key: cord-0797270-qlekpl0e authors: Tremblay, Nicolas; Freppel, Wesley; Sow, Aïssatou Aïcha; Chatel-Chaix, Laurent title: The Interplay between Dengue Virus and the Human Innate Immune System: A Game of Hide and Seek date: 2019-10-10 journal: Vaccines (Basel) DOI: 10.3390/vaccines7040145 sha: ac8b5e9b4a49a1062eddf4fc48a19778e66e9a78 doc_id: 797270 cord_uid: qlekpl0e With 40% of the world population at risk, infections with dengue virus (DENV) constitute a serious threat to public health. While there is no antiviral therapy available against this potentially lethal disease, the efficacy of the only approved vaccine is not optimal and its safety has been recently questioned. In order to develop better vaccines based on attenuated and/or chimeric viruses, one must consider how the human immune system is engaged during DENV infection. The activation of the innate immunity through the detection of viruses by cellular sensors is the first line of defence against those pathogens. This triggers a cascade of events which establishes an antiviral state at the cell level and leads to a global immunological response. However, DENV has evolved to interfere with the innate immune signalling at multiple levels, hence dampening antiviral responses and favouring viral replication and dissemination. This review elaborates on the interplay between DENV and the innate immune system. A special focus is given on the viral countermeasure mechanisms reported over the last decade which should be taken into consideration during vaccine development. Dengue is a neglected tropical disease caused by the dengue virus (DENV) that is transmitted to humans by Aedes aegypti or Aedes albopictus mosquitoes. In the recent years, it has become a public health concern, given that its global incidence has dramatically increased over the last 20 years and that no effective antiviral therapies are currently available. A dengue vaccine developed by Sanofi Pasteur, Dengvaxia, has been recently approved in over 20 countries around the globe. However, while the efficacy of this vaccine is not optimal for all DENV serotypes, its safety has been seriously questioned, especially for seronegative individuals. Epidemiological models estimate that about 390 million out of the 3.9 billion people living in endemic or epidemic areas contract the disease each year, making it the most prevalent arbovirus infection [1, 2] . About 60 million infected individuals per year will develop symptomatic dengue fever, resulting in 10 to 20 thousand fatalities and 1.14 million Disability-Adjusted Life Years (DALY) [3] . This increase in the disease burden is not evenly distributed across the globe, with Latin America and the Caribbean ending up since 1990 with an increase in dengue-related DALY of about 250% or 4-fold above the global average and with Southeast Asia accounting for 596,000 DALY. This might be due to the simultaneous occurrence of multiple factors in these low-to-middle income emerging countries which affect the complex balance of DENV transmission dynamics, leading to a rapid endemic-epidemic cycle. For instance, this includes an optimal climate for Aedes life cycle, a rapid increase in population density in urban centres, and the concomitant circulation of all DENV This induces the formation of MAVS aggregates that serve as an immune signalosome to activate through phosphorylation transcription factors IRF3 and NF-κB. In turn, these transcription factors translocate to the nucleus and elicit the transactivation of type I IFN. Once produced, type I IFN is secreted into the extracellular space and activates, in paracrine and autocrine fashions, the JAK-STAT pathway, leading to the amplification of the antiviral response via the activation of STAT1, STAT2, and IRF9, a transcription factor complex known as ISGF3. Infection also leads to mitochondrial stress and the release of mtDNA into the cytosol. mtDNA is recognized by cGAS, which activates STING, leading to the activation of IRF3/NF-κB and IFN production. DENV has developed various countermeasures to evade and/or engage the RLR and cGAS/STING pathways and to hinder innate immune signalling. These evasion mechanisms include (1) posttranscriptional modification of vRNA; (2) inhibition of 14-3-3ε or TRIM25 to hinder RIG-I activation; (3) MAVS aggregation; (4) alterations in endomembrane architecture to disrupt the MAVS signalosome; (5) the inhibition of TBK1 and IKBKE kinase activation, preventing the activation of transcription factors like IRF3; (6) the disruption of cGAS/STING signalling; and (7) the dampening of ISGF3 complex activation that is required for late innate antiviral responses. The RIG-I-Like Receptor (RLR) pathway has been classically studied for its role in the detection of viral ribonucleic acids and involves Retinoic Acid-Inducible gene I (RIG-I or DDX58), Melanoma Differentiation-Associated protein 5 (MDA5), and Laboratory of Genetics and Physiology 2 (LGP2/DHX58) as cytosolic sensors of an infection. They are ubiquitously expressed and are able to sense specific viral RNA moieties such as short uncapped 5′ triphosphorylated single-stranded (ss) RNA, short double-stranded (ds) RNA, U/A-rich 3′ regions of viral RNA, or long dsRNA [27] . Upon binding to vRNA, RIG-I and MDA5 are activated and translocated to the surface of the mitochondria where they interact with an adaptor protein, Mitochondrial Antiviral-Signalling Protein (MAVS), via their respective caspase activation and recruitment domains (CARD) (Figure 1, upper panel) . This proteins. This induces the formation of MAVS aggregates that serve as an immune signalosome to activate through phosphorylation transcription factors IRF3 and NF-κB. In turn, these transcription factors translocate to the nucleus and elicit the transactivation of type I IFN. Once produced, type I IFN is secreted into the extracellular space and activates, in paracrine and autocrine fashions, the JAK-STAT pathway, leading to the amplification of the antiviral response via the activation of STAT1, STAT2, and IRF9, a transcription factor complex known as ISGF3. Infection also leads to mitochondrial stress and the release of mtDNA into the cytosol. mtDNA is recognized by cGAS, which activates STING, leading to the activation of IRF3/NF-κB and IFN production. DENV has developed various countermeasures to evade and/or engage the RLR and cGAS/STING pathways and to hinder innate immune signalling. These evasion mechanisms include (1) posttranscriptional modification of vRNA; (2) inhibition of 14-3-3ε or TRIM25 to hinder RIG-I activation; (3) MAVS aggregation; (4) alterations in endomembrane architecture to disrupt the MAVS signalosome; (5) the inhibition of TBK1 and IKBKE kinase activation, preventing the activation of transcription factors like IRF3; (6) the disruption of cGAS/STING signalling; and (7) the dampening of ISGF3 complex activation that is required for late innate antiviral responses. The RIG-I-Like Receptor (RLR) pathway has been classically studied for its role in the detection of viral ribonucleic acids and involves Retinoic Acid-Inducible gene I (RIG-I or DDX58), Melanoma Differentiation-Associated protein 5 (MDA5), and Laboratory of Genetics and Physiology 2 (LGP2/DHX58) as cytosolic sensors of an infection. They are ubiquitously expressed and are able to sense specific viral RNA moieties such as short uncapped 5 triphosphorylated single-stranded (ss) RNA, short double-stranded (ds) RNA, U/A-rich 3 regions of viral RNA, or long dsRNA [27] . Upon binding to vRNA, RIG-I and MDA5 are activated and translocated to the surface of the mitochondria where they interact with an adaptor protein, Mitochondrial Antiviral-Signalling Protein (MAVS), via their respective caspase activation and recruitment domains (CARD) (Figure 1, upper panel) . This triggers MAVS oligomerization and the formation of prion-like structures required to transduce the signal to downstream effector proteins. This signalling platform, often called MAVS signalosome is believed to bring together to the mitochondrial surface both upstream PRRs and downstream regulatory subunits (TRAF2, TRAF5, TRAF6, and NEMO) and protein kinases (TBK1 and IKBKE, also known as IKKε). Once activated, this signalosome orchestrates the phosphorylation of the transcription factors IRF3 and NF-κB, resulting in their nuclear translocation and the induction of type 1 IFN [28-34]. Remarkably, the activity of the MAVS regulome is tightly regulated by the morphodynamics of mitochondria as well as their contacts with the ER (see below) [35] [36] [37] [38] . To sum up, mitochondria outer membrane is the scaffold of RLR signalling and, thus, plays an essential role in the initiation, the propagation, and the amplification of innate immunity against RNA viruses. To date, there are several lines of evidence suggesting that that DENV is preferentially and directly sensed by the RLR pathway. An early study using RIG-I dominant-negative mutants showed that RIG-I is required to trigger IRF-3-dependent antiviral immunity against DENV in A549 lung cancer cells [39] . This observation was then recapitulated in various cell types (Huh7 hepatocarcinoma cells, endothelial cells, and primary monocytes) and experimental settings [17, [40] [41] [42] . Combining affinity purification and next-generation sequencing, Chazal and colleagues recently showed that RIG-I is able to recognize 5 -ppp uncapped DENV vRNA, suggesting that detection of DENV nucleic acids occurs before the 5 -end maturation by capping is completed and that vRNA is afterwards hidden from PRR [43] . In stark contrast, the putative role of MDA5 in the detection of DENV nucleic acids remains unclear. Several groups have shown that silencing or interfering with MDA5 results in increased viral susceptibility and an altered innate immune response, but no clear mechanism of direct vRNA sensing has emerged despite this body of evidence. This might be due to the fact that all of these studies addressed the role of MDA5 in the early detection of vRNA. Strikingly, in the case of WNV, RIG-I and MDA5 have nonredundant roles at various stages of infection in vitro and in vivo [44] . Indeed, it was shown that MDA5 signalling to rely on the presence of WNV RNA PAMPs processed by cellular factors to become immunostimulatory but not on virion RNA that is initially uncoated upon viral entry. Overall, these studies support the view that RIG-I is a major player of the early detection of DENV RNA and of the initiation of the immune response. These further highlight that additional work is required to define the role of MDA5 during DENV infection. Toll-like receptors (TLR) constitute another important family of PRR. They are located at the plasma membrane or within late endosomes and can sense a variety of PAMPs such as bacterial lipopolysaccharide, dsDNA, ssRNA, and dsRNA [45, 46] . For DENV infection, TLR3 can recognize vRNA and its stimulation or overexpression dampens DENV replication in cellulo [47, 48] . Both TLR and RLR pathways converge to MAVS and stimulate IRF3, NF-κB, and type I IFN production. While it remains unknown whether DENV stimulates TLR3 in vivo, a study in rhesus macaques showed that the combined agonist-mediated stimulation of both TLR3 and TLR7/8 following DENV infection resulted in a decrease in viral replication and enhanced pro-inflammatory responses [49] . This highlights the antiviral potential of the TLR3 pathway against DENV in vivo. Converging to the same downstream effector molecules as the RLR pathway, that is, the TBK1/IRF3/IFN-β axis, the foreign DNA-sensing pathway relies on various intracellular DNA sensors, such as cyclic GMP-AMP Synthase (cGAS), which detect dsDNA danger signals from self or pathogen [50] . Upon binding, cGAS produces the second messenger cyclic GMP-AMP (cGAMP), which in turn activates Stimulator of Interferon Genes (STING). The engagement of the STING/cGAS pathway leads to the stimulation of TBK1 and ultimately to the production of type I IFN. Since its discovery, this pathway has been at the centre of many research interests in the fields of cancer, autoimmunity, inflammation, senescence, cell death, and viral infection, among others. Interestingly, while sensing DNA, the cGAS/STING pathway has been reported to be essential for the optimal innate immune response against RNA viruses [51] [52] [53] . Not surprisingly, several viruses such as influenza virus, hepatitis C virus, and coronavirus [52, [54] [55] [56] have developed targeted evasion mechanisms to interfere with this pathway. The involvement of cGAS and STING in the innate immune response against DENV has become evident over the last few years, with several reports showing that DENV actually modulates this pathway (see below) [54, [57] [58] [59] . While it remains unclear how a DNA-binding protein, like cGAS, can detect viral RNA, it was recently suggested that the sensing of DENV infection rather occurs indirectly. Indeed, virus-induced mitochondrial stress leads to the leakage of mitochondrial DNA (mtDNA) into the cytosol. It is subsequently sensed by cGAS, which in turn activates the type I IFN response [60] . More recently, it was shown that following inflammasome activation, secreted Interleukin (IL)-1β can induce mtDNA release, leading to the stimulation of the cGAS pathway and IFN production [61] . Considering that DENV infection triggers the activation of the inflammasome in human platelets and macrophages [62, 63] , it is plausible that this indirectly contributes to the mtDNA-dependent induction of early innate immunity. Lastly, the emerging concept of a cross talk between the RIG-I/MAVS and cGAS/STING pathways is surely interesting to understand DENV biology. Indeed, there is an accumulating literature that shows that the RLR pathway is able to potentiate the cGAS/STING pathway and vice versa during RNA virus infection [64] . This seems to occur through the physical connection between RIG, MAVS, and STING during viral infection [51, 65, 66] , the coactivation of both pathways [52, 53, 67] , or a transcriptional feedback loops [66, 68, 69] . For flaviviral infections, a study has shown that STING potentiates the RLR signalling via the assembly of a RIG-I/MAVS/STING complex following JEV nucleic acid recognition [66] . In addition, another study in mice has shown that cGAS is required for the optimal RLR-dependent IFN response against WNV. Indeed, cGAS mice had a higher viral load and lower ISG expression, resulting in a higher mortality when compared to wild-type controls [53] . Future studies will be required to assess the role of the cross talk between the RIG-I/MAVS and cGAS/STING during DENV infection. More generally, it will be required to investigate the exact nature, role, and distribution of the immunomodulatory molecules and signalling adaptors that are produced and expressed as a result of the pathogenesis of DENV infection. This will ultimately provide mechanistic insights on the exact contribution of the cGAS/STING pathway during DENV infection. DENV RNA synthesis and capping are believed to occur within the VPs since the viral replication dsRNA intermediate localizes within this ER-derived ultrastructure as shown by imaging studies using immunogold labelling and electron microscopy [16] . This confined environment is most probably important not only to concentrate the metabolites and proteins required for replication but also to exclude potentially inhibitory host factors. Considering this, it is tempting to speculate that neo-synthesized uncapped DENV RNAs "hide" from RIG-I in VPs although this model remains to be experimentally validated. The replicated DENV RNA molecule possesses several intrinsic features that allow its evasion from detection by the innate immune system. These sensing interference strategies rely on two specific modifications of vRNA: its 2 -O-methylation and its partial degradation by host nucleases (Figure 1 , bottom left panel). In addition to its RNA polymerase function, DENV NS5 has a methyltransferase (MTase) activity that generates the 5 7-methyl-guanosine cap and also methylates the 2 -OH position on the first nucleotide, forming a type 1 cap (m7GpppNm) structure [70, 71] . The 2 -O-methylated vRNAs mimic cellular mRNAs, thus evading the host immune system [72] [73] [74] [75] [76] (Figure 1) . Surprisingly, this mechanism is not necessarily strictly dependent on the capping of the vRNA since internal 2 -O-methylation can occur on vRNA lacking the 5 cap structure during infections with DENV and other flavivirus such as WNV [72] . The 2 -O-methylation has been shown to be important to avoid coronavirus infection recognition by MDA5 in mouse and human cells. Indeed, a DENV2 mutant deficient in the 2 -O MTase activity (NS5 E217A mutation) leads to an attenuated viral spread in IFN-competent cells [73, 74] . Consistent with this, it was later shown that the replication of the 2 -O-MTase mutant is attenuated at the very early stages of the life cycle, i.e. during the first 16 h of infection. As shown by a transcriptome kinetic analysis, this resulted in an increase in ISG transcription at the early time points postinfection with an enrichment in PRR genes (RIG-I, MDA5, and IFIT1). Altogether, these studies strongly support that DENV 2 -O-methylation aims to hide viral nucleic acids from early recognition by RLRs [76] . Importantly, a 2 -O-MTase mutant DENV2 strain was identified as a vaccine candidate since immunization with it conferred protection against subsequent DENV infection in mice and rhesus macaques [75] . However, it was proposed that exacerbated immune response might be due rather to a 2 -O-methylation-mediated evasion of the antiviral action of specific ISGs that enact downstream RLR and IFN signalling (see below). Another vRNA-related evasion mechanism relies on the partial degradation of vRNA by host factors, resulting in the generation of the subgenomic flavivirus RNA (sfRNA), a small noncoding RNA (Figure 1 ). sfRNAs are produced as byproducts of an incomplete degradation of flaviviral RNA due to unique secondary structures in the 3 UTR that causes XRN1, a 5 -3 exoribonuclease, to stall and cut the vRNA prematurely [77, 78] . Produced by all tested flaviviruses, the accumulation of biologically active sfRNA can contribute to virus-induced cytopathic effects [78, 79] ; to hijack mRNA processing machinery [80, 81] ; to promote optimal viral replication [82] ; and most relevant here, to help the virus to circumvent antiviral signalling [83] [84] [85] . Indeed, in the case of DENV, one study showed that DENV sfRNAs are able to inhibit TRIM25 deubiquitylation by USP15 [86] . This prevents the K63 polyubiquitylation of RIG-I, its dimerization via its CARD domains; and its subsequent interaction with downstream adaptor protein MAVS, resulting in hindered interferon signalling [31, 87, 88] . On the whole, vRNA methylation and degradation into sfRNA consist of two evolutionarily conserved flaviviral strategies to counteract innate antiviral immunity. In addition to interfering with RLR sensing of vRNA, DENV has developed other evasion strategies to inhibit the signalling cascade following RLR activation. Several DENV proteins modulate the pathway both upstream and downstream from MAVS. Notably, DENV NS3, through a conserved phosphomimetic RxEP motif, is able to prevent RIG-I translocation to the mitochondria by sequestering 14-3-3ε [89] (Figure 1 ). 14-3-3ε is required for efficient RIG-I/TRIM25 association and subsequent recruitment to mitochondria and interaction with MAVS [90] . Importantly, viruses expressing a mutated NS3 unable to associate with 14-3-3ε elicited a stronger immune response and replicated less efficiently in immune-competent cells [89] . Moreover, DENV NS4A is able to bind to MAVS CARD domains and to effectively prevent RIG-I/MAVS interaction through an unknown mechanism [91] . In order to sustain and maintain the immune synapse, MAVS needs to be physically accessible and at the proper position on the surface of the mitochondria; otherwise, antiviral signalling and response may not be optimal. Targeting this aspect of RLR-dependent innate immune signalling, DENV is also able to change the architecture of mitochondria in terms of morphodynamics and contacts with the endoplasmic reticulum ( Figure 1 ). First, mitochondria show an elongated morphology in DENV-infected cells and make contacts with DENV convoluted membranes [17, 92] . Interestingly, mitochondrial morphodynamics were reported to modulate antiviral signalling [35] [36] [37] . The morphology of mitochondria relies on an equilibrium between their fusion and fission regulated by dynamin-like GTPases. Briefly, these key players are mitofusins (MFN1 and MFN2) and Optic Atrophy 1 (OPA1), which mediate fusion (leading to elongation), and Dynamin-Related Protein 1 (DRP1), which mediates mitochondrial fission [93] . DENV-induced mitochondria elongation was attributed to NS4B since its overexpression alone recapitulated the phenotype. Furthermore, an inhibition of the phosphorylation-dependent activation of DRP1 was observed in both infected and NS4B-expressing cells, thus favouring mitochondrial elongation over fission [17] . Second, DENV infection resulted in a drastic disruption of the contacts between mitochondria and the ER (ERMC) [17] . Interestingly, the residual Mitochondria-Associated Membranes (MAM) appeared to be connected to CMs, suggesting that the biogenesis of this DENV RF substructure was responsible for ERMC disruption. Notably, MAMs are important for normal RLR signalling since they favour the tethering of MAVS required for optimal antiviral signalling [35] [36] [37] [38] . This suggests that the alteration of the reticulo-mitochondrial interface by DENV would contribute to the dampening of RLR signalling. In strong support to this, upon enforced mitochondria elongation via DRP1 expression knockdown in DENV-infected Huh7 cells, RIG-I was barely recruited to the MAMs, correlating with a dampened type I and III IFN mRNA transcription and an increased viral replication. This highlights the functional interplay between mitochondria morphodynamics and contact with ER. In another study, Yu and colleagues showed that DENV NS2B3 protease cleaves MFN1 and MFN2 [94] . Interestingly, MFN1 overexpression-mediated hyperfusion of mitochondria in the perinuclear region resulted in a decreased viral replication. Moreover, independent expression knockdown of MFN1 or MFN2 led to different phenotypes in viral replication, IFN-β induction, and cell death, illustrating the divergent roles of mitofusins despite their similarity. This is consistent with the different assumed functions of MFN1 in docking and fusion of the mitochondria and of MFN2 in the stabilization of the interactions between mitochondria [95, 96] . Furthermore, MFN2 localizes to MAMs and as an ER-resident protein tethers mitochondria to ER through homodimerization and heterotypic interactions with MFN1 [97] . This suggests that DENV might alter ERMCs by directly targeting MFN2 through NS2B3 protease activity without necessarily impacting on mitochondria fusion and elongation. Overall, these studies support the model that antiviral signalling is dependent on various spatiotemporal events and that alterations of the mitochondrial morphology and cytosolic interface regulate MAVS signalosome function. This concept of viral subversion of mitochondrial morphodynamics is not unique to DENV since related strategies have been observed for other viruses such as hepatitis B virus, hepatitis C virus, Severe acute respiratory syndrome-related coronavirus (SARS-CoV), and alphaherpesvirus [98] [99] [100] [101] . In addition to ER and mitochondria, DENV impairs the morphology of another membranous organelle functionally linked to MAVS-related signalling. Indeed, DENV as well as WNV and ZIKV induce a loss of cellular peroxisomes [102, 103] , which contain MAVS at their surface and constitute a transduction platform for innate signalling [104] [105] [106] . Notably, the expression knockdown of the peroxin Pex19, which is essential for peroxisome biogenesis [107] , led to a significant decrease in type III IFN gene transcription following the activation of RLR signalling with a synthetic dsRNA [102] . This highlights the importance of peroxisome integrity in innate immune signalling and the benefit for DENV of depleting them upon infection. Overall, these changes in cellular endomembrane morphology are believed to create a cytoplasmic microenvironment in which viral proteins are able to engage cellular host factors that are crucial to mount an adequate antiviral response. Finally, downstream of MAVS, DENV is also able to evade RLR signalling by interfering with the activity of effector proteins ( Figure 1 ). Indeed, NS2A, NS2B3, and NS4B are able to block IRF3 phosphorylation by interfering with serine kinase activity of TBK1 and IKBKE, which leads to a suboptimal type I IFN response [108, 109] . All these evasion strategies are consistent with the idea that DENV physically targets the MAVS signalosome in its entirety when it brings together upstream and downstream factors to the cytosolic side of the mitochondria. Interestingly, several DENV proteins involved in this co-opting, namely NS2B3, NS4A, and NS4B, all interact and partly localize within CMs [16] [17] [18] [110] [111] [112] [113] . Furthermore, CMs make with elongated mitochondria and there is strong evidence that they originate from MAMs, an important compartment for MAVS-dependent signalling [17] . Altogether, this supports a model in which CMs constitute a "hijacking" unit, which targets the whole MAVS signalosome at multiple levels. Rather than directly regulating vRNA replication, this viral substructure would instead "shutoff" potentially antiviral responses to infection, including early innate immunity, hence creating a cytoplasmic environment favourable to viral replication. The close spatial relationship between CMs and mitochondria would help to "trap" host targets inside CMs or to concentrate them near viral proteins in order to very efficiently downregulate the MAVS signalosome and maximize viral evasion. Nevertheless, this model requires experimental challenging. DENV also has the capacity to directly interfere with IFN induction that is triggered by the activated cGAS/STING pathway as a result of the release of mtDNA into the cytoplasm following DENV infection [114] (Figure 1) . Indeed, DENV NS2B marks cGAS for lysosomal degradation and DENV NS2B-NS3 protease cleaves STING to suppress type I IFN induction [54, 58, 114, 115] . Interestingly, DENV NS2B3 cannot process either mouse or nonhuman primate STING orthologs, suggesting that this pathogen has evolved towards an optimal pathogenicity in its natural hosts [54, 59] . Very recently, it has been reported that DRP1-mediated mitochondrial fission results in mitochondrial stress and the release of mtDNA in the cytosol [116] . Considering this, it will be interesting to investigate in the future whether DENV-induced mitochondrial elongation through DRP1 inhibition actually dampens not only RIG-I signal but also the activation of the STING/cGAS pathway and/or its cross talk with RLR signalling. In the same line of ideas, STING is a resident protein of MAMs [117] , and the alteration of these structures during DENV infection might contribute to targeting STING to NS2B3-rich CMs for degradation or spatiotemporal sequestration. In addition to the primary virus sensing pathway, DENV is also able to target several components of the downstream interferon-induced JAK-STAT signalling cascade that normally leads to the sustained expression of various ISGs following the secretion of IFN-α and -β ( Figure 1) . Early in vitro assays demonstrated that DENV NS4B is able to prevent STAT1 activation by a mechanism conserved in WNV and YFV that is yet to be elucidated [118, 119] . However, it can be rationalized that STAT1 inhibition most likely occurs either through prevention of its activation or by its dephosphorylation or by degradation of activated STAT1. On the matter of another STAT protein, DENV NS5 is able to bind STAT2 and to promote its E3 ubiquitin ligase UBR4-dependent degradation. This results in a drastic decrease of STAT2 basal expression level and, hence, to a hindered JAK-STAT signalling [120] [121] [122] . Interestingly, this STAT2-related evasion strategy is also conserved in other flaviviruses, such as ZIKV. Indeed, ZIKV is able to bind to STAT2 and to promote its proteasomal degradation but in an UBR4-independent manner [123, 124] . In addition to its role in STAT2 degradation, DENV NS5 protein is also able to interfere with ISG expression. First, both DENV and ZIKV NS5 selectively inhibit ISG expression by binding to and antagonizing the transcription factor PAF1C [125] . Second, DENV NS5 hijacks core components of the spliceosomal machinery, namely CD2BP2 and DDX23, resulting in an alteration of isoform abundance and stoichiometry of many antiviral factors such as RIG-I, ISG15, or IL-8, hence contributing to an environment favourable to viral replication [126] . At the posttranscriptional level, sfRNA has been shown to interfere with the translation of ISGs. Indeed, DENV sfRNA associates with the host RNA-binding proteins G3BP1, G3BP2, and CAPRIN1, which, as a result, are unable to stimulate as efficiently the translation of many ISGs such as PKR and IFITM2 for instance [127] . Consistently, sfRNA binding to these host factors protects DENV replication from IFN-β treatment. Interestingly, the 2 -O-methylation mechanism described above as a way to downregulate PRR early IFN response is also able to interfere with the functions of specific ISGs. Indeed, the replication of the NS5 E217A 2 -O-MTase DENV2 mutant is significantly more sensitive to IFN-β treatment than wt virus [75] , demonstrating that this modification may contribute to immunity evasion downstream of PRR signalling. More specifically for DENV but also for WNV, poxvirus, coronavirus, and JEV, the 2 -O-methylation of vRNA cap enables the viral nucleic acids to be marked as "self-RNA" and to evade from the antiviral function of IFN-Induced Protein with Tetratricopeptide Repeat (IFIT) proteins [128] [129] [130] [131] . Notably, IFIT1 was shown to associate with non-methylated RNAs with a higher affinity than the other IFITs. It was proposed that translation initiation factor eIF4E and IFIT1 compete for cap binding [129] . Hence, IFIT1 senses virus infection through the binding to foreign RNAs lacking 2 -O-methyls and inhibits the translation of viral RNA via the displacement of eIF4E. Consistently, IFIT1 overexpression in HEK293-DC-SIGN cells significantly decreases DENV2 replication in contrast to other IFITs [75] . In the case of WNV, the pathogenesis of the E218A MTase mutant is greatly attenuated in wt mice. Very interestingly, intracranial infection of Ifit1 knockout C57BL/6 mice with the E218A MTase mutant WNV restored lethality, highlighting the critical role of this IFIT in the sensing of the "2 -O-methyl-free" vRNA in vivo [128] . Considering the genetic and biological proximity with WNV, it is tantalizing to speculate that DENV RNA is able to evade IFIT antiviral activity in vivo by mimicking cellular mRNAs through the addition of 2 -O-methyls, one of the most common posttranscriptional modifications of RNA allowing vRNA to hide in plain sight among host mRNAs. As explained above, DENV sfRNA targets innate immune signalling in human cells at multiple levels (i.e. RIG-I activation and ISG expression). Of significant interest, several recent studies have highlighted the important role of sfRNA in the dissemination of DENV among infected insects [81, [132] [133] [134] . In fact, the DENV and YFV subgenomic RNAs are able to interfere with the Aedes mosquito innate system as well by targeting both Toll receptor and RNA interference (RNAi) pathways to favour optimal replication. In the mosquitoes, viral double-stranded RNA is processed into small siRNAs by RNase III DICER and is processed into viral siRNAs (vsiRNAs) by AGO2, which are able to target viral RNA for silencing [135, 136] . This shows that sfRNA plays two distinct roles, albeit using the same cellular factors, in the evasion of the innate immune response in mammalian hosts or arthropod vectors. This concept of different roles of host factors between the mammalian hosts or arthropod vectors will most likely be explored in depth in the future considering that a recent system biology study identified more than 45 shared pathway interactions between DENV-human and DENV-mosquito interactomes in the context of innate immune evasion and viral pathogenesis [125] . Multiple tetravalent vaccine candidates are being developed. These include live-attenuated vaccines, whole virus inactivated vaccines, protein-based vaccines, chimeric vaccines, and mRNA-based and synthetic virus-like particle vaccines [137] [138] [139] [140] [141] [142] . Among these vaccines, Dengvaxia (also called CYD-TDV), developed by Sanofi Pasteur, is the only one licensed for use in about 20 dengue-endemic countries in Asia, Latin America, and Oceania as well as in Europe. CYD-TDV is a tetravalent dengue chimeric live-attenuated vaccine which is based on the YFV 17D strain as a backbone. This vaccine is typically indicated for individuals aged from 9 to 45 years living in an endemic country and, hence, is not accessible to the population that is the most at risk to develop severe dengue-related symptoms. Unfortunately, the vaccine confers less protection against serotypes 1 and 2 than with serotypes 3 and 4 [143, 144] . Importantly, CYD-TDV efficacy was higher than pooled estimates in seropositive individuals but extremely low for seronegative children with risks of severe complications due to the vaccine. As a probable consequence of this, a global vaccination campaign in schools of the Philippines has resulted in the death of many children and the suspension of the program by the Department of Health in late 2017 [145] . As such, a pre-vaccination screening of previous exposure is now recommended prior to vaccination [146] . Lastly, a study shows that the CYD-TDV-elicited memory response decreases over time, as measured by low antibody titres in blood samples 5 years following an initial three-dose vaccine regimen. This suggests that a booster dose might be needed for sustained long-term immunological protection [147] . The development of new dengue vaccines which are safe and highly efficacious against all serotypes faces many challenges, including some involving the interplay between the virus and the immune system. Seminal studies by Albert Sabin, dating from the WWII era [148] , have demonstrated that volunteers exposed to DENV showed long-lived homotypic immunity but short-lived cross-protection against viruses of a different serotype. However, the mechanisms behind these observations are still not fully understood and prospective studies at the population level show that these conclusions cannot be generalized to inform rational vaccine design [149] . While a majority of DENV infections go unnoticed, some infected individuals progress along the disease spectrum and develop dengue hemorrhagic fever or dengue shock. Although no consensus exists about the pathophysiology behind this progression, it is most likely due to host-specific factors related to the presence of preexisting immunity such as antibody-dependent enhancements (ADE) and immune-mediated cytokine storm related to the antigenic sin phenomenon that occurs even past the peak in viremia in symptomatic individuals [10, 150] . Indeed, a second heterologous DENV infection is expected to elicit a strong memory recall of DENV-specific T and B cells that will produce an enhanced level of cytokines and neutralizing antibodies. However, this adaptive immune response might not lead to the control of viremia but rather quite the opposite, resulting in an antibody-dependent enhanced infection (virus opsonization and increased replication), mast cell activation (vascular permeability), and cellular cytotoxicity (cytokine storm) [151] . With this in mind, an effective antiviral therapy would need to be taken as a prophylactic treatment and would most likely be highly expensive given the number of individuals living in endemic areas. Thus, the only way to eradicate dengue worldwide would be to develop a safe, effective, and pan-serotypic vaccine that confers long-term sterilizing immunity. In summary, while CYD-TDV constitutes a safe and efficacious tool in the therapeutic arsenal against DENV when properly administered, it is plagued by shortcomings and controversies and does not completely meet the need for an effective dengue vaccine, especially in seronegative individuals. Thus, many challenges remain to be addressed to develop the "holy grail" of DENV vaccines and a better understanding of host-pathogen interactions is warranted to set up new vaccinal strategies or relevant animal models. In the following section, we discuss three examples illustrating how innate immunity research can inform vaccine design. The DENV∆30 is a live attenuated vaccine candidate that was engineered by deleting 30 nucleotides of the 3 UTR vRNA using reverse genetics and by subsequently creating modified dengue viruses for all four serotypes [152, 153] . Proof-of-concept studies showed that, despite attenuation, the DENV∆30 is highly immunogenic in both humans and rhesus monkeys for all tested serotypes and confers protection upon homologous rechallenge [153] [154] [155] [156] [157] [158] [159] [160] [161] . In addition, DENV∆30 appears to be less virulent in mosquitoes, limiting the risk of transmission from a vaccinated human to the Aedes vector [162] . For long, the molecular mechanism conferring attenuation of this vaccine candidate was largely unknown. However, it was shown that DENV4∆30-infected cells accumulated less sfRNA, resulting in an increased sensitivity to type I IFN [163] . Further studies will be required to assess if other evasion mechanisms are in play in DENV∆30 attenuation to fully understand how this strain is able to create a balanced environment that is favourable to antigen presentation and cytokine production. However, given the reported roles of sfRNA in both IFN production and response (see above), it is likely that the attenuation of DENV∆30 results from an increased innate immune response. As explained above, 2 -O-methylation is a modification that allows the vRNA to mimic cellular mRNAs and to evade the host innate immune system. Using reverse genetics, Züst and colleagues have generated DENV clones from serotypes 1 and 2 that harbour a single point mutation in the conserved KDKE motif of the NS5 methyltransferase catalytic site. These mutants (E216A for DENV1 or E217A for DENV2) are impaired in 2 -O-methylation but are still able to perform the N7-methylation of the cap. Importantly, they are more susceptible to IFN treatment or human IFIT1 overexpression than wild-type DENV, which is consistent with the idea that they are impaired in IFIT inhibition. Importantly, treatment of mice competent for T-cell responses with these viruses alone or in serotype combination conferred protection against a subsequent infection with a lethal mouse-adapted DENV strain. Remarkably, a single low dose of the DENV2 E217A mutant in monkeys was enough for a complete seroconversion and protection [75] . This approach was also applied to develop a JEV vaccine candidate. A E218A mutant JEV was attenuated and more sensitive to a type I IFN treatment as well [164] . A single dose of this live attenuated vaccine was sufficient to elicit a strong humoral response that conferred protection and survival following heterologous rechallenge in BALB/c mice. Ongoing studies are now trying to implement this vaccine rational design towards the development of a tetravalent, non-chimeric vaccine with therapeutic relevance. Vaccine development studies usually rely on large animal models during nonclinical and preclinical studies [165] . The use of small animal models such as mice usually requires genetic modifications that dampen their immune system in order to increase viral permissiveness and, thus, do not reflect the reality of an infection in the natural host with an intact immune response. In most of the cases, in vivo DENV studies classically involve immunodeficient mouse strains such as AG129 which do not express IFN-α/β and IFN-γ receptors. These mice are highly susceptible to DENV infection and generally die within two weeks after injection [166] [167] [168] . Following advancement of the knowledge about host-pathogen interactions and the advent of novel technologies (e.g., by CRISPR/Cas9 gene editing), a paradigm shift can be anticipated. As an example of this, using a gene knock-in approach and a mouse-adapted virus strain, Gorman and colleagues recently developed an immunocompetent transgenic mouse model of ZIVK infection by replacing the mouse STAT2 by the human STAT2 [169] . This genetic engineering was conceptually made possible thanks to previous studies which showed that ZIKV induces the degradation of human STAT2 but not of the mouse ortholog [123, 124, 170] . While it will be interesting to see if the use of humanized immunocompetent mouse model can be expanded to DENV studies, it definitely opens up many future research opportunities. DENV research will most likely remain a challenging and exciting field in times to come. In recent years, DENV infection has taken a front seat in the realm of global health concerns as it spreads outside the Western Pacific region and is now threatening more than 4 billion individuals. With no effective antiviral therapies or prophylactic vaccine, DENV is expected to cause disease and to inflict harm that amounts to more than 1.14 million DALY. As extensively discussed in this review, DENV is well equipped to evade the innate antiviral immune system through various antagonizing functions of viral NS proteins and RNA. From being able to hide in plain sight due to the 2 -O-methylation of the viral genome and to the disruption of MAVS signalosome activity and of the IFN signalling, it can be acknowledged that, despite a simple viral genome organization, DENV pathogenesis relies on a complex network of host-pathogen interactions. It is conceivable that better understanding this complexity will contribute to solving the puzzle of dengue control and/or eradication. Indeed, the interplay between DENV and the immune system can lead to rational vaccine design by providing attractive targets for antigen and adjuvant development and by filling the gaps in knowledge regarding how the innate immune system can be harnessed to support a long-lasting protective immune response. The global distribution and burden of dengue Refining the Global Spatial Limits of Dengue Virus Transmission by Evidence-Based Consensus The global burden of dengue: An analysis from the Global Burden of Disease Study Dengue in Latin America: Systematic Review of Molecular Epidemiological Trends The epidemiology of dengue in the americas over the last three decades: A worrisome reality Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control; World Health Organization Viraemia in patients with naturally acquired dengue infection Viremia en plasma como factor asociado a gravedad en la infección por el virus del dengue: Revisión sistemática de la literatura Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity Immune-mediated cytokine storm and its role in severe dengue Coupling of replication and assembly in flaviviruses Rewiring cellular networks by members of the Flaviviridae family The Multiples Fates of the Flavivirus RNA Genome During Pathogenesis Flaviviridae Replication Organelles: Oh, What a Tangled Web We Weave Dengue virus-and hepatitis C virus-induced replication and assembly compartments: The enemy inside-caught in the web Composition and three-dimensional architecture of the dengue virus replication and assembly sites Dengue Virus Perturbs Mitochondrial Morphodynamics to Dampen Innate Immune Responses The non-structural protein 4A of dengue virus is an integral membrane protein inducing membrane alterations in a 2K-regulated manner Direct triggering of the type I interferon system by virus infection: Activation of a transcription factor complex containing IRF-3 and CBP/p300 Virus infection induces the assembly of coordinately activated transcription factors on the IFN-beta enhancer in vivo Essential role of RIG-I in the activation of endothelial cells by dengue virus Inhibition of dengue and chikungunya virus infections by RIG-I-mediated type I interferon-independent stimulation of the innate antiviral response RIG-I Recognizes the 5 Region of Dengue and Zika Virus Genomes The Essential, Nonredundant Roles of RIG-I and MDA5 in Detecting and Controlling West Nile Virus Infection Molecular mechanisms of regulation of Toll-like receptor signaling Structures and recognition modes of toll-like receptors Human TLR3 recognizes dengue virus and modulates viral replication in vitro Activation of Toll-like receptor 3 impairs the dengue virus serotype 2 replication through induction of IFN-β in cultured hepatoma cells Decreased dengue replication and an increased anti-viral humoral response with the use of combined Toll-like receptor 3 and 7/8 agonists in macaques Cytosolic sensing of immuno-stimulatory DNA, the enemy within STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling Influenza A virus targets a cGAS-independent STING pathway that controls enveloped RNA viruses Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity DENV inhibits type I IFN production in infected cells by cleaving human STING Coronavirus papain-like proteases negatively regulate antiviral innate immune response through disruption of STING-mediated signaling Hepatitis C virus NS4B blocks the interaction of STING and TBK1 to evade host innate immunity Dengue virus targets the adaptor protein MITA to subvert host innate immunity Dengue virus NS2B protein targets cGAS for degradation and prevents mitochondrial DNA sensing during infection Dengue viruses cleave STING in humans but not in nonhuman primates Mitochondrial DNA stress primes the antiviral innate immune response Interleukin-1β Induces mtDNA Release to Activate Innate Immune Signaling via cGAS-STING CLEC5A is critical for dengue virus-induced inflammasome activation in human macrophages Platelets mediate increased endothelium permeability in dengue through NLRP3-inflammasome activation Crosstalk between Cytoplasmic RIG-I and STING Sensing Pathways The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation STING mediates neuronal innate immune response following Japanese encephalitis virus infection Interferon γ-inducible Protein (IFI) 16 Transcriptionally Regulates Type I Interferons and Other Interferon-stimulated Genes and Controls the Interferon Response to both DNA and RNA Viruses DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity RIG-I-Mediated STING Upregulation Restricts Herpes Simplex Virus 1 Infection Causes and Consequences of Flavivirus RNA Methylation Viral and cellular mRNA capping: Past and prospects 2 -O methylation of internal adenosine by flavivirus NS5 methyltransferase Live Cell Analysis and Mathematical Modeling Identify Determinants of Attenuation of Dengue Virus 2 -O-Methylation Mutant Ribose 2 -O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5 Rational Design of a Live Attenuated Dengue Vaccine: 2 -O-Methyltransferase Mutants Are Highly Attenuated and Immunogenic in Mice and Macaques Evasion of early innate immune response by 2 -O -methylation of dengue genomic RNA Functional non-coding RNAs derived from the flavivirus 3 untranslated region A highly structured, nuclease-resistant, noncoding RNA produced by flaviviruses is required for pathogenicity Dengue virus subgenomic RNA induces apoptosis through the Bcl-2-mediated PI3k/Akt signaling pathway A noncoding RNA produced by arthropod-borne flaviviruses inhibits the cellular exoribonuclease XRN1 and alters host mRNA stability Noncoding flavivirus RNA displays RNA interference suppressor activity in insect and Mammalian cells Small noncoding RNA modulates Japanese encephalitis virus replication and translation in trans Japanese encephalitis virus non-coding RNA inhibits activation of interferon by blocking nuclear translocation of interferon regulatory factor 3 West Nile virus noncoding subgenomic RNA contributes to viral evasion of the type I interferon-mediated antiviral response Full Genome Sequence and sfRNA Interferon Antagonist Activity of Zika Virus from Recife, Brazil Dengue subgenomic RNA binds TRIM25 to inhibit interferon expression for epidemiological fitness Influenza A virus NS1 targets the ubiquitin ligase TRIM25 to evade recognition by the host viral RNA sensor RIG-I TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity A phosphomimetic-based mechanism of dengue virus to antagonize innate immunity The mitochondrial targeting chaperone 14-3-3ε regulates a RIG-I translocon that mediates membrane association and innate antiviral immunity Dengue Virus Subverts Host Innate Immunity by Targeting Adaptor Protein MAVS Dengue virus induces mitochondrial elongation through impairment of Drp1-triggered mitochondrial fission Mitochondrial fission: Regulation and ER connection Dengue Virus Impairs Mitochondrial Fusion by Cleaving Mitofusins Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity Structural basis of mitochondrial tethering by mitofusin complexes Mitofusin 2 tethers endoplasmic reticulum to mitochondria Hepatitis B virus disrupts mitochondrial dynamics: Induces fission and mitophagy to attenuate apoptosis Hepatitis C virus triggers mitochondrial fission and attenuates apoptosis to promote viral persistence Alphaherpesvirus infection disrupts mitochondrial transport in neurons SARS-coronavirus open reading frame-9b suppresses innate immunity by targeting mitochondria and the MAVS/TRAF3/TRAF6 signalosome Flavivirus Infection Impairs Peroxisome Biogenesis and Early Antiviral Signaling Interplay between Zika Virus and Peroxisomes during Infection Peroxisomes are signaling platforms for antiviral innate immunity Diverse intracellular pathogens activate type III interferon expression from peroxisomes Activation of Type I and III Interferon Response by Mitochondrial and Peroxisomal MAVS and Inhibition by Hepatitis C Virus Pex19p, a farnesylated protein essential for peroxisome biogenesis Dengue virus subverts the interferon induction pathway via NS2B/3 protease-IκB kinase epsilon interaction Dengue Virus NS Proteins Inhibit RIG-I/MAVS Signaling by Blocking TBK1/IRF3 Phosphorylation: Dengue Virus Serotype 1 NS4A Is a Unique Interferon-Regulating Virulence Determinant A Combined Genetic-Proteomic Approach Identifies Residues within Dengue Virus NS4B Critical for Interaction with NS3 and Viral Replication Mapping the Interactions between the NS4B and NS3 proteins of dengue virus Characterization of dengue virus NS4A and NS4B protein interaction Dengue virus NS4B interacts with NS3 and dissociates it from single-stranded RNA Dengue virus activates cGAS through the release of mitochondrial DNA P122 Dengue virus targets the adaptor protein MITA to subvert host innate immunity Mitochondrial fission-induced mtDNA stress promotes tumor-associated macrophage infiltration and HCC progression STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity Inhibition of interferon signaling by dengue virus Inhibition of alpha/beta interferon signaling by the NS4B protein of flaviviruses Dengue virus co-opts UBR4 to degrade STAT2 and antagonize type I interferon signaling NS5 of dengue virus mediates STAT2 binding and degradation Dengue virus inhibits alpha interferon signaling by reducing STAT2 expression Zika virus inhibits type-I interferon production and downstream signaling Zika Virus Targets Human STAT2 to Inhibit Type I Interferon Signaling Comparative Flavivirus-Host Protein Interaction Mapping Reveals Mechanisms of Dengue and Zika Virus Pathogenesis The Dengue Virus NS5 Protein Intrudes in the Cellular Spliceosome and Modulates Splicing G3BP2 and CAPRIN1 Are Required for Translation of Interferon Stimulated mRNAs and Are Targeted by a Dengue Virus Non-coding RNA 2 -O methylation of the viral mRNA cap evades host restriction by IFIT family members Sequestration by IFIT1 impairs translation of 2 O-unmethylated capped RNA Ifit1 Inhibits Japanese Encephalitis Virus Replication through Binding to 5 Capped 2 -O Unmethylated RNA Inhibition of translation by IFIT family members is determined by their ability to interact selectively with the 5 -terminal regions of cap0-, cap1-and 5 ppp-mRNAs Dengue subgenomic flaviviral RNA disrupts immunity in mosquito salivary glands to increase virus transmission Flavivirus sfRNA suppresses antiviral RNA interference in cultured cells and mosquitoes and directly interacts with the RNAi machinery Noncoding Subgenomic Flavivirus RNA Is Processed by the Mosquito RNA Interference Machinery and Determines West Nile Virus Transmission by Culex pipiens Mosquitoes RNA interference-mediated antiviral defense in insects On the road to reading the RNA-interference code The successful induction of T-cell and antibody responses by a recombinant measles virus-vectored tetravalent dengue vaccine provides partial protection against dengue-2 infection Recent Developments in Recombinant Protein-Based Dengue Vaccines Dengue vaccines: Recent developments, ongoing challenges and current candidates Next generation designer virus-like particle vaccines for dengue Dengue viruses and promising envelope protein domain III-based vaccines Aiming at the heart: The capsid protein of dengue virus as a vaccine candidate Efficacy and Long-Term Safety of a Dengue Vaccine in Regions of Endemic Disease Clinical Efficacy, Safety, and Immunogenicity of a Live Attenuated Tetravalent Dengue Vaccine (CYD-TDV) in Children: A Systematic Review with Meta-analysis. Front The Lancet Infectious Diseases The dengue vaccine dilemma Dengue vaccine: WHO position paper Low antibody titers 5 years after vaccination with the CYD-TDV dengue vaccine in both pre-immune and naïve vaccinees Research on dengue during World War II Review article: Research on dengue during World War II revisited Highlights for Dengue Immunopathogenesis: Antibody-Dependent Enhancement Adaptive immune responses to primary and secondary dengue virus infections Development of a live attenuated dengue virus vaccine using reverse genetics Attenuation and immunogenicity in humans of a live dengue virus type-4 vaccine candidate with a 30 nucleotide deletion in its 3 -untranslated region Dengue virus type 3 vaccine candidates generated by introduction of deletions in the 3 untranslated region (3 -UTR) or by exchange of the DENV-3 3 -UTR with that of DENV-4. Vaccine A single dose of the DENV-1 candidate vaccine rDEN1∆30 is strongly immunogenic and induces resistance to a second dose in a randomized trial Genetically modified, live attenuated dengue virus type 3 vaccine candidates The Live Attenuated Dengue Serotype 1 Vaccine rDEN1∆30 is Safe and Highly Immunogenic in Healthy Adult Volunteers R. rDEN2/4∆30 (ME), a live attenuated chimeric dengue serotype 2 vaccine, is safe and highly immunogenic in healthy dengue-naïve adults A. rDEN4delta30, a live attenuated dengue virus type 4 vaccine candidate, is safe, immunogenic, and highly infectious in healthy adult volunteers Live-Attenuated Tetravalent Dengue Virus Vaccine Formulations Induce a Balanced, Broad, and Protective Neutralizing Antibody Response against Each of the Four Serotypes in Rhesus Monkeys The live attenuated dengue vaccine TV003 elicits complete protection against dengue in a human challenge model A live attenuated recombinant dengue-4 virus vaccine candidate with restricted capacity for dissemination in mosquitoes and lack of transmission from vaccinees to mosquitoes Decreased accumulation of subgenomic RNA in human cells infected with vaccine candidate DEN4∆30 increases viral susceptibility to type I interferon Rational design of a flavivirus vaccine by abolishing viral RNA 2 -O methylation Large animal models for vaccine development and testing New mouse model for dengue virus vaccine testing A lethal murine infection model for dengue virus 3 in AG129 mice deficient in type I and II interferon receptors leads to systemic disease Interferon-Dependent Immunity Is Essential for Resistance to Primary Dengue Virus Infection in Mice, Whereas T-and B-Cell-Dependent Immunity Are Less Critical An Immunocompetent Mouse Model of Zika Virus Infection Zika Virus Antagonizes Type I Interferon Responses during Infection of Human Dendritic Cells This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license We are grateful to Karine Boulay (Université de Montréal, Canada) for critical reading of the manuscript and helpful comments. The authors declare no conflict of interest. The funders had no role in the writing of the manuscript or in the decision to publish this review.