 2012; 287:4403–4410 [View Article] [PubMed]
    [Google Scholar]
  35. Zhang S, Li Z, Nian X, Wu F, Shen Z et al. Sequence analysis, expression profiles and function of thioredoxin 2 and thioredoxin reductase 1 in resistance to nucleopolyhedrovirus in Helicoverpa armigera . Sci Rep 2015; 5:15531 [View Article] [PubMed]
    [Google Scholar]
  36. Karpenko IL, Valuev-Elliston VT, Ivanova ON, Smirnova OA, Ivanov AV. Peroxiredoxins-The underrated actors during virus-induced oxidative stress. Antioxidants 2021; 10:977 [View Article] [PubMed]
    [Google Scholar]
  37. Chen T-H, Tang P, Yang C-F, Kao L-H, Lo Y-P et al. Antioxidant defense is one of the mechanisms by which mosquito cells survive dengue 2 viral infection. Virology 2011; 410:410–417 [View Article] [PubMed]
    [Google Scholar]
  38. Wang J-M, Cheng Y, Shi Z-K, Li X-F, Xing L-S et al. Aedes aegypti HPX8C modulates immune responses against viral infection. PLoS Negl Trop Dis 2019; 13:e0007287 [View Article]
    [Google Scholar]
  39. Caicedo PA, Serrato IM, Sim S, Dimopoulos G, Coatsworth H et al. Immune response-related genes associated to blocking midgut dengue virus infection in Aedes aegypti strains that differ in susceptibility. Insect Sci 2019; 26:635–648 [View Article] [PubMed]
    [Google Scholar]
  40. M’Angale PG, Staveley BE. Bax-inhibitor-1 knockdown phenotypes are suppressed by Buffy and exacerbate degeneration in a Drosophila model of Parkinson disease. PeerJ 2017; 5:e2974 [View Article] [PubMed]
    [Google Scholar]
  41. Labeau A, Simon-Loriere E, Hafirassou M-. L, Bonnet-Madin L, Tessier S et al. A Genome-wide CRISPR-Cas9 screen identifies the dolichol-phosphate mannose synthase complex as a host dependency factor for dengue virus infection. J Virol 2020; 94: [View Article] [PubMed]
    [Google Scholar]
  42. Zhao L, Pridgeon JW, Becnel JJ, Clark GG, Linthicum KJ. Identification of genes differentially expressed during heat shock treatment in Aedes aegypti . J Med Entomol 2009; 46:490–495 [View Article] [PubMed]
    [Google Scholar]
  43. Runtuwene LR, Kawashima S, Pijoh VD, Tuda JSB, Hayashida K et al. The lethal(2)-essential-for-life [L(2)EFL] gene family modulates dengue virus infection in Aedes aegypti . Int J Mol Sci 2020; 21:7520 [View Article] [PubMed]
    [Google Scholar]
  44. Waterhouse RM, Kriventseva EV, Meister S, Xi Z, Alvarez KS et al. Evolutionary dynamics of immune-related genes and pathways in disease-vector mosquitoes. Science 2007; 316:1738–1743 [View Article] [PubMed]
    [Google Scholar]
  45. Vilcinskas A. Evolutionary plasticity of insect immunity. J Insect Physiol 2013; 59:123–129 [View Article] [PubMed]
    [Google Scholar]
  46. 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 U S A 2013; 110:10276–10281 [View Article] [PubMed]
    [Google Scholar]
  47. Bhattacharya T, Yan L, Zaher H, Newton ILG, Hardy RW. Differential viral RNA methylation contributes to pathogen blocking in Wolbachia-colonized arthropod. bioRxiv 2021; 2021:
    [Google Scholar]
  48. Durdevic Z, Schaefer M. Dnmt2 methyltransferases and immunity: An ancient overlooked connection between nucleotide modification and host defense?. BioEssays 2013; 35:1044–1049 [View Article] [PubMed]
    [Google Scholar]
  49. Imam H, Kim G-. W, Siddiqui A. Epitranscriptomic(N6-methyladenosine) modification of viral RNA and virus-host interactions. Front Cell Infect Microbiol 2020; 10:584283 [View Article] [PubMed]
    [Google Scholar]
  50. Gokhale NS, McIntyre ABR, McFadden MJ, Roder AE, Kennedy EM et al. N6-methyladenosine in Flaviviridae viral RNA genomes regulates infection. Cell Host Microbe 2016; 20:654–665 [PubMed]
    [Google Scholar]
  51. Kang S, Shin D, Mathias DK, Londono-Renteria B, Noh MY et al. Homologs of human dengue-resistance genes, FKBP1B and ATCAY, confer antiviral resistance in Aedes aegypti mosquitoes. Insects 2019; 10:E46 [View Article] [PubMed]
    [Google Scholar]
  52. Cerritelli SM, Crouch RJ. Ribonuclease H: the enzymes in eukaryotes. FEBS J 2009; 276:1494–1505 [View Article] [PubMed]
    [Google Scholar]
  53. Hartmann G. Nucleic acid immunity. Adv Immunol 2017; 133:121–169 [PubMed]
    [Google Scholar]
  54. Majorek KA, Dunin-Horkawicz S, Steczkiewicz K, Muszewska A, Nowotny M et al. The RNase H-like superfamily: new members, comparative structural analysis and evolutionary classification. Nucleic Acids Res 2014; 42:4160–4179 [View Article] [PubMed]
    [Google Scholar]
  55. Moelling K, Broecker F, Russo G, Sunagawa S. RNase H as gene modifier, driver of evolution and antiviral defense. Front Microbiol 2017; 8:1745 [View Article] [PubMed]
    [Google Scholar]
  56. Laalami S, Zig L, Putzer H. Initiation of mRNA decay in bacteria. Cell Mol Life Sci 2014; 71:1799–1828 [View Article] [PubMed]
    [Google Scholar]
  57. Anupama K, Leela JK, Gowrishankar J. Two pathways for RNase E action in Escherichia coli in vivo and bypass of its essentiality in mutants defective for Rho-dependent transcription termination. Mol Microbiol 2019; 112:1371 [View Article] [PubMed]
    [Google Scholar]
  58. Hackett BA, Yasunaga A, Panda D, Tartell MA, Hopkins KC et al. RNASEK is required for internalization of diverse acid-dependent viruses. Proc Natl Acad Sci U S A 2015; 112:7797–7802 [View Article] [PubMed]
    [Google Scholar]
  59. Zhang G, Asad S, Khromykh AA, Asgari S. Cell fusing agent virus and dengue virus mutually interact in Aedes aegypti cell lines. Sci Rep 2017; 7:6935 [View Article] [PubMed]
    [Google Scholar]
  60. Stokes S, Almire F, Tatham MH, McFarlane S, Mertens P et al. The SUMOylation pathway suppresses arbovirus replication in Aedes aegypti cells. PLoS Pathog 2020; 16:e1009134 [View Article] [PubMed]
    [Google Scholar]
  61. Sigle LT, McGraw EA. Expanding the canon: Non-classical mosquito genes at the interface of arboviral infection. Insect Biochem Mol Biol 2019; 109:72–80 [View Article] [PubMed]
    [Google Scholar]
  62. Barón OL, Ursic-Bedoya RJ, Lowenberger CA, Ocampo CB. Differential gene expression from midguts of refractory and susceptible lines of the mosquito, Aedes aegypti, infected with Dengue-2 virus. J Insect Sci 2010; 10:41 [View Article] [PubMed]
    [Google Scholar]
  63. Li Y, Piermarini PM, Esquivel CJ, Price DP, Drumm HE et al. RNA-Seq comparison of larval and adult Malpighian tubules of the yellow fever mosquito Aedes aegypti reveals life stage-specific changes in renal function. Front Physiol 2017; 8:283 [View Article] [PubMed]
    [Google Scholar]
  64. Wimalasiri-Yapa BMCR, Barrero RA, Stassen L, Hafner LM, McGraw EA et al. Temperature modulates immune gene expression in mosquitoes during arbovirus infection. Open Biol 2021; 11:200246 [View Article] [PubMed]
    [Google Scholar]
  65. Reyes JIL, Suzuki Y, Carvajal T, Muñoz MNM, Watanabe K. Intracellular interactions between arboviruses and Wolbachia in Aedes aegypti . Front Cell Infect Microbiol 2021; 11:690087 [View Article] [PubMed]
    [Google Scholar]
  66. Xi Z, Gavotte L, Xie Y, Dobson SL. Genome-wide analysis of the interaction between the endosymbiotic bacterium Wolbachia and its Drosophila host. BMC Genomics 2008; 9:1
    [Google Scholar]
  67. Zhao L, Alto BW, Shin D. Transcriptional profile of Aedes aegypti leucine-rich repeat proteins in response to Zika and chikungunya viruses. IJMS 2019; 20:615 [View Article]
    [Google Scholar]
  68. Waterhouse RM, Povelones M, Christophides GK. Sequence-structure-function relations of the mosquito leucine-rich repeat immune proteins. BMC Genomics 2010; 11:531 [View Article] [PubMed]
    [Google Scholar]
  69. Ng A, Xavier RJ. Leucine-rich repeat (LRR) proteins: integrators of pattern recognition and signaling in immunity. Autophagy 2011; 7:1082–1084 [View Article] [PubMed]
    [Google Scholar]
  70. Angleró-Rodríguez YI, MacLeod HJ, Kang S, Carlson JS, Jupatanakul N et al. Aedes aegypti molecular responses to Zika Virus: modulation of infection by the Toll and Jak/Stat immune pathways and virus host factors. Front Microbiol 2017; 8:2050 [View Article] [PubMed]
    [Google Scholar]
  71. Mukherjee D, Das S, Begum F, Mal S, Ray U. The mosquito immune system and the life of dengue virus: what we know and do not know. Pathogens 2019; 8:E77 [View Article] [PubMed]
    [Google Scholar]
  72. Jupatanakul N, Sim S, Dimopoulos G. Aedes aegypti ML and Niemann-Pick type C family members are agonists of dengue virus infection. Dev Comp Immunol 2014; 43:1–9 [View Article] [PubMed]
    [Google Scholar]
  73. Ramirez JL, Dimopoulos G. The Toll immune signaling pathway control conserved anti-dengue defenses across diverse Ae. aegypti strains and against multiple dengue virus serotypes. Dev Comp Immunol 2010; 34:625–629 [View Article] [PubMed]
    [Google Scholar]
  74. Ramirez JL, Muturi EJ, Barletta ABF, Rooney AP. The Aedes aegypti IMD pathway is a critical component of the mosquito antifungal immune response. Dev Comp Immunol 2019; 95:1–9 [View Article] [PubMed]
    [Google Scholar]
  75. Sim S, Jupatanakul N, Ramirez JL, Kang S, Romero-Vivas CM et al. Transcriptomic profiling of diverse Aedes aegypti strains reveals increased basal-level immune activation in dengue virus-refractory populations and identifies novel virus-vector molecular interactions. PLoS Negl Trop Dis 2013; 7:e2295 [View Article] [PubMed]
    [Google Scholar]
  76. Russell TA, Ayaz A, Davidson AD, Fernandez-Sesma A, Maringer K. Imd pathway-specific immune assays reveal NF-κB stimulation by viral RNA PAMPs in Aedes aegypti Aag2 cells. PLoS Negl Trop Dis 2021; 15:e0008524 [View Article] [PubMed]
    [Google Scholar]
  77. Ding Y, Zhang Z, Liu Y, Shi C, Zhang J et al. Relationship of long noncoding RNA and viruses. Genomics 2016; 107:150–154 [View Article] [PubMed]
    [Google Scholar]
  78. Clark MB, Mattick JS. Long noncoding RNAs in cell biology. Semin Cell Dev Biol 2011; 22:366–376 [View Article] [PubMed]
    [Google Scholar]
  79. Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet 2009; 10:155–159 [View Article] [PubMed]
    [Google Scholar]
  80. Ahmad P, Bensaoud C, Mekki I, Rehman MU, Kotsyfakis M. Long non-coding RNAs and their potential roles in the vector-host-pathogen triad. Life 2021; 11:56 [View Article] [PubMed]
    [Google Scholar]
  81. Etebari K, Asad S, Zhang G, Asgari S. Identification of Aedes aegypti long intergenic non-coding RNAs and their association with Wolbachia and dengue virus infection. PLoS Negl Trop Dis 2016; 10:e0005069 [View Article] [PubMed]
    [Google Scholar]
  82. Azlan A, Obeidat SM, Theva Das K, Yunus MA, Azzam G. Genome-wide identification of Aedes albopictus long noncoding RNAs and their association with dengue and Zika virus infection. PLoS Negl Trop Dis 2021; 15:e0008351 [View Article] [PubMed]
    [Google Scholar]
  83. Shrinet J, Srivastava P, Sunil S. Transcriptome analysis of Aedes aegypti in response to mono-infections and co-infections of dengue virus-2 and chikungunya virus. Biochem Biophys Res Commun 2017; 492:617–623 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001694
Loading
/content/journal/jgv/10.1099/jgv.0.001694
Loading

Data & Media loading...

Supplements

Supplementary material 1

EXCEL

Supplementary material 2

EXCEL

Supplementary material 3

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error