Skip to main content
Tropical Medicine and Health logoLink to Tropical Medicine and Health
. 2011 Sep 13;39(4 Suppl):53–62. doi: 10.2149/tmh.2011-S06

Dengue and Soluble Mediators of the Innate Immune System

Lyre Anni Espada-Murao 1, Kouichi Morita 1,*
PMCID: PMC3317598  PMID: 22500137

Abstract

Huge emphasis has been placed on the role of the adaptive immune system in dengue pathogenesis. Yet there is increasing evidence for the importance of the innate immune system in regulating dengue infection and possibly influencing the disease. This review focuses on the interplay between the innate immune system and dengue and highlights the role of soluble immunological mediators. Type I and type II interferons of the innate immune system demonstrate non-overlapping roles in dengue infection. Furthermore, while some IFN responses to dengue are protective, others may exert disease-related effects on the host. But aside from interferons, a number of cytokines have also been implicated in dengue pathogenesis. Our expanding knowledge of cytokines indicates that these soluble mediators act upon a complicated network of events to provoke the disease. This cytokine storm is generally attributed to massive T cell activation as an outcome of secondary infection. However, there is reason to believe that innate immune response-derived cytokines also have contributory effects, especially in the context of severe cases of primary dengue infection. Another less popular but interesting perspective on dengue pathogenesis is the effect of mosquito feeding on host immune responses and viral infection. Various studies have shown that soluble factors from vector saliva have the capacity to alter immune reactions and thereby influence pathogen transmission and establishment. Hence, modulation of the innate immune system at various levels of infection is a critical component of dengue disease. In the absence of an approved drug or vaccine for dengue, soluble mediators of the innate immune system could be a strategic foothold for developing anti-viral therapeutics and improving clinical management.

Keywords: dengue, innate immunity, cytokines

Contents

  • 1. Introduction

  • 2. The innate immune response to dengue: to defend or to destroy

    • 2-1. Interferons

    • 2-2. Cytokines

  • 3. Mosquito-derived immunomodulators in flavivirus infection

  • 4. Prospects for dengue management and therapy

  • 5. Conclusion

1. Introduction

Dengue virus (DV) is transmitted to a human through the bite of an infected mosquito, particularly of the Aedes group. The period of incubation ranges from three to seven days [1]. Then viremia lasts for about one to seven days and can reach 107 to 109 mosquito infectious doses (MID50)/ml [2], which is more than sufficient to transmit the virus to another feeding mosquito.

After its injection into the skin, DV is presumed to undergo an initial round of replication in Langerhans dendritic cells (LDCs) [3, 4]. Infected LDCs migrate to draining lymph nodes [5], where infection spreads to monocytes and macrophages [6]. The lymphatic system may then play a key role in the ensuing viremia, through which the virus can be disseminated to other organs such as the spleen, liver and bone marrow [6].

The clinical manifestation of dengue ranges from mild febrile syndrome to fatal disease [1]. Dengue fever (DF) is an acute and self-limited illness manifested by fever, headache, myalgia and arthralgia, with physical evidence of rash. Laboratory tests reveal leukopenia, as well as varying degrees of thrombocytopenia and hemorrhage. The more severe dengue hemorrhagic fever (DHF) is complicated by plasma leakage that occurs around three to five days after the disease. A sudden and extensive plasma leakage may result in shock or death, a phenomenon called dengue shock syndrome (DSS). Typically, patients undergo a defervescence phase marked by an abrupt drop in body temperature, at which point the illness may either wane to recovery or proceed to serious complications [7].

Dengue is widely accepted to be an immunopathological disease. Compounding evidence also associates dengue severity with secondary infection of a heterologous serotype [811]. From this perspective, dengue pathogenesis can be explained through various hypotheses that implicate immune-related factors, including the: 1) enhancement of viral infection through cross-reactive antibodies, 2) activation of cross-reactive memory T cells, 3) cytokine storm, and 4) complement activation. Unfortunately, a huge part of dengue pathogenesis remains elusive despite years of arduous study. In particular, while the adaptive immune response has been central to dengue research, the initial immune events that lead to disease evolution also warrant consideration. Thus, we present a review of recent developments in the field of dengue and the innate immune system, with a focus on soluble mediators that might be involved in infection and pathogenesis.

2. The innate immune response to dengue: to defend or to destroy

At the onset of skin infection, DV is immediately confronted with the host innate immune system. Whether infection becomes limited or progresses to disease depends on the balance between the defensive and destructive effect of the immune response. Here we explore various soluble mediators of the innate immune system and their role in dengue infection. Elucidating such early events will be valuable in shedding light on dengue pathogenesis and may impart prognostic utility for predicting disease.

2-1. Interferons

The initial immune response to viral infection is mediated by interferons (IFN). Most cells produce type I IFNs (IFNα/β) to inhibit viral translation and replication. On the other hand, type II IFNs (IFNγ) derived from NK cells and activated T cells modulate the production of proinflammatory and antiviral molecules. An early innate immune response accompanied by strong upregulation of IFN-related genes has been described for dengue [1214]. Among the various pattern-recognition receptor (PRR) pathways utilized by the host for IFN activation, the IPS-1/Cardif system operated by retinoic acid-inducible gene I-like helicases (RLH) appears to be the most critical in initiating the innate immune response to dengue, as exhibited by delayed IFN production in lymphoid tissues of IPS-1-deficient mice, rendering them more susceptible to the virus [15]. RLH-dependent IFN activation also seems to be regulated by the intracellular localization of flavivirus double-stranded RNA (dsRNA). A recent study in our laboratory demonstrated dynamic changes in Japanese encephalitis virus dsRNA localization, which was initially concealed in intracellular membrane structures but eventually exposed to the cytosol [16]. This cytosolic exposure sets the stage for PRR recognition and IFN activation, and its timing occurs in a cell/species-specific manner, which ultimately determines cell permissiveness to infection [16]. We are currently exploring the applicability of this model to DV. Other PRRs such as toll-like receptors (TLRs) may also be involved, as TLR7 was demonstrated to be essential for DV-induced production of IFN in plasmacytoid dendritic cells [17].

Experiments using knock-out mice were conducted to examine the contribution of IFN to viral clearance and disease. Mice deficient in receptors for type I and type II IFN were extremely vulnerable to dengue infection, while B or T cell-deficient mice displayed a normal ability to resist infection, demonstrating the predominant role of IFN over the adaptive immune system in controlling primary infection by DV [18]. Although the protective effect of type I and type II IFNs is synergistic, their respective roles are non-overlapping. For instance, viral distribution was widespread in IFNα/β receptor-knockout mice, in contrast to a benign viral titer in IFNγ receptor-knockout mice, suggesting that type I IFNs are specifically responsible for limiting the initial replication and/or spread of dengue [18]. However, IFNγ receptor-knockout mice exhibited a lower survival rate than IFNα/β receptor-knockout mice, indicating the protective role of IFNγ, but not IFNα/β, against the disease [18].

Nevertheless, type I IFNs and other IFN-stimulated genes have been associated with disease severity in several clinical studies. These studies are characterized by the consistent theme of highly activated type I IFN response in uncomplicated cases of dengue, in contrast to a blunted IFN profile in severe cases [1921]. Clearly, IFN serves as an early barrier against infection in mild cases of dengue, but is overridden or no longer functional in severe forms of the disease. A dampened IFN response could additionally cause the enhanced viremia associated with severe dengue [2]. This differential immune response can be accounted for by an altered profile of IFN-producing innate immune cells during disease progression. For example, expansion of TLR-expressing monocytes was observed in mild, but not severe forms of dengue [22], and plasmacytoid dendritic cells decreased at an accelerated rate in DHF compared to DF patients [23]. It is not clear how these immune cells are differentially modulated, but it would certainly be an interesting aspect of dengue research. Suppressed IFN expression in severe dengue can also be explained by the antibody-dependent enhancement theory. According to Ubol et al. [24], DV-antibody complexes trigger negative regulators that disable IFN production in monocytes. Consistent with this finding is the observation that IFNβ, RLH and IPS-1 levels were downregulated in peripheral blood mononuclear cells of secondary DHF patients but not in secondary DF patients [24]. But since both groups have pre-existing antibodies to DV, other components such as antibody type may factor in on the repressed type I IFN profile of severe secondary dengue infections. For example, the overall effect on IFN response may vary from patient to patient, depending on the combination of enhancing and neutralizing antibodies present in the host.

For the type II IFN response, the correlation between IFNγ levels and disease severity is not so clear. Bozza et al. [7] observed higher IFNγ levels in severe versus mild dengue, and suggested its predictive utility for disease severity. Kadhiravan et al. [25] and Restrepo et al. [26] also observed higher IFNγ levels in DHF/DSS patients. However, in another study, IFNγ was higher in primary versus secondary infections but did not correlate with severity [27]. This inconsistency may be due to differences in study design, population sampling, and timing of cytokine measurements, which is difficult to standardize. Thus, sequential measurement of cytokine levels is a better option for a thorough analysis of IFN levels. Using this approach, Libraty et al. [28] and Priyadarshini et al. [29] observed earlier peak plasma IFNγ among DHF patients compared to DF patients. Furthermore, a significant number of DHF cases with plasma leakage had increased levels of IFNγ, indicating its role in dengue pathogenesis [29]. Possible sources of IFNγ are NK cells and T cells, which are highly activated in DHF versus DF patients [30, 31]. IFNγ modulates the microenvironment of immune cells by enhancing the activation of DV-infected dendritic cells (DC) and the release of IL-12, a T-cell activating cytokine [32]. IFNγ has also been shown to augment antibody-mediated dengue infection of monocytic cells and DCs [3234]. Furthermore, IFNγ directly affects endothelial cell permeability [35, 36]. A combination of these effects is a fine recipe for a dysfunctional immune system. Hence, although IFNγ is originally designed to protect against disease [18], it may potentially inflict damage on the host, depending on the timing and level of production, and in concert with other pathogenic events of dengue infection.

A novel insight into the relationship between IFN and plasma leakage has been explored in an in vitro study on endothelial cells [37]. DV infection imparted a protective effect against TNFα-mediated hyperpermeability of endothelial cells at early periods of infection, but this protective effect diminished after several days of infection. The authors further proved that IFNβ mediates this protective activity against vascular permeability. But as IFNβ production wanes at later phases of infection, endothelial cells become more sensitized to TNFα-induced permeability. Indeed, the addition of recombinant IFNβ during late-stage infection restored the endothelial cells to normal permeability [37]. Type I IFNs have been shown to stabilize the vascular barrier in various studies [3841]. Moreover, IFNα and IFNβ protect against IFNγ-induced endothelial permeability [42]. Based on these findings, it is tempting to speculate on a model for IFN-mediated pathogenesis of dengue. IFNγ, as well as other unknown factors, may be responsible for plasma leakage in dengue patients. However, a strong type I IFN response in mild dengue protects against vascular permeability, aside from controlling viral replication. On the other hand, a muted type I IFN response in severe dengue is unable to block either infection or plasma leakage. While this hypothesis sounds promising, it is challenged by the lack of an appropriate animal model for dengue disease [43].

2-2. Cytokines

The cytokine storm theory proposed for dengue claims that a dysregulated production of cytokines during DV infection contributes to the disease. These cytokines are believed to be a product of massive T cell activation [44, 45]. According to the “original antigenic sin” proposed by Mongkolsapaya et al. [46], preferential expansion of memory T cells from primary infection over high-affinity T cells for the current infection promotes cytokine responses that imperil rather than protect the host. However, this does not explain DHF/DSS in infants with primary infection. Upregulated levels of cytokines have also been reported for dengue patients at the age of less than 1 year [4749], indicating that altered cytokine profiles are not solely directed by the cell-mediated immune response. It can be hypothesized that instead of T cells, maternal antibody-enhanced viremia is responsible for the increased cytokine response in infant cases of dengue [47, 48], most likely from cells of the innate immune system which are targets for initial replication of DV. For example, in vitro experiments show that monocytes and DCs could act as important sources of cytokines during DV infection [5052]. Accordingly, Chen et al. [51] proposed a model for dengue pathogenesis in which cytokine production by monocytes during early DV infection triggers a cascade of events that eventually leads to an augmented cytokine response, thereby causing vascular permeability.

The current challenge of dengue research is to identify soluble factors that mediate pathogenesis, and a number of candidate cytokines have been identified. The reader is referred to other articles for a more comprehensive overview regarding these molecules [5355]. Despite these leads, the definite cytokine determinant of dengue severity is still not established. Thus, an alternative approach is to identify the central regulator of cytokine expression during DV infection. So far, two such putative regulators have been identified. DV infection of macrophage inhibitory factor (MIF)-deficient mice produced a less severe clinical disease, with reduced proinflammatory cytokine levels [56]. Moreover, anti-MIF antibodies reduced cytokine expression in DV-infected macrophages [56]. These results suggest the involvement of MIF in the amplification of cytokine responses that inflict damage on the host. Elevated levels of MIF have been detected in severe dengue [56, 57], and macrophages and hepatocytes were identified as sources of this cytokine [56]. CLEC5A is another molecule implicated in dengue pathogenesis. CLEC5A is a C-type lectin expressed exclusively on monocytes and macrophages. Knockdown of CLEC5A suppressed monocyte production of proinflammatory cytokines without any effect on viral entry [58]. Furthermore, treatment of anti-CLEC5A antibodies in DV-infected STAT-deficient mice abrogated pro-inflammatory cytokine expression without affecting viral replication, prevented hemorrhage and vascular permeability, and reduced mortality [58]. Hence, CLEC5A may also act as a central regulator of pro-inflammatory cytokine responses during DV infection. However, this hypothesis contrasts with a recent study, which reports that CLEC5A expression is downregulated in dengue patients [59]. Hence, more extensive clinical data is required to confirm the importance of CLEC5A in dengue pathogenesis.

Another aspect of the cytokine storm theory that requires evaluation is how the cytokine storm develops and causes disease. One relevant hypothesis is that cytokines may not necessarily behave in a linear fashion but rather as a complex network of events. Chen et al. [51] proposed that cytokine production during DV infection occurs in a hierarchichal manner, progressing from a local gradient derived from monocytic cells and expanding further as other immune cells are recruited and activated. This complex nature of cytokines makes it difficult, if not impossible, to pinpoint the exact mediator of dengue severity.

To complicate things further, cytokines may not act alone but instead exert a synergistic effect in unison. For example, IL-4 exerts a synergistic effect on endothelial cell permeability when combined with either TNFα or IFNγ [60], and increased expression of all three cytokines has been reported in severe dengue [7, 25, 26, 52, 61, 62]. Hence, aside from increased cytokine levels, the cytokine profile of dengue patients should also be investigated. When patients with severe dengue were compared, the primary infection group (infants less than 1 year of age) and the secondary infection group (older children) similarly had elevated levels of IFNγ and IL-10, but IL-6 and TNFα were additionally upregulated in infants [48]. A specific combination of cytokines, instead of a solo cytokine, may be a prerequisite for disease development.

The timing of cytokine production also plays a role in the evolution of disease. To illustrate, IL-6 and IL-8 levels increased earlier in DHF compared to DF cases [29]. IL-6 levels were associated with the presence of pleural effusion/ascites in DHF, while IL-8 correlated with thrombocytopenia and increased serum alanine transaminase levels, an indicator of hepatic injury [29]. IL-8 has also been shown to mediate vascular permeability during DV infection [63]. The early appearance of these cytokines in DHF could enhance or hasten immunopathological events that lead to disease progression.

Finally, viral load also has an influence on the induction potential of cytokines. The minimum viral inoculation level required for cytokine expression in monocytes varies to a significant degree. While IL-8, MIP-1α and RANTES could be induced by a small viral input, TNFα and IL-1β demanded high doses of DV [51]. Thus, certain cytokines are readily inducible during the initial phases of DV infection, while others remain unexpressed or unaltered until high-titer viremia or tissue viral load is established. This probably explains the hierarchical expression of cytokines during DV infection, and thereby justifies the comprehensive analysis of cytokine profiles at different periods of illness to improve our understanding of dengue pathogenesis.

3. Mosquito-derived immunomodulators in flavivirus infection

When the host is bitten by a pathogen-transmitting arthropod, it also encounters saliva-associated factors. Arthropod saliva has been shown to manipulate host hemostasis in order to facilitate blood feeding. Almost all blood-feeding arthropods studied so far have at least one anti-clotting, one vasodilator, and one anti-platelet compound [64]. But research also shows that the saliva of ticks, blackflies, sand flies and mosquitoes have the capacity to regulate the host immune system [6469], indicating that immunomodulation may be common among hematophagous arthropods. For arthropods that live in long-term, close association with their host (e.g. ticks), immune regulation serves the obvious purpose of averting host reactions that impede feeding and survival. But for rapid feeders like mosquitoes, the arthropod has already terminated contact with the host by the time the immune defense reaches its climax. Thus, Schneider and Higgs [70] proposed two hypotheses to explain the immunomodulatory activity of mosquito saliva. Since the host is frequently exposed to mosquitoes, immune reactions to a previous exposure must be modeled to allow subsequent feeding. Alternatively, these inflammation and immune responses are by-products of mosquito anti-hemostatic activities, since these three physiological pathways are closely intertwined. In the context of arthropod-borne infections, vector saliva may alter immune reactions in a way that can influence pathogen transmission and establishment, as has been demonstrated for Leishmania major, Plasmodium yoleii, Cache Valley virus and vesicular stomatitis virus [7174]. Thus, mosquito saliva has the potential to direct the course of flavivirus infections, specifically by interrupting the innate immune response. This section evaluates the impact of soluble mediators from mosquito saliva on the earliest process of dengue infection, as the virus initially establishes itself in the host.

Although research has been done on various mosquitoes, this review focuses on Aedes aegypti, the most important vector for dengue, with reference to other vectors. Mosquito feeding or saliva from A. aegypti creates a shift from Th1 cytokine to Th2 cytokine expression. Th1 cytokines such as IFNγ and IL-2 were downregulated [68, 7578], while Th2 cytokines such as IL-4, IL-5 and IL-10 were either upregulated or unaffected [68, 75, 76, 78, 79], although in one case IL-10 was downregulated [77]. This is attributable to differences in preparation and/or dose of salivary gland extract (SGE) and the mice used. Culex pipiens feeding also favored Th2 cytokine responses [78]. A Th1-to-Th2 shift in cytokine response has been observed in severe dengue [25, 80], but there is no direct evidence so far linking this to mosquito bites. Th1 cytokines stimulate a proinflammatory response to kill intracellular parasites. On the other hand, Th2 cytokines have a counteractive effect on Th1 cytokines, and their function is targeted toward extracellular pathogens. Hence, a Th2-skewed cytokine response during mosquito feeding would benefit viral transmission and initial establishment in the host. In support of this, reconstitution of Th1 cytokines during vector feeding restored innate immunity and restricted infection by Borrelia burgdorferi, an intracellular spirochete, in mice [81]. Furthermore, other antiviral mediators such as IFNβ, iNOS and TLR3 were suppressed by mosquito feeding/saliva [75, 76, 79].

T cells are also largely affected by mosquito saliva. For example, A. aegypti SGE restricted splenocyte proliferation by inducing cell death and arresting cell division, with T lymphocytes as the most susceptible population [68, 77]. Dermal recruitment of T cells was also inhibited when mice were subjected to mosquito feeding prior to WNV infection [79]. Thus, mosquito bite-mediated T cell suppression can be exploited by DV for transmission and survival. In contrast, SGE from Culex quinquefasciatus did not affect T cell proliferation [77], although its effect on other immune cells was not investigated. This differential activity is probably due to biological differences between the two vectors, which may eventually influence host preference of the transmitted pathogen. For example, Aedes-transmitted viruses primarily cycle between mosquitoes and primates, and cause hemorrhagic disease. On the other hand, Culex-transmitted viruses usually cycle between mosquitoes and birds, and cause neurological disease in humans, who serve as dead-end hosts. Therefore, further studies are needed to look into the role of mosquito-dependent immunomodulation on host tropism and pathogenesis of arbovirus infections.

An increasing number of molecules responsible for blood feeding have already been identified in mosquito saliva, but the immunomodulatory components have yet to be determined. In the case of Anopheles stephensi, Owhashi and his group [82, 83] were able to isolate a neutrophil chemotactic factor and an eosinophil chemotactic factor. Zeidner et al. [78] attributed the cytokine-modulating activity of A. aegypti and C. pipiens saliva to sialokinins. Sialokinins are vasodilatory molecules released into the saliva. However, these molecules were also able to inhibit the production of IL-2 and IFNγ while enhancing IL-10 and IL-4 expression [78]. Thus, aside from promoting blood feeding, sialokinins may have a secondary function in immune modulation. Another mosquito salivary protein with immunomodulatory property was recently isolated from A. aegypti and identified as SAAG-4. SAAG-4 reduced IFNγ but enhanced IL-4 response in CD4+ T cells [84]. Interestingly, both sialokinin and SAAG-4 direct a Th2-skewed pattern of cytokine response. Hence, certain substances from mosquito saliva have the capacity for host immunoregulation.

Such immunological activity in mosquito saliva compels us to contemplate on its implications for pathogen infection and disease development. Scientific investigations along this line have been initiated for various arthropod-borne pathogens, including flaviviruses. Unfortunately, the majority of flavivirus work is conducted on West Nile Virus (WNV). Nevertheless, these findings should lay a foundation for our knowledge regarding mosquito-flavivirus-host interactions. Mosquito feeding/saliva-mediated enhancement of WNV infection has been demonstrated in mice using Aedes aegypti and Culex tarsalis [79, 85, 86], and in chicken using C. pipiens [87]. In the mouse studies, mosquito feeding increased viremia and accelerated neuroinvasion compared to needle injection. When mosquito feeding was artificially mimicked in mice using SGE treatment prior to needle infection, the enhanced infection was similarly achieved [85, 86]. But when SGE was applied at a site distal from the infection, the enhancing effect was lost [86]. These results indicate that mosquito saliva is responsible for the augmented infection and that this activity is mediated locally. However, the effect of mosquito saliva on mortality is not clear. Schneider et al. [85] described a lower survival rate among mosquito fed-WNV infected mice, but Styer et al. [86] did not observe any differences in morbidity, mortality, onset of disease or survival time. This discrepancy may be due to differences in the mosquitoes or mice used or to the experimental methods employed. But it is also possible that mosquito feeding may enhance viral replication without affecting the outcome of disease. Further studies should clarify the role of mosquito-virus-host interactions on immunopathogenesis of flaviviral infections.

Other findings do not support this mosquito saliva-mediated enhancement theory of flavivirus infection. Mosquito feeding did not alter WNV infection in hamsters [88], nor did it affect the viremia and antibody response of St. Louis encephailitis virus-infected chicken and house finches [89]. These findings can be easily explained by differences in experimental methodology. Alternatively, saliva-mediated enhancement may be determined by the combination of mosquito, virus and host species. Hence, saliva-mediated enhancement may be or not be a universal feature of flavivirus infections. In the case of DV, only one study on mosquito saliva has been conducted. A. aegypti saliva inhibited DV infection of human myeloid DCs, and this effect was augmented when cells were pre-sensitized with saliva, leading the authors to conclude that mosquito saliva has instead an antiviral function [90]. At this point, it is difficult to generalize since the effect of mosquito saliva on DV infection in other susceptible cells or in vivo has not yet been investigated. To resolve this, more extensive research on mosquito saliva and its role in DV infection and pathogenesis is recommended. Nevertheless, mosquito saliva has immunomodulatory effects that can be exploited by DV to enhance transmission and infection. Although there is no compelling evidence for this as yet, the findings based on WNV emphasize the impact of vector feeding on early events of flavivirus infection.

4. Prospects for dengue management and therapy

The identification of soluble factors required for dengue infection and pathogenesis would facilitate the development of anti-viral and disease management strategies. Until now, no therapeutic drug has been developed for dengue, and the success of hospital treatment relies heavily on careful observation and supportive care. With up to 50 million DV infections every year [91], the race is on to find an improved intervention method for dengue.

The success of IFN in hepatitis B and C virus therapy prompted some researchers to look into its utility for dengue. IFNα has been shown to inhibit DV replication in vitro using various cell lines [9294]. To assess its effectiveness in vivo, two types of rIFN-α-2a (nonpegylated and pegylated) were administered in DV-infected rhesus monkeys one day after the onset of viremia [92]. The pegylated form has a covalently conjugated 40kD branched methoxy-polyethylene glycol (PEG) molecule, which prolongs the systemic prevalence of rIFN-α-2a. A single administration of either preparations inhibited DV replication to some extent, but not completely [92]. The nonpegylated form delayed viremia, while the pegylated form slightly reduced viral titers. In future studies, a combination of the two forms is recommended to potentiate absolute clearance of the virus. rIFN-α-2a surpasses other antiviral drugs for its clinical safety and tolerability, especially among children [95]. Furthermore, if a single dose administration is proven to be effective, IFN treatment would be simple and affordable. A combination therapy with other antiviral drugs may also increase its potency. However, since dengue viremia peaks within the first 72 hours of illness [2, 32], early intervention is an important prerequisite for the success of this strategy.

Another therapeutic approach is to neutralize cytokines/cytokine mediators that promote disease. For example, antibody treatment against MIF and CLEC5a has been suggested as a method of intervention. Using this strategy in mice, cytokine production was reduced, and the clinical outcome of DV infection was improved, suggesting its potential for application in human dengue patients [56, 58]. Other soluble factors are also being considered for similar purposes, but the complex nature of cytokines precludes the identification of a single target that can generate promising results. Moreover, the intricate networks that link cytokines raise the possibility of undesirable physiological effects on the human body. Finally, whether the host is still capable of viral clearance when certain cytokines are neutralized is a risk that should be given proper consideration. Alternatively, identification of soluble factors that mediate dengue severity could be applied as clinical and laboratory tools for predicting disease, which is integral to dengue management strategies, especially in endemic areas.

5. Conclusion

Dengue pathogenesis is a multifaceted phenomenon, initially dictated by complex interactions between the virus, vector and host, thereby resulting in a modulated immune response. Although the adaptive immune response is usually associated with dengue pathogenesis, the innate immune system has much to contribute as well, especially in early events of infection. The innate immune reaction to dengue is characterized by the production of soluble factors that shape the early events of infection to favor or counter the virus. Soluble mediators may also influence disease evolution, most likely by operating on an intricate network of reactions. Since the host operates on a critical balance of immune responses, a disrupted equilibrium mounted by such soluble mediators could facilitate in disease development. The current challenge is to advance the conventional simplistic approach of dengue immunopathogenesis research towards a holistic strategy, which could better assist in the development of rational, practical and effective methods for dengue diagnosis and intervention.

References

  • 1.Thomas S, Strickman D, Vaughn DW. Dengue epidemiology: virus epidemiology, ecology, and emergence. In: Chambers TJ and Monath TP, ed. Advances in virus research: The flaviviruses: detection, diagnosis and vaccine development. 1st ed. vol. 61. San Diego, California: Elsevier Academic Press; 2003. pp. 235-290. [DOI] [PubMed]
  • 2.Vaughn DW, Green S, Kalayanarooj S, Innis BL, Nimmannitya S, Suntayakorn S, Endy TP, Raengsakulrach B, Rothman AL, Ennis FA, Nisalak A. Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity. J Infect Dis 2000; 181: 2–9 [DOI] [PubMed] [Google Scholar]
  • 3.Marovich M, Grouard-Vogel G, Louder M, Eller M, Sun W, Wu SJ, Putvatana R, Murphy G, Tassaneetrithep B, Burgess T, Birx D, Hayes C, Schlesinger-Frankel S, Mascola J. Human dendritic cells as targets of dengue virus infection. J Investig Dermatol Symp Proc 2001; 6: 219–224 [DOI] [PubMed] [Google Scholar]
  • 4.Wu SJ, Grouard-Vogel G, Sun W, Mascola JR, Brachtel E, Putvatana R, Louder MK, Filgueira L, Marovich MA, Wong HK, Blauvelt A, Murphy GS, Robb ML, Innes BL, Birx DL, Hayes CG, Frankel SS. Human skin Langerhans cells are targets of dengue virus infection. Nat Med 2000; 6: 816–820 [DOI] [PubMed] [Google Scholar]
  • 5.Johnston LJ, Halliday GM, King NJ. Langerhans cells migrate to local lymph nodes following cutaneous infection with an arbovirus. J Invest Dermatol 2000; 114: 560–568 [DOI] [PubMed] [Google Scholar]
  • 6.Jessie K, Fong MY, Devi S, Lam SK, Wong KT. Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization. J Infect Dis 2004; 189: 1411–1418 [DOI] [PubMed] [Google Scholar]
  • 7.Bozza FA, Cruz OG, Zagne SMO, Azeredo EL, Nogueira RMR, Assis EF, Bozza PT, Kubelka CF. Multiplex cytokine profile from dengue patients: MIP-1beta and IFN-gamma as predictive factors for severity. BMC Infectious Diseases 2008; 8: 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Burke DS, Nisalak A, Johnson DE, Scott RM. A Prospective Study of Dengue Infections in Bangkok. Am J Trop Med Hyg 1988; 38: 172–180 [DOI] [PubMed] [Google Scholar]
  • 9.Halstead SB, Nimmannitya S, Cohen SN. Observations related to pathogenesis of dengue hemorrhagic fever. IV. Relation of disease severity to antibody response and virus recovered. Yale J Biol Med 1970; 42: 311–328 [PMC free article] [PubMed] [Google Scholar]
  • 10.Russell PK, Yuill TM, Nisalak A, Udomsakdi S, Gould DJ, Winter PE. An insular outbreak of dengue hemorrhagic fever: II. Virologic and serologic studies. Am J Trop Med Hyg 1968; 17: 600–608 [DOI] [PubMed] [Google Scholar]
  • 11.Sangkawibha N, Rojanasuphot S, Ahandrik S, Viriyapongse S, Jatanasen S, Salitul V, Phanthumachinda B, Halstead SB. Risk factors in dengue shock syndrome: a prospective epidemiologic study in Rayong, Thailand. I. The 1980 outbreak. Am J Epidemiol 1984; 120: 653–669 [DOI] [PubMed] [Google Scholar]
  • 12.Becquart P, Wauquier N, Nkoghe D, Ndjoyi-Mbiguino A, Padilla C, Souris M, Leroy EM. Acute dengue virus 2 infection in Gabonese patients is associated with an early innate immune response, including strong interferon alpha production. BMC Infectious Diseases 2010; 10: 356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hoang LT, Lynn DJ, Henn M, Birren BW, Lennon NJ, Le PT, Duong KT, Nguyen TT, Mai LN, Farrar JJ, Hibberd ML, Simmons CP. The early whole-blood transcriptional signature of dengue virus and features associated with progression to dengue shock syndrome in Vietnamese children and young Adults. J Virol 2010; 84: 12982–12994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kurane I, Innis BL, Nimmannitya S, Nisalak A, Meager A, Ennis FA. High levels of interferon alpha in the sera of children with dengue virus infection. Am J Trop Med Hyg 1993; 48: 222–229 [DOI] [PubMed] [Google Scholar]
  • 15.Perry ST, Prestwood TR, Lada SM, Benedict CA, Shresta S. Cardif-mediated signaling controls the initial innate response to dengue virus in vivo. J Virol 2009; 83: 8276–8281 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Espada-Murao LA, Morita K. Delayed cytosolic exposure of Japanese encephalitis virus double-stranded RNA impedes interferon activation and enhances viral dissemination in porcine cells. J Virol 2011; 85: 6736–6749 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sun P, Fernandez S, Marovich MA, Palmer DR, Celluzzi CM, Boonnak K, Liang Z, Subramanian H, Porter KR, Sun W, Burgess TH. Functional characterization of ex vivo blood myeloid and plasmacytoid dendritic cells after infection with dengue virus. Virol 2009; 383: 207–215 [DOI] [PubMed] [Google Scholar]
  • 18.Shresta S, Kyle JL, Snider HM, Basavapatna M, Beatty PR, Harris E. Interferon-dependent immunity is essential for resistance to primary dengue virus infection in mice, whereas T- and B-cell-dependent immunity are less critical. J Virol 2004; 78: 2701–2710 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Long HT, Hibberd ML, Hien TT, Dung NM, Van Ngoc T, Farrar J, Wills B, Simmons CP. Patterns of gene transcript abundance in the blood of children with severe or uncomplicated dengue highlight differences in disease evolution and host response to dengue virus infection. J Infect Dis 2009; 199: 537–546 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Simmons CP, Popper S, Dolocek C, Chau TN, Griffiths M, Dung NT, Long TH, Hoang DM, Chau NV, Thao le TT, Hien TT, Relman DA, Farrar J. Patterns of host genome-wide gene transcript abundance in the peripheral blood of patients with acute dengue hemorrhagic fever. J Infect Dis 2007; 195: 1097–1107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ubol S, Masrinoul P, Chaijaruwanich J, Kalayanarooj S, Charoensirisuthikul T, Kasisith J. Differences in global gene expression in peripheral blood mononuclear cells indicate a significant role of the innate responses in progression of dengue fever but not dengue hemorrhagic fever. J Infect Dis 2008; 197: 1459–1467 [DOI] [PubMed] [Google Scholar]
  • 22.Azeredo EL, Neves-Souza PC, Alvarenga AR, Reis SRNI, Torrentes-Carvalho A, Zagne S-MO, Nogueira RMR, Oliveira-Pinto LM, Kubelka CF. Differential regulation of toll-like receptor-2, toll-like receptor-4, CD16 and human leucocyte antigen-DR on peripheral blood monocytes during mild and severe dengue fever. Immunology 2010; 130: 202–216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pichyangkul S, Endy TP, Kalayanarooj S, Nisalak A, Yongvanitchit K, Green S, Rothman AL, Ennis FA, Libraty DH. A blunted blood plasmacytoid dendritic cell response to an acute systemic viral infection is associated with increased disease severity. J Immunol 2003; 171: 5571–5578 [DOI] [PubMed] [Google Scholar]
  • 24.Ubol S, Phuklia W, Kalayanarooj S, Modhiran N. Mechanisms of immune evasion induced by a complex of dengue virus and preexisting enhancing antibodies. J Infect Dis 2010; 201: 923–935 [DOI] [PubMed] [Google Scholar]
  • 25.Kadhiravan T, Saxena A, Singh A, Broor S, Sharma SK, Mitra DK. Association of intracellular TH1-TH2 Balance in CD4+ T-cells and MIP-1α in CD8+ T-cells with disease severity in adults with dengue. Immune Netw 2010; 10: 164–172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Restrepo BN, Ramirez RE, Arboleda M, Alvarez G, Ospina M, Diaz FJ. Serum levels of cytokines in two ethnic groups with dengue virus infection. Am J Trop Med Hyg 2008; 79: 673–677 [PubMed] [Google Scholar]
  • 27.Chakravarti A, Kumaria R. Circulating levels of tumour necrosis factor-alpha & interferon-gamma in patients with dengue & dengue haemorrhagic fever during an outbreak. Indian J Med Res 2006; 123: 25–30 [PubMed] [Google Scholar]
  • 28.Libraty DH, Endy TP, Houng HS, Green S, Kalayanarooj S, Suntayakorn S, Chansiriwongs W, Vaughn DW, Nisalak AA, Ennis FA, Rothman AL. Differing influences of virus burden and immune activation on disease severity in secondary dengue-3 virus infections. J Infect Dis 2002; 185: 1213–1221 [DOI] [PubMed] [Google Scholar]
  • 29.Priyadarshini D, Gadia RR, Tripathy A, Gurukumar KR, Bhagat A, Patwardhan S, Mokashi N, Vaidya D, Shah PS, Cecilia D. Clinical findings and pro-inflammatory cytokines in dengue patients in Western India: a facility-based study. PLoS One 2010; 5: e8709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Green S, Pichyangkul S, Vaughn DW, Kalayanarooj S, Nimmannitya S, Nisalak A, Kurane I, Rothman AL, Ennis FA. Early CD69 expression on peripheral blood lymphocytes from children with dengue hemorrhagic fever. J Infect Dis 1999; 180: 1429–1435 [DOI] [PubMed] [Google Scholar]
  • 31.Kurane I, Innis BL, Nimmannitya S, Nisalak A, Meager A, Janus J, Ennis FA. Activation of T lymphocytes in dengue virus infections. High levels of soluble interleukin 2 receptor, soluble CD4, soluble CD8, interleukin 2, and interferon-gamma in sera of children with dengue. J Clin Invest 1991; 88: 1473–1480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Libraty DH, Pichyangkul S, Ajariyakhajorn C, Endy TP, Ennis FA. Human dendritic cells are activated by dengue virus infection: enhancement by gamma interferon and implications for disease pathogenesis. J Virol 2001; 75: 3501–3508 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kontny U, Kurane I, Ennis FA. Gamma interferon augments Fc gamma receptor-mediated dengue virus infection of human monocytic cells. J Virol 1988; 62: 3928–3933 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kurane I, Innis BL, Nimmannitya S, Nisalak A, Rothman AL, Livingston PG, Janus J, Ennis FA. Human immune responses to dengue viruses. Southeast Asian J Trop Med Public Health 1990; 21: 658–662 [PubMed] [Google Scholar]
  • 35.Blum MS, Toninelli E, Anderson JM, Balda MS, Zhou J, O’Donnell L, Pardi R, Bender JR. Cytoskeletal rearrangement mediates human microvascular endothelial tight junction modulation by cytokines. Am J Physiol 1997; 273: H286–H294 [DOI] [PubMed] [Google Scholar]
  • 36.Dewi BE, Takasaki T, Kurane I. In vitro assessment of human endothelial cell permeability: effects of inflammatory cytokines and dengue virus infection. J Virol Methods 2004; 121: 171–180 [DOI] [PubMed] [Google Scholar]
  • 37.Liu P, Woda M, Ennis FA, Libraty DH. Dengue Virus Infection Differentially Regulates Endothelial Barrier Function over Time through Type I Interferon Effects. J Infect Dis 2009; 200: 191–201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.de Boer AG, Gaillard PJ. Blood-brain barrier dysfunction and recovery. J Neural Transm 2006; 113: 455–462 [DOI] [PubMed] [Google Scholar]
  • 39.Kiss J, Yegutkin GG, Koskinen K, Savunen T, Jalkanen S, Salmi M. IFN-beta protects from vascular leakage via up-regulation of CD73. Eur J Immunol 2007; 37: 3334–3338 [DOI] [PubMed] [Google Scholar]
  • 40.Kraus J, Ling AK, Hamm S, Voigt K, Oschmann P, Engelhardt B. Interferon-beta stabilizes barrier characteristics of brain endothelial cells in vitro. Ann Neurol 2004; 56: 192–205 [DOI] [PubMed] [Google Scholar]
  • 41.Kraus J, Voigt K, Schuller AM, Scholz M, Kim KS, Schilling M, Schäbitz WR, Oschmann P, Engelhardt B. Interferon-beta stabilizes barrier characteristics of the blood-brain barrier in four different species in vitro. Mult Scler 2008; 14: 843–852 [DOI] [PubMed] [Google Scholar]
  • 42.Minagar A, Long A, Ma T, Jackson TH, Kelley RE, Ostanin DV, Sasaki M, Warren AC, Jawahar A, Cappell B, Alexander JS. Interferon (IFN)-beta 1a and IFN-beta 1b block IFN-gamma-induced disintegration of endothelial junction integrity and barrier. Endothelium 2003; 10: 299–307 [DOI] [PubMed] [Google Scholar]
  • 43.Bente DA, Rico-Hesse R. Models of dengue virus infection. Drug Discov Today Dis Models 2006; 3: 97–103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Dong T, Moran E, Vinh Chau N, Simmons C, Luhn K, Peng Y, Wills B, Phuong Dung N, Thi Thu Thao L, Hien TT, McMichael A, Farrar J, Rowland-Jones S. High pro-inflammatory cytokine secretion and loss of high avidity cross-reactive cytotoxic T-cells during the course of secondary dengue virus infection. PLoS One 2007; 2: e1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rothman AL. Cellular immunology of sequential dengue virus infection and its role in disease pathogenesis. Curr Top Microbiol Immunol 2010; 338: 83–98 [DOI] [PubMed] [Google Scholar]
  • 46.Mongkolsapaya J, Dejnirattisai W, Xu XN, Vasanawathana S, Tangthawornchaikul N, Chairunsri A, Sawasdivorn S, Duangchinda T, Dong T, Rowland-Jones S, Yenchitsomanus PT, McMichael A, Malasit P, Screaton G. Original antigenic sin and apoptosis in the pathogenesis of dengue hemorrhagic fever. Nat Med 2003; 9: 921–927 [DOI] [PubMed] [Google Scholar]
  • 47.Chau TN, Quyen NT, Thuy TT, Tuan NM, Hoang DM, Dung NT, Lien le B, Quy NT, Hieu NT, Hieu LT, Hien TT, Hung NT, Farrar J, Simmons CP. Dengue in Vietnamese infants-results of infection-enhancement assays correlate with age-related disease epidemiology, and cellular immune responses correlate with disease severity. J Infect Dis 2008; 198: 516–524 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nguyen TH, Lei HY, Nguyen TL, Lin YS, Huang KJ, Le BL, Lin CF, Yeh TM, Do QH, Vu TQ, Chen LC, Huang JH, Lam TM, Liu CC, Halstead SB. Dengue hemorrhagic fever in infants: a study of clinical and cytokine profiles. J Infect Dis 2004; 189: 221–232 [DOI] [PubMed] [Google Scholar]
  • 49.Restrepo BN, Isaza DM, Salazar CL, Ramírez R, Ospina M, Alvarez LG. Serum levels of interleukin-6, tumor necrosis factor-alpha and interferon-gamma in infants with and without dengue. Rev Soc Bras Med Trop 2008; 41: 6–10 [DOI] [PubMed] [Google Scholar]
  • 50.Becerra A, Warke RV, Martin K, Xhaja K, de Bosch N, Rothman AL, Bosch I. Gene expression profiling of dengue infected human primary cells identifies secreted mediators in vivo. J Med Virol 2009; 81: 1403–1411 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chen YC, Wang SY. Activation of terminally differentiated human monocytes/macrophages by dengue virus: productive infection, hierarchical production of innate cytokines and chemokines, and the synergistic effect of lipopolysaccharide. J Virol 2002; 76: 9877–9887 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Levy A, Valero N, Espina LM, Añez G, Arias J, Mosquera J. Increment of interleukin 6, tumour necrosis factor alpha, nitric oxide, C-reactive protein and apoptosis in dengue. Trans R Soc Trop Med Hyg 2010; 104: 16–23 [DOI] [PubMed] [Google Scholar]
  • 53.Fink J, Gu F, Vasudevan SG. Role of T cells, cytokines and antibody in dengue fever and dengue haemorrhagic fever. Rev Med Virol 2006; 16: 263–275 [DOI] [PubMed] [Google Scholar]
  • 54.Martina BEE, Koraka P, Osterhaus ADME. Dengue virus pathogenesis: an integrated view. Clin Microbiol Rev 2009; 22: 564–581 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Pang T, Cardosa MJ, Guzman MG. Of cascades and perfect storms: the immunopathogenesis of dengue haemorrhagic fever-dengue shock syndrome (DHF/DSS). Immunol Cell Biol 2007; 85: 43–45 [DOI] [PubMed] [Google Scholar]
  • 56.Assunção-Miranda I, Amaral FA, Bozza FA, Fagundes CT, Sousa LP, Souza DG, Pacheco P, Barbosa-Lima G, Gomes RN, Bozza PT, Da Poian AT, Teixeira MM, Bozza MT. Contribution of macrophage migration inhibitory factor to the pathogenesis of dengue virus infection. FASEB J 2010; 24: 218–228 [DOI] [PubMed] [Google Scholar]
  • 57.Chen LC, Lei HY, Liu CC, Shiesh SC, Chen SH, Liu HS, Lin YS, Wang ST, Shyu HW, Yeh TM. Correlation of serum levels of macrophage migration inhibitory factor with disease severity and clinical outcome in dengue patients. Am J Trop Med Hyg 2006; 74: 142–147 [PubMed] [Google Scholar]
  • 58.Chen ST, Lin YL, Huang MT, Wu MF, Cheng SC, Lei HY, Lee CK, Chiou TW, Wong CH, Hsieh SL. CLEC5A is critical for dengue-virus-induced lethal disease. Nature 2008; 453: 672–676 [DOI] [PubMed] [Google Scholar]
  • 59.Gomes AL, Wee LJ, Khan AM, Gil LH, Marques ET, Jr, Calzavara-Silva CE, Tan TW. Classification of dengue fever patients based on gene expression data using support vector machines. PLoS One 2010; 5: e11267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Beynon HL, Haskard DO, Davies KA, Haroutunian R, Walport MJ. Combinations of low concentrations of cytokines and acute agonists synergize in increasing the permeability of endothelial monolayers. Clin Exp Immunol 1993; 91: 314–319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Gagnon SJ, Mori M, Kurane I, Green S, Vaughn DW, Kalayanarooj S, Suntayakorn S, Ennis FA, Rothman AL. Cytokine gene expression and protein production in peripheral blood mononuclear cells of children with acute dengue virus infections. J Med Virol 2002; 67: 41–46 [DOI] [PubMed] [Google Scholar]
  • 62.Houghton-Triviño N, Salgado DM, Rodríguez JA, Bosch I, Castellanos JE. Levels of soluble ST2 in serum associated with severity of dengue due to tumour necrosis factor alpha stimulation. J Gen Virol 2010; 91: 697–706 [DOI] [PubMed] [Google Scholar]
  • 63.Talavera D, Castillo AM, Dominguez MC, Gutierrez AE, Meza I. IL8 release, tight junction and cytoskeleton dynamic reorganization conducive to permeability increase are induced by dengue virus infection of microvascular endothelial monolayers. J Gen Virol 2004; 85: 1801–1813 [DOI] [PubMed] [Google Scholar]
  • 64.Ribeiro JM, Makoul GT, Levine J, Robinson DR, Spielman A. Antihemostatic, antiinflammatory, and immunosuppressive properties of the saliva of a tick, Ixodes dammini. J Exp Med 1985; 161: 332–344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ribeiro JM, Francischetti IM. Role of arthropod saliva in blood feeding: sialome and post-sialome perspectives. Annu Rev Entomol 2003; 48: 73–88 [DOI] [PubMed] [Google Scholar]
  • 66.Bissonnette EY, Rossignol PA, Befus AD. Extracts of mosquito salivary gland inhibit tumour necrosis factor alpha release from mast cells. Parasite Immunol 1993; 15: 27–33 [DOI] [PubMed] [Google Scholar]
  • 67.Cross ML, Cupp MS, Cupp EW, Galloway AL, Enriquez FJ. Modulation of murine immunological responses by salivary gland extract of Simulium vittatum (Diptera: Simuliidae). J Med Entomol 1993; 30: 928–935 [DOI] [PubMed] [Google Scholar]
  • 68.Cross ML, Cupp EW, Enriquez FJ. Differential modulation of murine cellular immune responses by salivary gland extract of Aedes aegypti. Am J Trop Med Hyg 1994; 51: 690–696 [DOI] [PubMed] [Google Scholar]
  • 69.Gillespie RD, Mbow ML, Titus RG. The immunomodulatory factors of bloodfeeding arthropod saliva. Parasite Immunol 2000; 22: 319–331 [DOI] [PubMed] [Google Scholar]
  • 70.Schneider BS, Higgs S. The enhancement of arbovirus transmission and disease by mosquito saliva is associated with modulation of the host immune response. Trans R Soc Trop Med Hyg 2008; 102: 400–408 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Belkaid Y, Kamhawi S, Modi G, Valenzuela J, Noben-Trauth N, Rowton E, Ribeiro J, Sacks DL. Development of a natural model of cutaneous leishmaniasis: powerful effects of vector saliva and saliva preexposure on the long-term outcome of Leishmania major infection in the mouse ear dermis. J Exp Med 1998; 188: 1941–1953 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Donovan MJ, Messmore AS, Scrafford DA, Sacks DL, Kamhawi S, McDowell MA. Uninfected mosquito bites confer protection against infection with malaria parasites. Infect Immun 2007; 75: 2523–2530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Edwards JF, Higgs S, Beaty BJ. Mosquito feeding-induced enhancement of Cache Valley Virus (Bunyaviridae) infection in mice. J Med Entomol 1998; 35: 261–265 [DOI] [PubMed] [Google Scholar]
  • 74.Limesand KH, Higgs S, Pearson LD, Beaty BJ. Potentiation of vesicular stomatitis New Jersey virus infection in mice by mosquito saliva. Parasite Immunol 2000; 22: 461–467 [DOI] [PubMed] [Google Scholar]
  • 75.Schneider BS, Soong L, Zeidner NS, Higgs S. Aedes aegypti salivary gland extracts modulate anti-viral and TH1/TH2 cytokine responses to sindbis virus infection. Viral Immunol 2004; 17: 565–573 [DOI] [PubMed] [Google Scholar]
  • 76.Thangamani S, Higgs S, Ziegler S, Vanlandingham D, Tesh R, Wikel S. Host immune response to mosquito-transmitted chikungunya virus differs from that elicited by needle inoculated virus. PLoS One 2010; 5: e12137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wanasen N, Nussenzveig RH, Champagne DE, Soong L, Higgs S. Differential modulation of murine host immune response by salivary gland extracts from the mosquitoes Aedes aegypti and Culex quinquefasciatus. Med Vet Entomol 2004; 18: 191–199 [DOI] [PubMed] [Google Scholar]
  • 78.Zeidner NS, Higgs S, Happ CM, Beaty BJ, Miller BR. Mosquito feeding modulates Th1 and Th2 cytokines in flavivirus susceptible mice: an effect mimicked by injection of sialokinins, but not demonstrated in flavivirus resistant mice. Parasite Immunol 1999; 21: 35–44 [DOI] [PubMed] [Google Scholar]
  • 79.Schneider BS, Soong L, Coffey LL, Stevenson HL, McGee CE, Higgs S. Aedes aegypti saliva alters leukocyte recruitment and cytokine signaling by antigen-presenting cells during West Nile virus infection. PLoS One 2010; 5: e11704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chaturvedi UC. Shift to Th2 cytokine response in dengue haemorrhagic fever. Indian J Med Res 2009; 129: 1–3 [PubMed] [Google Scholar]
  • 81.Zeidner NS, Schneider BS, Rutherford JS, Dolan MC. Suppression of Th2 cytokines reduces tick-transmitted Borrelia burgdorferi load in mice. J Parasitol 2008; 94: 767–769 [DOI] [PubMed] [Google Scholar]
  • 82.Owhashi M, Harada M, Suguri S, Ohmae H, Ishii A. The role of saliva of Anopheles stephensi in inflammatory response: identification of a high molecular weight neutrophil chemotactic factor. Parasitol Res 2001; 87: 376–382 [DOI] [PubMed] [Google Scholar]
  • 83.Owhashi M, Harada M, Suguri S, Omae H, Ishii A. Identification of an eosinophil chemotactic factor from anopheline mosquitoes as a chitinase family protein. Parasitol Res 2008; 102: 357–363 [DOI] [PubMed] [Google Scholar]
  • 84.Boppana VD, Thangamani S, Adler AJ, Wikel SK. SAAG-4 is a novel mosquito salivary protein that programmes host CD4 T cells to express IL-4. Parasite Immunol 2009; 31: 287–295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Schneider BS, McGee CE, Jordan JM, Stevenson HL, Soong L, Higgs S. Prior exposure to uninfected mosquitoes enhances mortality in naturally-transmitted West Nile virus infection. PLoS One 2007; 2: e1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Styer LM, Lim PY, Louie KL, Albright RG, Kramer LD, Bernard KA. Mosquito saliva causes enhancement of West Nile virus infection in mice. J Virol 2011; 85: 1517–1527 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Styer LM, Bernard KA, Kramer LD. Enhanced early West Nile virus infection in young chickens infected by mosquito bite: effect of viral dose. Am J Trop Med Hyg 2006; 75: 337–345 [PubMed] [Google Scholar]
  • 88.Sbrana E, Tonry JH, Xiao SY, da Rosa AP, Higgs S, Tesh RB. Oral transmission of West Nile virus in a hamster model. Am J Trop Med Hyg 2005; 72: 325–329 [PubMed] [Google Scholar]
  • 89.Reisen WK, Chiles RE, Kramer LD, Martinez VM, Eldridge BF. Method of infection does not alter response of chicks and house finches to western equine encephalomyelitis and St. Louis encephalitis viruses. J Med Entomol 2000; 37: 250–258 [DOI] [PubMed] [Google Scholar]
  • 90.Ader DB, Celluzzi C, Bisbing J, Gilmore L, Gunther V, Peachman KK, Rao M, Barvir D, Sun W, Palmer DR. Modulation of dengue virus infection of dendritic cells by Aedes aegypti saliva. Viral Immunol 2004; 17: 252–265 [DOI] [PubMed] [Google Scholar]
  • 91.Halstead SB. Pathogenesis of dengue: challenges to molecular biology. Science 1988; 239: 476–481 [DOI] [PubMed] [Google Scholar]
  • 92.Ajariyakhajorn C, Mammen MP, Jr, Endy TP, Gettayacamin M, Nisalak A, Nimmannitya S, Libraty DH. Randomized, placebo-controlled trial of nonpegylated and pegylated forms of recombinant human alpha interferon 2a for suppression of dengue virus viremia in rhesus monkeys. Antimicrob Agents Chemother 2005; 49: 4508–4514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Diamond MS, Roberts TG, Edgil D, Lu B, Ernst J, Harris E. Modulation of Dengue virus infection in human cells by alpha, beta, and gamma interferons. J Virol 2000; 74: 4957–4966 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Vithanomsat S, Wasi C, Harinasuta C, Thongcharoen P. The effect of interferon on flaviviruses in vitro: a preliminary study. Southeast Asian J Trop Med Public Health 1984; 15: 27–31 [PubMed] [Google Scholar]
  • 95.Haria M, Benfield P. Interferon-alpha-2a. A review of its pharmacological properties and therapeutic use in the management of viral hepatitis. Drugs 1995; 50: 873–896 [DOI] [PubMed] [Google Scholar]

Articles from Tropical Medicine and Health are provided here courtesy of BMC

RESOURCES