Revisiting dengue virus-mosquito interactions: molecular insights into viral fitness Open Access

Abstract

Dengue virus (DENV), like other viruses, closely interacts with the host cell machinery to complete its life cycle. Over the course of infection, DENV interacts with several host factors with pro-viral activities to support its infection. Meanwhile, it has to evade or counteract host factors with anti-viral activities which inhibit its infection. These molecular virus-host interactions play a crucial role in determining the success of DENV infection. Deciphering such interactions is thus paramount to understanding viral fitness in its natural hosts. While DENV-mammalian host interactions have been extensively studied, not much has been done to characterize DENV-mosquito host interactions despite its importance in controlling DENV transmission. Here, to provide a snapshot of our current understanding of DENV-mosquito interactions, we review the literature that identified host factors and cellular processes related to DENV infection in its mosquito vectors, and , with a particular focus on DENV-mosquito omics studies. This knowledge provides fundamental insights into the DENV life cycle, and could contribute to the development of novel antiviral strategies.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001693
2021-11-30
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jgv/102/11/jgv001693.html?itemId=/content/journal/jgv/10.1099/jgv.0.001693&mimeType=html&fmt=ahah

References

  1. Kraemer MU, Sinka ME, Duda KA, Mylne AQ, Shearer FM et al. The global distribution of the arbovirus vectors Aedes aegypti and Ae. albopictus. eLife 2015; 4: [View Article]
    [Google Scholar]
  2. Brady OJ, Gething PW, Bhatt S, Messina JP, Brownstein JS et al. Refining the global spatial limits of dengue virus transmission by evidence-based consensus. PLoS Negl Trop Dis 2012; 6:e1760 [View Article] [PubMed]
    [Google Scholar]
  3. World Health Organization (WHO Dengue and severe dengue: Fact Sheet; 2019
  4. World Health Organization Revised SAGE recommendation on use of dengue vaccine; 2018
  5. Utarini A, Indriani C, Ahmad RA, Tantowijoyo W, Arguni E et al. Efficacy of wolbachia-infected mosquito deployments for the control of dengue. N Engl J Med 2021; 384:2177–2186 [View Article] [PubMed]
    [Google Scholar]
  6. Jupatanakul N, Sim S, Angleró-Rodríguez YI, Souza-Neto J, Das S et al. Engineered Aedes aegypti JAK/STAT pathway-mediated immunity to dengue virus. PLoS Negl Trop Dis 2017; 11:e0005187 [View Article] [PubMed]
    [Google Scholar]
  7. Chiu HC, Hannemann H, Heesom KJ, Matthews DA, Davidson AD. High-throughput quantitative proteomic analysis of dengue virus type 2 infected A549 cells. PLoS One 2014; 9:e93305 [View Article] [PubMed]
    [Google Scholar]
  8. Hafirassou ML, Meertens L, Umaña-Diaz C, Labeau A, Dejarnac O et al. A global interactome map of the dengue virus NS1 identifies virus restriction and dependency host factors. Cell Reports 2017; 21:3900–3913 [View Article]
    [Google Scholar]
  9. Marceau CD, Puschnik AS, Majzoub K, Ooi YS, Brewer SM et al. Genetic dissection of Flaviviridae host factors through genome-scale CRISPR screens. Nature 2016; 535:159–163 [View Article] [PubMed]
    [Google Scholar]
  10. Ooi YS, Majzoub K, Flynn RA, Mata MA, Diep J et al. An RNA-centric dissection of host complexes controlling flavivirus infection. Nat Microbiol 2019; 4:2369–2382 [View Article]
    [Google Scholar]
  11. Shah PS, Link N, Jang GM, Sharp PP, Zhu T et al. Comparative flavivirus-host protein interaction mapping reveals mechanisms of dengue and zika virus pathogenesis. Cell 2018; 175:1931–1945S0092-8674(18)31553-8 [View Article] [PubMed]
    [Google Scholar]
  12. Wongtrakul J, Thongtan T, Pannengpetch S, Wikan N, Kantamala D et al. Phosphoproteomic analysis of dengue virus infected U937 cells and identification of pyruvate kinase M2 as a differentially phosphorylated phosphoprotein. Sci Rep 2020; 10:14493 [View Article] [PubMed]
    [Google Scholar]
  13. Acosta EG, Kumar A, Bartenschlager R. Revisiting dengue virus-host cell interaction: New insights into molecular and cellular virology. In Advances in Virus Research Cambridge, Massachusetts: Academic Press; 2014 pp 1–109
    [Google Scholar]
  14. Campos RK, Garcia-Blanco MA, Bradrick SS. Roles of pro-viral host factors in mosquito-borne flavivirus infections. In Current Topics in Microbiology and Immunology Springer, Cham; 2018
    [Google Scholar]
  15. Krishnan MN, Garcia-Blanco MA. Targeting host factors to treat West Nile and dengue viral infections. Viruses 2014; 6:683–708 [View Article] [PubMed]
    [Google Scholar]
  16. Salazar MI, del Angel RM, Lanz-Mendoza H, Ludert JE, Pando-Robles V. The role of cell proteins in dengue virus infection. J Proteomics 2014; 111:6–15 [View Article] [PubMed]
    [Google Scholar]
  17. Trammell CE, Goodman AG. Host factors that control mosquito-borne viral infections in humans and their vector. Viruses 2021; 13:748 [View Article]
    [Google Scholar]
  18. Choy MM, Ng DHL, Siriphanitchakorn T, Ng WC, Sundstrom KB et al. A non-structural 1 protein G53D substitution attenuates a clinically tested live dengue vaccine. Cell Reports 2020; 31:107617 [View Article]
    [Google Scholar]
  19. Pompon J, Manuel M, Ng GK, Wong B, Shan C et al. Dengue subgenomic flaviviral RNA disrupts immunity in mosquito salivary glands to increase virus transmission. PLoS Pathog 2017; 13:e1006535 [View Article]
    [Google Scholar]
  20. Sessions OM, Barrows NJ, Souza-Neto JA, Robinson TJ, Hershey CL et al. Discovery of insect and human dengue virus host factors. Nature 2009; 458:1047–1050 [View Article] [PubMed]
    [Google Scholar]
  21. Nene V, Wortman JR, Lawson D, Haas B, Kodira C et al. Genome sequence of Aedes aegypti, a major arbovirus vector. Science 2007; 316:1718–1723 [View Article]
    [Google Scholar]
  22. Chen X-G, Jiang X, Gu J, Xu M, Wu Y et al. Genome sequence of the Asian tiger mosquito, aedes albopictus, reveals insights into its biology, genetics, and evolution. Proc Natl Acad Sci U S A 2015; 112:E5907–15 [View Article] [PubMed]
    [Google Scholar]
  23. Luplertlop N, Surasombatpattana P, Patramool S, Dumas E, Wasinpiyamongkol L et al. Induction of a peptide with activity against a broad spectrum of pathogens in the Aedes aegypti salivary gland, following infection with dengue virus. PLoS Pathog 2011; 7:e1001252 [View Article]
    [Google Scholar]
  24. Sim S, Ramirez JL, Dimopoulos G, Diamond MS. Dengue virus infection of the Aedes aegypti Salivary gland and chemosensory apparatus induces genes that modulate infection and blood-feeding behavior. PLoS Pathog 2012; 8:e1002631 [View Article]
    [Google Scholar]
  25. Xi Z, Ramirez JL, Dimopoulos G, Schneider DS. The Aedes aegypti toll pathway controls dengue virus infection. PLoS Pathog 2008; 4:e1000098 [View Article]
    [Google Scholar]
  26. 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]
  27. 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]
  28. Li MJ, Lan CJ, Gao HT, Xing D, Gu ZY et al. Transcriptome analysis of aedes aegypti aag2 cells in response to dengue virus-2 infection. Parasit Vectors 2020; 13:421
    [Google Scholar]
  29. Sim S, Dimopoulos G. Dengue virus inhibits immune responses in Aedes aegypti cells. PLoS One 2010; 5:e10678 [View Article] [PubMed]
    [Google Scholar]
  30. Li M, Xing D, Su D, Wang D, Gao H et al. Transcriptome analysis of responses to dengue virus 2 infection in aedes albopictus (Skuse) c6/36 cells. Viruses 2021; 13:343 [View Article]
    [Google Scholar]
  31. Behura SK, Gomez-Machorro C, Harker BW, deBruyn B, Lovin DD et al. Global cross-talk of genes of the mosquito Aedes aegypti in response to dengue virus infection. PLoS Negl Trop Dis 2011; 5:e1385 [View Article] [PubMed]
    [Google Scholar]
  32. Souza-Neto JA, Sim S, Dimopoulos G. An evolutionary conserved function of the JAK-STAT pathway in anti-dengue defense. Proc Natl Acad Sci U S A 2009; 106:17841–17846 [View Article] [PubMed]
    [Google Scholar]
  33. Liu J, Liu Y, Nie K, Du S, Qiu J et al. Flavivirus NS1 protein in infected host sera enhances viral acquisition by mosquitoes. Nat Microbiol 2016; 1: [View Article]
    [Google Scholar]
  34. Paradkar PN, Duchemin JB, Voysey R, Walker PJ. Dicer-2-dependent activation of culex vago occurs via the TRAF-Rel2 signaling pathway. PLoS Negl Trop Dis 2014; 8:e2823 [View Article] [PubMed]
    [Google Scholar]
  35. Bonning BC, Saleh MC. The Interplay between viruses and RNAi pathways in insects. Annu Rev Entomol 2021; 66:61–79 [View Article] [PubMed]
    [Google Scholar]
  36. Kingsolver MB, Huang Z, Hardy RW. Sect antiviral innate immunity: Pathways, effectors, and connections. J Mol Biol 2013; 425:4921–4936S0022-2836(13)00632-3 [View Article] [PubMed]
    [Google Scholar]
  37. 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]
    [Google Scholar]
  38. 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]
    [Google Scholar]
  39. Franz AWE, Sanchez-Vargas I, Adelman ZN, Blair CD, Beaty BJ et al. Engineering RNA interference-based resistance to dengue virus type 2 in genetically modified Aedes aegypti. Proceedings of the National Academy of Sciences 2006; 103:4198–4203 [View Article]
    [Google Scholar]
  40. Mathur G, Sanchez-Vargas I, Alvarez D, Olson KE, Marinotti O et al. Transgene-mediated suppression of dengue viruses in the salivary glands of the yellow fever mosquito, Aedes aegypti. Insect Mol Biol 2010; 19:753–763 [View Article] [PubMed]
    [Google Scholar]
  41. Chauhan C, Behura SK, deBruyn B, Lovin DD, Harker BW et al. Comparative expression profiles of midgut genes in dengue virus refractory and susceptible Aedes aegypti across critical period for virus infection. PLoS ONE 2012; 7:e47350 [View Article]
    [Google Scholar]
  42. 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]
    [Google Scholar]
  43. Patramool S, Surasombatpattana P, Luplertlop N, Sévéno M, Choumet V et al. Proteomic analysis of an Aedes albopictus cell line infected with Dengue serotypes 1 and 3 viruses. Parasites Vectors 2011; 4: [View Article]
    [Google Scholar]
  44. Tchankouo-Nguetcheu S, Khun H, Pincet L, Roux P, Bahut M et al. Differential protein modulation in midguts of Aedes aegypti infected with chikungunya and dengue 2 viruses. PLoS One 2010; 5:e13149 [View Article] [PubMed]
    [Google Scholar]
  45. Mehrbod P, Ande SR, Alizadeh J, Rahimizadeh S, Shariati A et al. The roles of apoptosis, autophagy and unfolded protein response in arbovirus, influenza virus, and HIV infections. Virulence 2019; 10:376–413 [View Article] [PubMed]
    [Google Scholar]
  46. Chisenhall DM, Christofferson RC, McCracken MK, Johnson AMF, Londono-Renteria B et al. fection with dengue-2 virus alters proteins in naturally expectorated saliva of Aedes aegypti mosquitoes. Parasites Vectors 2014; 7: [View Article]
    [Google Scholar]
  47. Fontaine A, Diouf I, Bakkali N, Missé D, Pagès F et al. Implication of haematophagous arthropod salivary proteins in host-vector interactions. Parasites Vectors 2011; 4: [View Article]
    [Google Scholar]
  48. Chee HY, AbuBakar S. Identification of a 48kDa tubulin or tubulin-like C6/36 mosquito cells protein that binds dengue virus 2 using mass spectrometry. Biochem Biophys Res Commun 2004; 320:11–17 [View Article] [PubMed]
    [Google Scholar]
  49. Mercado-Curiel RF, Esquinca-Avilés HA, Tovar R, Díaz-Badillo A, Camacho-Nuez M et al. The four serotypes of dengue recognize the same putative receptors in Aedes aegypti midgut and Ae. albopictus cells. BMC Microbiol 2006; 6:85 [View Article]
    [Google Scholar]
  50. Muñoz M de L, Limón-Camacho G, Tovar R, Diaz-Badillo A, Mendoza-Hernández G et al. Proteomic identification of dengue virus binding proteins in Aedes aegypti mosquitoes and Aedes albopictus cells. BioMed Research International 2013; 2013:1–11 [View Article]
    [Google Scholar]
  51. Paingankar MS, Gokhale MD, Deobagkar DN. Dengue-2-virus-interacting polypeptides involved in mosquito cell infection. Arch Virol 2010; 155:1453–1461 [View Article]
    [Google Scholar]
  52. Vega-Almeida TO, Salas-Benito M, De Nova-Ocampo MA, Del Angel RM, Salas-Benito JS. Surface proteins of C6/36 cells involved in dengue virus 4 binding and entry. Arch Virol 2013; 158:1189–1207 [View Article]
    [Google Scholar]
  53. Colpitts TM, Cox J, Nguyen A, Feitosa F, Krishnan MN et al. Use of a tandem affinity purification assay to detect interactions between West Nile and dengue viral proteins and proteins of the mosquito vector. Virology 2011; 417:179–187 [View Article]
    [Google Scholar]
  54. Perera R, Riley C, Isaac G, Hopf-Jannasch AS, Moore RJ et al. Dengue virus infection perturbs lipid homeostasis in infected mosquito cells. PLoS Pathog 2012; 8:e1002584 [View Article]
    [Google Scholar]
  55. den Boon JA, Ahlquist P. Organelle-like membrane compartmentalization of positive-strand RNA virus replication factories. Annu Rev Microbiol 2010; 64:241–256 [View Article] [PubMed]
    [Google Scholar]
  56. Vial T, Tan W-L, Wong Wei Xiang B, Missé D, Deharo E et al. Dengue virus reduces AGPAT1 expression to alter phospholipids and enhance infection in Aedes aegypti. PLoS Pathog 2019; 15:e1008199 [View Article]
    [Google Scholar]
  57. Vial T, Tan WL, Deharo E, Missé D, Marti G et al. Mosquito metabolomics reveal that dengue virus replication requires phospholipid reconfiguration via the remodeling cycle. Proc Natl Acad Sci U S A 2020; 117:27627–27636 [View Article] [PubMed]
    [Google Scholar]
  58. Koh C, Islam MN, Ye YH, Chotiwan N, Graham B et al. Dengue virus dominates lipid metabolism modulations in Wolbachia-coinfected Aedes aegypti. Commun Biol 2020; 3:518 [View Article] [PubMed]
    [Google Scholar]
  59. 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; 43:1–9S0145-305X(13)00293-0 [View Article] [PubMed]
    [Google Scholar]
  60. Guo X, Xu Y, Bian G, Pike AD, Xie Y et al. Response of the mosquito protein interaction network to dengue infection. BMC Genomics 2010; 11:380 [View Article]
    [Google Scholar]
  61. Doolittle JM, Gomez SM. Mapping protein interactions between dengue virus and its human and insect hosts. PLoS Negl Trop Dis 2011; 5:e954 [View Article] [PubMed]
    [Google Scholar]
  62. Bustos-Arriaga J, Gromowski GD, Tsetsarkin KA, Firestone C-. Y, Castro-Jiménez T et al. Decreased accumulation of subgenomic RNA in human cells infected with vaccine candidate DEN4Δ30 increases viral susceptibility to type I interferon. Vaccine 2018; 36:3460–3467S0264-410X(18)30607-8 [View Article]
    [Google Scholar]
  63. Manokaran G, Finol E, Wang C, Gunaratne J, Bahl J et al. Dengue subgenomic RNA binds TRIM25 to inhibit interferon expression for epidemiological fitness. Science 2015; 350:217–221 [View Article]
    [Google Scholar]
  64. Rodpai A, Pulmanausahakul R, Midoeng P, Ubol S, Yoksan S. Contribution of monocyte-derived dendritic cells infected with virulent- or attenuateddengue virus to tnf-α reactogenicity and association of il-1β and il-8 with attenuated phenotype 2018 p 49
    [Google Scholar]
  65. Sessions OM, Tan Y, Goh KC, Liu Y, Tan P et al. Host cell transcriptome profile during wild-type and attenuated dengue virus infection. PLoS Negl Trop Dis 2013; 7:e2107 [View Article] [PubMed]
    [Google Scholar]
  66. Syenina A, Vijaykrishna D, Gan ES, Tan HC, Choy MM et al. Positive epistasis between viral polymerase and the 3’ untranslated region of its genome reveals the epidemiologic fitness of dengue virus. Proc Natl Acad Sci U S A 2020; 117:11038–11047 [View Article] [PubMed]
    [Google Scholar]
  67. Vignuzzi M, Stone JK, Arnold JJ, Cameron CE, Andino R. Quasispecies diversity determines pathogenesis through cooperative interactions in a viral population. Nature 2006; 439:344–348 [View Article] [PubMed]
    [Google Scholar]
  68. Dolan PT, Taguwa S, Rangel MA, Acevedo A, Hagai T et al. Principles of dengue virus evolvability derived from genotype-fitness maps in human and mosquito cells. Elife; 2021
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001693
Loading
/content/journal/jgv/10.1099/jgv.0.001693
Loading

Data & Media loading...

Most cited Most Cited RSS feed