Background

Mosquito-borne diseases are one of the most significant public health burdens [1,2,3,4,5]. Human activities, urbanization, and climate change are increasingly bringing more human hosts in contact with disease vectors [2, 6,7,8,9]. Currently, half of the global population is at risk for dengue virus (DENV) infection [2, 8, 10]. In the aftermath of the 2015–2016 Zika virus (ZIKV) outbreak, which exposed more than 130 million people to infection, the virus remains endemic to tropical and subtropical regions [11].

Sharing a common vector in Aedes aegypti, DENV and ZIKV endemicity may expand in concert, resulting in widespread co-circulation [12, 13]. Thus, DENV-ZIKV synergy presents a bleak outlook for the near future with a growing proportion of the global population living under threat of simultaneous infection by both flaviviruses. As DENV-ZIKV co-circulation expands worldwide to affect currently low-risk or virus-free regions, mosquito vectors will have increased opportunities to receive and transmit both viruses [5]. Recent reports of DENV-ZIKV co-infection corroborate this prediction and reflect a proliferating synergy that has been overlooked. This may be the result of systemic underreporting engendered by difficulties in the differential diagnosis and detection of asymptomatic infections [14,15,16].

One cross-sectional study of the ZIKV epidemic in Colombia detected 8.8% of the DENV-ZIKV serotype among 34 co-infection cases [15]. Another study in southern Mexico randomly sampled a cohort of pregnant women during a non-epidemic period and found a relatively high proportion (2%) with DENV-ZIKV co-infection [17]. A previous report revealed that DENV infections occurred during the same period, highlighting the concerning extent of silent transmission of ZIKV with DENV [17, 18]. Besides underreporting, underlying this phenomenon may be antibody-dependent enhancement (ADE) between DENV and ZIKV in co-endemic areas. Studies support ADE of ZIKV infection by anti-DENV antibodies [4, 19,20,21], which not only may increase disease severity, but also may drive ZIKV transmission into primarily DENV-endemic areas. Similar results have been suggested for DENV, in which ZIKV-mediated ADE increases the propensity for severe disease and enables DENV to persist and proliferate in primarily ZIKV-endemic regions [

Fig. 3
figure 3

The replicating DENV2 genome interacts with the ZIKV-NS5 protein in co-infected mosquito cells. ZIKV-NS5 interacting with DENV2 and ZIKV genomic RNA was precipitated from cell lysates at 4 days using CLIP-PCR, followed by RT-PCR with primers specific for DENV and ZIKV genomes. Ribosomal protein S7 was used as a loading control

Co-infection of Ae. aegypti with DENV2 and ZIKV results in differential viral genome expression

Finding significant enhancement of viral replication by DENV2-ZIKV co-infection in vitro possibly arising from cross-species interactions, we challenged adult female Ae. aegypti sequentially with both viruses to quantify the effects of DENV2-ZIKV co-infection on vector competence in vivo. So far, existing studies into DENV-ZIKV co-infection have only challenged Ae. aegypti simultaneously, presenting both viruses in the same blood meal (BM). Contrarily, we elected to challenge mosquitos with DENV2 and ZIKV sequentially to maximize ecological validity. It is much more likely that in a DENV-ZIKV co-endemic area, a female mosquito would acquire co-infection in sequential feeding episodes: by feeding first on a DENV-infected host, then a ZIKV-infected host, or vice versa. The possibility of a mosquito obtaining both viruses from a DENV-ZIKV co-viremic human host through a single BM is much lower. Critically, we also consider the findings of studies reporting that non-infectious BM prior to subsequent viral BM can promote viral replication, as the initial non-infectious BM induces physiological changes in the midgut epithelium, rendering it more permissible to dissemination through the basal lamina [28, 29, 33, 34].

Thus, we included cohorts of mosquitoes presented with an initial naïve BM before challenge with DENV2 or ZIKV to account for this possible confounder in evaluating co-infection effects on viral replication (Fig. 4A). Overall, two co-infection schemes were used in vivo, in which Ae. aegypti were either challenged first with DENV2 and then ZIKV on a second BM (DENV2 → ZIKV), or vice versa (ZIKV → DENV2). These cohorts were compared against mosquitos single-infected with DENV and ZIKV, through either one or two BMs, as well as a mock cohort (BM-BM).

Fig. 4
figure 4

Co-infection of Ae. aegypti mosquitos with DENV and ZIKV results in differential viral genome expression. A Time course of experimental oral challenge with DENV2 and ZIKV. Mosquitos were captured 12 h ahead of day 0 and presented with blood meal (BM), DENV2, ZIKV, or maintained on sugar feeding (Sugar). On day 5, one group was given a second BM (BM-BM), and others were challenged with DENV2 or ZIKV. At 7 dpi (day 12), whole mosquito bodies were collected and homogenized. Relative viral genome expression of B DENV2 and C ZIKV was determined by qRT-PCR analysis, with normalization to the endosomal S7 protein. Each of six oral challenge schemes consisted of at least five biologically independent cohorts, and post hoc comparisons between groups were performed using Tukey’s multiple comparisons test; **P < 0.01; ***P < 0.001; ****P < 0.0001

Collecting mosquitos at 7 dpi, which we previously determined to be an optimal time point for viral genomic analysis, we quantified via qRT-PCR the relative expression of DENV2 and ZIKV genomes, respectively (Fig. 4A). We found that, contrary to our observations in vitro, DENV2 expression was significantly downregulated in both co-infection scenarios, particularly when compared with those challenged twice (DENV2 → DENV2) with DENV2 (Fig. 4B). Also, a significant difference in expression between DENV2 single-infected mosquitos presented with (BM → DENV2), and without (sugar-fed → DENV2), an initial naïve BM suggests that BM-induced modifications do indeed promote viral replication by encouraging dissemination from the midgut. Genomic expression of ZIKV was significantly elevated in both co-infection scenarios, even in comparison to mosquitos infected twice (ZIKV → ZIKV) with ZIKV (Fig. 4C). Between the two co-infection groups, ZIKV → DENV2 mosquitos expressed genomic ZIKV at significantly higher levels than did their DENV2 → ZIKV counterparts. There was also a significant difference in expression between sugar-fed → ZIKV and BM → ZIKV individuals, as with DENV2, again highlighting the initial BM’s ability to promote viral replication in and past the midgut. Moving forward, we evaluated the implications of co-infection on vector competence, quantifying the infectivity of virus particles produced in vivo.

Virus production and vector susceptibility are enhanced in Ae. aegypti challenged with both DENV2 and ZIKV

Having characterized the genomic expression profiles of co-infected mosquitos, we next assessed the quantity and infectivity of viruses produced therein. Performing plaque assays on isolated virus from individuals collected at 7 dpi from each treatment group (Fig. 4A), we found that viral titers were significantly higher in co-infected mosquitos than in the DENV single-infected cohort (Fig. 5A). Notably, these increases were observed for both DENV2 → ZIKV and ZIKV → DENV2 co-infection groups against all three DENV single-infection cohorts: those challenged either once (sugar/BM → DENV2) or twice (DENV2 → DENV2) with DENV2. Accordingly, the corresponding infection rates were markedly higher in co-infected mosquitos, increasing by as much as 36.7% (Fig. 5B). Compared with ZIKV single-infected cohorts, viral titers were also higher in co-infected individuals. Infection rates also saw increases by about 10% except when compared with the sugar → ZIKV cohort, a result likely attributable to individual variability. Despite the unexpectedly high infection rate, the median viral titer for this cohort was at least twofold lower than those of co-infected cohorts. Overall, mosquitos challenged sequentially with DENV and ZIKV produced more infectious virus, which induced a greater extent of infection ex vivo.

Fig. 5
figure 5

Virus production and vector susceptibility are enhanced in DENV2/ZIKV co-infected mosquitos. A Mosquitos were challenged with virus as described in Fig. 4, and then collected at 7 dpi (day 12) for plaque assay. Geometric means (PFU/ml) are plotted, and each of eight viral challenge schemes comprised at least three biological cohorts. B Sample size n, infection rate, and median PFU/ml corresponding to the experimental groups. Infected samples had positive (> 0) PFU/ml values; uninfected, negative samples are represented on the log scale as positive (PFU/ml = 1) only for visual interpretation. Comparison between groups post hoc was performed using Dunn’s multiple comparisons test; *P < 0.05; **P < 0.01; ***P < 0.001

Mosquitos twice-challenged with ZIKV produced higher viral titers than those co-infected, an unsurprising result following consecutive ZIKV infection. Secondary infection is greatly assisted by the existing RC infrastructure and an infected, compromised midgut epithelium and basal lamina (ZIKV → ZIKV). Indeed, viral titers were higher in co-infected mosquitos compared with those challenged once (sugar/BM → ZIKV). Infection rates of co-infected mosquitos also increased, suggesting greater susceptibility to infection and virus propagation, i.e., enhanced vector competence. As for mosquitos twice-challenged with DENV2, the clear enhancement of viral replication by co-infection is much more readily apparent, as these cohorts were entirely refractory to infection (0% infected, DENV2 → DENV2). Across DENV2 single-infected cohorts, infection rates peaked at 11.1%. The fact that vector infection rates were elevated to 44.1% and 46.7%, respectively, in co-infected cohorts indicates that DENV2-ZIKV co-infection positively, mutually modulates viral replication.

Of note, mosquitos given an initial naïve BM neither produced significantly higher viral titers nor were infected at higher rates than those directly challenged with DENV2/ZIKV. This suggests that while an initial non-infectious BM may correlate with higher viral genomic expression by assisting virus dissemination from the midgut, it ultimately neither enhances the production of viable, infectious particles nor promotes co-infected Ae. aegypti susceptibility to infection.

Taken together, our results show that DENV2-ZIKV co-infection significantly enhances virus production and vector susceptibility to infection. At the cellular level, viral replication is mutually enhanced owing to the overlap of highly homologous flaviviral replication machinery. We observed therein that DENV2 transcripts engage ZIKV-NS5. Overall, our findings raise grave concerns about DENV2 and ZIKV co-circulation, which threatens to strain healthcare resources and exacerbate transmission and disease as mosquitos vector these flaviviruses with greater efficiency.