Abstract
Summary: West Nile Virus was introduced into the Western Hemisphere during the late summer of 1999 and has been causing significant and sometimes severe human diseases since that time. This article briefly touches upon the biology of the virus and provides a comprehensive review regarding recent discoveries about virus transmission, virus acquisition, and human infection and disease.
INTRODUCTION
West Nile virus (WNV) is a neurotropic human pathogen that is the causative agent of West Nile fever and encephalitis. WNV was introduced into the Western Hemisphere during the late summer of 1999, when infected individuals were diagnosed in New York State (104, 125). In 2000, the epizootic expanded to 12 states and the District of Columbia (125), and WNV can now be found in many avian and mosquito species throughout North America (72, 73). From 1999 to 2010, more than 2.5 million people were infected, with over 12,000 reported cases of encephalitis or meningitis and over 1,300 deaths (93).
The purpose of this review is to present and summarize recent discoveries about the acquisition and transmission of WNV by mosquitoes as well as insights into human infection. We discuss and review data collected and presented over the last decade, and we present future directions of research.
BIOLOGY
Flaviviridae
The family Flaviviridae contain 3 genera: the flaviviruses, which include WNV, dengue virus (DENV), and yellow fever virus (YFV); the hepaciviruses, which include hepatitis B and C viruses; and the pestiviruses, which affect hoofed mammals. Within the Flavivirus genus, which contains more than 70 viruses, viruses can be further classified into tick-borne and mosquito-borne virus groups. The mosquito-borne viruses may be roughly sorted into the encephalitic clade, or the JE serocomplex, which includes WNV and Japanese encephalitis virus (JEV), and the nonencephalitic or hemorrhagic fever clade, which includes DENV and YFV, and there are 10 serologic/genetic complexes (30, 101, 118). The geographic distribution of the mosquito-borne flaviviruses largely depends on the habitat of the preferred mosquito vector, with Culex mosquitoes transmitting encephalitic flaviviruses mainly in the Northern Hemisphere.
Structure and Proteins
WNV is an enveloped virion containing a single-stranded, positive-sense RNA genome. The genome consists of a single open reading frame of approximately 11 kb with no polyadenylation tail at the 3′ end. Both the 5′ and 3′ noncoding regions of the genome form stem-loop structures that aid in replication, transcription, translation, and packaging (63, 92, 196). The viral RNA is translated as a single polyprotein that is post- and cotranslationally cleaved by both host and viral proteases, resulting in three structural (capsid, envelope, and premembrane) and seven nonstructural (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) proteins (174). The 5′ end of the genome encodes the structural proteins, which are necessary for virus entry and fusion as well as encapsidation of the viral genome during assembly (118). The nonstructural proteins have many diverse functions, which is understandable as the virus has a very limited number of proteins and they must each serve multiple purposes during infection. NS1 has both a “cellular” form and a secreted form and is highly immunogenic but has no described role in virion assembly, though it has been suggested to play a role in replication (234). NS3 is the viral protease responsible for cleaving other nonstructural proteins from the viral polyprotein and encodes enzyme activities, and these functions have been widely characterized (118). The NS5 protein serves as the viral polymerase and encodes a methyltransferase, and it is necessary for viral replication (117, 174). Several of the nonstructural proteins, including NS2A, NS2B, NS4A, and NS4B, have been shown to inhibit one or more components of the innate immune response against viral infection (116, 121, 122, 139).
The West Nile virus virion is an icosahedral particle with the capsid protein associating with the RNA genome to form the nucleocapsid, which is surrounded by a lipid bilayer. A high proportion of capsid protein localizes to the nucleus, while viral assembly takes place in the cytoplasm, with budding in the endoplasmic reticulum (ER) (17, 41, 183). Although the nuclear functions of capsid are not fully understood, recent evidence suggests a role in gene regulation through binding with histone proteins (41). During virus assembly, the envelope protein embeds in the lipid bilayer of the virus and is exposed to the virion surface. The envelope protein is responsible for binding the receptor on the cell surface for viral entry (134). The prM protein is also known to embed in the lipid bilayer and is thought to protect E from undergoing premature fusion upon virus exocytosis to the cell surface. During infection, the virus population contains both mature and immature virus particles containing a varying number of immature prM protein molecules on the surface (57, 239).
Life Cycle
Entry of WNV is through receptor-mediated endocytosis after virus attachment to the cell surface. Several molecules have been implicated as receptors for West Nile virus, including DC-SIGN, mannose receptor, and several glycosaminoglycans (52, 110, 211). The virus-containing endosome matures during internalization from the cell surface, with the pH dropping from neutral to slightly acidic in the early endosome and becoming more acidic during maturation to the late endosome. Within the late endosome, the envelope protein will undergo a conformational change resulting in fusion of the viral lipid membrane with the endocytic membrane and the release of the viral RNA genome into the cell cytoplasm (134). Following capsid disassociation, the RNA genome is replicated and virus assembly is initiated following a well-documented program (118). The viral polyprotein is translated and processed on intracellular membranes, resulting in the expression of the 10 viral proteins. The original viral RNA is replicated by viral and cellular proteins into multiple copies to be used in the production of new virions. The structural proteins assemble onto membranes in the endoplasmic reticulum, associate with the nucleocapsid, and bud into the cytoplasm via the Golgi network. The virus travels to the cell surface in an exocytic vesicle and matures as cellular enzymes cleave the prM, resulting in the release of mature virus from the cell surface (174).
There has been a recent spike in interest in the role of partially or fully immature flavivirus particles during infection. These immature flavivirus particles form when there is inefficient cleavage of the prM protein from the virion surface during maturation and budding (237). Immature or partially mature flavivirus particles of both DENV and WNV have been shown to account for up to as much as 40% of the total virus population in a given infection (135). While they were traditionally thought to be noninfectious, several recent studies have shown that immature WNV particles can be highly immunogenic and infectious in vitro and in vivo when bound by antibodies against the E or prM protein (43, 51, 179). These antibody-bound immature virus particles enter immune cells via the Fc receptor, resulting in productive infection. Further work remains to be done to determine the role that immature particles play in viral pathogenesis and disease in both vector and mammalian hosts.
VECTOR-VIRUS RELATIONSHIP
Vector Preference
The ability of different mosquito species to acquire and transmit WNV is highly variable. Culex mosquitoes are accepted as the primary global transmission vector; C. tarsalis is a main mosquito vector of WNV in the western United States and can feed on a variety of avian and mammalian species (95, 163). Other vectors shown to have competence for both infection and transmission of West Nile virus are C. quinquefasciatus, C. stigmatosoma, C. thriambus, C. pipiens, and C. nigripalpus; to date, over 65 mosquito species have been shown to be infected by WNV (79, 164, 222). There is evidence that C. pipiens in the eastern United States may feed on mammals and humans instead of birds during the late summer and early fall, and this “host switching” has also been reported with C. tarsalis in the western United States (96, 212). There are several reports of WNV in Aedes mosquitoes, though they are not considered a primary vector in nature (46, 58, 83, 184, 216, 221). WNV has also been detected in field-collected male A. triseriatus and C. salinarius (219), which not only points to vertical transmission of virus, as only females feed on animal blood, but also further supports that WNV has the ability to infect Aedes mosquitoes in nature. The ability to infect various mosquito species, the geographic range of mosquito species, and the ability of mosquitoes to feed on and transmit virus to particular hosts all play a role in WNV vector preference.
Host Reservoirs
WNV is maintained in nature in a cycle between mosquitoes and animal hosts (Fig. 1 shows a schematic of mosquito-mammal transmission), with the predominant and preferred reservoir being birds (3, 75, 136, 162). Birds of some species become ill, show symptoms of disease, and may die, while others become infected and serve as carriers without showing signs of disease. Although house sparrows and crows are highly susceptible to WNV, they make up a small fraction of analyzed mosquito blood meals and may be of minor importance in transmission. The American robin is instead thought to be the main host species responsible for the maintenance and transmission of WNV in the United States due to the feeding preference for robins by the dominant viral vectors (80, 91, 94). Bird-bird transmission has been demonstrated in the laboratory, with several species proving to be capable of contact transmission (99). Humans are considered “dead-end” hosts for WNV, as the low level of viremia in mammals is usually not sufficient to be transmitted to mosquitoes, thereby ending the transmission cycle (20). The ability of mammals to act as hosts could change, though, should Aedes mosquitoes, which feed primarily on humans, become primary transmission vectors for WNV.
Fig 1.
Schematic of West Nile virus transmission from mosquito to mammal and host factors known to be involved in infection.
Vector Acquisition
Mosquitoes acquire WNV after taking a blood meal from a viremic animal. The stages of infection and replication in the mosquito have been well described (68, 126, 175). The virus must then infect and replicate in cells of the mosquito midgut as the blood meal is being processed. After replication in the midgut epithelia, the virus travels through the mosquito hemolymph to the salivary glands. Accumulation of virus in the salivary glands will eventually result in high viremia in the saliva, from where it can then be transmitted to mammalian hosts during feeding. The mosquito midgut can serve as a barrier to infection due to the presence of certain chitins and other proteins as well as a strong immune response to the virus (194). The peritrophic matrix, which consists of chitin microfibrils embedded in a proteoglycan matrix, has been shown to play a role in reducing pathogen invasion of the midgut epithelium, though its role in flavivirus infection is not entirely understood (89). A recent study looking at alteration in midgut gene expression in C. pipiens quinquefasciatus during WNV infection found 21 genes to be upregulated and 5 genes downregulated after mosquitoes fed on infected blood. Most of the genes were not canonical immune genes, though a putative Toll-like receptor (TLR) with increased expression during infection was identified (201). Proteins that have significantly increased or reduced levels in the mosquito midgut during WNV infection may play a role in disease acquisition or viral spread throughout the mosquito, and many are under active investigation as virulence factors. For example, a recent study found that a C-type lectin from mosquitoes facilitated WNV entry into mosquito cells by directly binding the virion and aiding interaction with a mosquito CD45 receptor homolog on the cell surface (36). These molecules may prove to be important for virus acquisition in the mosquito midgut. In mosquitoes that are refractory to infection, apoptosis in infected midgut epithelial cells has been proposed to limit the dissemination of WNV throughout the mosquito body (220). There is also evidence of a midgut barrier to secondary flavivirus infection, where mosquitoes which acquired more than one virus showed no evidence of dissemination of the second virus, which would prevent transmission (151). Research supports the existence of both physical and immune midgut barriers to WNV infection, and the list of genes both required for and inhibitory to acquisition is sure to increase with further experimentation.
Vector Response to Infection
There have been many recent studies aimed at elucidating the transcriptomic and proteomic response to flavivirus infection in the mosquito vector. Although WNV establishes a persistent infection in mosquito cells in vitro and in live mosquitoes, there is growing evidence that the mosquito does mount some immune response to virus infection. Most of what is known about the insect immune system comes from experiments with Drosophila melanogaster, though current examination of the mosquito immune response is starting to reveal corresponding proteins and pathways. The mosquito antiviral response is thought to consist of two pathways: the innate immune pathway and the RNA interference (RNAi) pathway (7). The innate immune response is comprised of three signaling pathways: Toll, JAK-STAT, and IMD. The Toll and IMD pathways both culminate in NF-κB-mediated expression of antimicrobial peptides (AMPs), and IMD signaling has been shown to control RNA virus infection in Drosophila (44). Not much is known regarding the role of mosquito AMPs in antiviral immunity, though their expression is often induced by viral infection. Both Toll signaling and the JAK-STAT pathways have been shown to play a role in the control of DENV infection in Aedes aegypti (161, 202) and may also be significant during infection of Culex with WNV. The RNAi pathway in mosquitoes is activated by viral double-stranded RNA and has been shown to be crucial for controlling alphavirus infection in both Aedes and Anopheles (32, 90). The RNAi pathway is known to be induced during WNV infection of Culex pipiens (21), and another RNAi pathway, PIWI, may participate in the mosquito response to virus, as it was shown to be involved in limiting WNV infection in Drosophila (39). Infection with dengue virus was also found to actively suppress mosquito immune responses in vitro (200).
Evidence for a transcriptomic signature of flavivirus infection was found during a comprehensive study of Aedes aegypti infected with WNV, DENV, and YFV (42). Genes involved in transcription and ion binding were found to be downregulated, and genes coding for proteases and cuticle proteins were found to be upregulated, during infection with all three viruses (42). Serine proteases had previously been shown to be important for viral propagation and blood digestion, though there have been varying reports regarding their impact on flaviviral infection in the mosquito (22, 138). Another global study of flaviviral infection in Drosophila identified many insect host factors relevant during dengue virus infection of the mosquito, including a putative NADH dehydrogenase and proteins involved in vesicular transport and endocytosis (193). Adding to our knowledge of the mosquito response to WNV infection, a recent transcriptomic analysis of Culex quinquefasciatus revealed that many genes involved in metabolism and transport are upregulated during infection (14). Given that the virus must infect a variety of cell types and organs in the mosquito vector, as well as optimize the cellular environment to benefit its life cycle, there are likely a large number of differentially regulated genes, proteins, and other host factors important to WNV infection of the mosquito that have yet to be discovered.
Transmission to Vertebrate Host
WNV is transmitted to its vertebrate hosts by an infected mosquito vector during the probing process of blood feeding. Mosquitoes probe host skin using their proboscis in order to inject pharmacologically active saliva proteins and to locate a blood source (84, 171, 172). Although many hematophagous insects can obtain a blood meal without functional salivary glands, the efficiency of blood feeding is severely limited (84, 171, 172). In order to combat the host's hemostatic system, all hematophagous insects inject at least one vasodilator, one coagulation inhibitor, and one platelet inhibitor, and often the saliva includes immunomodulatory, digestive, and antimicrobial proteins as well (167, 169, 170, 186). While numerous proteins in the saliva of hematophagous insects have been described, many remain that have not been characterized, especially with respect to viral infection.
During probing, mosquito saliva is injected mostly extravascularly in the skin's dermal layer (205). Dermal blood vessels are the targets for hematophagous insects. In order to locate these structures, the proboscis must navigate through a very elastic environment that has a high tensile strength. To efficiently move through this environment, mosquito saliva may contain components that liquefy the bite site. A salivary endonuclease with a proposed function to facilitate probing in host skin has been identified in C. quinquefasciatus (31).
Host skin acts as an important barrier to many infections, though WNV antigen has been detected in skin at multiple phases of infection. WNV replication was observed in skin tissue at the inoculation site at 1 and 3 days postinfection (189), and WNV has also been shown to spread to areas of skin contralateral to the site of inoculation (27). Infectious WNV has been shown to persist in skin at the inoculation site for at least 14 days postinfection (5). Many reports document that both keratinocytes and fibroblasts are permissive to WNV infection in vitro and in vivo (8, 37, 38, 55, 60, 62, 86, 87, 102, 109, 115, 165, 185, 195, 233). By immunohistochemistry and fluorescence-activated cell sorter (FACS) analysis, WNV antigen was detected in keratinocytes at 4 and 5 days postinfection, and virus presence in a small subset of skin cells that lacked the keratin marker K10 suggests that skin cells other than keratinocytes may also be important early reservoirs (115).
Mosquito Saliva Factors
Saliva from hematophagous insects has been shown to alter the transmissibility of many pathogens (1, 50, 160, 178, 187, 206, 223, 231). Saliva from both A. aegypti and C. tarsalis has been shown to alter transmissibility in a WNV mouse model (189, 206). Specifically, when mice were fed on by uninfected A. aegypti prior to intradermal inoculation with WNV, more progressive infection, higher viremia, and accelerated neuroinvasion occurred. Even at a low dose of infection, mice that were previously fed on by mosquitoes had a lower survival rate after WNV infection (189). Similar experiments with C. tarsalis showed that mice infected with WNV through the bite of a single mosquito had viremia and tissue titers that were 5- to 10-fold higher postinoculation and showed faster neuroinvasion than those in animals infected by syringe inoculation (206). Enhanced early infection was also observed when mice were inoculated with WNV mixed with mosquito salivary gland extract (SGE). Importantly, enhanced viremia was not observed when SGE was inoculated in a distal site, supporting that mosquito saliva exerts its effect locally (206).
Due to the complex nature of mosquito saliva, multiple activities may lead to the enhancement of early virus infection. Further, due to the intense selective pressures exerted on mosquito saliva proteins by the host immune systems, successful viruses likely coevolve with their mosquito vectors in order to coopt unique saliva protein activities. For example, A. aegypti SGE reduced murine splenocyte proliferation and production of both Th1 and Th2 cytokines while C. quinquefasciatus SGE did not have this activity (224). These data suggest that the reduction of splenocyte proliferation and Th1/Th2 cytokine production may be critical for virus transmission and predict that C. quinquefasciatus would be less efficient at transmitting virus. The adaptation that has taken place between a virus and its vector's saliva proteins may contribute to vector competence, although these mechanisms remain poorly defined.
Multiple reports have suggested that immunomodulatory activities in mosquito saliva could result in enhanced early infection (45, 188, 190, 224, 231). These reports suggest that saliva modulates skin-resident immune cells. In one report, A. aegypti saliva was able to decrease beta interferon (IFN-β) and inducible nitric oxide synthase in macrophages ex vivo (188). Recruitment of T cells was also reduced when WNV was inoculated during mosquito feeding, rather than by syringe, suggesting that saliva hinders infiltration of these cells into the inoculation site (188). These effects correlated with enhanced expression of interleukin-10 (IL-10), which has anti-inflammatory activities, including the downregulation of Th1 cytokines, major histocompatibility complex (MHC) class II molecules, and costimulatory molecules on macrophages (188). While this study is limited by the use of A. aegypti SGE, which is not the primary vector for WNV, it is likely that some Culex salivary proteins act to enhance WNV infection.
It is unknown whether Culex sp. SGEs have similar immunomodulatory activities; however, C. pipiens SGE was able to enhance Cache Valley fever virus infection, and C. tarsalis saliva was able to enhance WNV infection in a mouse model (56, 206). Additionally, saliva from C. tarsalis and C. pipiens was able to enhance WNV infection in chickens (204). The fact that saliva from multiple species in both the Aedes and Culex genera was able to enhance virus infectivity would suggest either that the relevant saliva proteins are highly conserved or that a similar activity has convergently evolved in multiple mosquito vectors. If all Culex spp. modulate a specific component of the host immune system to facilitate blood feeding, WNV may have evolved to benefit from this universal mosquito saliva activity. In addition, differences in salivary gland protein activities could alter the ability of a mosquito species to enhance pathogen transmission. Multiple activities that differ between Aedes and Culex mosquitoes have been noted (166, 169, 170, 173, 223). Since such dramatically different saliva activities exist between Aedes and Culex spp., direct comparisons of mosquito saliva activities that are responsible for the enhancement of WNV transmission need to be performed for each Culex sp. that is able to vector WNV.
Though mosquito saliva has been shown to enhance WNV infection, the precise mechanisms as well as the specific saliva proteins involved remain to be investigated. In one example, hyaluronidase from sand fly saliva was found to be important for the enhancement of Leishmania infectivity in mice (223). Saliva hyaluronidase may enlarge the feeding lesion and serve as a spreading factor for other pharmacologically active factors present in saliva (223). This activity was also found in C. quinquefasciatus saliva and may also affect the spread of WNV and other saliva components as well as influence the local host immune response (168, 223). In another example, Salp15 from tick saliva was able to directly interact with the surface of Borrelia burgdorferi and facilitated evasion from host B cell-mediated immunity (160), and immunization against Salp15 protected mice from Lyme disease (50). Another study identified two tick saliva proteins that functioned to inhibit polymorphonuclear leukocyte recruitment during infection of mice with Borrelia burgdorferi, likely increasing the spirochete burden and enhancing infection (77). Identification of proteins in mosquito saliva that are responsible for the enhancement of WNV transmission is under way, and these investigations may provide novel nonvirus targets for vaccine design.
Multiple negative salivary gland factors that limit flavivirus transmission have been identified (42, 124). In one example, microarray analysis of DENV-infected and uninfected salivary gland mRNAs showed an upregulation of a putative antibacterial, cecropin-like peptide (i.e., AAEL000598), which showed antiviral activity against both DENV and Chikungunya virus (124). A recent comparative microarray analysis of mRNAs from DENV-, YFV-, and WNV-infected and uninfected whole A. aegypti identified multiple genes that were downregulated by all three viruses (42). Genes downregulated by day 14 postinfection likely play a role in salivary gland invasion or virus transmission. Among those, a recombinant pupal cuticle protein was able to directly interact with WNV envelope protein and inhibit infection in vitro and prevent lethal WNV encephalitis in mice (42). Although these proteins were expressed in salivary glands, they have yet to be formally identified in saliva.
Transgenic traits and introduced factors can also alter the transmission of vector-borne pathogens and may play a role in the future control of virus-infected mosquito populations. Transgenic mosquito populations that can be selected to either block transmission, block acquisition, decrease host seeking, decrease probing and biting, increase background mortality, or increase mosquito infection-induced mortality are in development (1, 59, 98, 128, 147, 148). To date, most studies have focused on producing transgenic mosquitoes that block transmission. For example, experimental strains of A. aegypti that inhibit flavivirus replication in the midgut and consequent migration to the salivary gland have been engineered (59, 98, 147, 148). Another gene that is responsible for host seeking behavior has been identified (203). Many strategies that lead to increased background mortality have been implemented, and field trials have already begun to test the effectiveness of these transgenic mosquitoes in reducing wild mosquito populations (9, 64, 65, 214). Laboratory infection with Wolbachia bacteria also reduces the life span of mosquitoes (127). This strategy has also been tested in field trials to reduce wild mosquito populations (82). The release of insect-specific densoviruses also shows high mortality in mosquito populations and may be used as a control strategy (34). The advantage of using Wolbachia or Densovirus infection as opposed to insecticide treatment is that these pathogens are expected to replicate and spread through the wild mosquito populations (128).
MAMMALIAN INFECTION
Epidemiology and Clinical Features
The emergence of WNV in North America was first documented in the fall of 1999 in New York City following an outbreak of mosquito-borne encephalitis responsible for the death of humans, birds, and horses (3, 26, 104, 145, 232). Over the next decade, WNV spread throughout the United States and into Canada, Mexico, and the Caribbean (75). From 2005 to 2009, 12,975 cases were reported to the CDC, including 496 fatalities, and 35% of reported cases were the more severe forms of neuroinvasive disease, including encephalitis (119). As detailed above, in most cases the virus is transmitted by the Culex mosquito vector (4), but transmission may occur through blood transfusion, organ transplantation, breast-feeding, or intrauterine exposure, and laboratory-acquired infection has also been reported (35a, 81, 85, 103, 177).
Infections in humans are predominantly subclinical, but reported infection manifestations may range from fever and myalgias to meningoencephalitis and death (152). Encephalitis occurs in only a small subset of patients; progression to severe neurological illness may induce acute flaccid paralysis after meningitis or encephalitis, with rapidly progressing symptoms that may involve all four limbs (111). Severe poliomyelitis-like syndrome can occur and has a poor long-term outcome (191). Elderly individuals are more susceptible to neurological involvement that may result in death, and among those older adults who survive, as many as 50% may have significant postillness morbidity for at least a year following infection (33) and may have an increased risk of death for up to 3 years after acute illness (120). Among individuals over 70 years of age, the case-fatality rate ranges from 15% to 29% (152). Higher fatality is also seen in infected infants and immunocompromised patients (73). Risk factors for encephalitis and death include being homeless, a history of cardiovascular disease or chronic renal disease, hepatitis C virus infection, and immunosuppression (140, 192). In addition, in some cases convalescent patients may have persistent or chronic infection detected through PCR of the urine, which suggested ongoing viral replication in renal tissue (141, 143). Although persistence of WNV has also been noted in several animal models (156, 199, 213), it has not been uniformly evident in assays of urine (66).
Diagnostics
The diagnosis of WNV infection is based largely on clinical criteria and testing for antibody responses (28). The incubation period for WNV infection is thought to range from about 2 to 14 days (143). The presence of anti-WNV IgM, particularly from cerebrospinal fluid (CSF), is used for diagnosis. Cross-reactivity with related flaviviruses (Japanese encephalitis virus, St. Louis encephalitis virus, YFV, and DENV), if suspected, can be assessed through plaque neutralization assays (143). Replication of WNV has been documented in human monocytes in vitro and with even higher efficiency in polymorphonuclear leukocytes; this could lead to transmission via transfusion of blood (10, 177). Thus, several rapid tests have been developed for blood donor screening using nucleic acid testing (NAT), an amplification-based transcription technique, which identifies WNV-infected individuals before they become symptomatic and may be used to safeguard the blood supply (238). Of note, 45% of NAT-positive subjects were subsequently not confirmed, and in one study, only 4 to 5% of the patients received a diagnosis of WNV infection (238).
Antibody testing in patients follows an expected timetable of median times of 3.9 days from RNA detection to IgM seroconversion and 7.7 days from RNA detection to IgG seroconversion (28). RNA generally became undetectable after 13.2 days, although it rarely was found to persist for >40 days. IgM and IgA antibodies fell significantly, although not universally, while the IgG level remained elevated for >1 year after detection of viremia (28, 141, 149, 180). Antibody to WNV NS5 persists in vivo, and thus NS5 antibody cannot be used to distinguish recent from past WNV infection (157).
Immune Response
Control of WNV infection by the human and murine hosts has been investigated for both innate and adaptive immune responses. Through integrating these results, a picture of critical elements in immune responses to WNV is emerging (Tables 1 and 2). Sensing WNV pathogen-associated molecular patterns through pathogen recognition receptors such as Toll-like receptors (TLRs) and cytoplasmic RNA helicases is critical for early detection and activation of innate immune pathways that facilitate early control of viral replication (48, 61, 208–210, 218, 226, 230). This early response is mediated largely by macrophages; WNV infection of macrophage-depleted mice results in increased mortality, higher and extended viremia, and substantially shortened survival. Moreover, in mice, even a nonneurotrophic WNV strain may cross the blood-brain barrier (BBB) in the absence of macrophage clearance of virus (16). Macrophages express TLRs, mediate clearance of opsonized viral particles, produce proinflammatory cytokines, and upregulate costimulatory proteins that link innate to adaptive immune responses (114a, 215). Macrophages are also a major component in inflamed central nervous system (CNS) tissues and are considered protective against WNV infection. The control of WNV by macrophages has been linked both to constitutive expression of innate immune genes, such as those for RIG-I, MDA5, PKR, and RNase L, and to direct effector mechanisms such as the production of radical oxygen species and type I IFN (47, 49, 61, 67, 181, 182, 208a, 226, 229).
Table 1.
In vivo function of murine genes in WNV infection
Gene | Survivala | Viremia | Brain viral load | Remarks | Reference(s) |
---|---|---|---|---|---|
Myd88−/− | S | Up | Up | Reduced leukocytes in CNS | 209, 218 |
Tlr3−/− | R | Up | Up | Reduced viral entry into CNS | 230 |
Tlr7−/− | S | Up | Up | Defective leukocyte homing | 218 |
Il10−/− | R | Down | Down | Enhanced antiviral response | 9a |
Irf7−/− | S | Up | Up | Defective type I IFN production | 48 |
Casp12−/− | S | Up | Up | Defective type I IFN production | 226 |
IfnαβR−/− | S | Up | Up | 181 | |
Ifnβ−/− | S | Up | Up | 109 | |
IfnγR−/− or Ifnγ−/− | S | Up | Up | 53a | |
Ips1−/− | S | Up | Up | Lack of regulatory T cells | 208 |
Pkr−/− or RnaseL−/− | S | Up | Up | 182, 185 | |
Mmp9−/− | R | Equivalent | Down | Reduced viral entry into CNS | 227 |
Irf3−/− | S | Up | Up | Impaired IFN-stimulated gene expression in macrophages | 47 |
C3−/− | S | Up | Up | Impaired CD8+ T/antibodies | 131 |
Compl R1/2−/− | S | Up | Up | Impaired protective antibodies | 129 |
Ccr5−/− | S | Equivalent | Up | Reduced T cells, NK cells, macrophages in CNS | 69 |
Cxcr2−/− | R | Down | Down | 10 | |
Cxcr3−/− | S | Equivalent | Up | Impaired CD8+ T cell recruitment to brain | 97a |
Cxcl10−/− | S | Equivalent | Up | Impaired CD8+ T cell recruitment to brain | 114a |
Ccr2−/− | S | Equivalent | Up | Fewer inflammatory monocytes in CNS | 197a |
sIgM−/− | S | Up | Up | Reduced WNV-specific IgG, no IgM | 197b |
Casp3−/− | R | Equivalent | Equivalent | Reduced neuron apoptosis | 182 |
Icam1−/− | R | Equivalent | Down | Reduced viral entry into CNS | 50 |
Cd8a−/− MHCclass1a−/− | S | Equivalent | Up | Increased viral loads (spleen), persistent infection in surviving mice | 197 |
Cd4−/− MHCclass2−/− | S | Equivalent | Up | Impaired WNV-specific IgM and IgG production, persistent infection | 200a |
Cd40−/− | S | Equivalent | Up | Impaired WNV-specific IgM/IgG production, reduced CD8+ cells in CNS | 200b |
Il22−/− | R | Equivalent | Down | Reduced viral entry into the CNS | 225 |
Dhx58−/− | S | Equivalent | Up | Reduced CD8+ T cell expansion | 208a |
TRAIL−/− | S | Equivalent | Equivalent | CD8+ T cells use TRAIL to limit infection | 236a |
R and S, mice are more resistant or susceptible, respectively, to lethal WNV infection than their wild-type controls.
Table 2.
Genes and corresponding SNPs important in human WNV infection
Gene(s) | SNP(s) | Comparison groups (n) | Study results | Reference |
---|---|---|---|---|
OASL | rs3213545 | WNV+ cases (33) vs healthy controls (16) | Associated with increased susceptibility to WNV infection | 236 |
CCR5 | Δ32 deletion | WNV+ cases (395) vs WNV− (1,463) | Increased risk of symptomatic WNV infection | 69 |
WNV+ cases (224) vs healthy controls (1,318) | Increased risk of symptomatic WNV infection | 113 | ||
WNV+ cases (634) vs WNV− (422) | Not a risk factor for WNV initial infection; associated with symptomatic WNV infection | 114 | ||
OAS1 | rs10774671 | WNV+ cases (501) vs healthy controls (552) | A risk factor for initial infection with WNV | 112 |
IRF3, MX1, OAS1 | rs2304207, rs7280422, rs34137742 | Symptomatic cases (422) vs asymptomatic cases (331) | Associated with symptomatic WNV infection | 19 |
RFC1, SCN1A, ANPEP | rs2066786, rs2298771, rs25651 | Severe WNV cases (560) vs mild WNV cases (950) | Associated with neuroinvasive disease in patients infected with WNV | 123 |
Although cellular immune mechanisms remain incompletely explained, innate immunity and in particular interferon responses have been shown to be critical in resistance to WNV (7, 9a, 181, 197b). Patients who mount a robust IFN-α response show lower viral loads, even before IgM seroconversion, concomitant with significant upregulation of IFN-γ during the viremic phase (217). Permeability of the blood-brain barrier (BBB), which is enhanced by cytokine responses, has been shown in murine models to be critical to resistance to WNV infection (230), and elements which decrease the integrity of the BBB contribute to susceptibility to infection with WNV (7, 226, 227). Entry to the CNS may be afforded by trafficking of infected CD45+ leukocytes and CD11b macrophages (218), T cells (228), or neutrophils (225). Mice lacking TLR3 show improved survival over wild-type animals due to a lower cytokine response and protection from BBB permeability (100, 230). Human studies show a role for CXCL10 and CCL2 in control of early infection and an important role for IFN-mediated innate immunity in resolving acute WNV infection (217). RNAi studies in human cell lines have indicated that interferon-inducible transmembrane protein (IFITM) inhibits the early replication of WNV (23).
Infection with flaviviruses leads to upregulation of MHC class I, MHC class II, and adhesion molecules, which may enhance infection through reducing NK cell activity, or enhance a transient autoimmunity in early infection (97). It is clear that CD8+ T cells are critical in the response to flavivirus infections. Overall T cell responses in humans revealed that multiple peptide regions of WNV proteins are recognized by T cells, with a subset of 8 peptides predominating, and the highest magnitude of specific T cell responses was from CD8+ cells (105). The immunodominant T cell epitopes which elicited both highest-frequency and highest-magnitude responses included sequences from WNV M, E, NS3, and NS4 proteins and, furthermore, were equivalent between symptomatic and asymptomatic subjects in this cohort (105). During infection with WNV, CD8+ T cells expand dramatically and migrate to the site of CNS infection (97, 236a). Examination of immune responses from WNV patients shows that memory T cell responses to WNV are mainly due to CD8+ T cells with a defined set of epitopes; these were quite constant over 12 months of observation and were not apparently related to disease severity (150). Examination of memory T cells from 40 patients months after infection showed persistence of the memory phenotype and WNV-specific polyfunctional CD8+ T cell responses. More cytolytic memory T cells were found in patients with neurological disease (154). Indeed, CD8+ T cells have been shown to be important for control of viral load in mouse models of WNV infection, at least in part due to a role for perforin (97a, 197, 197a, 200a). WNV-specific murine CD4 T cells produced IFN-γ and IL-2 and also showed direct antiviral activity (25, 197b, 200b). Tregs play an important role in protecting against severe disease, and it has been shown in both human patients and animal models that symptomatic patients show a lower frequency of Tregs despite having similar systemic T cell responses (108).
Complement has also been indicated as an important component of the host innate immune response to flavivirus infection. However, while complement traditionally limits the spread of many pathogens, it appears to have both protective and pathogenic roles during flavivirus infection. Whether or not complement is protective or pathogenic depends on a variety of factors, including the specific virus, the phase of infection, and the underlying immune status of the host (40, 130, 131).
A paradoxical role for polymorphonuclear cells (PMNs) in WNV infection has been described, where PMNs are recruited to the site inoculated with WNV (10). It was determined by depleting PMNs prior to WNV infection that recruitment of PMNs to the inoculation site was associated with enhanced WNV replication. However, if PMNs were depleted after WNV infection, mice developed higher viremia and mortality. Thus, infiltrating PMNs may serve as an early reservoir of WNV replication (10). Dendritic cells express DC-SIGN, suggesting that they may also be early cellular targets in host skin (211). WNV infection of dendritic cells leads to production of IFN. Interestingly, studies with dendritic cells from human donors showed that type I IFN expression in response to WNV in vivo is lower in cells from older donors than in those from younger donors, which may contribute to older individuals being more susceptible to WNV disease (159).
These innate pathways are critical not only for immediate antiviral defense pathways such as the upregulation of type I interferons but also for the generation of an effective adaptive T and B cell-mediated sustained immune response (24, 53a, 129, 131, 155, 181, 198). The γδ T cell population rapidly expands after WNV infection. Mice that lack γδ T cells have higher viremia and increased mortality (229). Soon after infection, γδ T cells produce IFN-γ, which correlates with an increase in perforin expression in splenic T cells. Bone marrow chimera reconstitution experiments in mice support that IFN-γ production by γδ T cells is critical for the early control of WNV infection (229). γδ T cells also promote a protective adaptive immune response by facilitating dendritic cell maturation, providing an important link between the innate and adaptive immune responses against WNV infection (229).
Genetic Determinants of Disease
Specific human genetic factors that influence the severity of infection with WNV and the antiviral innate immune response have been identified (Table 2). Certain HLA types appear to be associated with risk of a more severe outcome (HLA-A*68 and C*08) or better resistance to infection (B*40 and C*03) (107). Single nucleotide polymorphism (SNP) studies have detected SNPs in key regulators of immune function, including interferon pathway elements. In particular, polymorphisms in IRF3 and MX-1 were associated with symptomatic infection, and an SNP in the oligoadenylate synthetase 1b (OAS-1) gene, an interferon-regulated gene involved in RNA degradation, was associated with an increased risk for initial infection with WNV and severe neurological disease (>750 subjects) (19, 112). Notably, the 2′,5′-oligoadenylate synthase (2′-5′-OAS) gene has also been identified as a susceptibility factor in WNV in horses and as a contributing factor for severity of neurological disease in tick-borne encephalitis virus (13, 176, 236). A dominant negative splice variant of RNase L, which functions in the antiproliferative roles of interferon, was detected more often in WNV patients than in control patients (236). Another genomic study investigated >1,500 symptomatic subjects (with severe versus mild disease) and showed more severe neurological disease to be associated with SNPs in the genes for RFC1 (a replication factor), SCN1a (a sodium channel), and ANPEP (an aminopeptidase), although even more differences might have been revealed when comparing asymptomatic and symptomatic cases (123). In addition, a deletion in CCR5, which is known to be protective in infection with HIV, while not associated with susceptibility to WNV, did correspond to severity of infection, presumably due to reduced function of CCR5 pathways in infected hosts (69, 113, 114). As more host factors are identified, there are sure to be a number of new determinants of WNV infection.
Therapeutics
Current therapeutic options against WNV are mainly supportive; there are no FDA-approved vaccines or treatments available (54). Investigations to identify individual susceptibility markers, recombinant antibodies, peptides, RNA interference, and small molecules with the ability to directly or indirectly neutralize WNV have been reported; however, an effective drug is still lacking (6, 12, 70, 71, 74, 146, 158). There are currently four USDA-licensed vaccines available for equines (two are inactivated whole WNV, one is a nonreplicating live canary pox recombinant vector vaccine, and one is an inactivated flavivirus chimeric vaccine). Though passive immunization has been used in a few cases, it has serious limitations, such as inadvertent transfer of blood-borne pathogens, inconsistent quality of the donor antisera, cost, and allergic reactions (78). A case study of two WNV encephalitis patients treated with alpha interferon, the standard of care for infection with the related flavivirus hepatitis C virus, showed substantial improvement and an improved convalescence course (88).
Several approaches are being pursued for the development of a vaccine in humans that may prove valuable for use by targeted populations. Investigations include live attenuated vaccines, recombinant subunit vaccines, vectorized vaccines, DNA vaccines with constructs that express the WNV E protein, live recombinant vaccines, and an attenuated strain based on nonglycosylated E and mutant NS1 proteins (15, 235). A neutralizing, WNV-specific monoclonal antibody, E16 (MGAWN1), which penetrates the CNS in animal models, produced neutralizing antibodies in phase I trials (15). Very promising results were seen with a chimeric vaccine based on the WNV prM and E proteins inserted into the yellow fever 17D vaccine moiety (ChimeriVax-WN02). It was shown to be safe and immunogenic in phase II clinical trials, with high seroconversion rates, but it is no longer available (18).
CONCLUSIONS AND FUTURE DIRECTIONS
WNV has now persisted and become established in North America. Of particular significance is the expansion of the mosquito vectors harboring WNV to include Aedes albopictus, a common mammal-biting mosquito (2, 73, 136). It is hoped that the increase in our knowledge of the interactions of WNV with the mosquito vector will lead to new avenues for therapeutics and preventative measures. Mosquito responses at the levels of protein and gene expression as well as a more complete understanding of viral pathogenesis in the vector, especially with regard to the immune response, may point to novel targets to focus our efforts to inhibit or block WNV infection in both mosquitoes and mammals.
For example, a single-chain human monoclonal antibody developed through phage display directed against the fusion loop of the envelope protein showed both pan-flaviviral protection and therapeutic efficacy when tested in the murine model (71, 207). Recent advances in nanoparticle technology have also been employed in vaccination studies of murine WNV infection and show promising efficacy of TLR9-targeted biodegradable nanoparticles, which produce a high number of circulating effector T cells and antigen-specific lymphocytes (53). Potential relevant viral susceptibility mechanisms, including host antagonism of chemokine responses as has been noted in infection with the related flavivirus hepatitis C virus (35), may reveal infectious mechanisms used by WNV and other mosquito-borne flaviviruses. The pace of discovery of vector, virus, and host components of pathogenesis continues to provide critical insights for the successful development of controls and treatments for WNV.
ACKNOWLEDGMENTS
We are grateful to our long-standing colleague John F. Anderson of the Connecticut Agricultural Experiment Station and to members of the Yale IMAGIN and HIPC teams for valuable discussions.
This work was supported in part by the NIH (grants HHS N272201100019C, U19AI089992, and U01AI 070343). E.F. is an Investigator of the Howard Hughes Medical Institute. T.M.C. is supported by grant 2T32HL007974-11, and M.J.C is supported by grant 5T32AI07019-35.
Biographies
Tonya M. Colpitts received a B.A. from the University of Hawaii and a Ph.D. in microbiology and immunology from the University of Texas Medical Branch, where she showed that the cellular endocytic machinery is highly evolutionarily conserved between mosquitoes and mammals and that alphaviruses require functional endocytosis and low pH for entry into both cell types. She is currently a postdoctoral associate in the laboratory of Professor Erol Fikrig in the Section of Infectious Diseases at Yale University School of Medicine. Her research focuses on the exploration of the interactions between mosquitoes and flaviviruses, the identification of human host factors that bind flaviviral proteins, and the examination of how flavivirus infection affects proteins and pathways of human cells. She is also researching the interactions of flavivirus capsid protein with nuclear and cytoplasmic proteins as well as the role of capsid in the nucleus of the cell during infection.
Michael J. Conway received a B.S. from Northern Michigan University and a Ph.D. in microbiology and immunology in the laboratory of Professor Craig Meyers at the Pennsylvania State University College of Medicine, where he studied differentiation-dependent mechanisms of the human papillomavirus life cycle, including capsid assembly and maturation. He currently is a postdoctoral research fellow in the laboratory of Professor Erol Fikrig in the Infectious Diseases Section of the Department of Internal Medicine at Yale University School of Medicine. His current research interests involve vector-virus-host interactions that occur as disease vectors deposit salivary components and pathogens into the host.
Ruth R. Montgomery received a B.A. from the University of Pennsylvania and a Ph.D. from Rockefeller University, where she studied with Zanvil Cohn and Carl Nathan. After postdoctoral work on macrophage endocytosis with Ira Mellman, she remained at Yale, where she is now Associate Professor of Medicine. The focus of her lab is human innate immunity, specifically the interaction of macrophages, neutrophils, and dendritic cells with pathogens such as West Nile virus and the agent of Lyme disease, Borrelia burgdorferi, including elucidating effects of vector saliva on phagocyte function. In studies of the pathogenesis of West Nile virus, the Montgomery lab has described inhibition of macrophage function, an unexpected biphasic role for PMNs in infection, and effects of aging on innate immunity, including dysregulation of TLR3 responses in macrophages and reduced responses of dendritic cells to infection with West Nile virus.
Erol Fikrig received a B.A. in chemistry from Cornell University and an M.D. from Cornell University Medical College. Dr. Fikrig did a residency in internal medicine at Vanderbilt University School of Medicine and was a fellow in infectious diseases and immunobiology at Yale University School of Medicine. He is currently a Professor of Medicine, Microbial Pathogenesis, and Epidemiology and Public Health, a Waldemar Von Zedtwitz Professor of Medicine, Chief of Infectious Diseases, and an investigator with the Howard Hughes Medical Institute. He currently leads a research group studying the immunopathogenesis of arthropod-borne diseases. Lyme disease, human granulocytic anaplasmosis, and West Nile encephalitis are areas of particular interest. Studies are directed at understanding the interactions between pathogen, host, and vector that result in virulence and transmission and the molecular basis of disease in animal models and patient populations.
REFERENCES
- 1. Ader DB, et al. 2004. Modulation of dengue virus infection of dendritic cells by Aedes aegypti saliva. Viral Immunol. 17:252–265 [DOI] [PubMed] [Google Scholar]
- 2. Anderson JF, Andreadis TG, Main AJ, Ferrandino FJ, Vossbrinck CR. 2006. West Nile virus from female and male mosquitoes (Diptera: Culicidae) in subterranean, ground, and canopy habitats in Connecticut. J. Med. Entomol. 43:1010–1019 [DOI] [PubMed] [Google Scholar]
- 3. Anderson JF, et al. 1999. Isolation of West Nile virus from mosquitoes, crows, and a Cooper's hawk in Connecticut. Science 286:2331–2333 [DOI] [PubMed] [Google Scholar]
- 4. Andreadis TG, Anderson JF, Vossbrinck CR, Main AJ. 2004. Epidemiology of West Nile virus in Connecticut: a five-year analysis of mosquito data 1999–2003. Vector Borne Zoonotic Dis. 4:360–378 [DOI] [PubMed] [Google Scholar]
- 5. Appler KK, et al. 2010. Persistence of West Nile virus in the central nervous system and periphery of mice. PLoS One 5:e10649 doi:10.1371/journal.pone.0010649 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Arjona A, et al. 2007. Abrogation of macrophage migration inhibitory factor decreases West Nile virus lethality by limiting viral neuroinvasion. J. Clin. Invest. 117:3059–3066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Arjona A, Wang P, Montgomery RR, Fikrig E. 2011. Innate immune control of West Nile virus infection. Cell. Microbiol. 13:1648–1658 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Arnold SJ, Osvath SR, Hall RA, King NJ, Sedger LM. 2004. Regulation of antigen processing and presentation molecules in West Nile virus-infected human skin fibroblasts. Virology 324:286–296 [DOI] [PubMed] [Google Scholar]
- 9. Atkinson MP, et al. 2007. Analyzing the control of mosquito-borne diseases by a dominant lethal genetic system. Proc. Natl. Acad. Sci. U. S. A. 104:9540–9545 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9a. Bai F, et al. 2009. IL-10 signaling blockade controls murine West Nile virus infection. PLoS Pathog. 5:e1000610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Bai F, et al. 2010. A paradoxical role for neutrophils in the pathogenesis of West Nile virus J. Infect. Dis. 202:1804–1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Reference deleted.
- 12. Bai F, et al. 2007. Antiviral peptides targeting the West Nile virus envelope protein. J. Virol. 81:2047–2055 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Barkhash AV, et al. 2010. Variability in the 2′-5′-oligoadenylate synthetase gene cluster is associated with human predisposition to tick-borne encephalitis virus-induced disease. J. Infect. Dis. 202:1813–1818 [DOI] [PubMed] [Google Scholar]
- 14. Bartholomay LC, et al. 2010. Pathogenomics of Culex quinquefasciatus and meta-analysis of infection responses to diverse pathogens. Science 330:88–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Beasley DW. 2011. Vaccines and immunotherapeutics for the prevention and treatment of infections with West Nile virus. Immunotherapy 3:269–285 [DOI] [PubMed] [Google Scholar]
- 16. Ben-Nathan D, Huitinga I, Lustig S, van Rooijen N, Kobiler D. 1996. West Nile virus neuroinvasion and encephalitis induced by macrophage depletion in mice. Arch. Virol. 141:459–469 [DOI] [PubMed] [Google Scholar]
- 17. Bhuvanakantham R, Chong MK, Ng ML. 2009. Specific interaction of capsid protein and importin-alpha/beta influences West Nile virus production. Biochem. Biophys. Res. Commun. 389:63–69 [DOI] [PubMed] [Google Scholar]
- 18. Biedenbender R, Bevilacqua J, Gregg AM, Watson M, Dayan G. 2011. Phase II, randomized, double-blind, placebo-controlled, multicenter study to investigate the immunogenicity and safety of a West Nile virus vaccine in healthy adults. J. Infect. Dis. 203:75–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Bigham AW, et al. 2011. Host genetic risk factors for West Nile virus infection and disease progression. PLoS One 6:e24745 doi:10.1371/journal.pone.0024745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Bowen RA, Nemeth NM. 2007. Experimental infections with West Nile virus. Curr. Opin. Infect. Dis. 20:293–297 [DOI] [PubMed] [Google Scholar]
- 21. Brackney DE, Beane JE, Ebel GD. 2009. RNAi targeting of West Nile virus in mosquito midguts promotes virus diversification. PLoS Pathog. 5:e1000502 doi:10.1371/journal.ppat.1000502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Brackney DE, Foy BD, Olson KE. 2008. The effects of midgut serine proteases on dengue virus type 2 infectivity of Aedes aegypti. Am. J. Trop. Med. Hyg. 79:267–274 [PMC free article] [PubMed] [Google Scholar]
- 23. Brass AL, et al. 2009. The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and Dengue virus. Cell 139:1243–1254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Brien JD, et al. 2011. Interferon regulatory factor-1 (IRF-1) shapes both innate and CD8(+) T cell immune responses against West Nile virus infection. PLoS Pathog. 7:e1002230 doi:10.1371/journal.ppat.1002230 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Brien JD, Uhrlaub JL, Nikolich-Zugich J. 2008. West Nile virus-specific CD4 T cells exhibit direct antiviral cytokine secretion and cytotoxicity and are sufficient for antiviral protection. J. Immunol. 181:8568–8575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Briese T, Jia XY, Huang C, Grady LJ, Lipkin WI. 1999. Identification of a Kunjin/West Nile-like flavivirus in brains of patients with New York encephalitis. Lancet 354:1261–1262 [DOI] [PubMed] [Google Scholar]
- 27. Brown AN, Kent KA, Bennett CJ, Bernard KA. 2007. Tissue tropism and neuroinvasion of West Nile virus do not differ for two mouse strains with different survival rates. Virology 368:422–430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Busch MP, et al. 2008. Virus and antibody dynamics in acute West Nile virus infection. J. Infect. Dis. 198:984–993. [DOI] [PubMed] [Google Scholar]
- 29. Reference deleted.
- 30. Calisher CH, et al. 1989. Antigenic relationships between flaviviruses as determined by cross-neutralization tests with polyclonal antisera. J. Gen. Virol. 70:37–43 [DOI] [PubMed] [Google Scholar]
- 31. Calvo E, Ribeiro JM. 2006. A novel secreted endonuclease from Culex quinquefasciatus salivary glands. J. Exp. Biol. 209:2651–2659 [DOI] [PubMed] [Google Scholar]
- 32. Campbell CL, et al. 2008. Aedes aegypti uses RNA interference in defense against Sindbis virus infection. BMC Microbiol. 8:47 doi:10.1186/1471-2180-8-47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Campbell GL, Marfin AA, Lanciotti RS, Gubler DJ. 2002. West Nile virus. Lancet Infect. Dis. 2:519–529 [DOI] [PubMed] [Google Scholar]
- 34. Carlson J, Suchman E, Buchatsky L. 2006. Densoviruses for control and genetic manipulation of mosquitoes. Adv. Virus Res. 68:361–392 [DOI] [PubMed] [Google Scholar]
- 35. Casrouge A, et al. 2011. Evidence for an antagonist form of the chemokine CXCL10 in patients chronically infected with HCV. J. Clin. Invest. 121:308–317 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35a. Centers for Disease Control and Prevention 2002. Laboratory-acquired West Nile virus infections—United States, 2002. MMWR Morb. Mortal. Wkly. Rep. 51:1133–1135 [PubMed] [Google Scholar]
- 36. Cheng G, et al. 2010. A C-type lectin collaborates with a CD45 phosphatase homolog to facilitate West Nile virus infection of mosquitoes. Cell 142:714–725 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Cheng Y, King NJ, Kesson AM. 2004. Major histocompatibility complex class I (MHC-I) induction by West Nile virus: involvement of 2 signaling pathways in MHC-I up-regulation. J. Infect. Dis. 189:658–668 [DOI] [PubMed] [Google Scholar]
- 38. Cheng Y, King NJ, Kesson AM. 2004. The role of tumor necrosis factor in modulating responses of murine embryo fibroblasts by flavivirus, West Nile. Virology 329:361–370 [DOI] [PubMed] [Google Scholar]
- 39. Chotkowski HL, et al. 2008. West Nile virus infection of Drosophila melanogaster induces a protective RNAi response. Virology 377:197–206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Chung KM, et al. 2006. West Nile virus nonstructural protein NS1 inhibits complement activation by binding the regulatory protein factor H. Proc. Natl. Acad. Sci. U. S. A. 103:19111–19116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Colpitts TM, Barthel S, Wang P, Fikrig E. 2011. Dengue virus capsid protein binds core histones and inhibits nucleosome formation in human liver cells. PLoS One 6:e24365 doi:10.1371/journal.pone.0024365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Colpitts TM, et al. 2011. Alterations in the Aedes aegypti transcriptome during infection with West Nile, dengue and yellow fever viruses. PLoS Pathog. 7:e1002189 doi:10.1371/journal.ppat.1002189 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Colpitts TM, et al. 2011. prM-antibody renders immature West Nile virus infectious in vivo. J. Gen. Virol. 92:2281–2285 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Costa A, Jan E, Sarnow P, Schneider D. 2009. The Imd pathway is involved in antiviral immune responses in Drosophila. PLoS One 4:e7436 doi:10.1371/journal.pone.0007436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Cross ML, Cupp EW, Enriquez FJ. 1994. Differential modulation of murine cellular immune responses by salivary gland extract of Aedes aegypti. Am. J. Trop. Med. Hyg. 51:690–696 [DOI] [PubMed] [Google Scholar]
- 46. Cupp EW, et al. 2007. West Nile virus infection in mosquitoes in the mid-south USA, 2002-2005. J. Med. Entomol. 44:117–125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Daffis S, Samuel MA, Keller BC, Gale M, Jr, Diamond MS. 2007. Cell-specific IRF-3 responses protect against West Nile virus infection by interferon-dependent and -independent mechanisms. PLoS Pathog. 3:e106 doi:10.1371/journal.ppat.0030106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Daffis S, Samuel MA, Suthar MS, Gale M, Jr, Diamond MS. 2008. Toll-like receptor 3 has a protective role against West Nile virus infection. J. Virol. 82:10349–10358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Daffis S, et al. 2008. Interferon regulatory factor IRF-7 induces the antiviral alpha interferon response and protects against lethal West Nile virus infection. J. Virol. 82:8465–8475 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Dai J, et al. 2009. Antibodies against a tick protein, Salp15, protect mice from the Lyme disease agent. Cell Host Microbe 6:482–492 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. da Silva Voorham JM, et al. 2012. Antibodies against the envelope glycoprotein promote infectivity of immature dengue virus serotype 2. PLoS One 7:e29957 doi:10.1371/journal.pone.0029957 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Davis CW, et al. 2006. West Nile virus discriminates between DC-SIGN and DC-SIGNR for cellular attachment and infection. J. Virol. 80:1290–1301 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Demento SL, et al. 2010. TLR9-targeted biodegradable nanoparticles as immunization vectors protect against West Nile encephalitis. J. Immunol. 185:2989–2997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53a. Diamond MS, Shrestha B, Marri A, Mahan D, Engle M. 2003. B cells and antibody play critical roles in the immediate defense of disseminated infection by West Nile encephalitis virus. J. Virol. 77:2578–2586 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Diamond MS. 2005. Development of effective therapies against West Nile virus infection. Expert Rev. Anti Infect. Ther. 3:931–944 [DOI] [PubMed] [Google Scholar]
- 55. Douglas MW, Kesson AM, King NJ. 1994. CTL recognition of West Nile virus-infected fibroblasts is cell cycle dependent and is associated with virus-induced increases in class I MHC antigen expression. Immunology 82:561–570 [PMC free article] [PubMed] [Google Scholar]
- 56. Edwards JF, Higgs S, Beaty BJ. 1998. Mosquito feeding-induced enhancement of Cache Valley virus (Bunyaviridae) infection in mice. J. Med. Entomol. 35:261–265 [DOI] [PubMed] [Google Scholar]
- 57. Elshuber S, Allison SL, Heinz FX, Mandl CW. 2003. Cleavage of protein prM is necessary for infection of BHK-21 cells by tick-borne encephalitis virus. J. Gen. Virol. 84:183–191 [DOI] [PubMed] [Google Scholar]
- 58. Farajollahi A, Nelder MP. 2009. Changes in Aedes albopictus (Diptera: Culicidae) populations in New Jersey and implications for arbovirus transmission. J. Med. Entomol. 46:1220–1224 [DOI] [PubMed] [Google Scholar]
- 59. Franz AW, et al. 2006. Engineering RNA interference-based resistance to dengue virus type 2 in genetically modified Aedes aegypti. Proc. Natl. Acad. Sci. U. S. A. 103:4198–4203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Fredericksen BL, Gale M., Jr 2006. West Nile virus evades activation of interferon regulatory factor 3 through RIG-I-dependent and -independent pathways without antagonizing host defense signaling. J. Virol. 80:2913–2923 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Fredericksen BL, Keller BC, Fornek J, Katze MG, Gale M., Jr 2008. Establishment and maintenance of the innate antiviral response to West Nile Virus involves both RIG-I and MDA5 signaling through IPS-1. J. Virol. 82:609–616 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Fredericksen BL, Smith M, Katze MG, Shi PY, Gale M., Jr 2004. The host response to West Nile Virus infection limits viral spread through the activation of the interferon regulatory factor 3 pathway. J. Virol. 78:7737–7747 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Friebe P, Harris E. 2010. Interplay of RNA elements in the dengue virus 5′ and 3′ ends required for viral RNA replication. J. Virol. 84:6103–6118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Fu G, et al. 2007. Female-specific insect lethality engineered using alternative splicing. Nat. Biotechnol. 25:353–357 [DOI] [PubMed] [Google Scholar]
- 65. Fu G, et al. 2010. Female-specific flightless phenotype for mosquito control. Proc. Natl. Acad. Sci. U. S. A. 107:4550–4554 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Gibney KB, et al. 2011. West Nile virus RNA not detected in urine of 40 people tested 6 years after acute West Nile virus disease. J. Infect. Dis. 203:344–347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Gilfoy FD, Mason PW. 2007. West Nile virus-induced interferon production is mediated by the double-stranded RNA-dependent protein kinase PKR. J. Virol. 81:11148–11158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Girard YA, Popov V, Wen J, Han V, Higgs S. 2005. Ultrastructural study of West Nile virus pathogenesis in Culex pipiens quinquefasciatus (Diptera: Culicidae). J. Med. Entomol. 42:429–444 [DOI] [PubMed] [Google Scholar]
- 69. Glass WG, et al. 2006. CCR5 deficiency increases risk of symptomatic West Nile virus infection. J. Exp. Med. 203:35–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Goodell JR, Puig-Basagoiti F, Forshey BM, Shi PY, Ferguson DM. 2006. Identification of compounds with anti-West Nile virus activity. J. Med. Chem. 49:2127–2137 [DOI] [PubMed] [Google Scholar]
- 71. Gould HL, et al. 2005. Protective and therapeutic capacity of human single chain Fv-Fc fusion proteins against West Nile virus. J. Virol. 79:14606–14613 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Gould LH, Fikrig E. 2004. West Nile virus: a growing concern? J. Clin. Invest. 113:1102–1107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Granwehr BP, et al. 2004. West Nile virus: where are we now? Lancet Infect. Dis. 4:547–556 [DOI] [PubMed] [Google Scholar]
- 74. Gu B, et al. 2006. Discovery of small molecule inhibitors of West Nile virus using a high-throughput sub-genomic replicon screen. Antiviral Res. 70:39–50 [DOI] [PubMed] [Google Scholar]
- 75. Gubler DJ. 2007. The continuing spread of West Nile virus in the western hemisphere. Clin. Infect. Dis. 45:1039–1046. [DOI] [PubMed] [Google Scholar]
- 76. Reference deleted.
- 77. Guo X, et al. 2009. Inhibition of neutrophil function by two tick salivary proteins. Infect. Immun. 77:2320–2329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Hamdan A, et al. 2002. Possible benefit of intravenous immunoglobulin therapy in a lung transplant recipient with West Nile virus encephalitis. Transpl. Infect. Dis. 4:160–162 [DOI] [PubMed] [Google Scholar]
- 79. Hamer GL, et al. 2008. Culex pipiens (Diptera: Culicidae): a bridge vector of West Nile virus to humans. J. Med. Entomol. 45:125–128 [DOI] [PubMed] [Google Scholar]
- 80. Hamer GL, et al. 2009. Host selection by Culex pipiens mosquitoes and West Nile virus amplification. Am. J. Trop. Med. Hyg. 80:268–278 [PubMed] [Google Scholar]
- 81. Hayes EB, O'Leary DR. 2004. West Nile virus infection: a pediatric perspective. Pediatrics 113:1375–1381 [DOI] [PubMed] [Google Scholar]
- 82. Hoffmann AA, et al. 2011. Successful establishment of Wolbachia in Aedes populations to suppress dengue transmission. Nature 476:454–457 [DOI] [PubMed] [Google Scholar]
- 83. Holick J, Kyle A, Ferraro W, Delaney RR, Iwaseczko M. 2002. Discovery of Aedes albopictus infected with west nile virus in southeastern Pennsylvania. J. Am. Mosq. Control Assoc. 18:131. [PubMed] [Google Scholar]
- 84. Hudson A, Bowman L, Orr CW. 1960. Effects of absence of saliva on blood feeding by mosquitoes. Science 131:1730–1731 [DOI] [PubMed] [Google Scholar]
- 85. Iwamoto M, et al. 2003. Transmission of West Nile virus from an organ donor to four transplant recipients. N. Engl. J. Med. 348:2196–2203 [DOI] [PubMed] [Google Scholar]
- 86. Jarman RV, Morgan PN, Duffy CE. 1968. Persistence of West Nile virus in L-929 mouse fibroblasts. Proc. Soc. Exp. Biol. Med. 129:633–637 [DOI] [PubMed] [Google Scholar]
- 87. Kajaste-Rudnitski A, et al. 2006. The 2′,5′-oligoadenylate synthetase 1b is a potent inhibitor of West Nile virus replication inside infected cells. J. Biol. Chem. 281:4624–4637 [DOI] [PubMed] [Google Scholar]
- 88. Kalil AC, et al. 2005. Use of interferon-alpha in patients with West Nile encephalitis: report of 2 cases. Clin. Infect. Dis. 40:764–766 [DOI] [PubMed] [Google Scholar]
- 89. Kato N, et al. 2008. Evaluation of the function of a type I peritrophic matrix as a physical barrier for midgut epithelium invasion by mosquito-borne pathogens in Aedes aegypti. Vector Borne Zoonotic Dis. 8:701–712 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Keene KM, et al. 2004. RNA interference acts as a natural antiviral response to O'nyong-nyong virus (Alphavirus; Togaviridae) infection of Anopheles gambiae. Proc. Natl. Acad. Sci. U. S. A. 101:17240–17245 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Kent R, Juliusson L, Weissmann M, Evans S, Komar N. 2009. Seasonal blood-feeding behavior of Culex tarsalis (Diptera: Culicidae) in Weld County, Colorado, 2007. J. Med. Entomol. 46:380–390 [DOI] [PubMed] [Google Scholar]
- 92. Khromykh AA, Meka H, Guyatt KJ, Westaway EG. 2001. Essential role of cyclization sequences in flavivirus RNA replication. J. Virol. 75:6719–6728 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Kilpatrick AM. 2011. Globalization, land use, and the invasion of West Nile virus. Science 334:323–327 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Kilpatrick AM, Daszak P, Jones MJ, Marra PP, Kramer LD. 2006. Host heterogeneity dominates West Nile virus transmission. Proc. Biol. Sci. 273:2327–2333 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Kilpatrick AM, et al. 2005. West Nile virus risk assessment and the bridge vector paradigm. Emerg. Infect. Dis. 11:425–429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Kilpatrick AM, Kramer LD, Jones MJ, Marra PP, Daszak P. 2006. West Nile virus epidemics in North America are driven by shifts in mosquito feeding behavior. PLoS Biol. 4:e82 doi:10.1371/journal.pbio.0040082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. King NJ, et al. 2007. Immunopathology of flavivirus infections. Immunol. Cell Biol. 85:33–42 [DOI] [PubMed] [Google Scholar]
- 97a. Klein RS, et al. 2005. Neuronal CXCL10 directs CD8+ T-cell recruitment and control of West Nile virus encephalitis. J. Virol. 79:11457–11466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Kokoza V, et al. 2000. Engineering blood meal-activated systemic immunity in the yellow fever mosquito, Aedes aegypti. Proc. Natl. Acad. Sci. U. S. A. 97:9144–9149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Komar N, et al. 2003. Experimental infection of North American birds with the New York 1999 strain of West Nile virus. Emerg. Infect. Dis. 9:311–322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Kong KF, et al. 2008. Dysregulation of TLR3 impairs the innate immune response to West Nile virus in the elderly. J. Virol. 82:7613–7623 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Kuno G, Chang GJ, Tsuchiya KR, Karabatsos N, Cropp CB. 1998. Phylogeny of the genus Flavivirus. J. Virol. 72:73–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Kurane I, Janus J, Ennis FA. 1992. Dengue virus infection of human skin fibroblasts in vitro production of IFN-beta, IL-6 and GM-CSF. Arch. Virol. 124:21–30 [DOI] [PubMed] [Google Scholar]
- 103. Kusne S, Smilack J. 2005. Transmission of West Nile virus by organ transplantation. Liver Transpl. 11:239–241 [DOI] [PubMed] [Google Scholar]
- 104. Lanciotti RS, et al. 1999. Origin of the West Nile virus responsible for an outbreak of encephalitis in the northeastern United States. Science 286:2333–2337 [DOI] [PubMed] [Google Scholar]
- 105. Lanteri MC, et al. 2008. Comprehensive analysis of West Nile virus-specific T cell responses in humans. J. Infect. Dis. 197:1296–1306. [DOI] [PubMed] [Google Scholar]
- 106. Reference deleted.
- 107. Lanteri MC, et al. 2011. Association between HLA class I and class II alleles and the outcome of West Nile virus infection: an exploratory study. PLoS One 6:e22948 doi:10.1371/journal.pone.0022948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Lanteri MC, et al. 2009. Tregs control the development of symptomatic West Nile virus infection in humans and mice. J. Clin. Invest. 119:3266–3277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Lazear HM, Pinto AK, Vogt MR, Gale M, Jr, Diamond MS. 2011. Beta interferon controls West Nile virus infection and pathogenesis in mice. J. Virol. 85:7186–7194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Lee E, Hall RA, Lobigs M. 2004. Common E protein determinants for attenuation of glycosaminoglycan-binding variants of Japanese encephalitis and West Nile viruses. J. Virol. 78:8271–8280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Li J, et al. 2003. Asymmetric flaccid paralysis: a neuromuscular presentation of West Nile virus infection. Ann. Neurol. 53:703–710 [DOI] [PubMed] [Google Scholar]
- 112. Lim JK, et al. 2009. Genetic variation in OAS1 is a risk factor for initial infection with West Nile virus in man. PLoS Pathog. 5:e1000321 doi:10.1371/journal.ppat.1000321 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Lim JK, et al. 2008. Genetic deficiency of chemokine receptor CCR5 is a strong risk factor for symptomatic West Nile virus infection: a meta-analysis of 4 cohorts in the US epidemic. J. Infect. Dis. 197:262–265 [DOI] [PubMed] [Google Scholar]
- 114. Lim JK, et al. 2010. CCR5 deficiency is a risk factor for early clinical manifestations of West Nile virus infection but not for viral transmission. J. Infect. Dis. 201:178–185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114a. Lim JK, et al. 2011. Chemokine receptor Ccr2 is critical for monocyte accumulation and survival in West Nile virus encephalitis. J. Immunol. 186:471–478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Lim PY, Behr MJ, Chadwick CM, Shi PY, Bernard KA. 2011. Keratinocytes are cell targets of West Nile virus in vivo. J. Virol. 85:5197–5201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Lin CW, et al. 2008. Interferon antagonist function of Japanese encephalitis virus NS4A and its interaction with DEAD-box RNA helicase DDX42. Virus Res. 137:49–55 [DOI] [PubMed] [Google Scholar]
- 117. Lindenbach BD, Rice CM. 2003. Molecular biology of flaviviruses. Adv. Virus Res. 59:23–61 [DOI] [PubMed] [Google Scholar]
- 118. Lindenbach DR, CM 2001. Flaviviridae: the viruses and their replication, p 991–1041 In Knipe DM, Howley PM. (ed), Fields virology, 4th ed Lippincott Williams and Wilkins, Philadelphia, PA [Google Scholar]
- 119. Lindsey NP, et al. 2007. West Nile virus activity—United States, 2006. JAMA 298:619–621 [Google Scholar]
- 120. Lindsey NP, Sejvar JJ, Bode AV, Pape WJ, Campbell GL. 2012. Delayed mortality in a cohort of persons hospitalized with West Nile virus disease in Colorado in 2003. Vector Borne Zoonotic Dis. 12:230–235 [DOI] [PubMed] [Google Scholar]
- 121. Liu WJ, Chen HB, Wang XJ, Huang H, Khromykh AA. 2004. Analysis of adaptive mutations in Kunjin virus replicon RNA reveals a novel role for the flavivirus nonstructural protein NS2A in inhibition of beta interferon promoter-driven transcription. J. Virol. 78:12225–12235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Liu WJ, et al. 2006. A single amino acid substitution in the West Nile virus nonstructural protein NS2A disables its ability to inhibit alpha/beta interferon induction and attenuates virus virulence in mice. J. Virol. 80:2396–2404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Loeb M, Eskandarian S, Rupp M. 2011. Genetic variants and susceptibility to neurological complications following West Nile virus infection. J. Infect. Dis. 204:1031–1037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Luplertlop N, et al. 2011. Induction of a peptide with activity against a broad spectrum of pathogens in the Aedes aegypti salivary gland, following infection with dengue virus. PLoS Pathog. 7:e1001252 doi:10.1371/journal.ppat.1001252 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Marfin AA, Gubler DJ. 2001. West Nile encephalitis: an emerging disease in the United States. Clin. Infect. Dis. 33:1713–1719 [DOI] [PubMed] [Google Scholar]
- 126. McGee CE, et al. 2010. Infection, dissemination, and transmission of a West Nile virus green fluorescent protein infectious clone by Culex pipiens quinquefasciatus mosquitoes. Vector Borne Zoonotic Dis. 10:267–274 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. McMeniman CJ, et al. 2009. Stable introduction of a life-shortening Wolbachia infection into the mosquito Aedes aegypti. Science 323:141–144 [DOI] [PubMed] [Google Scholar]
- 128. Medlock J, Luz PM, Struchiner CJ, Galvani AP. 2009. The impact of transgenic mosquitoes on dengue virulence to humans and mosquitoes. Am. Nat. 174:565–577 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Mehlhop E, Diamond MS. 2006. Protective immune responses against West Nile virus are primed by distinct complement activation pathways. J. Exp. Med. 203:1371–1381 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Mehlhop E, et al. 2009. Complement protein C1q reduces the stoichiometric threshold for antibody-mediated neutralization of West Nile virus. Cell Host Microbe 6:381–391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Mehlhop E, et al. 2005. Complement activation is required for induction of a protective antibody response against West Nile virus infection. J. Virol. 79:7466–7477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Reference deleted.
- 133. Reference deleted.
- 134. Modis Y, Ogata S, Clements D, Harrison SC. 2004. Structure of the dengue virus envelope protein after membrane fusion. Nature 427:313–319 [DOI] [PubMed] [Google Scholar]
- 135. Moesker B, Rodenhuis-Zybert IA, Meijerhof T, Wilschut J, Smit JM. 2010. Characterization of the functional requirements of West Nile virus membrane fusion. J. Gen. Virol. 91:389–393 [DOI] [PubMed] [Google Scholar]
- 136. Molaei G, Andreadis TG, Armstrong PM, Anderson JF, Vossbrinck CR. 2006. Host feeding patterns of Culex mosquitoes and West Nile virus transmission, northeastern United States. Emerg. Infect. Dis. 12:468–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Reference deleted.
- 138. Molina-Cruz A, et al. 2005. Effect of mosquito midgut trypsin activity on dengue-2 virus infection and dissemination in Aedes aegypti. Am. J. Trop. Med. Hyg. 72:631–637 [PubMed] [Google Scholar]
- 139. Munoz-Jordan JL, et al. 2005. Inhibition of alpha/beta interferon signaling by the NS4B protein of flaviviruses. J. Virol. 79:8004–8013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Murray K, et al. 2006. Risk factors for encephalitis and death from West Nile virus infection. Epidemiol. Infect. 134:1325–1332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Murray K, et al. 2010. Persistent infection with West Nile virus years after initial infection. J. Infect. Dis. 201:2–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Reference deleted.
- 143. Murray KO, Walker C, Gould E. 2011. The virology, epidemiology, and clinical impact of West Nile virus: a decade of advancements in research since its introduction into the Western Hemisphere. Epidemiol. Infect. 139:807–817. [DOI] [PubMed] [Google Scholar]
- 144. Reference deleted.
- 145. Nash D, et al. 2001. The outbreak of West Nile virus infection in the New York City area in 1999. N. Engl. J. Med. 344:1807–1814 [DOI] [PubMed] [Google Scholar]
- 146. Oliphant T, et al. 2005. Development of a humanized monoclonal antibody with therapeutic potential against West Nile virus. Nat. Med. 11:522–530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Olson KE, et al. 2002. Developing arbovirus resistance in mosquitoes. Insect Biochem. Mol. Biol. 32:1333–1343 [DOI] [PubMed] [Google Scholar]
- 148. Olson KE, et al. 1996. Genetically engineered resistance to dengue-2 virus transmission in mosquitoes. Science 272:884–886 [DOI] [PubMed] [Google Scholar]
- 149. Papa A, Danis K, Athanasiadou A, Delianidou M, Panagiotopoulos T. 2011. Persistence of West Nile virus immunoglobulin M antibodies, Greece. J. Med. Virol. 83:1857–1860 [DOI] [PubMed] [Google Scholar]
- 150. Parsons R, et al. 2008. The memory T cell response to West Nile virus in symptomatic humans following natural infection is not influenced by age and is dominated by a restricted set of CD8+ T cell epitopes. J. Immunol. 181:1563–1572 [DOI] [PubMed] [Google Scholar]
- 151. Pesko K, Mores CN. 2009. Effect of sequential exposure on infection and dissemination rates for West Nile and St. Louis encephalitis viruses in Culex quinquefasciatus. Vector Borne Zoonotic Dis. 9:281–286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Petersen LR, Marfin AA. 2002. West Nile virus: a primer for the clinician. Ann. Intern. Med. 137:173–179. [DOI] [PubMed] [Google Scholar]
- 153. Reference deleted.
- 154. Piazza P, et al. 2010. Surface phenotype and functionality of WNV specific T cells differ with age and disease severity. PLoS One 5:e15343 doi:10.1371/journal.pone.0015343 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Pinto AK, et al. 2011. A temporal role of type I interferon signaling in CD8+ T cell maturation during acute West Nile virus infection. PLoS Pathog. 7:e1002407 doi:10.1371/journal.ppat.1002407 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Pogodina VV, et al. 1983. Study on West Nile virus persistence in monkeys. Arch. Virol. 75:71–86 [DOI] [PubMed] [Google Scholar]
- 157. Prince HE, Lape-Nixon M, Yeh C, Tobler LH, Busch MP. 2008. Persistence of antibodies to West Nile virus nonstructural protein 5. J. Clin. Virol. 43:102–106 [DOI] [PubMed] [Google Scholar]
- 158. Puig-Basagoiti F, et al. 2006. Triaryl pyrazoline compound inhibits flavivirus RNA replication. Antimicrob. Agents Chemother. 50:1320–1329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Qian F, et al. 2011. Impaired interferon signaling in dendritic cells from older donors infected in vitro with West Nile virus. J. Infect. Dis. 203:1415–1424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Ramamoorthi N, et al. 2005. The Lyme disease agent exploits a tick protein to infect the mammalian host. Nature 436:573–577 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Ramirez JL, Dimopoulos G. 2010. The Toll immune signaling pathway control conserved anti-dengue defenses across diverse Ae. aegypti strains and against multiple dengue virus serotypes. Dev. Comp. Immunol. 34:625–629 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Rappole JH, Derrickson SR, Hubalek Z. 2000. Migratory birds and spread of West Nile virus in the Western Hemisphere. Emerg. Infect. Dis. 6:319–328 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Reisen WK, Fang Y, Martinez VM. 2005. Avian host and mosquito (Diptera: Culicidae) vector competence determine the efficiency of West Nile and St. Louis encephalitis virus transmission. J. Med. Entomol. 42:367–375 [DOI] [PubMed] [Google Scholar]
- 164. Reisen WK, Fang Y, Martinez VM. 2006. Vector competence of Culiseta incidens and Culex thriambus for West Nile virus. J. Am. Mosq. Control Assoc. 22:662–665 [DOI] [PubMed] [Google Scholar]
- 165. Rezepova AI, Kuz'mina SV, Kulikova KS, Unanov SS. 1971. Cultivation of various arboviruses in new transplantable lines of mouse fibroblasts. Vopr. Virusol. 16:704–707 [PubMed] [Google Scholar]
- 166. Ribeiro JM. 2000. Blood-feeding in mosquitoes: probing time and salivary gland anti-haemostatic activities in representatives of three genera (Aedes, Anopheles, Culex). Med. Vet. Entomol. 14:142–148 [DOI] [PubMed] [Google Scholar]
- 167. Ribeiro JM, et al. 2007. An annotated catalogue of salivary gland transcripts in the adult female mosquito, Aedes aegypti. BMC Genomics 8:6 doi:10.1186/1471-2164-8-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Ribeiro JM, Charlab R, Rowton ED, Cupp EW. 2000. Simulium vittatum (Diptera: Simuliidae) and Lutzomyia longipalpis (Diptera: Psychodidae) salivary gland hyaluronidase activity. J. Med. Entomol. 37:743–747 [DOI] [PubMed] [Google Scholar]
- 169. Ribeiro JM, Charlab R, Valenzuela JG. 2001. The salivary adenosine deaminase activity of the mosquitoes Culex quinquefasciatus and Aedes aegypti. J. Exp. Biol. 204:2001–2010 [DOI] [PubMed] [Google Scholar]
- 170. Ribeiro JM, Francischetti IM. 2001. Platelet-activating-factor-hydrolyzing phospholipase C in the salivary glands and saliva of the mosquito Culex quinquefasciatus. J. Exp. Biol. 204:3887–3894 [DOI] [PubMed] [Google Scholar]
- 171. Ribeiro JM, Rossignol PA, Spielman A. 1985. Aedes aegypti: model for blood finding strategy and prediction of parasite manipulation. Exp. Parasitol. 60:118–132 [DOI] [PubMed] [Google Scholar]
- 172. Ribeiro JM, Rossignol PA, Spielman A. 1984. Role of mosquito saliva in blood vessel location. J. Exp. Biol. 108:1–7 [DOI] [PubMed] [Google Scholar]
- 173. Ribeiro JM, Valenzuela JG. 2003. The salivary purine nucleosidase of the mosquito, Aedes aegypti. Insect Biochem. Mol. Biol. 33:13–22 [DOI] [PubMed] [Google Scholar]
- 174. Rice CM. 1996. Flaviviradae: the viruses and their replication, p 931–959 In Fields BN, Knipe DM, Howley PM. (ed), Fields virology, 3rd ed Lippincott-Raven, Philadelphia, PA [Google Scholar]
- 175. Richards SL, Anderson SL, Lord CC, Smartt CT, Tabachnick WJ. 2012. Relationships between infection, dissemination, and transmission of West Nile virus RNA in Culex pipiens quinquefasciatus (Diptera: Culicidae). J. Med. Entomol. 49:132–142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Rios JJ, et al. 2010. OAS1 polymorphisms are associated with susceptibility to West Nile encephalitis in horses. PLoS One 5:e10537 doi:10.1371/journal.pone.0010537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Rios M, et al. 2006. Monocytes-macrophages are a potential target in human infection with West Nile virus through blood transfusion. Transfusion 46:659–667 [DOI] [PubMed] [Google Scholar]
- 178. Rocha AC, Braga EM, Araujo MS, Franklin BS, Pimenta PF. 2004. Effect of the Aedes fluviatilis saliva on the development of Plasmodium gallinaceum infection in Gallus (gallus) domesticus. Mem. Inst. Oswaldo Cruz 99:709–715 [DOI] [PubMed] [Google Scholar]
- 179. Rodenhuis-Zybert IA, et al. 2010. Immature dengue virus: a veiled pathogen? PLoS Pathog. 6:e1000718 doi:10.1371/journal.ppat.1000718 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Roehrig JT, et al. 2003. Persistence of virus-reactive serum immunoglobulin m antibody in confirmed West Nile virus encephalitis cases. Emerg. Infect. Dis. 9:376–379 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Samuel MA, Diamond MS. 2005. Alpha/beta interferon protects against lethal West Nile virus infection by restricting cellular tropism and enhancing neuronal survival. J. Virol. 79:13350–13361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Samuel MA, et al. 2006. PKR and RNase L contribute to protection against lethal West Nile Virus infection by controlling early viral spread in the periphery and replication in neurons. J. Virol. 80:7009–7019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Sangiambut S, et al. 2008. Multiple regions in dengue virus capsid protein contribute to nuclear localization during virus infection. J. Gen. Virol. 89:1254–1264 [DOI] [PubMed] [Google Scholar]
- 184. Sardelis MR, Turell MJ, O'Guinn ML, Andre RG, Roberts DR. 2002. Vector competence of three North American strains of Aedes albopictus for West Nile virus. J. Am. Mosq. Control Assoc. 18:284–289 [PubMed] [Google Scholar]
- 185. Scherbik SV, Stockman BM, Brinton MA. 2007. Differential expression of interferon (IFN) regulatory factors and IFN-stimulated genes at early times after West Nile virus infection of mouse embryo fibroblasts. J. Virol. 81:12005–12018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Schneider BS, Higgs S. 2008. 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. 102:400–408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Schneider BS, et al. 2007. Prior exposure to uninfected mosquitoes enhances mortality in naturally-transmitted West Nile virus infection. PLoS One 2:e1171 doi:10.1371/journal.pone.0001171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Schneider BS, et al. 2010. Aedes aegypti saliva alters leukocyte recruitment and cytokine signaling by antigen-presenting cells during West Nile virus infection. PLoS One 5:e11704 doi:10.1371/journal.pone.0011704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Schneider BS, et al. 2006. Potentiation of West Nile encephalitis by mosquito feeding. Viral Immunol. 19:74–82 [DOI] [PubMed] [Google Scholar]
- 190. Schneider BS, Soong L, Zeidner NS, Higgs S. 2004. Aedes aegypti salivary gland extracts modulate anti-viral and TH1/TH2 cytokine responses to sindbis virus infection. Viral Immunol. 17:565–573 [DOI] [PubMed] [Google Scholar]
- 191. Sejvar JJ, et al. 2003. Neurologic manifestations and outcome of West Nile virus infection. JAMA 290:511–515 [DOI] [PubMed] [Google Scholar]
- 192. Sejvar JJ, Lindsey NP, Campbell GL. 2011. Primary causes of death in reported cases of fatal West Nile Fever, United States, 2002-2006. Vector Borne Zoonotic Dis. 11:161–164 [DOI] [PubMed] [Google Scholar]
- 193. Sessions OM, et al. 2009. Discovery of insect and human dengue virus host factors. Nature 458:1047–1050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Shao L, Devenport M, Jacobs-Lorena M. 2001. The peritrophic matrix of hematophagous insects. Arch. Insect Biochem. Physiol. 47:119–125 [DOI] [PubMed] [Google Scholar]
- 195. Shen J, Devery JM, King NJ. 1995. Early induction of interferon-independent virus-specific ICAM-1 (CD54) expression by flavivirus in quiescent but not proliferating fibroblasts—implications for virus-host interactions. Virology 208:437–449 [DOI] [PubMed] [Google Scholar]
- 196. Shi PY, Brinton MA, Veal JM, Zhong YY, Wilson WD. 1996. Evidence for the existence of a pseudoknot structure at the 3′ terminus of the flavivirus genomic RNA. Biochemistry 35:4222–4230 [DOI] [PubMed] [Google Scholar]
- 197. Shrestha B, Diamond MS. 2004. Role of CD8+ T cells in control of West Nile virus infection. J. Virol. 78:8312–8321 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197a. Shrestha B, Pinto AK, Green S, Bosch I, Diamond MS. 2012. CD8+ T cells use TRAIL to restrict West Nile virus pathogenesis by controlling infection in neurons. J. Virol. 86:8937–8948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197b. Shrestha B, et al. 2006. Gamma interferon plays a crucial early antiviral role in protection against West Nile virus infection. J. Virol. 80:5338–5348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Shrestha B, Zhang B, Purtha WE, Klein RS, Diamond MS. 2008. Tumor necrosis factor alpha protects against lethal West Nile virus infection by promoting trafficking of mononuclear leukocytes into the central nervous system. J. Virol. 82:8956–8964 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Siddharthan V, et al. 2009. Persistent West Nile virus associated with a neurological sequela in hamsters identified by motor unit number estimation. J. Virol. 83:4251–4261 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Sim S, Dimopoulos G. 2010. Dengue virus inhibits immune responses in Aedes aegypti cells. PLoS One 5:e10678 doi:10.1371/journal.pone.0010678 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200a. Sitati E, McCandless EE, Klein RS, Diamond MS. 2007. CD40-CD40 ligand interactions promote trafficking of CD8+ T cells into the brain and protection against West Nile virus encephalitis. J. Virol. 81:9801–9811 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200b. Sitati EM, Diamond MS. 2006. CD4+ T-cell responses are required for clearance of West Nile virus from the central nervous system. J. Virol. 80:12060–12069 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Smartt CT, Richards SL, Anderson SL, Erickson JS. 2009. West Nile virus infection alters midgut gene expression in Culex pipiens quinquefasciatus Say (Diptera: Culicidae). Am. J. Trop. Med. Hyg. 81:258–263 [PMC free article] [PubMed] [Google Scholar]
- 202. Souza-Neto JA, Sim S, Dimopoulos G. 2009. An evolutionary conserved function of the JAK-STAT pathway in anti-dengue defense. Proc. Natl. Acad. Sci. U. S. A. 106:17841–17846 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Stracker TH, Thompson S, Grossman GL, Riehle MA, Brown MR. 2002. Characterization of the AeaHP gene and its expression in the mosquito Aedes aegypti (Diptera: Culicidae). J. Med. Entomol. 39:331–342 [DOI] [PubMed] [Google Scholar]
- 204. Styer LM, Bernard KA, Kramer LD. 2006. Enhanced early West Nile virus infection in young chickens infected by mosquito bite: effect of viral dose. Am. J. Trop. Med. Hyg. 75:337–345 [PubMed] [Google Scholar]
- 205. Styer LM, et al. 2007. Mosquitoes inoculate high doses of West Nile virus as they probe and feed on live hosts. PLoS Pathog. 3:1262–1270 doi:10.1371/journal.ppat.0030132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Styer LM, et al. 2011. Mosquito saliva causes enhancement of West Nile virus infection in mice. J. Virol. 85:1517–1527 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Sultana H, et al. 2009. Fusion loop peptide of the West Nile virus envelope protein is essential for pathogenesis and is recognized by a therapeutic cross-reactive human monoclonal antibody. J. Immunol. 183:650–660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Suthar MS, et al. 2010. IPS-1 is essential for the control of West Nile virus infection and immunity. PLoS Pathog. 6:e1000757 doi:10.1371/journal.ppat.1000757 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208a. Suthar MS, et al. 2012. The RIG-I-like receptor LGP2 controls CD8+ T cell survival and fitness. Immunity 37:235–248 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Szretter KJ, et al. 2010. The innate immune adaptor molecule MyD88 restricts West Nile virus replication and spread in neurons of the central nervous system. J. Virol. 84:12125–12138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Szretter KJ, et al. 2009. The immune adaptor molecule SARM modulates tumor necrosis factor alpha production and microglia activation in the brainstem and restricts West Nile Virus pathogenesis. J. Virol. 83:9329–9338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Tassaneetrithep B, et al. 2003. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J. Exp. Med. 197:823–829 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Tempelis CH, Francy DB, Hayes RO, Lofy MF. 1967. Variations in feeding patterns of seven culicine mosquitoes on vertebrate hosts in Weld and Larimer Counties, Colorado. Am. J. Trop. Med. Hyg. 16:111–119 [DOI] [PubMed] [Google Scholar]
- 213. Tesh RB, et al. 2005. Persistent West Nile virus infection in the golden hamster: studies on its mechanism and possible implications for other flavivirus infections. J. Infect. Dis. 192:287–295 [DOI] [PubMed] [Google Scholar]
- 214. Thomas DD, Donnelly CA, Wood RJ, Alphey LS. 2000. Insect population control using a dominant, repressible, lethal genetic system. Science 287:2474–2476 [DOI] [PubMed] [Google Scholar]
- 215. Thoma-Uszynski S, et al. 2001. Induction of direct antimicrobial activity through mammalian Toll-like receptors. Science 291:1544–1547 [DOI] [PubMed] [Google Scholar]
- 216. Tiawsirisup S, Platt KB, Evans RB, Rowley WA. 2005. A comparision of West Nile Virus transmission by Ochlerotatus trivittatus (COQ.), Culex pipiens (L.), and Aedes albopictus (Skuse). Vector Borne Zoonotic Dis. 5:40–47 [DOI] [PubMed] [Google Scholar]
- 217. Tobler LH, et al. 2008. Interferon and interferon-induced chemokine expression is associated with control of acute viremia in West Nile virus-infected blood donors. J. Infect. Dis. 198:979–983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Town T, et al. 2009. Toll-like receptor 7 mitigates lethal West Nile encephalitis via interleukin 23-dependent immune cell infiltration and homing. Immunity 30:242–253 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Unlu I, Mackay AJ, Roy A, Yates MM, Foil LD. 2010. Evidence of vertical transmission of West Nile virus in field-collected mosquitoes. J. Vector Ecol. 35:95–99 [DOI] [PubMed] [Google Scholar]
- 220. Vaidyanathan R, Scott TW. 2006. Apoptosis in mosquito midgut epithelia associated with West Nile virus infection. Apoptosis 11:1643–1651 [DOI] [PubMed] [Google Scholar]
- 221. Vanlandingham DL, et al. 2007. Relative susceptibilties of South Texas mosquitoes to infection with West Nile virus. Am. J. Trop. Med. Hyg. 77:925–928 [PubMed] [Google Scholar]
- 222. Vitek CJ, Richards SL, Mores CN, Day JF, Lord CC. 2008. Arbovirus transmission by Culex nigripalpus in Florida, 2005. J. Med. Entomol. 45:483–493 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Volfova V, Hostomska J, Cerny M, Votypka J, Volf P. 2008. Hyaluronidase of bloodsucking insects and its enhancing effect on leishmania infection in mice. PLoS Negl. Trop. Dis. 2:e294.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Wanasen N, Nussenzveig RH, Champagne DE, Soong L, Higgs S. 2004. Differential modulation of murine host immune response by salivary gland extracts from the mosquitoes Aedes aegypti and Culex quinquefasciatus. Med. Vet. Entomol. 18:191–199 [DOI] [PubMed] [Google Scholar]
- 225. Wang P. IL-22 signaling contributes to West Nile encephalitis pathogenesis. PLoS One, in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Wang P, et al. 2010. Caspase-12 controls West Nile virus infection via the viral RNA receptor RIG-I. Nat. Immunol. 11:912–919 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Wang P, et al. 2008. Matrix metalloproteinase 9 facilitates West Nile virus entry into the brain. J. Virol. 82:8978–8985 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Wang S, et al. 2008. Drak2 contributes to West Nile virus entry into the brain and lethal encephalitis. J. Immunol. 181:2084–2091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Wang T, et al. 2003. IFN-gamma-producing gamma delta T cells help control murine West Nile virus infection. J. Immunol. 171:2524–2531 [DOI] [PubMed] [Google Scholar]
- 230. Wang T, et al. 2004. Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis. Nat. Med. 10:1366–1373 [DOI] [PubMed] [Google Scholar]
- 231. Wasserman HA, Singh S, Champagne DE. 2004. Saliva of the Yellow Fever mosquito, Aedes aegypti, modulates murine lymphocyte function. Parasite Immunol. 26:295–306 [DOI] [PubMed] [Google Scholar]
- 232. Weiss D, et al. 2001. Clinical findings of West Nile virus infection in hospitalized patients, New York and New Jersey, 2000. Emerg. Infect. Dis. 7:654–658 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Welte T, et al. 2009. Toll-like receptor 7-induced immune response to cutaneous West Nile virus infection. J. Gen. Virol. 90:2660–2668 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Westaway EG, Mackenzie JM, Khromykh AA. 2002. Replication and gene function in Kunjin virus. Curr. Top. Microbiol. Immunol. 267:323–351 [DOI] [PubMed] [Google Scholar]
- 235. Whiteman MC, et al. 2010. Development and characterization of non-glycosylated E and NS1 mutant viruses as a potential candidate vaccine for West Nile virus. Vaccine 28:1075–1083 [DOI] [PubMed] [Google Scholar]
- 236. Yakub I, et al. 2005. Single nucleotide polymorphisms in genes for 2′-5′-oligoadenylate synthetase and RNase L inpatients hospitalized with West Nile virus infection. J. Infect. Dis. 192:1741–1748 [DOI] [PubMed] [Google Scholar]
- 236a. Zhang B, Chan YK, Lu B, Diamond MS, Klein RS. 2008. CXCR3 mediates region-specific antiviral T cell trafficking within the central nervous system during West Nile virus encephalitis. J. Immunol. 180:2641–2649 [DOI] [PubMed] [Google Scholar]
- 237. Zhang Y, Kaufmann B, Chipman PR, Kuhn RJ, Rossmann MG. 2007. Structure of immature West Nile virus. J. Virol. 81:6141–6145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Zou S, Foster GA, Dodd RY, Petersen LR, Stramer SL. 2010. West Nile fever characteristics among viremic persons identified through blood donor screening. J. Infect. Dis. 202:1354–1361 [DOI] [PubMed] [Google Scholar]
- 239. Zybert IA, van der Ende-Metselaar H, Wilschut J, Smit JM. 2008. Functional importance of dengue virus maturation: infectious properties of immature virions. J. Gen. Virol. 89:3047–3051 [DOI] [PubMed] [Google Scholar]