Abstract

Transposable elements are mobile repeated sequences found in all genomes. Transposable elements are controlled by RNA interference pathways in most organisms, and this control involves the PIWI-interacting RNA pathway and the small interfering RNA pathway, which is also known to be the first line of antiviral defense in invertebrates. Using Drosophila, we recently showed that viral infections result in the modulation of transposable element transcript levels through modulation of the small RNA repertoire. The Aedes aegypti mosquito is of particular interest because almost half of its genome is made of transposable elements, and it is described as a major vector of viruses (such as the dengue [DENV], Zika [ZIKV], and chikungunya [CHIKV] arboviruses). Moreover, Aedes mosquitoes are unique among insects in that the PIWI-interacting RNA pathway is also involved in the somatic antiviral response, in addition to the transposable element control and PIWI-interacting RNA pathway genes expanded in the mosquito genome. For these reasons, we studied the impacts of viral infections on transposable element transcript levels in A. aegypti samples. We retrieved public datasets corresponding to RNA-seq data obtained from viral infections by DENV, ZIKV, and CHIKV in various tissues. We found that transposable element transcripts are moderately modulated following viral infection and that the direction of the modulation varies greatly across tissues and viruses. These results highlight the need for an in-depth investigation of the tightly intertwined interactions between transposable elements and viruses.

Significance

Transposable elements are genomic parasites that are found in all genomes. In insects, similar RNA interference pathways control transposable element activity and fight against viral infections. In this study, we focused on A. aegypti, the yellow fever mosquito, which harbors almost 50% transposable elements in its genome and is known to be a major vector of arboviruses. Using various publicly available RNA-seq datasets, we showed that arboviral infections induce moderate modulations of transposable element transcript levels in mosquito tissues. In addition, we revealed that the direction of the modulation strongly depends on the virus. These results highlight the need for an in-depth investigation of the tightly intertwined interactions between transposable elements and viruses.

Introduction

Transposable elements (TEs) are repeated DNA sequences found in all genomes, in which their amounts can vary due to their capacity to move within chromosomes. For example, TEs constitute approximately 3%, 47%, and 78% of the genomes of the honey bee Apis mellifera (Honeybee Genome Sequencing Consortium 2006; Elsik et al. 2014), the mosquito Aedes aegypti (Nene et al. 2007), and the arctic krill Euphausia superba (Shao et al. 2023), respectively. The ability of TEs to transpose within host chromosomes and their high abundance in many organisms have made TEs drivers of evolution (Biémont 2010). TE insertions are generally neutral or slightly deleterious. They may induce double-strand DNA breaks, ectopic recombination or disruption of coding sequences (Goodier and Kazazian 2008; Zamudio et al. 2015), but they are also involved in adaptation and genetic innovations (Casacuberta and González 2013; Gilbert et al. 2021; Galbraith and Hayward 2023; Lawson et al. 2023). TEs can be classified into two principal classes according to their transposition intermediates. Class I TEs or retrotransposons have an RNA intermediate and transpose via a “copy-and-paste” mechanism. Class II TEs are TEs with DNA intermediates called DNA transposons and are mainly mobilized through a “cut-and-paste” mechanism (Wells and Feschotte 2020). These classes are further subdivided into subclasses, families, and subfamilies according to their replication mechanism, structure, and sequence similarity (Wicker et al. 2007).

Regulatory mechanisms that provide protection from the deleterious effects of TEs have been selected in hosts. Among these, RNA interference (RNAi) is one of the major processes involved in TE control and acts at the transcriptional or posttranscriptional level through the action of small RNAs. In Drosophila, two RNAi pathways participate in TE control and rely on distinct RNA molecules: small interfering RNAs (siRNAs) and PIWI-interacting RNAs (piRNAs). They have their own RNA precursors and involve particular molecular effectors. RNAi is also known to be the main antiviral mechanism in insects (Obbard et al. 2009; Mussabekova et al. 2017).

piRNAs are 23 to 30 nucleotide-long sequences processed from single-stranded RNA precursors that are enriched in TE sequences and transcribed from genomic loci called piRNA clusters (Siomi et al. 2011; Senti et al. 2015; Suzuki et al. 2020). piRNAs are loaded onto PIWI proteins, members of the AGO protein family. The corresponding RNA-induced silencing complex (RISC) subsequently targets TE RNAs through sequence complementarity and induces the transcriptional or posttranscriptional silencing of TE sequences (Ozata et al. 2019). The Dicer-2 endonuclease plays a central role in the siRNA pathway. This protein slices double-stranded RNA (dsRNA) precursors into 21 nucleotides, and one strand is then loaded onto Argonaute 2, constituting the RISC that degrades any TE or viral sequence complementary to the loaded siRNA (Campbell et al. 2008; Palmer et al. 2018). In Drosophila melanogaster, in which they have been extensively studied, piRNAs are involved in TE control in both germline and gonadal somatic cells to prevent TE transposition, whereas siRNAs fight against TEs and viruses both in the soma and gonads (Siomi et al. 2011; Bronkhorst and van Rij 2014).

Given that the siRNA pathway targets both TEs and viruses, it seems legitimate to wonder how organisms cope when they are challenged from both sides. In particular, one can ask whether and how TE regulation is affected by virus infection. Two scenarios can be envisioned (Roy et al. 2020). The first one posits that there is a trade-off between TE control by the host and its ability to fight a viral infection so that an organism cannot achieve both effectively at the same time. If this is the case, then one would expect an increase in TE transcript amounts upon viral infection. This scenario is in line with the idea that the Dicer-2 protein, which is the siRNA producer, can be saturated (Durdevic et al. 2013). The second scenario involves a synergistic interaction between antiviral and TE silencing responses: viral infections can activate small RNA pathways and thus trigger a strong TE silencing response and conversely. In a recent study, Roy et al. (2020) investigated the effects of viral infections on TE transcript levels in Drosophila. They showed that during Sindbis virus (SINV) infection in Drosophila simulans and D. melanogaster carcasses, TE transcript and TE-derived small RNA modulation support the scenario of an amplification between TE control and antiviral immunity: upon infection, TE transcript levels are downmodulated, which is associated with an increase in TE-derived small RNAs. In a subsequent study, Roy et al. (2021) performed similar experiments and showed that when D. simulans was infected with D. melanogaster sigma virus (DMelSV), modulation resulted in a different scenario: TE transcripts levels increased in carcasses, in line with a trade-off scenario. Thus, depending on the Drosophila species and the virus, TE transcripts and associated small RNAs are differentially modulated. Data from other organisms are thus clearly needed to determine whether a general pattern emerges for the impact on both piRNA and siRNA pathways and to determine the extent to which TEs are modulated during viral infection. This comparative genomics approach is all the more needed given that D. melanogaster appears to be an exception rather than the rule among arthropods in terms of its very low abundance of somatic piRNAs (Lewis et al. 2018). Here, we focused on TE transcript modulation upon viral infection by DENV, ZIKV, and CHIKV in A. aegypti.

Aedes aegypti is a dipteran species of the Culicidae family, which diverged from Drosophilidae approximately 260 million years ago (Gaunt and Miles 2002). It is a major vector of arboviruses, such as Zika virus (ZIKV), yellow fever virus (YFV), dengue virus (DENV serotypes 1 to 4), and chikungunya virus (CHIKV). The first three viruses belong to the Flavivirus genus and have single-stranded positive and nonpolyadenylated RNA genomes of approximately 10 kb (Markoff 2003; Kuno and Chang 2007). These genomes encode a unique long open reading frame (ORF) corresponding to three structural proteins and seven nonstructural proteins (Chambers et al. 1990). CHIKV is an Alphavirus of the Togaviridae family that has a single-stranded positive and polyadenylated RNA genome (Strauss and Strauss 1994; Solignat et al. 2009). It encodes two ORFs corresponding to five structural proteins and four nonstructural proteins (Solignat et al. 2009). In addition, some viral products act as viral suppressors of RNAi, such as the nonstructural protein NS2A of DENV-2 (Qiu et al. 2020), NS4B of all DENV serotypes (Kakumani et al. 2013), or NSP2 and NSP3 of CHIKV (Mathur et al. 2016). Flaviviruses are also known to produce subgenomic flavivirus RNAs (sfRNAs) that antagonize the antiviral response of mosquitoes (Schnettler et al. 2012, Moon et al. 2015, Göertz et al. 2019). In addition to being a major vector of arboviruses, A. aegypti is known for its large genome, which carries many TEs (∼47%) (Nene et al. 2007). The most abundant TEs are non-long terminal repeat (LTR) retrotransposons, followed by DNA transposons and LTR retrotransposons (Matthews et al. 2018). Some authors have proposed that this ability to vector viruses and this high TE content are linked with a particular piRNA pathway in Aedes mosquitoes. Indeed, A. aegypti appears to be an exception among arthropods for producing antiviral piRNAs (Hess et al. 2011; Morazzani et al. 2012; Miesen et al. 2015, 2016; Lewis et al. 2018). In addition, in the Aedes genus, there has been an expansion of PIWI genes. Indeed, Campbell et al. (2008) found eight proteins of the PIWI subfamily in A. aegypti with specific localizations and functions, compared to only three in Drosophila (supplementary fig. S1, Supplementary Material online). The authors propose that this expansion could be adaptive for the control of a larger TE burden. Miesen et al. (2015) identified four distinct groups of A. aegypti TE families based on the different PIWI proteins involved in their regulation. In addition, Varjak et al. (2017) showed that Piwi4 interacts with effectors of both the siRNA and piRNA pathways in A. aegypti, establishing a link between these pathways that has remained elusive in Drosophila. Moreover, the A. aegypti genome also contains endogenous viral elements (EVEs), which are remnants of ancient viral infections and behave as piRNA clusters (Whitfield et al. 2017).

For these reasons, we investigated the impacts of viral infections on TE control in A. aegypti. The aim of the present study was to test whether DENV, ZIKV, or CHIKV infection affects TE transcript levels and whether the impacts vary according to the virus and tissue. We exploited RNA-seq data already available in public databases. Our analyses show that these viral infections have only a modest effect on TE transcripts and that the direction of the modulations varies greatly across viruses, tissues, and TE superfamilies. The datasets also allowed us to test the effect of temperature on TE transcript levels upon viral infection and thus to determine the magnitude of the impact of these factors.

Results

TE Transcript Levels Are Modulated upon Arboviral Infection in A. aegypti

We analyzed 11 publicly available RNA-seq datasets, corresponding to 371 samples, which covered a wide variety of tissues (whole body, midgut, ovary, and cell culture), incubation times (from 3 h postinfection [hpi] to 14 d postinfection [dpi]), mosquito strains and serotypes for three viruses (ZIKV, DENV, and CHIKV) (Table 1). Across the different samples, the ratios of TE counts to gene counts were consistently very low, ranging from 0.01 to 0.05, except 0.11 for PRJNA545086 (ZIKV, whole bodies) (supplementary table S1, Supplementary Material online). For each modality, which corresponds to a single combination of viruses, tissues, time after infection, and additional factors, we computed the log2-fold change (lg2fc) of the normalized read counts attributed to TEs in the infected samples compared to those in the corresponding noninfected samples (mock) (supplementary fig. S2 to S16, Supplementary Material online). As an example, we show the results obtained upon DENV-2 infection in samples prepared from whole bodies at 18 hpi (Fig. 1). We detected a global decrease in TE transcript levels upon infection for this particular modality (mean lg2fc = −0.41, Wilcoxon paired test, V = 920,198, P = 1e−54). Separate analyses splitting TEs into three categories (DNA transposons, LTR retrotransposons, and non-LTR retrotransposons) revealed consistent patterns: mean lg2fc = −0.45 for DNA transposons (Wilcoxon paired test, V = 8,068, P = 6e−17), −0.41 for LTR retrotransposons (Wilcoxon paired test, V = 442,841, P = 1e−27), and −0.37 for non-LTR retrotransposons (Wilcoxon paired test, V = 39,807, P = 1e−14) (Fig. 1). This global decrease is, however, associated with a large dispersion of lg2fc values across TE families, especially for LTR and non-LTR retrotransposons.

TE transcript modulation upon DENV-2 infection in samples prepared from whole bodies at 18 hpi, PRJNA476553 (Kang et al. 2018). Log2 of the ratio (infected counts/mock counts) for each annotated TE family of DNA transposons (left panel), LTR retrotransposons (central panel) and non-LTR retrotransposons (right panel). P-values are for Wilcoxon paired tests. Dashed lines correspond to the mean of all TE families.
Fig. 1.

TE transcript modulation upon DENV-2 infection in samples prepared from whole bodies at 18 hpi, PRJNA476553 (Kang et al. 2018). Log2 of the ratio (infected counts/mock counts) for each annotated TE family of DNA transposons (left panel), LTR retrotransposons (central panel) and non-LTR retrotransposons (right panel). P-values are for Wilcoxon paired tests. Dashed lines correspond to the mean of all TE families.

Table 1

Summary of the analyzed datasets

VirusTissueTime post infectionMosquito strainViral strainNumber of analyzed samplesAccession numberReference
ZIKVMidgut1 or 2 dpiChiapas, Mexico; field samplingZIKV MEX1-4436PRJNA615972Ferreira et al. (2020)
2 dpiCaliforniaZIKV BR1511aPRJNA818687Louie et al. (2022)
7 dpiRockefellerZIKV FSS130253bPRJNA379149Angleró-Rodríguez et al. (2017)
Whole bodies10 dpiColony from AustraliaZIKV MR7669PRJNA545086Slonchak et al. (2020)
2, 7 and 14 dpiGalvestonZIKV Mex 1-718PRJNA399504Etebari et al. (2017)
DENVMidgut1 or 4 dpiWild type population from ThailandDENV-1 KDH0030A47PRJNA386455Raquin et al. (2017); Suzuki et al. (2017)
7 dpiRockefellerDENV-2 NGC6PRJNA379149Angleró-Rodríguez et al. (2017)
Ovaries3 dpiRockefellerDENV-2 NGC6PRJNA786000Feitosa-Suntheimer et al. (2022)
Whole bodies3 or 18 hpiTrinidad field isolateDENV-2 JAM140924PRJNA476553Kang et al. (2018)
CHIKVAag2 cells1 dpiAag2 cellsCHIKV IND-10-DEL1 E14PRJNA802350Dubey et al. (2022)
2 dpiAag2 cellsCHIKV LS312PRJNA885496Rosendo Machado et al. (2022)
Whole bodies3 or 7 dpiColony from AustraliaCHIKV Asian genotype70PRJNA630779Wimalasiri-Yapa et al. (2021)
VirusTissueTime post infectionMosquito strainViral strainNumber of analyzed samplesAccession numberReference
ZIKVMidgut1 or 2 dpiChiapas, Mexico; field samplingZIKV MEX1-4436PRJNA615972Ferreira et al. (2020)
2 dpiCaliforniaZIKV BR1511aPRJNA818687Louie et al. (2022)
7 dpiRockefellerZIKV FSS130253bPRJNA379149Angleró-Rodríguez et al. (2017)
Whole bodies10 dpiColony from AustraliaZIKV MR7669PRJNA545086Slonchak et al. (2020)
2, 7 and 14 dpiGalvestonZIKV Mex 1-718PRJNA399504Etebari et al. (2017)
DENVMidgut1 or 4 dpiWild type population from ThailandDENV-1 KDH0030A47PRJNA386455Raquin et al. (2017); Suzuki et al. (2017)
7 dpiRockefellerDENV-2 NGC6PRJNA379149Angleró-Rodríguez et al. (2017)
Ovaries3 dpiRockefellerDENV-2 NGC6PRJNA786000Feitosa-Suntheimer et al. (2022)
Whole bodies3 or 18 hpiTrinidad field isolateDENV-2 JAM140924PRJNA476553Kang et al. (2018)
CHIKVAag2 cells1 dpiAag2 cellsCHIKV IND-10-DEL1 E14PRJNA802350Dubey et al. (2022)
2 dpiAag2 cellsCHIKV LS312PRJNA885496Rosendo Machado et al. (2022)
Whole bodies3 or 7 dpiColony from AustraliaCHIKV Asian genotype70PRJNA630779Wimalasiri-Yapa et al. (2021)

dpi: days postinfection, hpi: hours postinfection.

aOnly antibiotic treatments during the pupal stage and blood-fed samples were analyzed.

bBoth DENV- and ZIKV-infected samples from Angleró-Rodriguez et al. (2017) shared the same mock condition samples, which are only indicated for DENV infection in the table.

Table 1

Summary of the analyzed datasets

VirusTissueTime post infectionMosquito strainViral strainNumber of analyzed samplesAccession numberReference
ZIKVMidgut1 or 2 dpiChiapas, Mexico; field samplingZIKV MEX1-4436PRJNA615972Ferreira et al. (2020)
2 dpiCaliforniaZIKV BR1511aPRJNA818687Louie et al. (2022)
7 dpiRockefellerZIKV FSS130253bPRJNA379149Angleró-Rodríguez et al. (2017)
Whole bodies10 dpiColony from AustraliaZIKV MR7669PRJNA545086Slonchak et al. (2020)
2, 7 and 14 dpiGalvestonZIKV Mex 1-718PRJNA399504Etebari et al. (2017)
DENVMidgut1 or 4 dpiWild type population from ThailandDENV-1 KDH0030A47PRJNA386455Raquin et al. (2017); Suzuki et al. (2017)
7 dpiRockefellerDENV-2 NGC6PRJNA379149Angleró-Rodríguez et al. (2017)
Ovaries3 dpiRockefellerDENV-2 NGC6PRJNA786000Feitosa-Suntheimer et al. (2022)
Whole bodies3 or 18 hpiTrinidad field isolateDENV-2 JAM140924PRJNA476553Kang et al. (2018)
CHIKVAag2 cells1 dpiAag2 cellsCHIKV IND-10-DEL1 E14PRJNA802350Dubey et al. (2022)
2 dpiAag2 cellsCHIKV LS312PRJNA885496Rosendo Machado et al. (2022)
Whole bodies3 or 7 dpiColony from AustraliaCHIKV Asian genotype70PRJNA630779Wimalasiri-Yapa et al. (2021)
VirusTissueTime post infectionMosquito strainViral strainNumber of analyzed samplesAccession numberReference
ZIKVMidgut1 or 2 dpiChiapas, Mexico; field samplingZIKV MEX1-4436PRJNA615972Ferreira et al. (2020)
2 dpiCaliforniaZIKV BR1511aPRJNA818687Louie et al. (2022)
7 dpiRockefellerZIKV FSS130253bPRJNA379149Angleró-Rodríguez et al. (2017)
Whole bodies10 dpiColony from AustraliaZIKV MR7669PRJNA545086Slonchak et al. (2020)
2, 7 and 14 dpiGalvestonZIKV Mex 1-718PRJNA399504Etebari et al. (2017)
DENVMidgut1 or 4 dpiWild type population from ThailandDENV-1 KDH0030A47PRJNA386455Raquin et al. (2017); Suzuki et al. (2017)
7 dpiRockefellerDENV-2 NGC6PRJNA379149Angleró-Rodríguez et al. (2017)
Ovaries3 dpiRockefellerDENV-2 NGC6PRJNA786000Feitosa-Suntheimer et al. (2022)
Whole bodies3 or 18 hpiTrinidad field isolateDENV-2 JAM140924PRJNA476553Kang et al. (2018)
CHIKVAag2 cells1 dpiAag2 cellsCHIKV IND-10-DEL1 E14PRJNA802350Dubey et al. (2022)
2 dpiAag2 cellsCHIKV LS312PRJNA885496Rosendo Machado et al. (2022)
Whole bodies3 or 7 dpiColony from AustraliaCHIKV Asian genotype70PRJNA630779Wimalasiri-Yapa et al. (2021)

dpi: days postinfection, hpi: hours postinfection.

aOnly antibiotic treatments during the pupal stage and blood-fed samples were analyzed.

bBoth DENV- and ZIKV-infected samples from Angleró-Rodriguez et al. (2017) shared the same mock condition samples, which are only indicated for DENV infection in the table.

We performed the same analyses for all considered datasets (Fig. 2). Wilcoxon paired tests revealed significant, global modulations of TE transcript amounts in most conditions (after Bonferroni correction for multiple tests), but in directions that varied according to the virus and tissue. The mean lg2fc ranged from −0.45 to 0.79 when all TEs were considered simultaneously. The clearest pattern was observed for DENV infections, which induced either no effect or a decrease in TE transcript amounts (mean lg2fc: from −0.41 to 0.06). The greatest increase in TE transcript levels was observed upon CHIKV infection in whole-body samples (mean lg2fc: from 0.24 to 0.79), while CHIKV infection in Aag2 cell cultures was associated with a decrease in TE transcript levels (mean lg2fc: from −0.45 to −0.05). Regarding ZIKV infections, the results showed greater contrast, and the directions of the modulations were highly variable across studies and samples (mean lg2fc: from −0.22 to 0.24) (Fig. 2).

Mean TE transcript modulation in all analyzed datasets. The different shades in the background allow to distinguish the different studies. The horizontal line indicates lg2fc = 0, i.e. absence of modulation. Stars at the top (*) indicate significant global TE transcript modulation (Wilcoxon paired tests, Bonferroni correction). Wh. bodies = whole bodies, Ov. = ovaries, Midg. = midgut, Aag2 c. = Aag2 cells, antib. = antibiotic treatment, sfRNA-def. = sfRNA-deficient ZIKV, stress = larval-stage crowding stress, Dhx15 = Dhx15 knockdown. 1. Ferreira et al. 2020. 2. Louie et al. 2022. 3. Angleró-Rodriguez et al. 2017. 4. Etebari et al. 2017. 5. Slonchak et al. 2020. 6. Feitosa-Suntheimer et al. 2022. 7. Suzuki et al. 2017; Raquin et al. 2017. 8. Kang et al. 2018. 9. Dubey et al. 2022. 10. Rosendo Machado et al. 2022. 11. Wimalasiri-Yapa et al. 2021.
Fig. 2.

Mean TE transcript modulation in all analyzed datasets. The different shades in the background allow to distinguish the different studies. The horizontal line indicates lg2fc = 0, i.e. absence of modulation. Stars at the top (*) indicate significant global TE transcript modulation (Wilcoxon paired tests, Bonferroni correction). Wh. bodies = whole bodies, Ov. = ovaries, Midg. = midgut, Aag2 c. = Aag2 cells, antib. = antibiotic treatment, sfRNA-def. = sfRNA-deficient ZIKV, stress = larval-stage crowding stress, Dhx15 = Dhx15 knockdown. 1. Ferreira et al. 2020. 2. Louie et al. 2022. 3. Angleró-Rodriguez et al. 2017. 4. Etebari et al. 2017. 5. Slonchak et al. 2020. 6. Feitosa-Suntheimer et al. 2022. 7. Suzuki et al. 2017; Raquin et al. 2017. 8. Kang et al. 2018. 9. Dubey et al. 2022. 10. Rosendo Machado et al. 2022. 11. Wimalasiri-Yapa et al. 2021.

TE Transcript Levels Are More Sensitive to Temperature Changes than to Arboviral Infections

Some RNA-seq datasets included samples corresponding to different temperatures (for ZIKV and CHIKV). In these different studies, RNA-seq data were obtained at optimal mosquito temperature (28 °C) or at cold (either 18 °C or 20 °C) or hot (either 32 °C or 36 °C) temperatures. Using an ANOVA approach on total TE counts, we found no interaction between temperature and infection status (supplementary table S2, Supplementary Material online). Temperature had a significant effect in all studies (two-way ANOVA P-values for temperature effect: ZIKV 1 dpi: 1.21e−04, 2 dpi: 9.97e−06, CHIKV 3 dpi: 1.21e−08, 7 dpi: 6.53e−05; Fig. 3 and supplementary table S2, Supplementary Material online). Cold temperatures were always found to be associated with the highest total TE counts. In contrast, these ANOVAs of total TE counts revealed a significant impact of the infection status in CHIKV samples only (two-way ANOVA P-values for infection effect: CHIKV 3 dpi: 5.06e−04, 7 dpi: 7.13e−05). The ratios of the effect sizes of temperature to infection status were always greater than 1, indicating that temperature had a much greater effect than infection status (effect size ratio: ZIKV 1 dpi: 5.65; ZIKV 2 dpi: 4.77; CHIKV 3 dpi: 2.64; and CHIKV 7 dpi: 1.37).

Effects of temperature and infection status on TE transcript levels. Sum of normalized TE counts. Red points correspond to means across replicates. Cold = 18 °C (CHIKV) or 20 °C (ZIKV), Optimal = 28 °C (CHIKV and ZIKV), Hot = 32 °C (CHIKV) or 36 °C (ZIKV). A) ZIKV infection, midgut, 1 dpi, B) 2 dpi (Ferreira et al. 2020), C) CHIKV infection, whole bodies, 3 dpi, and D) 7 dpi (Wimalasiri-Yapa et al. 2021).
Fig. 3.

Effects of temperature and infection status on TE transcript levels. Sum of normalized TE counts. Red points correspond to means across replicates. Cold = 18 °C (CHIKV) or 20 °C (ZIKV), Optimal = 28 °C (CHIKV and ZIKV), Hot = 32 °C (CHIKV) or 36 °C (ZIKV). A) ZIKV infection, midgut, 1 dpi, B) 2 dpi (Ferreira et al. 2020), C) CHIKV infection, whole bodies, 3 dpi, and D) 7 dpi (Wimalasiri-Yapa et al. 2021).

TE Responsiveness to Viral Infection Varies across Superfamilies

Although mosquito strains differed across datasets and TE landscapes are known to vary across natural populations, we investigated whether the patterns of differentially expressed (DE) TE families were consistent across viruses. Because small-RNA pathways target TE RNAs through sequence complementarity (Campbell et al. 2008; Palmer et al. 2018; Ozata et al. 2019), we assume that TE families belonging to the same superfamily are regulated by related mechanisms and thus share the same sensitivity to homoeostasis perturbations. Hence, we tested whether the different TE superfamilies displayed similar distributions of DE versus non-DE families upon infection across the different conditions. We detected significant differences among superfamilies for the three viruses (chi-squared tests, ZIKV infection: chi-squared = 210.65, simulated P < 4.8e−10; DENV infection: chi-squared = 93.49, simulated P = 6.0e−08; and CHIKV infection: chi-squared = 121.73, simulated P < 4.8e−10) (Fig. 4A–C). The largest contributions to the chi-squared statistics came from particularly infection-responsive superfamilies, i.e. more often DE than expected. Moreover, DNAt (superfamily), Loa, RTE, and Sola2 had more DE TE families than expected, whereas families from BEL superfamilies were less frequently DE than expected upon infection with all three viruses. Some superfamilies displayed contrasting patterns across viruses. For instance, Crack TE families were particularly insensitive to flavivirus infections (DENV and ZIKV). CR1 displayed an increased number of DE TE families upon CHIKV infection. TE families from Mariner/Tc1 were more often DE than expected upon flavivirus infection. The same was true for R1 upon DENV or CHIKV infection. In contrast, the numbers of DE TE families from Gypsy, I, and Tx1 followed random expectations in all conditions (Fig. 4A–C). In conclusion, it appears that there is a strong interaction between the TE superfamily and the virus in terms of responsiveness to infection.

Observed and expected numbers of differentially expressed TE families within each TE superfamily upon infection with A) ZIKV, B) DENV, and C) CHIKV. Total: Total number of observed TE families (DE and not DE); Obs: Observed number of DE TE families; Exp: Expected number of DE TE families under the homogeneity hypothesis; C: Contribution to chi-squared statistics under the null hypothesis of a homogeneous distribution of DE and non-DE TE families, i.e. (Obs−Exp)2/Exp; R: Relative deviation from homogeneity, i.e. (Obs−Exp)/Exp.
Fig. 4.

Observed and expected numbers of differentially expressed TE families within each TE superfamily upon infection with A) ZIKV, B) DENV, and C) CHIKV. Total: Total number of observed TE families (DE and not DE); Obs: Observed number of DE TE families; Exp: Expected number of DE TE families under the homogeneity hypothesis; C: Contribution to chi-squared statistics under the null hypothesis of a homogeneous distribution of DE and non-DE TE families, i.e. (Obs−Exp)2/Exp; R: Relative deviation from homogeneity, i.e. (Obs−Exp)/Exp.

As families could be up- or downmodulated upon infection in different studies, we further investigated the consistency of the response of TE superfamilies across the different modalities by plotting the mean of the mean lg2fc across the different modalities over the variation coefficient of the mean lg2fc across studies for each superfamily. This allowed us to spot superfamilies displaying a strong response in the same direction in all the modalities separately for each virus. Because some superfamilies were found only in a few samples, they could be associated with a small variation coefficient, as was the case for the P element (Fig. 5A). In order to display this bias, we adjusted the size of the dots according to the presence of the TE superfamily across datasets. The global TE superfamily modulation patterns differed across the three viruses, in agreement with our observations (Fig. 2). Considering the most represented TE superfamilies across datasets, Zator, Kolobok, and Harbinger were particularly upmodulated and L2B, Jockey, Outcast, Crack, and ISL2EU were the most consistently downmodulated superfamilies between modalities upon ZIKV infection (Fig. 5A). With regard to DENV infection, Helitron and ISL2EU were globally the most upmodulated TE superfamilies across datasets, whereas Crack, MITE, Outcast, or Zator were generally downmodulated (Fig. 5B). Finally, CryptonI, Nimb, Tx1, Helitron, and Kiri were mostly upmodulated upon CHIKV infection in all the datasets (Fig. 5C).

Mean of the TE superfamily mean lg2fc across the different modalities according to the variation coefficient. A) ZIKV, B) DENV, and C) CHIKV. The larger the dot is, the greater the number of families per superfamily. TE superfamilies most consistently responsive to infection across datasets are in bold.
Fig. 5.

Mean of the TE superfamily mean lg2fc across the different modalities according to the variation coefficient. A) ZIKV, B) DENV, and C) CHIKV. The larger the dot is, the greater the number of families per superfamily. TE superfamilies most consistently responsive to infection across datasets are in bold.

Discussion

TE Transcripts Are Modulated upon Viral Infection

Overall, the analyses that we performed on a collection of diverse publicly available RNA-seq datasets allowed us to uncover significant effects of arboviral infections on TE transcript levels in A. aegypti. We found that TE transcript levels were modulated upon arboviral infection in most studies. Nevertheless, the mean lg2fc values were lower than one, suggesting that the biological impacts of such modulations are expected to be mild, especially since TE read counts account for only a small fraction of the total gene counts (supplementary table S1, Supplementary Material online).

Although direct quantitative comparisons are difficult to perform, we recall that our previous studies of TE transcripts in Drosophila revealed mean values of −0.25 and −0.32 for lg2fc upon SINV infection in D. simulans and D. melanogaster, respectively (Roy et al. 2020), and 0.32 upon DMelSV infection in D. simulans (Roy et al. 2021). Apart from the CHIKV data, the lg2fc values that we measured here were generally lower, between −0.25 and 0.25 (Fig. 2), which suggests stronger TE transcript homeostasis upon viral infection in A. aegypti than in Drosophila. This could be related to the amplification of piRNA pathway genes in the mosquito genome (Campbell et al. 2008) and their involvement in both TE control and antiviral immunity (Morazzani et al. 2012; Lewis et al. 2018). Alternatively, it is possible that we did not detect any strong impact of arboviral infections on TE transcript amounts simply because arboviral infections globally have a modest impact on the mosquito transcriptome, which is consistent with the fact that arboviruses are known to have only a modest effect on mosquito fitness (Lambrechts and Scott 2009). This idea is reinforced by our analysis of the effect of temperature changes, which have greater impacts on the TE transcriptome than the arboviral infection status. Our study revealed contrasting TE transcript modulations and sensitivities depending on viral infection.

DENV Infection

The clearest pattern was observed for DENV infections; in most studies, DENV infection was associated with TE downmodulation (Fig. 2). Because DENV has tissue tropism for the midgut (Salazar et al. 2007; Angleró-Rodríguez et al. 2017), we expected stronger TE modulation in this tissue. However, this is not what we observed. This may be related to the recent observation that the siRNA pathway controls systemic DENV and ZIKV dissemination in mosquitoes but not in the midgut, although the pathway is fully functional against TEs in this tissue (Olmo et al. 2018). This global decrease in TE transcript levels upon DENV infection is similar to the pattern we observed in Drosophila upon SINV infection, for which we proposed that the antiviral response led to an increase in TE control (Roy et al. 2020). Indeed, the dsRNA uptake pathway was demonstrated to participate in the systemic spread of the RNAi antiviral response (Saleh et al. 2009), and we proposed that dsRNA molecules corresponding to TE sequences—resulting from antisense transcription or from inverted repeat pairing—hijack the process, leading to a concomitant increase in virus-derived siRNAs and TE-derived small RNAs. Interestingly, the study by Kang et al. (2018) revealed lower viral replication in individuals subjected to larval-stage crowding stress, which we found to be associated with a less pronounced decrease in TE transcript levels. This highlights the concomitant variation of the host antiviral response and TE control.

CHIKV Infection

With regard to CHIKV infections, TE transcripts were globally downmodulated in somatic Aag2 cells but clearly upmodulated in whole-body samples, with the highest observed mean lg2fc of all datasets. This discrepancy between the two studies is difficult to explain. Cell cultures are known to be subject to frequent genome rearrangements (Lee et al. 2014), which can have impacts on TE landscapes. CHIKV data in whole bodies all come from the same study, and results from additional studies are needed to further confirm our conclusion. However, these findings are in line with what we previously observed in Drosophila upon DMelSV infection: we observed a global increase in TE transcript levels in fly carcasses. The underlying molecular mechanisms are not understood, but a scenario involving a trade-off between antiviral immunity and TE control may be proposed.

ZIKV Infection

ZIKV infection led to more variable patterns across datasets, and we observed that TE transcript levels decreased as well as increased, depending on the study. Thus far, it has been very difficult to identify the factors determining the direction of the modulation. Moreover, the different viral strains used to infect mosquitoes all originated from the Asian lineage (Faria et al. 2016; Lanciotti et al. 2016). In comparative studies, strains from the Asian lineage presented more similar infection characteristics than strains from the African lineage (Roundy et al. 2017; Ou et al. 2021). Thus, even if mosquitoes were infected by different ZIKV strains, it cannot be used as a strong argument to explain the variability of the modulation observed (Fig. 2).

Some TE Superfamilies Are Particularly Infection Responsive

We analyzed in detail the patterns of TE modulation upon viral infection at the superfamily level. Indeed, families belonging to the same superfamily display some degree of structural and sequence similarity, enabling common regulatory mechanisms to some extent. Across the different studies, we found that the TE superfamilies DNAt, Loa, RTE, and Sola2 were the most often impacted by the three viral infections compared to other superfamilies, whereas BEL was very insensitive to arboviral infections (Fig. 4A–C). In addition, our results showed that some superfamilies displayed particularly homogeneous modulations across all the corresponding families in different studies of the same virus, as is the case for Helitron upon DENV or CHIKV infection or L2B upon ZIKV infection (Fig. 5A–C). However, we cannot exclude the possibility that this is due to the lower number of families in these superfamilies. Indeed, a large number of families increases the probability of detecting heterogeneity across study modalities.

Variations Across Studies Are Explained by Various Biotic Factors

Similar to what we described in Drosophila, our results clearly show that the direction and extent of TE transcript modulation depend on the virus. Lambrechts and Scott (2009) showed that Alphaviruses have a greater impact on mosquito fitness than other viral genera, such as Flaviviruses. Consistent with this, in the present analysis, we found that the Alphavirus (CHIKV) had the strongest impact on TE transcript modulation. Finally, the study by Angleró-Rodriguez et al. (2017) allows to compare the impacts of DENV-2 and ZIKV on TEs using the same Rockefeller mosquito strain, and the same infection protocol at 7 dpi. While their data did not allow us to detect any significant effect on TEs upon DENV infection, we observed an upmodulation upon ZIKV infection. Even though ZIKV and DENV are Flaviviruses with shared features, Williams et al. (2020) showed that A. aegypti is less resistant to ZIKV infection than to DENV-2 in terms of viral suppression effectiveness, possibly because ZIKV is less susceptible to the siRNA pathway. We assume that this lower resistance to the virus can be reflected in various host responses, such as stronger modulation of TE transcripts.

As described above, the tissue under consideration may be relevant. Indeed, (i) depending on viral tropism, viral replication is not homogeneous in the mosquito body, and thus, the interaction with TE control, if any, is expected to vary, and (ii) the expansion of the PIWI genes in A. aegypti, as previously mentioned, is accompanied by both spatial and functional specializations of the different genes in the mosquito body, e.g. TE control, antiviral immunity or both. Piwi 1 to 3 are specific to the germline, Piwi 4 to 6 and Ago3 are specific to adult somatic tissues, and Piwi 7 is localized in early embryos (Akbari et al. 2013; Miesen et al. 2015) (supplementary fig. S1, Supplementary Material online). Thus, according to the tissue and function of the Piwi protein, the interaction between TE control and antiviral immunity, if any, is expected to differ. As mentioned above, flaviviruses are known to produce sfRNAs, which facilitate infection, suppress antiviral RNAi, and are required for efficient viral transmission (Schnettler et al. 2012; Moon et al. 2015; Göertz et al. 2019; Slonchak et al. 2020). Thus, it is possible that upon DENV and ZIKV infection, TE-derived piRNAs and siRNAs are also impacted by sfRNAs, which subsequently induces changes in TE transcripts. In line with this idea, the results obtained in whole-body samples at 10 dpi by mutated or not ZIKV for sfRNA production show that sfRNAs could have an impact on TE transcript regulation because when the virus cannot produce sfRNAs, the downmodulation of TE transcripts is milder (Fig. 2) (Slonchak et al. 2020).

Limitations

We have to acknowledge that the present approach suffers from several limitations. In particular, while the analysis of publicly available datasets allows the sensible use of research efforts and the exploration of a wealth of data, the corresponding experimental setups may not be perfectly suited to all research questions. Most notably, cross-study comparisons are not straightforward since many factors can vary or lack control. Of major impact, mosquito genotypes and virus strains are known to determine the outcome of arboviral infections (Lambrechts 2011; Lambrechts et al. 2013; Lequime et al. 2016; Souza-Neto et al. 2019; Ekwudu et al. 2020). In addition, considering that we found that the different TE superfamilies are not evenly sensitive to viral infections and that the TE content is known to vary across natural populations (Vieira et al. 1999, 2002; Goubert et al. 2017; Mérel et al. 2021), it is expected that different mosquito genotypes will display different modulations of their TE transcriptomes depending on the TE composition of their genomes. The tissue under study is also of fundamental importance since viral replication does not occur homogeneously within the whole body; therefore, the molecular interactions with TEs may differ. Moreover, some studies have been performed in cultures of cells, whose genomes are known to suffer frequent rearrangements (Lee et al. 2014), which may have an impact on TE distribution (Potter et al. 1979; Ilyin et al. 1980). In addition, the infection status with regard to bacterial or chronic viral infections is not known in most studies, but it was shown that they could interfere with the antiviral response (Ramirez et al. 2012; Parry and Asgari 2018; Souza-Neto et al. 2019; Olmo et al. 2023). Wolbachia bacteria are known to reduce the replication of many viruses by, e.g. (i) modifying gene regulation (Zhang et al. 2013) or (ii) increasing the level of piRNAs and Ago3 transcripts (Mayoral et al. 2014). Current evidence suggests that A. aegypti does not harbor Wolbachia naturally (Ross et al. 2020); however, the bacteria invaded some A. aegypti populations after release in the wild of artificially infected mosquitoes (Moreira et al. 2009; Hoffmann et al. 2011, 2014; Walker et al. 2011; Ahmad et al. 2021). The infection status of the populations under study is not known, and even though it is highly unlikely, some of them may have been affected because certain releases were carried out before mosquito sampling and in nearby geographical areas, as is the case for Queensland in Australia (PRJNA630779 in Wimalasiri-Yapa et al. (2021) and PRJNA545086 in Slonchak et al. (2020)). Other parameters, such as the route of infection (Mondotte and Saleh 2018) or the viral dose, may also impact the outcome of the infection and therefore the potential effect on the TE transcriptome. However, despite these limitations, the large number of analyzed samples allowed us to identify general trends in the interaction between arboviral infections and TE control.

Conclusion

The results of our analyses show that TE transcript levels are modulated upon arboviral infection in A. aegypti. However, we revealed that the direction of the modulation depends on the virus and potentially on some unidentified factors. Overall, the extent of the modulations that we found appears to be particularly low, raising doubts about their biological impacts. However, further studies are needed to evaluate this phenomenon. Nevertheless, even subtle changes in TE transcript levels may result in differences in TE transposition rates, and, therefore, in somatic genetic diversity (Faulkner and Garcia-Perez 2017; Chang et al. 2019), or even in genome mutation rates if the ovarian transposition rate is affected.

Moreover, it would be particularly interesting to test whether TEs have an impact on viral replication in mosquitoes. This idea is suggested by recent research that revealed the role of EVEs in mosquitoes. These are remnants of ancient viral infections, which are acquired through recombination with LTR retrotransposons, behave as piRNA clusters, and participate in viral immunity against exogenous viruses (Whitfield et al. 2017). EVEs are known to vary across mosquito populations and are proposed to explain differences in vector competence (Suzuki et al. 2017; Whitfield et al. 2017). The obvious similarities between EVEs and TEs suggest that this is a relevant direction for future research.

Materials and Methods

Datasets

We retrieved publicly available RNA-seq datasets corresponding to A. aegypti samples infected with DENV, CHIKV, or ZIKV and their corresponding noninfected samples using the prefetch and fastq-dump tools of the sra-toolkit (http://github.com/ncbi/sra-tools/wiki/01.Downloading-SRA-Toolkit). All datasets were produced following Illumina procedures. We only retained datasets that displayed at least three biological replicates. Sequencing quality was assessed using the fastqc-0.11.9 tool (www.bioinformatics.babraham.ac.uk/projects/fastqc/), and poor-quality samples were removed (SRR17843159 and SRR17843156 in PRJNA802350). When the datasets had technical replicates, they were concatenated into a single fastq file. SRR18435178 (PRJNA818687), SRR11440744 (PRJNA615972), and SRR6818585 and SRR6820666 (PRJNA386455) were also removed because they were potentially corrupted.

We used version 5.0 of the A. aegypti reference genome (AaegL5.0) obtained by long-read sequencing via PacBio technology (Matthews et al. 2018). This reference genome corresponds to the LVP_AGWG strain of A. aegypti. After masking TEs using RepeatMasker-4.1.2-p1 (http://www.repeatmasker.org) to remove them from genes, we retrieved the gff file corresponding to gene models in AaegL5.0 (GCF_002204515.2_AaegL5.0_genomic.gff.gz) and used getfasta-2.30.0 from bedtools (Quinlan and Hall 2010) to create a multifasta file of gene sequences devoid of TEs. For the analysis of TEs, we considered A. aegypti sequences retrieved from the Repbase-25.08 database of eukaryotic repetitive elements (Jurka et al. 2005), corresponding to 3,013 TE families classified into 40 superfamilies. We formatted these sequences to analyze the TE data using the TEcount module of TEtools (Lerat et al. 2017).

RNA-seq Analysis

Sequencing adaptors were removed using Trimmomatic version 0.39 (Bolger et al. 2014). Alignments against the masked multifasta of reference genes and gene counts were performed using kallisto-0.48.0 (Bray et al. 2016). TE transcript counts were obtained using the TEcount module of TEtools (Lerat et al. 2017) and the list of TE sequences described above.

Gene and TE count tables were concatenated and further analyzed using the R package DESeq2-1.34.0 (Love et al. 2014). For each study and tissue, normalization was done globally on the concatenated table using the DESeq function. We considered a minimum threshold of ten normalized counts for TEs in both infected and mock conditions. Global TE transcript modulation was assessed by Wilcoxon paired tests at the TE family or superfamily levels across mean counts in infected and control conditions. Bonferroni correction was applied across the different datasets.

The effect of temperature on the log-transformed sums of the normalized TE counts of all TE families was assessed using two-way ANOVA, and the factors temperature, infection status, and temperature × infection status interaction were considered. The assumptions of normality and homogeneity were met (Shapiro, Bartlett, and Levene tests), except for samples corresponding to CHIKV infection in whole bodies at 3 dpi. The effect size of temperature was determined by averaging the means of both the control and infected conditions for each temperature, followed by computing the difference between the two most extreme means. Similarly, the effect size of infection was assessed across all three temperatures by computing the difference between the averages of all means from the infected condition on one side and all means from the mock condition on the other side.

TE responsiveness to viral infection was achieved after extracting TE families DE or not between infected and mock conditions from DESeq tables. A TE family was considered non-DE if it had an adjusted P > 0.05 across all modalities in the DESeq2 analysis; it was considered DE if it showed an adjusted P < 0.05 in at least one modality. Chi-squared tests for homogeneity with Monte Carlo simulations for the P-value performing 2.1e + 09 iterations were used to assess whether the proportions of DE and non-DE families were the same in all the superfamilies.

The consistency of TE superfamily responses across the different study modalities was further analyzed by computing the mean lg2fc for each TE superfamily expressed in each modality after aggregating the TE family lg2fc. Only families displaying at least ten counts in one condition (infected or mock) were kept for analysis. Variation coefficients were computed from these means for each study modality. The global mean lg2fc for each TE superfamily across all the modalities was also computed. Thus, high magnitude (either positive or negative) for mean lg2fc and a low variation coefficient of lg2fc corresponded to TE superfamilies displaying a strong response in the same direction across all the modalities.

Supplementary Material

Supplementary material is available at Genome Biology and Evolution online.

Acknowledgments

This work was performed using the computing facilities of Laboratoire de Biométrie Biologie Évolutive/Pôle Rhône-Alpes de Bioinformatique (CC LBBE/PRABI). We thank Philippe Veber, Natacha Kremer, Séverine Chambeyron, Guillaume Minard, François Sabot, Olivier Terrier, Alicia Reyes Ramirez, Cristina Vieira, William Vilas Boas Nunes, Camille Mayeux, Daniel Siqueira de Oliveira, Miriam Merenciano, and Anaïs Larue for helpful discussions. We also thank Clément Goubert for providing technical assistance. We thank the editor for her positive feedback that helped us improve the manuscript.

Funding

This work was supported by Agence Nationale de la Recherche (ANR) LongevitY ANR-20-CE02-0015, MosquiTEs ANR-21-CE02-0013, and TEMIT ANR-19-CE02-0022 grants and Universite Claude Bernard Lyon1.

Data Availability

There are no new data associated with this article. The scripts used for the analyses are available in Supplementary material.

Literature Cited

Ahmad
 
NA
,
Mancini
 
M-V
,
Ant
 
TH
,
Martinez
 
J
,
Kamarul
 
GMR
,
Nazni
 
WA
,
Hoffmann
 
AA
,
Sinkins
 
SP
.
Wolbachia strain w AlbB maintains high density and dengue inhibition following introduction into a field population of Aedes aegypti
.
Philos Trans R Soc Lond B Biol Sci
.
2021
:
376
(
1818
):
20190809
. https://doi.org/10.1098/rstb.2019.0809.

Akbari
 
OS
,
Antoshechkin
 
I
,
Amrhein
 
H
,
Williams
 
B
,
Diloreto
 
R
,
Sandler
 
J
,
Hay
 
BA
.
The developmental transcriptome of the mosquito Aedes aegypti, an invasive species and major arbovirus vector
.
G3 (Bethesda)
.
2013
:
3
(
9
):
1493
1509
. https://doi.org/10.1534/g3.113.006742.

Angleró-Rodríguez
 
YI
,
MacLeod
 
HJ
,
Kang
 
S
,
Carlson
 
JS
,
Jupatanakul
 
N
,
Dimopoulos
 
G
.
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
. https://doi.org/10.3389/fmicb.2017.02050.

Biémont
 
C
.
A brief history of the status of transposable elements: from junk DNA to major players in evolution
.
Genetics
.
2010
:
186
(
4
):
1085
1093
. https://doi.org/10.1534/genetics.110.124180.

Bolger
 
AM
,
Lohse
 
M
,
Usadel
 
B
.
Trimmomatic: a flexible trimmer for illumina sequence data
.
Bioinformatics
.
2014
:
30
(
15
):
2114
2120
. https://doi.org/10.1093/bioinformatics/btu170.

Bray
 
NL
,
Pimentel
 
H
,
Melsted
 
P
,
Pachter
 
L
.
Near-optimal probabilistic RNA-Seq quantification
.
Nat Biotechnol
.
2016
:
34
(
5
):
525
527
. https://doi.org/10.1038/nbt.3519.

Bronkhorst
 
AW
,
van Rij
 
RP
.
The long and short of antiviral defense: small RNA-based immunity in insects
.
Curr Opin Virol.
 
2014
:
7
:
19
28
. https://doi.org/10.1016/j.coviro.2014.03.010.

Campbell
 
CL
,
Black
 
WC
,
Hess
 
AM
,
Foy
 
BD
.
Comparative genomics of small RNA regulatory pathway components in vector mosquitoes
.
BMC Genomics
.
2008
:
9
:
425
. https://doi.org/10.1186/1471-2164-9-425.

Casacuberta
 
E
,
González
 
J
.
The impact of transposable elements in environmental adaptation
.
Mol Ecol
.
2013
:
22
(
6
):
1503
1517
. https://doi.org/10.1111/mec.12170.

Chambers
 
TJ
,
Hahn
 
CS
,
Galler
 
R
,
Rice
 
CM
.
Flavivirus genome organization, expression, and replication
.
Annu Rev Microbiol.
 
1990
:
44
:
649
688
. https://doi.org/10.1146/annurev.mi.44.100190.003245.

Chang
 
Y-H
,
Keegan
 
RM
,
Prazak
 
L
,
Dubnau
 
J
.
Cellular labeling of endogenous retrovirus replication (CLEVR) reveals de novo insertions of the gypsy retrotransposable element in cell culture and in both neurons and glial cells of aging fruit flies
.
PLoS Biol
.
2019
:
17
(
5
):
e3000278
. https://doi.org/10.1371/journal.pbio.3000278.

Dubey
 
SK
,
Mehta
 
D
,
Chaudhary
 
S
,
Hasan
 
A
,
Sunil
 
S
.
An E3 ubiquitin ligase scaffolding protein is proviral during chikungunya virus infection in Aedes aegypti
.
Microbiol Spectr
.
2022
:
10
(
3
):
e00595-22
. https://doi.org/10.1128/spectrum.00595-22.

Durdevic
 
Z
,
Mobin
 
MB
,
Hanna
 
K
,
Lyko
 
F
,
Schaefer
 
M
.
The RNA methyltransferase Dnmt2 is required for efficient Dicer-2-dependent siRNA pathway activity in Drosophila
.
Cell Rep.
 
2013
:
4
(
5
):
931
937
. https://doi.org/10.1016/j.celrep.2013.07.046.

Ekwudu
 
O
,
Marquart
 
L
,
Webb
 
L
,
Lowry
 
KS
,
Devine
 
GJ
,
Hugo
 
LE
,
Frentiu
 
FD
.
Effect of serotype and strain diversity on dengue virus replication in Australian mosquito vectors
.
Pathogens
.
2020
:
9
(
8
):
668
. https://doi.org/10.3390/pathogens9080668.

Elsik
 
CG
,
Worley
 
KC
,
Bennett
 
AK
,
Beye
 
M
,
Camara
 
F
,
Childers
 
CP
,
de Graaf
 
DC
,
Debyser
 
G
,
Deng
 
J
,
Devreese
 
B
, et al.  
Finding the missing honey bee genes: lessons learned from a genome upgrade
.
BMC Genomics
.
2014
:
15
:
86
. https://doi.org/10.1186/1471-2164-15-86.

Etebari
 
K
,
Hegde
 
S
,
Saldaña
 
MA
,
Widen
 
SG
,
Wood
 
TG
,
Asgari
 
S
,
Hughes
 
GL
.
Global transcriptome analysis of Aedes aegypti mosquitoes in response to Zika virus infection
.
mSphere
.
2017
:
2
:
e00456-17
. https://doi.org/10.1128/mSphere.00456-17.

Faria
 
NR
,
Azevedo
 
RDSDS
,
Kraemer
 
MUG
,
Souza
 
R
,
Cunha
 
MS
,
Hill
 
SC
,
Thézé
 
J
,
Bonsall
 
MB
,
Bowden
 
TA
,
Rissanen
 
I
, et al.  
Zika virus in the Americas: early epidemiological and genetic findings
.
Science
.
2016
:
352
(
6283
):
345
349
. https://doi.org/10.1126/science.aaf5036.

Faulkner
 
GJ
,
Garcia-Perez
 
JL
.
L1 mosaicism in mammals: extent, effects, and evolution
.
Trends Genet.
 
2017
:
33
(
11
):
802
816
. https://doi.org/10.1016/j.tig.2017.07.004.

Feitosa-Suntheimer
 
F
,
Zhu
 
Z
,
Mameli
 
E
,
Dayama
 
G
,
Gold
 
AS.
,
Broos-Caldwell
 
A
,
Troupin
 
A
,
Rippee-Brooks
 
M
,
Corley
 
RB
,
Lau
 
NC
, et al.  
Dengue virus-2 infection affects fecundity and elicits specific transcriptional changes in the ovaries of Aedes aegypti mosquitoes
.
Front Microbiol.
 
2022
:
13
:
886787
. https://doi.org/10.3389/fmicb.2022.886787.

Ferreira
 
PG
,
Tesla
 
B
,
Horácio
 
ECA
,
Nahum
 
LA
,
Brindley
 
MA
,
de Oliveira Mendes
 
TA
,
Murdock
 
CC
.
Temperature dramatically shapes mosquito gene expression with consequences for mosquito–Zika virus interactions
.
Front Microbiol.
 
2020
:
11
:
901
. https://doi.org/10.3389/fmicb.2020.00901.

Galbraith
 
JD
,
Hayward
 
A
.
The influence of transposable elements on animal colouration
.
Trends Genet.
 
2023
:
39
(
8
):
624
638
. https://doi.org/10.1016/j.tig.2023.04.005.

Gaunt
 
MW
,
Miles
 
MA
.
An insect molecular clock dates the origin of the insects and accords with palaeontological and biogeographic landmarks
.
Mol Biol Evol.
 
2002
:
19
(
5
):
748
761
. https://doi.org/10.1093/oxfordjournals.molbev.a004133.

Gilbert
 
C
,
Peccoud
 
J
,
Cordaux
 
R
.
Transposable elements and the evolution of insects
.
Annu Rev Entomol.
 
2021
:
66
:
355
372
. https://doi.org/10.1146/annurev-ento-070720-074650.

Göertz
 
GP
,
van Bree
 
JWM
,
Hiralal
 
A
,
Fernhout
 
BM
,
Steffens
 
C
,
Boeren
 
S
,
Visser
 
TM
,
Vogels
 
CBF
,
Abbo
 
SR
,
Fros
 
JJ
, et al.  
Subgenomic flavivirus RNA binds the mosquito DEAD/H-box helicase ME31B and determines Zika virus transmission by Aedes aegypti
.
Proc Natl Acad Sci USA.
 
2019
:
116
(
38
):
19136
19144
. https://doi.org/10.1073/pnas.1905617116.

Goodier
 
JL
,
Kazazian
 
HH
.
Retrotransposons revisited: the restraint and rehabilitation of parasites
.
Cell
.
2008
:
135
(
1
):
23
35
. https://doi.org/10.1016/j.cell.2008.09.022.

Goubert
 
C
,
Henri
 
H
,
Minard
 
G
,
Valiente Moro
 
C
,
Mavingui
 
P
,
Vieira
 
C
,
Boulesteix
 
M
.
High-throughput sequencing of transposable element insertions suggests adaptive evolution of the invasive Asian tiger mosquito towards temperate environments
.
Mol Ecol
.
2017
:
26
(
15
):
3968
3981
. https://doi.org/10.1111/mec.14184.

Hess
 
AM
,
Prasad
 
AN
,
Ptitsyn
 
A
,
Ebel
 
GD
,
Olson
 
KE
,
Barbacioru
 
C
,
Monighetti
 
C
,
Campbell
 
CL
.
Small RNA profiling of dengue virus-mosquito interactions implicates the PIWI RNA pathway in anti-viral defense
.
BMC Microbiol
.
2011
:
11
:
45
. https://doi.org/10.1186/1471-2180-11-45.

Hoffmann
 
AA
,
Iturbe-Ormaetxe
 
I
,
Callahan
 
AG
,
Phillips
 
BL
,
Billington
 
K
,
Axford
 
JK
,
Montgomery
 
B
,
Turley
 
AP
,
O'Neill
 
SL
.
Stability of the wMel Wolbachia infection following invasion into Aedes aegypti populations
.
PLoS Negl Trop Dis
.
2014
:
8
(
9
):
e3115
. https://doi.org/10.1371/journal.pntd.0003115.

Hoffmann
 
AA
,
Montgomery
 
BL
,
Popovici
 
J
,
Iturbe-Ormaetxe
 
I
,
Johnson
 
PH
,
Muzzi
 
F
,
Greenfield
 
M
,
Durkan
 
M
,
Leong
 
YS
,
Dong
 
Y
, et al.  
Successful establishment of Wolbachia in Aedes populations to suppress dengue transmission
.
Nature
.
2011
:
476
(
7361
):
454
457
. https://doi.org/10.1038/nature10356.

Honeybee Genome Sequencing Consortium
.
Insights into social insects from the genome of the honeybee Apis mellifera
.
Nature
.
2006
:
443
(
7114
):
931
949
. https://doi.org/10.1038/nature05260.

Ilyin
 
YV
,
Chmeliauskaite
 
VG
,
Ananiev
 
EV
,
Georgiev
 
GP
.
Isolation and characterization of a new family of mobile dispersed genetic elements, mdg3, in Drosophila melanogaster
.
Chromosoma
.
1980
:
81
:
27
53
. https://doi.org/10.1007/BF00292421.

Jurka
 
J
,
Kapitonov
 
VV
,
Pavlicek
 
A
,
Klonowski
 
P
,
Kohany
 
O
,
Walichiewicz
 
J
.
Repbase update, a database of eukaryotic repetitive elements
.
Cytogenet Genome Res
.
2005
:
110
(
1-4
):
462
467
. https://doi.org/10.1159/000084979.

Kakumani
 
PK
,
Ponia
 
SS
,
S
 
RK
,
Sood
 
V
,
Chinnappan
 
M
,
Banerjea
 
AC
,
Medigeshi
 
GR
,
Malhotra
 
P
,
Mukherjee
 
SK
,
Bhatnagar
 
RK
.
Role of RNA interference (RNAi) in dengue virus replication and identification of NS4B as an RNAi suppressor
.
J Virol
.
2013
:
87
(
16
):
8870
8883
. https://doi.org/10.1128/JVI.02774-12.

Kang
 
DS
,
Barron
 
MS
,
Lovin
 
DD
,
Cunningham
 
JM
,
Eng
 
MW
,
Chadee
 
DD
,
Li
 
J
,
Severson
 
DW
.
A transcriptomic survey of the impact of environmental stress on response to dengue virus in the mosquito, Aedes aegypti
.
PLoS Negl Trop Dis
.
2018
:
12
(
6
):
e0006568
. https://doi.org/10.1371/journal.pntd.0006568.

Kuno
 
G
,
Chang
 
G-JJ
.
Full-length sequencing and genomic characterization of Bagaza, Kedougou, and Zika viruses
.
Arch Virol
.
2007
:
152
(
4
):
687
696
. https://doi.org/10.1007/s00705-006-0903-z.

Lambrechts
 
L
.
Quantitative genetics of Aedes aegypti vector competence for dengue viruses: towards a new paradigm?
 
Trends Parasitol.
 
2011
:
27
(
3
):
111
114
. https://doi.org/10.1016/j.pt.2010.12.001.

Lambrechts
 
L
,
Quillery
 
E
,
Noël
 
V
,
Richardson
 
JH
,
Jarman
 
RG
,
Scott
 
TW
,
Chevillon
 
C
.
Specificity of resistance to dengue virus isolates is associated with genotypes of the mosquito antiviral gene Dicer-2
.
Proc R Soc B
.
2013
:
280
(
1751
):
20122437
. https://doi.org/10.1098/rspb.2012.2437.

Lambrechts
 
L
,
Scott
 
TW
.
Mode of transmission and the evolution of arbovirus virulence in mosquito vectors
.
Proc R Soc B
.
2009
:
276
(
1660
):
1369
1378
. https://doi.org/10.1098/rspb.2008.1709.

Lanciotti
 
RS
,
Lambert
 
AJ
,
Holodniy
 
M
,
Saavedra
 
S
,
Signor
 
LDCC
.
Phylogeny of Zika virus in western hemisphere, 2015
.
Emerg Infect Dis
.
2016
:
22
(
5
):
933
935
. https://doi.org/10.3201/eid2205.160065.

Lawson
 
HA
,
Liang
 
Y
,
Wang
 
T
.
Transposable elements in mammalian chromatin organization
.
Nat Rev Genet
.
2023
:
24
(
10
):
712
723
. https://doi.org/10.1038/s41576-023-00609-6.

Lee
 
H
,
McManus
 
CJ
,
Cho
 
D-Y
,
Eaton
 
M
,
Renda
 
F
,
Somma
 
MP
,
Cherbas
 
L
,
May
 
G
,
Powell
 
S
,
Zhang
 
D
, et al.  
DNA copy number evolution in Drosophila cell lines
.
Genome Biol
.
2014
:
15
(
8
):
R70
. https://doi.org/10.1186/gb-2014-15-8-r70.

Lequime
 
S
,
Fontaine
 
A
,
Ar Gouilh
 
M
,
Moltini-Conclois
 
I
,
Lambrechts
 
L.
 
2016
.
Genetic drift, purifying selection and vector genotype shape dengue virus intra-host genetic diversity in mosquitoes
.
PLoS Genet
.
12
(
6
):
e1006111
. https://doi.org/10.1371/journal.pgen.1006111.

Lerat
 
E
,
Fablet
 
M
,
Modolo
 
L
,
Lopez-Maestre
 
H
,
Vieira
 
C
.
TEtools facilitates big data expression analysis of transposable elements and reveals an antagonism between their activity and that of piRNA genes
.
Nucleic Acids Res
.
2017
:
45
(
4
):
e17
. https://doi.org/10.1093/nar/gkw953.

Lewis
 
SH
,
Quarles
 
KA
,
Yang
 
Y
,
Tanguy
 
M
,
Frézal
 
L
,
Smith
 
SA
,
Sharma
 
PP
,
Cordaux
 
R
,
Gilbert
 
C
,
Giraud
 
I
, et al.  
Pan-arthropod analysis reveals somatic piRNAs as an ancestral defence against transposable elements
.
Nat Ecol Evol
.
2018
:
2
(
1
):
174
181
. https://doi.org/10.1038/s41559-017-0403-4.

Louie
 
W
,
Ramírez
 
AL
,
Mack
 
LK
,
Kelly
 
ET
,
Attardo
 
GM
,
Coffey
 
LL.
Microbes reduce susceptibility of Aedes aegypti to Zika virus by enhancing blood digestion and limiting midgut cell infection. bioRxiv 2022.11.10.516021. https://doi.org/10.1101/2022.11.10.516021, 10 November 2022, preprint: not peer reviewed.

Love
 
MI
,
Huber
 
W
,
Anders
 
S
.
Moderated estimation of fold change and dispersion for RNA-Seq data with DESeq2
.
Genome Biol
.
2014
:
15
:
550
. https://doi.org/10.1186/s13059-014-0550-8.

Markoff
 
L
.
5′- and 3′-noncoding regions in flavivirus RNA
.
Adv Virus Res
.
2003
:
59
:
177
228
. https://doi.org/10.1016/S0065-3527(03)59006-6.

Mathur
 
K
,
Anand
 
A
,
Dubey
 
SK
,
Sanan-Mishra
 
N
,
Bhatnagar
 
RK
,
Sunil
 
S
.
Analysis of chikungunya virus proteins reveals that non-structural proteins nsP2 and nsP3 exhibit RNA interference (RNAi) suppressor activity
.
Sci Rep
.
2016
:
6
:
38065
. https://doi.org/10.1038/srep38065.

Matthews
 
BJ
,
Dudchenko
 
O
,
Kingan
 
SB
,
Koren
 
S
,
Antoshechkin
 
I
,
Crawford
 
JE
,
Glassford
 
WJ
,
Herre
 
M
,
Redmond
 
SN.
,
Rose
 
NH
, et al.  
Improved reference genome of Aedes aegypti informs arbovirus vector control
.
Nature
.
2018
:
563
(
7732
):
501
507
. https://doi.org/10.1038/s41586-018-0692-z.

Mayoral
 
JG
,
Hussain
 
M
,
Joubert
 
DA
,
Iturbe-Ormaetxe
 
I
,
O’Neill
 
SL
,
Asgari
 
S
.
Wolbachia small noncoding RNAs and their role in cross-kingdom communications
.
Proc Natl Acad Sci USA.
 
2014
:
111
(
52
):
18721
18726
. https://doi.org/10.1073/pnas.1420131112.

Mérel
 
V
,
Gibert
 
P
,
Buch
 
I
,
Rodriguez Rada
 
V
,
Estoup
 
A
,
Gautier
 
M
,
Fablet
 
M
,
Boulesteix
 
M
,
Vieira
 
C
.
The worldwide invasion of Drosophila suzukii is accompanied by a large increase of transposable element load and a small number of putatively adaptive insertions
.
Mol Biol Evol.
 
2021
:
38
(
10
):
4252
4267
. https://doi.org/10.1093/molbev/msab155.

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
(13
):
6545
6556
. https://doi.org/10.1093/nar/gkv590.

Miesen
 
P
,
Joosten
 
J
,
van Rij
 
RP
.
PIWIs go viral: arbovirus-derived piRNAs in vector mosquitoes
.
PLoS Pathog
.
2016
:
12
(
12
):
e1006017
. https://doi.org/10.1371/journal.ppat.1006017.

Mondotte
 
JA
,
Saleh
 
M-C.
 
2018
.
Antiviral immune response and the route of infection in Drosophila melanogaster
.
Adv. Virus Res
.
100
:
247
278
. https://doi.org/10.1016/bs.aivir.2017.10.006.

Moon
 
SL
,
Dodd
 
BJT
,
Brackney
 
DE
,
Wilusz
 
CJ
,
Ebel
 
GD
,
Wilusz
 
J
.
Flavivirus sfRNA suppresses antiviral RNA interference in cultured cells and mosquitoes and directly interacts with the RNAi machinery
.
Virology
.
2015
:
485
:
322
329
. https://doi.org/10.1016/j.virol.2015.08.009.

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
. https://doi.org/10.1371/journal.ppat.1002470.

Moreira
 
LA
,
Iturbe-Ormaetxe
 
I
,
Jeffery
 
JA
,
Lu
 
G
,
Pyke
 
AT
,
Hedges
 
LM
,
Rocha
 
BC
,
Hall-Mendelin
 
S
,
Day
 
A
,
Riegler
 
M
, et al.  
A Wolbachia symbiont in Aedes aegypti limits infection with dengue, chikungunya, and plasmodium
.
Cell
.
2009
:
139
(
7
):
1268
1278
. https://doi.org/10.1016/j.cell.2009.11.042.

Mussabekova
 
A
,
Daeffler
 
L
,
Imler
 
J-L
.
Innate and intrinsic antiviral immunity in Drosophila
.
Cell Mol Life Sci
.
2017
:
74
(
11
):
2039
2054
. https://doi.org/10.1007/s00018-017-2453-9.

Nene
 
V
,
Wortman
 
JR
,
Lawson
 
D
,
Haas
 
B
,
Kodira
 
C
,
Tu
 
ZJ
,
Loftus
 
B
,
Xi
 
Z
,
Megy
 
K
,
Grabherr
 
M
, et al.  
Genome sequence of Aedes aegypti, a major arbovirus vector
.
Science
.
2007
:
316
(
5832
):
1718
1723
. https://doi.org/10.1126/science.1138878.

Obbard
 
DJ
,
Gordon
 
KHJ
,
Buck
 
AH
,
Jiggins
 
FM
.
The evolution of RNAi as a defence against viruses and transposable elements
.
Philos Trans R Soc Lond B Biol Sci
.
2009
:
364
(
1513
):
99
115
. https://doi.org/10.1098/rstb.2008.0168.

Olmo
 
RP
,
Ferreira
 
AGA
,
Izidoro-Toledo
 
TC
,
Aguiar
 
ERGR
,
de Faria
 
IJS
,
de Souza
 
KPR
,
Osório
 
KP
,
Kuhn
 
L
,
Hammann
 
P
,
de Andrade
 
EG
, et al.  
Control of dengue virus in the midgut of Aedes aegypti by ectopic expression of the dsRNA-binding protein Loqs2
.
Nat Microbiol
.
2018
:
3
(
12
):
1385
1393
. https://doi.org/10.1038/s41564-018-0268-6.

Olmo
 
RP
,
Todjro
 
YMH
,
Aguiar
 
ERGR
,
de Almeida
 
JPP
,
Ferreira
 
FV
,
Armache
 
JN
,
de Faria
 
IJS
,
Ferreira
 
AGA
,
Amadou
 
SCG
,
Silva
 
ATS
, et al.  
Mosquito vector competence for dengue is modulated by insect-specific viruses
.
Nat Microbiol
.
2023
:
8
(
1
):
135
149
. https://doi.org/10.1038/s41564-022-01289-4.

Ou
 
TP
,
Auerswald
 
H
,
In
 
S
,
Peng
 
B
,
Pang
 
S
,
Boyer
 
S
,
Choeung
 
R
,
Dupont-Rouzeyrol
 
M
,
Dussart
 
P
,
Duong
 
V
.
Replication variance of African and Asian lineage Zika virus strains in different cell lines, mosquitoes and mice
.
Microorganisms
.
2021
:
9
(
6
):
1250
. https://doi.org/10.3390/microorganisms9061250.

Ozata
 
DM
,
Gainetdinov
 
I
,
Zoch
 
A
,
O’Carroll
 
D
,
Zamore
 
PD
.
PIWI-interacting RNAs: small RNAs with big functions
.
Nat Rev Genet
.
2019
:
20
(
2
):
89
108
. https://doi.org/10.1038/s41576-018-0073-3.

Palmer
 
W
,
Varghese
 
F
,
van Rij
 
R
.
Natural variation in resistance to virus infection in dipteran insects
.
Viruses
.
2018
:
10
(
3
):
118
. https://doi.org/10.3390/v10030118.

Parry
 
R
,
Asgari
 
S
.
Aedes anphevirus: an insect-specific virus distributed worldwide in Aedes aegypti mosquitoes that has complex interplays with Wolbachia and dengue virus infection in cells
.
J Virol
.
2018
:
92
(
17
):
e00224-18
. https://doi.org/10.1128/JVI.00224-18.

Potter
 
SS
,
Brorein
 
WJJR
,
Dunsmuir
 
P
,
Rubin
 
GM
.
Transposition of elements of the 412, copia and 297 dispersed repeated gene families in Drosophila
.
Cell
.
1979
:
17
(
2
):
415
427
. https://doi.org/10.1016/0092-8674(79)90168-5.

Qiu
 
Y
,
Xu
 
Y-P
,
Wang
 
M
,
Miao
 
M
,
Zhou
 
H
,
Xu
 
J
,
Kong
 
J
,
Zheng
 
D
,
Li
 
R-T
,
Zhang
 
R-R
, et al.  
Flavivirus induces and antagonizes antiviral RNA interference in both mammals and mosquitoes
.
Sci Adv.
 
2020
:
6
(
6
):
eaax7989
. https://doi.org/10.1126/sciadv.aax7989.

Quinlan
 
AR
,
Hall
 
IM
.
BEDTools: a flexible suite of utilities for comparing genomic features
.
Bioinformatics
.
2010
:
26
(
6
):
841
842
. https://doi.org/10.1093/bioinformatics/btq033.

Ramirez
 
JL
,
Souza-Neto
 
J
,
Torres Cosme
 
R
,
Rovira
 
J
,
Ortiz
 
A
,
Pascale
 
JM
,
Dimopoulos
 
G
.
Reciprocal tripartite interactions between the Aedes aegypti midgut microbiota, innate immune system and dengue virus influences vector competence
.
PLoS Negl Trop Dis
.
2012
:
6
(
3
):
e1561
. https://doi.org/10.1371/journal.pntd.0001561.

Raquin
 
V
,
Merkling
 
SH
,
Gausson
 
V
,
Moltini-Conclois
 
I
,
Frangeul
 
L
,
Varet
 
H
,
Dillies
 
M-A
,
Saleh
 
M-C
,
Lambrechts
 
L
.
Individual co-variation between viral RNA load and gene expression reveals novel host factors during early dengue virus infection of the Aedes aegypti midgut
.
PLoS Negl Trop Dis
.
2017
:
11
(
12
):
e0006152
. https://doi.org/10.1371/journal.pntd.0006152.

Rosendo Machado
 
S
,
Qu
 
J
,
Koopman
 
WJH
,
Miesen
 
P
.
The DEAD-box RNA helicase Dhx15 controls glycolysis and arbovirus replication in Aedes aegypti mosquito cells
.
PLoS Pathog
.
2022
:
18
(
11
):
e1010694
. https://doi.org/10.1371/journal.ppat.1010694.

Ross
 
PA
,
Callahan
 
AG
,
Yang
 
Q
,
Jasper
 
M
,
Arif
 
MAK
,
Afizah
 
AN
,
Nazni
 
WA
,
Hoffmann
 
AA
.
An elusive endosymbiont: does Wolbachia occur naturally in Aedes aegypti?
 
Ecol Evol.
 
2020
:
10
(
3
):
1581
1591
. https://doi.org/10.1002/ece3.6012.

Roundy
 
CM
,
Azar
 
SR
,
Rossi
 
SL
,
Huang
 
JH
,
Leal
 
G
,
Yun
 
R
,
Fernandez-Salas
 
I
,
Vitek
 
CJ
,
Paploski
 
IAD
,
Kitron
 
U
, et al.  
Variation in Aedes aegypti mosquito competence for Zika virus transmission
.
Emerg Infect Dis
.
2017
:
23
(
4
):
625
632
. https://doi.org/10.3201/eid2304.161484.

Roy
 
M
,
Viginier
 
B
,
Mayeux
 
CA
,
Ratinier
 
M
,
Fablet
 
M
.
Infections by transovarially transmitted DMelSV in Drosophila have no impact on ovarian transposable element transcripts but increase their amounts in the soma
.
Genome Biol Evol.
 
2021
:
13
(
9
):
evab207
. https://doi.org/10.1093/gbe/evab207.

Roy
 
M
,
Viginier
 
B
,
Saint-Michel
 
É
,
Arnaud
 
F
,
Ratinier
 
M
,
Fablet
 
M
.
Viral infection impacts transposable element transcript amounts in Drosophila
.
Proc Natl Acad Sci USA
.
2020
:
117
(
22
):
12249
12257
. https://doi.org/10.1073/pnas.2006106117.

Salazar
 
MI
,
Richardson
 
JH
,
Sánchez-Vargas
 
I
,
Olson
 
KE
,
Beaty
 
BJ
.
Dengue virus type 2: replication and tropisms in orally infected Aedes aegypti mosquitoes
.
BMC Microbiol
.
2007
:
7
(
1
):
9
. https://doi.org/10.1186/1471-2180-7-9.

Saleh
 
M-C
,
Tassetto
 
M
,
van Rij
 
RP
,
Goic
 
B
,
Gausson
 
V
,
Berry
 
B
,
Jacquier
 
C
,
Antoniewski
 
C
,
Andino
 
R
.
Antiviral immunity in Drosophila requires systemic RNA interference spread
.
Nature
.
2009
:
458
(
7236
):
346
350
. https://doi.org/10.1038/nature07712.

Schnettler
 
E
,
Sterken
 
MG
,
Leung
 
JY
,
Metz
 
SW
,
Geertsema
 
C
,
Goldbach
 
RW
,
Vlak
 
JM
,
Kohl
 
A
,
Khromykh
 
AA
,
Pijlman
 
GP
 
Noncoding flavivirus RNA displays RNA interference suppressor activity in insect and mammalian cells
.
J Virol
.
2012
:
86
(
24
):
13486
13500
. https://doi.org/10.1128/JVI.01104-12.

Senti
 
K-A
,
Jurczak
 
D
,
Sachidanandam
 
R
,
Brennecke
 
J
.
piRNA-guided slicing of transposon transcripts enforces their transcriptional silencing via specifying the nuclear piRNA repertoire
.
Genes Dev
.
2015
:
29
:
1747
1762
. https://doi.org/10.1101/gad.267252.115.

Shao
 
C
,
Sun
 
S
,
Liu
 
K
,
Wang
 
J
,
Li
 
S
,
Liu
 
Q
,
Deagle
 
BE
,
Seim
 
I
,
Biscontin
 
A
,
Wang
 
Q
, et al.  
The enormous repetitive Antarctic krill genome reveals environmental adaptations and population insights
.
Cell
.
2023
:
186
(
6
):
1279
1294.e19
. https://doi.org/10.1016/j.cell.2023.02.005.

Siomi
 
MC
,
Sato
 
K
,
Pezic
 
D
,
Aravin
 
AA
.
PIWI-interacting small RNAs: the vanguard of genome defence
.
Nat Rev Mol Cell Biol
.
2011
:
12
(
4
):
246
258
. https://doi.org/10.1038/nrm3089.

Slonchak
 
A
,
Hugo
 
LE
,
Freney
 
ME
,
Hall-Mendelin
 
S
,
Amarilla
 
AA
,
Torres
 
FJ
,
Setoh
 
YX
,
Peng
 
NYG
,
Sng
 
JDJ
,
Hall
 
RA
, et al.  
Zika virus noncoding RNA suppresses apoptosis and is required for virus transmission by mosquitoes
.
Nat Commun
.
2020
:
11
(
1
):
2205
. https://doi.org/10.1038/s41467-020-16086-y.

Solignat
 
M
,
Gay
 
B
,
Higgs
 
S
,
Briant
 
L
,
Devaux
 
C
.
Replication cycle of chikungunya: a re-emerging arbovirus
.
Virology
.
2009
:
393
(
2
):
183
197
. https://doi.org/10.1016/j.virol.2009.07.024.

Souza-Neto
 
JA
,
Powell
 
JR
,
Bonizzoni
 
M
.
Aedes aegypti vector competence studies: a review
.
Infect Genet Evol.
 
2019
:
67
:
191
209
. https://doi.org/10.1016/j.meegid.2018.11.009.

Strauss
 
JH
,
Strauss
 
EG
.
The alphaviruses: gene expression, replication, and evolution
.
Microbiol Rev
.
1994
:
58
(
3
):
491
562
. https://doi.org/10.1128/mr.58.3.491-562.1994.

Suzuki
 
Y
,
Baidaliuk
 
A
,
Miesen
 
P
,
Frangeul
 
L
,
Crist
 
AB
,
Merkling
 
SH
,
Fontaine
 
A
,
Lequime
 
S
,
Moltini-Conclois
 
I
,
Blanc
 
H
, et al.  
Non-retroviral endogenous viral element limits cognate virus replication in Aedes aegypti ovaries
.
Curr Biol.
 
2020
:
30
(
18
):
3495
3506.e6
. https://doi.org/10.1016/j.cub.2020.06.057.

Suzuki
 
Y
,
Frangeul
 
L
,
Dickson
 
LB
,
Blanc
 
H
,
Verdier
 
Y
,
Vinh
 
J
,
Lambrechts
 
L
,
Saleh
 
M-C
.
Uncovering the repertoire of endogenous flaviviral elements in Aedes mosquito genomes
.
J Virol
.
2017
:
91
(
15
):
e00571-17
. https://doi.org/10.1128/JVI.00571-17.

Varjak
 
M
,
Maringer
 
K
,
Watson
 
M
,
Sreenu
 
VB
,
Fredericks
 
AC
,
Pondeville
 
E
,
Donald
 
CL
,
Sterk
 
J
,
Kean
 
J
,
Vazeille
 
M
, et al.  
Aedes aegypti Piwi4 is a noncanonical PIWI protein involved in antiviral responses
.
mSphere
.
2017
:
2
(
3
):
e00144-17
. https://doi.org/10.1128/mSphere.00144-17.

Vieira
 
C
,
Lepetit
 
D
,
Dumont
 
S
,
Biemont
 
C
.
Wake up of transposable elements following Drosophila simulans worldwide colonization
.
Mol Biol Evol.
 
1999
:
16
(
9
):
1251
1255
. https://doi.org/10.1093/oxfordjournals.molbev.a026215.

Vieira
 
C
,
Nardon
 
C
,
Arpin
 
C
,
Lepetit
 
D
,
Biémont
 
C
.
Evolution of genome size in Drosophila. Is the invader's genome being invaded by transposable elements?
 
Mol Biol Evol.
 
2002
:
19
(
7
):
1154
1161
. https://doi.org/10.1093/oxfordjournals.molbev.a004173.

Walker
 
T
,
Johnson
 
PH
,
Moreira
 
LA
,
Iturbe-Ormaetxe
 
I
,
Frentiu
 
FD
,
McMeniman
 
CJ
,
Leong
 
YS
,
Dong
 
Y
,
Axford
 
J
,
Kriesner
 
P
, et al.  
The wMel Wolbachia strain blocks dengue and invades caged Aedes aegypti populations
.
Nature
.
2011
:
476
(
7361
):
450
453
. https://doi.org/10.1038/nature10355.

Wells
 
JN
,
Feschotte
 
C
.
A field guide to eukaryotic transposable elements
.
Annu Rev Genet.
 
2020
:
54
(
1
):
539
561
. https://doi.org/10.1146/annurev-genet-040620-022145.

Whitfield
 
ZJ
,
Dolan
 
PT
,
Kunitomi
 
M
,
Tassetto
 
M
,
Seetin
 
MG
,
Oh
 
S
,
Heiner
 
C
,
Paxinos
 
E
,
Andino
 
R
.
The diversity, structure, and function of heritable adaptive immunity sequences in the Aedes aegypti genome
.
Curr Biol.
 
2017
:
27
(
22
):
3511
3519.e7
. https://doi.org/10.1016/j.cub.2017.09.067.

Wicker
 
T
,
Sabot
 
F
,
Hua-Van
 
A
,
Bennetzen
 
JL
,
Capy
 
P
,
Chalhoub
 
B
,
Flavell
 
A
,
Leroy
 
P
,
Morgante
 
M
,
Panaud
 
O
, et al.  
A unified classification system for eukaryotic transposable elements
.
Nat Rev Genet
.
2007
:
8
(
12
):
973
982
. https://doi.org/10.1038/nrg2165.

Williams
 
AE
,
Sanchez-Vargas
 
I
,
Reid
 
WR
,
Lin
 
J
,
Franz
 
AWE
,
Olson
 
KE
.
The antiviral small-interfering RNA pathway induces Zika virus resistance in transgenic Aedes aegypti
.
Viruses
.
2020
:
12
(
11
):
1231
. https://doi.org/10.3390/v12111231.

Wimalasiri-Yapa
 
BMCR
,
Barrero
 
RA
,
Stassen
 
L
,
Hafner
 
LM
,
McGraw
 
EA
,
Pyke
 
AT
,
Jansen
 
CC
,
Suhrbier
 
A
,
Yakob
 
L
,
Hu
 
W
, et al.  
Temperature modulates immune gene expression in mosquitoes during arbovirus infection
.
Open Biol
.
2021
:
11
(
1
):
200246
. https://doi.org/10.1098/rsob.200246.

Zamudio
 
N
,
Barau
 
J
,
Teissandier
 
A
,
Walter
 
M
,
Borsos
 
M
,
Servant
 
N
,
Bourc'his
 
D
.
DNA methylation restrains transposons from adopting a chromatin signature permissive for meiotic recombination
.
Genes Dev
.
2015
:
29
(
12
):
1256
1270
. https://doi.org/10.1101/gad.257840.114.

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
(
25
):
10276
10281
. https://doi.org/10.1073/pnas.1303603110.

Author notes

Contributed equally

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected] for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact [email protected].
Associate Editor: Rachel Mueller
Rachel Mueller
Associate Editor
Search for other works by this author on:

Supplementary data