ABSTRACT
The mosquito-borne West Nile virus (WNV) is responsible for outbreaks of viral encephalitis in humans, horses, and birds, with particularly virulent strains causing recent outbreaks of disease in eastern Europe, the Middle East, North America, and Australia. Previous studies have phylogenetically separated WNV strains into two main genetic lineages (I and II) containing virulent strains associated with neurological disease. Several WNV-like strains clustering outside these lineages have been identified and form an additional five proposed lineages. However, little is known about whether these strains have the potential to induce disease. In a comparative analysis with the highly virulent lineage I American strain (WNVNY99), the low-pathogenicity lineage II strain (B956), a benign Australian strain, Kunjin (WNVKUN), the African WNV-like Koutango virus (WNVKOU), and a WNV-like isolate from Sarawak, Malaysia (WNVSarawak), were assessed for neuroinvasive properties in a murine model and for their replication kinetics in vitro. While WNVNY99 replicated to the highest levels in vitro, in vivo mouse challenge revealed that WNVKOU was more virulent, with a shorter time to onset of neurological disease and higher morbidity. Histological analysis of WNVKOU- and WNVNY99-infected brain and spinal cords demonstrated more prominent meningoencephalitis and the presence of viral antigen in WNVKOU-infected mice. Enhanced virulence of WNVKOU also was associated with poor viral clearance in the periphery (sera and spleen), a skewed innate immune response, and poor neutralizing antibody development. These data demonstrate, for the first time, potent neuroinvasive and neurovirulent properties of a WNV-like virus outside lineages I and II.
IMPORTANCE In this study, we characterized the in vitro and in vivo properties of previously uncharacterized West Nile virus strains and West Nile-like viruses. We identified a West Nile-like virus, Koutango virus (WNVKOU), that was more virulent than a known virulent lineage I virus, WNVNY99. The enhanced virulence of WNVKOU was associated with poor viral clearance and the induction of a poor neutralizing antibody response. These findings provide new insights into the pathogenesis of West Nile virus.
INTRODUCTION
West Nile virus (WNV) is a mosquito-transmitted, single-stranded, positive-sense flavivirus that has emerged as an important causative agent of viral encephalitis in humans and horses in many parts of the world. Outbreaks of potentially fatal neurological syndromes traditionally have been documented in Europe and Africa (1). However, in recent times strains of WNV have caused large outbreaks of encephalitis in the New World, involving humans and equines in the United States and equines in Australia (2–10). There have also been recent incursions of new, virulent strains in Europe (8–10). In the summer of 2012, the United States saw the second highest number of WNV cases on record with concurrent outbreaks in several European countries, highlighting the continuing public health threat of WNV to humans (11). In Australia, an indigenous strain of WNV, WNV Kunjin (WNVKUN), historically has caused only infrequent and mild symptoms in humans and horses. However, a large outbreak of encephalitis in horses in 2011 saw the emergence of the first virulent strain of WNVKUN in Australia, associated with the acquisition of at least two known molecular markers of WNV virulence not found in the prototype WNVKUN (4), demonstrating ongoing evolution even among low-virulence WNV strains.
Phylogenetic analysis has suggested that WNV emerged in Africa and subsequently dispersed through avian migration and can be separated into two main lineages (I and II), with an additional 5 lineages proposed (12). Lineage I contains WNVKUN isolates and WNV isolates from north, west, and central Africa, southern and eastern Europe, India, the Middle East, and North America. Lineage I can be further divided into 3 clades, with clade 1a containing WNV isolates from around the world, the Australian WNVKUN isolates forming clade 1b, and clade 1c containing isolates from India (previously described as lineage V [13]). Lineage II comprises WNV isolates from west, central, and east Africa and Madagascar (14, 15). Historically, lineage II strains were associated with fever and mild symptoms until 2008, when the emergence of lineage II strains was responsible for outbreaks of neurological disease in Greece, Hungary, and Italy (6, 8, 10, 16).
Studies comparing the virulence of various WNV strains in mice have identified several viral motifs residing in both structural and nonstructural genes as well as in the 5′- and 3′-untranslated regions that were associated with enhanced invasion of the central nervous system (CNS) and onset of neurological disease in this species (17–23). One example of these virulence determinants is N-linked glycosylation at a conserved site in the E protein (residues 154 to 156) of WNV that has been shown to increase virulence of lineage I WNV strains (19, 24) and which likely is mediated via enhanced assembly and/or secretion of virus particles (21, 25). However, the biological influence of N-linked glycosylation on viruses that branch outside lineages I and II has not been investigated.
WNV infection remains subclinical in most humans, but ∼20% may develop symptoms of disease ranging from a mild flu-like illness, known as West Nile fever, to more serious neurological complications, including meningitis and encephalitis. Postneurologic sequelae are common (26). In both humans and mice, WNV encephalitis is characterized by the reaction of resident cells in the CNS and infiltration of inflammatory leukocytes, including monocytes and T cells, in the perivascular space and parenchyma. Although increased age and immunosuppression are risk factors for severe WNV infection in humans, little is known about the mechanisms accountable for this (27, 28). A mouse model of infection is widely used in studies of WNV pathogenesis, and various components of the innate and adaptive immune response, including antibodies, T cells (particularly CD8+ T cells), monocytes/macrophages, and gamma delta T cells, have been identified as being critical for viral clearance. In this model, CD4+ and CD8+ T cells, as well as natural killer cells and infiltrating monocyte/macrophages, accumulate within the CNS in aggregates that colocalize primarily with WNV-infected neurons (29). The majority of murine studies have focused on the virulent lineage I North American strain of WNV, namely, NY99 (WNVNY99), or the lineage II Sarafend strain (30–33), and little is known about the pathogenesis of other strains of WNV, especially those viruses that branch outside lineages I and II.
This study aimed to characterize the pathogenic properties of multiple WNV strains and West Nile-like viruses to identify both virus and host factors leading to disease development. In particular, two previously uncharacterized West Nile-like viruses, WNVSarawak, isolated in 1966 from Borneo, and Koutango virus (WNVKOU), isolated in 1968 from Senegal, were investigated to determine phenotypic characteristics in vitro and virulence in vivo. Aside from phylogenetic data on these two viruses, little information was known prior to this study. Replication kinetics in cells confirmed the importance of N-linked glycosylation in the total amount of infectious virus at different time points. Studies using an adult mouse model of disease demonstrated for the first time that a West Nile-like virus, WNVKOU, branching outside the main lineages, was more virulent than the North American lineage I strain, with the ability to induce more prominent neuropathology. The enhanced virulence of WNVKOU was associated with poor viral clearance in extraneural tissues and a delayed and poor neutralizing antibody response.
MATERIALS AND METHODS
Cell culture and virus production.
African green monkey kidney (Vero) cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 2% fetal bovine serum (FBS), 50 U ml−1 penicillin, 50 μg ml−1 streptomycin, and 2 mM l-glutamine. Human brain microvascular endothelial cells (HBMECs) were cultured in RPMI 1640 medium supplemented with 20% FBS and nonessential amino acids (Invitrogen, Victoria, Australia) (34). HBMECs were plated on collagen-coated 24-well plates for growth kinetics studies. Cells were cultured at 37°C with 5% CO2. Aedes albopictus cells (C6/36) were cultured in RPMI 1640 supplemented with 10% FBS, 50 U ml−1 penicillin, 50 μg ml−1 streptomycin, and 2 mM l-glutamine. These cells were cultured at 28°C.
West Nile virus and West Nile-like viruses used in this study are listed with their sources of isolation in Table 1 and included B956, NY99 (GenBank accession number HQ596519), Sarafend, Koutango (accession number EU082200), Sarawak, MRM16, and Hu6774 viruses. These viruses were selected to represent the two main lineages of WNV (I and II), as well as those proposed to branch outside these lineages, namely, WNVKOU (proposed to represent lineage VII) and WNVSarawak (proposed to represent lineage VI). Strains representing lineage I include the North American strain NY99 (WNVNY99), known to be highly neurovirulent in humans, horses, birds, and mice, as well as two strains of WNVKUN, the only known human isolate of WNVKUN, Hu6774, and a mosquito isolate, MRM16, both thought to have low virulence (Table 1). Lineage II was represented by the prototype strain B956 and Sarafend viruses (31, 35). WNV strain NY99-4132 was obtained from the Division of Vector Borne Infectious Diseases, Centers for Disease Control, Fort Collins, CO. Virus stocks were produced by infecting a subconfluent monolayer of C6/36 cells with virus at a multiplicity of infection (MOI) of 0.1 to 1 in RPMI containing 2% FBS (2% FBS-RPMI). Culture supernatant was harvested and clarified at 72 to 96 h postinfection and stored at −80°C. The concentration of infectious virus in stocks was determined by titration on Vero cells in 96-well plates by serial 10-fold dilutions in 2% FBS-DMEM. After 5 days of incubation, wells exhibiting cytopathic effects (CPE) were identified and viral titers determined by the method of Reed and Muench and expressed as a 50% tissue culture infectious dose (TCID50) (36).
TABLE 1.
Summary of West Nile virus and West Nile-like viruses included in this study
Virus | Proposed lineage | Yr of isolation | Place of isolation | Source of isolation | Known neurovirulence property(ies) |
---|---|---|---|---|---|
NY99-4132 | Ia | 1999 | New York City, NY | Human, mosquito | Highly virulent in humans, mice, birdsb |
MRM16 | Ib | 1960 | Mitchell River Mission, Australia | Mosquito (Culex annulirostris) | Low virulence in humans, miced |
Hu6774 | Ib | 1991 | Southern NSW, Australia | Human, mosquito | Low virulence in humansd |
Sarafend | II | Unknown | Unknown | Unknown | Virulent in micec |
B956a | II | 1937 | Uganda | Human | Low virulence in humans, micee |
Sarawak (MP502-66) | VI | 1966 | Sarawak, Borneo, Malaysia | Mosquito (Culex pseudovishnui) | None |
Koutango-DakAad 5443f | VII | 1968 | Senegal, Africa | Rodent (Tatera kempi) | None |
Viral growth kinetics.
Cells were cultured in 24-well plates, allowing one well for each sample collected. All time points were collected in triplicate. When the cells were 70% confluent they were infected with virus at an MOI of 0.1 and diluted in 2% FBS-DMEM. After 1 h of incubation at 37°C, virus was removed and the cells were washed with phosphate-buffered saline (PBS) before addition of 1 ml 2% FBS-DMEM. Samples were removed every 12 or 24 h postinfection (p.i.) and stored at −80°C. The virus titer in each sample was determined as TCID50s on Vero cells by the method of Reed and Muench (36).
Plaque assays.
Vero cells in 6-well plates were infected with a series of 10-fold dilutions as described above for TCID50 assays. Cells were overlaid with 0.75% low-melting-point agarose in DMEM containing 2% FBS and incubated at 37°C. After 4 days, monolayers were fixed with 10% formaldehyde and stained with 0.2% crystal violet. The diameter of plaques of each virus tested were measured and compared to each other.
Assessment of glycosylation status by enzyme digestion and nucleotide sequence analysis.
Glycosylation of the E protein was examined by digestion with N-glycosidase F (PNGase F; Roche) using the protocol described by Adams and colleagues (37). Proteins in the viral supernatant were denatured with 10% (vol/vol) SDS at 98°C for 3 min and were digested with 1.2 μl of 0.5 M EDTA and 3 μl of 20% n-octyl-β-d-glucopyranoside. The mixture was digested overnight at 37°C with 1 U PNGase F. Proteins were separated and analyzed by Western blotting. Samples were loaded with nonreducing SDS-PAGE NuPage LDS sample buffer (Invitrogen) on a 12% NuPage gel (Invitrogen). Electrophoresed proteins were electroblotted onto nitrocellulose paper (Hybond C; Amersham) and immunostained with monoclonal antibody (MAb) 4G2 (anti-E) at a dilution of 1:5 as previously described (37). The genomic sequence encompassing amino acid residues 154 to 156 was confirmed by sequence analysis using reverse transcription-PCR (RT-PCR) and Sanger sequencing. Briefly, viral RNA was extracted using the RNeasy minikit (Qiagen) by following the manufacturer's instructions, followed by RT-PCR using a SuperScript III OneStep RT-PCR system with Platinum Taq DNA polymerase (Invitrogen) with a degenerate primer set (WNEglyF, 5′-GRAGCATYGACACATG-3′; WNEglyR, 5′-GTYTCYCKGTTCCTCCA-3′). The following RT-PCR cycling conditions were carried out: 1 cycle of 45°C for 20 min; 1 cycle of 94°C for 2 min; 40 cycles of 94°C (30 s), 45°C (1 min), and then 68°C (1 min); and a final extension at 68°C for 5 min. The expected band (400 bp) was resolved, and gel was purified from a 2% agarose gel using a NucleoSpin gel and PCR cleanup kit (Macherey-Nagel). Sequencing was completed at the Australian Genome Research Facility, The University of Queensland.
Mouse virulence.
All animal procedures received prior approval from The University of Queensland Animal Ethics Committee and, where necessary, were performed under ketamine-xylazil anesthesia. Four- to 5-week-old Swiss outbred mice CD-1 (Animal Resources Centre, Murdoch, Western Australia, Australia) were challenged intraperitoneally (i.p.) with a range of doses of WNV strains and West Nile-like viruses (described in Table 1). Mice were kept on clean bedding and given food and water ad libitum. Infected animals were monitored daily for the onset of disease and culled when the first signs of encephalitis (hunching, lethargy, eye closure, or hind-leg flaccid paralysis) were apparent. Surviving mice were bled by cardiac puncture at the end of the experiment (day 21), and the sera were tested for evidence of seroconversion to WNV using a fixed-cell ELISA as previously described (38). To assess differences in the induction of neutralizing antibodies postinfection between viral strains, 6-week-old Swiss outbred mice were injected i.p. with a sublethal dose of 1 infectious unit (IU) and bled on days 7, 14, and 21 days postinfection. Blood was separated and assayed for neutralizing antibodies in a virus neutralization assay.
Titrations of virus in tissue samples.
To measure the amount of infectious virus in peripheral (spleen and sera) and CNS (brain and spinal cord) tissues, samples were extracted, weighed, snap-frozen on dry ice, and stored at −80°C until viral titration assays were performed. At the time of these titrations, 10% (wt/vol) homogenates of each sample were prepared in 2% FBS-DMEM, and serial 10-fold dilutions of each homogenate were assayed by TCID50 on monolayers of Vero cells. The results presented are the means ± standard errors of the means (SEM) of the log10 IU per gram of tissue derived from three animals at each time point. The amount of infectious virus in serum also was determined. In this case, blood samples were collected via cardiac puncture, stored on ice until the blood had separated and serum could be collected, and stored at −80°C. Infectious virus in serum samples was assayed on Vero cells as described above.
Virus neutralization assay.
Heat-inactivated test sera were titrated in doubling dilutions from 1:10 to 1:10,240 in DMEM without fetal bovine serum (FBS) in wells of a 96-well microtiter plate (Costar, Corning). Approximately 100 IU of WNVNY99 or WNVKOU diluted in DMEM with 2% FBS was added to each well containing diluted serum, and plates were incubated at 37°C for an hour. Test sera were tested in a virus neutralization assay against both the homologous virus and heterologous virus. Low-passage-number Vero cells (passage 14 to 15) then were added to each well at a density of 2 × 105 cells/plate, and the plates were incubated for 5 days at 37°C in a humidified CO2 incubator before microscopic examination for CPE. Cells also were added to a 1:10 dilution of each serum in the absence of virus to ensure that the samples were not toxic to the cells. The neutralization titer was expressed as the reciprocal of the highest serum dilution where CPE was recorded (39). The cells were fixed in situ in the plates and subjected to an ELISA protocol using an antiflaviviral antibody, MAb 6B6C1 conjugated to horseradish peroxidase (HRP) (40), to confirm viral replication.
Assessment of inflammatory mediators.
A commercial BD cytometric bead array (CBA) mouse inflammation kit (catalog no. 552364; BD Biosciences Pharmingen, San Diego, CA) was used to determine the levels (pg/ml) of interleukin-6 (IL-6), IL-10, monocyte chemotactic protein-1 (MCP-1), gamma interferon (IFN-γ), tumor necrosis factor (TNF), and IL-12p70 in serum samples and brain homogenates of mice infected with 100 IU and terminated on days 1, 3, 5, and 7 postinfection. Fluorescence was analyzed using a flow cytometer (BD Accuri C6; BD Biosciences, San Jose, CA), and cytokine levels were determined using BD Accuri C6 software.
Histology.
Animals infected with 100 IU and designated for histological analyses sequentially were perfused with chilled PBS and 4% paraformaldehyde in PBS via a transcardial approach. Brain and spinal cord then were postfixed in 4% paraformaldehyde overnight at 4°C, after which they were routinely processed for paraffin embedding. Five-micron sections were stained with hematoxylin and eosin (HE) and Luxol fast blue (FLB) and examined on a Nikon Eclipse 51E microscope equipped with a Nikon DS-Fi1 camera with a DS-U2 unit and NIS elements F software. Microphotographs are reproduced without manipulation other than cropping and adjustment of light intensity.
IHC.
Immunohistochemistry (IHC) detection of flavivirus antigen was carried out as previously described (41), with a modification of the antigen retrieval protocol. Briefly, 5-μm sections of the paraformaldehyde-fixed, paraffin-embedded brain and spinal cord specimens were collected onto charged slides, heat treated, and deparaffinized. The sections were subjected to antigen retrieval by heating in an EDTA-Tris solution, pH 9.0 (Target Retrieval; Dako, Carpentaria, CA, USA), at 96°C for 25 min and allowed to cool to room temperature, followed by three blocking steps at room temperature: (i) 10-min incubation with Dako hydrogen peroxidase block, (ii) 15-min incubation with 0.1 M glycine in PBS, and (iii) 30-min incubation with Dako antibody dilution agent (Dako), with brief Tris-buffered saline-Tween 20 (TBST) rinses between each step. This was followed by 2 h of incubation with neat 4G4-hybridoma culture supernatant (flavivirus anti-nonstructural protein 1) (38) at room temperature. After multiple TBST rinses over a 10- to 15-min period, antibody binding was visualized using a Dako Envision anti-mouse kit. IHC for the apoptosis marker activated caspase 3 was performed as previously described (42). The slides were examined and micrographed as described above.
Data analysis.
Levels of infectious virus in growth kinetics were assessed using analysis of variance (ANOVA). Mean virus titers were compared between viruses using the Tukey method for pairwise multiple comparisons. The significance of clinical differences in vivo between groups was calculated by Kaplan-Meier analysis and analyzed by log-rank test. Differences in viral burden were analyzed by Mann-Whitney test. All statistical analysis was performed in Graphpad Prism 5.0 (GraphPad Software Inc., San Diego, CA).
RESULTS
Glycosylation status of WNV strains and West Nile-like viruses.
N-linked glycosylation of the envelope protein has been established as an important determinant of WNV neuroinvasion in murine models (19). Therefore, the presence of a carbohydrate molecule on the E protein, indicative of a glycosylated virus, was tested for all WNV strains and West Nile-like viruses included in this study by both sequence analysis over this region (residues 154 to 156) (Table 2) and PNGase F digestion of infected cell lysates (results not shown). The viral strains, NY99, Hu6774, and Sarafend, contained an NYS sequence at residues 154 to 156 of the E protein, which exhibited a band shift in Western blotting following PNGase F digestion indicative of a glycosylated E protein. Other viruses contained either an NYF (MRM16), NYP (Sarawak), or NFP (Koutango) sequence, and the E protein in lysates from these viruses did not exhibit a band shift following PNGase F digestion, consistent with an unglycosylated E protein. The B956 strain contained a deletion across residues 154 to 156, which also leads to an unglycosylated E protein (Table 2).
TABLE 2.
Envelope protein glycosylation and plaque phenotype of West Nile virus and West Nile-like viruses
Virus | Sequence | Glycosylation statusb | Avg plaque sizea (mm) |
---|---|---|---|
MRM16 | NYF | − | 1.3 |
Hu6774 | NYS | + | 2.2 |
Sarawak | NYP | − | 1.0 |
NY99 | NYS | + | 3.0 |
Koutango | NFP | − | 1.2 |
B956 | Deletion | − | 0.8 |
Sarafend | NYS | + | 2.2 |
Plaque size represents the diameter at 4 days postinfection.
Glycosylation motif located at amino acid positions 154 to 156 of the envelope protein. Glycosylation was determined by PNGase F digestion and sequence analysis.
Glycosylated viral strains grow more efficiently in vitro.
To determine the ability of WNV and West Nile-like viruses to replicate in vitro, growth kinetics of all viral strains were assessed in Vero cells (Fig. 1A) and a clinically relevant cell line derived from human brain microvascular endothelial cells (HBMECs) (Fig. 1B). In general, the glycosylated strains (WNVNY99, WNVSarafend, and WNVHu6774) grew more efficiently than the unglycosylated viruses (WNVB956, WNVKOU, and WNVMRM16), with the most notable difference in replication observed between 12 and 36 h postinfection. During this time period, glycosylated viruses replicated in Vero cells to approximately 7 logs IU/ml, whereas the unglycosylated virus replication levels were significantly lower, at approximately 4 logs IU/ml (P < 0.05 at 24 h postinfection). The exception was the unglycosylated virus WNVSarawak, which appeared to grow as efficiently as the glycosylated strains in both Vero cells and HBMECs. Glycosylated viruses also exhibited a large-plaque phenotype (2.2 to 3 mm) compared to unglycosylated viruses (0.8 to 1.3 mm) (Table 2). Overall, glycosylated viruses were able to grow more efficiently in cells in vitro.
FIG 1.
Growth kinetics of WNV strains in Vero (A) and human brain microvascular endothelial cells (HBMECs) (B). Cells were infected at an MOI of 0.1, and the titer of the virus in the supernatant was determined as TCID50 on Vero cells every 12 h until 72 h p.i. Growth curves were statistically different (P < 0.05) in HBMECs between NY99 and Koutango, NY99 and B956, Koutango and Sarafend, Koutango and Hu6774, and B956 and Hu6774 viruses.
Virulence properties of WNV strains and West Nile-like viruses in mice.
We have previously demonstrated that a young adult mouse model allows differentiation between virulent and attenuated strains of WNV (4). Four- to five-week-old mice were injected with a variety of doses, from 0.1 to 10,000 IU of each viral strain, to assess their virulence phenotype (Table 3). Interestingly, WNVKOU was consistently the most virulent of all strains tested, with the shortest time to death of 6.8 days and inducing 100% mortality at 100 IU after three independent experiments (Table 3 and Fig. 2). WNVKOU-infected mice appeared to succumb to seizures prior to humane endpoints. This symptom was observed repeatedly from day 4 postinfection and always in mice that ultimately were euthanized due to the onset of CNS symptoms (e.g., hind-limb paralysis). Seizures were observed in routine health checks, and occasionally WNVKOU-infected mice experienced severe seizures leading to death. Despite WNVKOU being significantly more virulent than WNVNY99 (P < 0.05 at 100 IU), producing more morbidity in a shorter time, further experiments testing additional doses determined the viruses had similar LD50 values of 0.5 and 0.7, respectively (Table 3). WNVNY99 and WNVSarafend both were virulent in this mouse model, causing 100% mortality at a dose of 100 IU (Fig. 2A) and 70% and 80% mortality, respectively, at a dose of 1,000 IU (Fig. 2B), in line with what has been reported previously (17, 35). WNVSarafend was comparatively less virulent than WNVNY99, as demonstrated by a longer time to death of 9.2 days at 100 IU (Table 3). WNVKUN strains MRM16 and Hu6774, WNVSarawak, as well as WNVB956 all were attenuated in this model at all doses tested (Table 3 and Fig. 2). However, all viruses that displayed an attenuated phenotype in the adult model were neuroinvasive in the more sensitive weanling mouse model (18 to 19 days old; data not shown), consistent with our previous observations with attenuated WNV strains (4). Since the West Nile-like virus WNVSarawak was not virulent in the young adult model, further characterization of this strain was not pursued. Instead, we focused on WNVKOU to determine a mechanism(s) for its enhanced virulence. Collectively, these data have identified a virulent strain of WNV outside the two major lineages.
TABLE 3.
Virulence of West Nile virus and West Nile-like viruses included in the study in 4- to 5-week-old Swiss mice following i.p. injection
Virus and dose (IUa) | Mortality (no. died/total no. [%]) | ASTb (days) | LD50c |
---|---|---|---|
NY99 | |||
1,000 | 7/10 (70) | 11.8 | 0.49 |
100 | 30/30 (100) | 8.2d | |
10 | 10/10 (100) | 8.9 | |
1 | 7/10 (70) | 12.9 | |
0.1 | 8/20 (40) | 17 | |
MRM16 | |||
10,000 | 3/10 (30) | 17.2 | >10,000 |
1,000 | 2/10 (20) | 19 | |
100 | 2/10 (20) | 18.7 | |
Hu6774 | |||
10,000 | 2/10 (20) | 18.2 | >10,000 |
1,000 | 3/10 (30) | 18.2 | |
100 | 1/10 (10) | 19.5 | |
Sarafend | |||
1,000 | 8/10 (80) | 11.2 | 1 |
100 | 10/10 (100) | 9.2 | |
10 | 9/10 (90) | 10.9 | |
1 | 5/10 (50) | 15.4 | |
B956 | |||
10,000 | 0/10 (0) | 21 | >10,000 |
1,000 | 0/10 (0) | 21 | |
100 | 2/10 (20) | 19.6 | |
Sarawak | |||
10,000 | 2/10 (20) | 19 | >10,000 |
1,000 | 0/10 (0) | 21 | |
100 | 1/10 (10) | 19.6 | |
Koutango | |||
1,000 | 10/10 (100) | 7.6 | 0.72 |
100 | 30/30 (100) | 6.8d | |
10 | 10/10 (100) | 7.6 | |
1 | 8/10 (70) | 11.4 | |
0.1 | 6/20 (30) | 18.4 |
IU, infectious units.
AST, average survival time.
LD50, 50% lethal dose.
P = 0.01 (comparison between NY99 and Koutango strains at 100 IU).
FIG 2.
Survival curves of 4- to 5-week-old Swiss mice following intraperitoneal infection with 100 IU (A) or 1,000 IU (B) per mouse (n = 10 per group). Mice were monitored for 21 days after injection for signs of encephalitis and then euthanized. Survival differences at the dose of 1,000 IU were statistically significant between NY99 and MRM16 as well as NY99 and Hu6774 (P < 0.05); NY99 and Sarawak, NY99 and B956, Sarafend and B956, Sarafend and MRM16, and Sarafend and Hu6774 (P < 0.01); and Koutango and Sarawak, Koutango and B956, Koutango and MRM16, and Koutango and Hu6774 (P < 0.0001) viruses. Survival differences at the dose of 100 IU were statistically significant between NY99 and Sarawak, NY99 and B956, NY99 and MRM16, NY99 and MRM16, NY99 and Hu6774, Koutango and Sarafend, Koutango and Sarawak, Koutango and B956, Koutango and MRM16, Koutango and Hu6774, Sarafend and B956, Sarafend and MRM16, and Sarafend and Hu6774 (P < 0.0001); and NY99 and Koutango (P < 0.05) viruses.
Viral clearance of WNVKOU is delayed in extraneural tissues.
To investigate the contribution of virus replication to overall virulence, 4- to 5-week-old mice were challenged with selected viruses at a dose of 100 IU in order to determine levels of virus replication and clearance in CNS and peripheral tissues (Fig. 3). WNVNY99 caused a detectable viremia from day 1 postinfection that peaked at day 3, with virus cleared from the blood by day 5 postinfection (Fig. 3A). A similar pattern was observed for WNVSarafend; however, levels of virus were lower than those of WNVNY99, consistent with WNVSarafend exhibiting comparatively longer average survival times (Table 3). The attenuated prototype strain WNVB956 was included as a nonneuroinvasive control, and no virus was detected at any time point assessed. Interestingly, WNVKOU caused a delayed but prolonged viremia which peaked at day 5 postinfection, but virus was still present at the last time point sampled, day 7 postinfection (Fig. 3A). A similar pattern also was seen in the spleen, where similar levels of virus were present on both days 3 and 5 postinfection for WNVKOU (Fig. 3C), indicating that there was delayed viral clearance from this organ. Conversely, while virus was present on day 3 in both WNVNY99- and WNVSarafend-challenged mice, levels of virus were significantly reduced by day 5 in the case of WNVNY99 or were no longer detectable in the case of WNVSarafend. These results suggest an inability of WNVKOU-infected mice to clear the virus, in contrast to WNVNY99- or WNVSarafend-infected mice, indicative of different virus-host interactions between these virulent viruses.
FIG 3.
Replication of selected WNV and WNV-like strains (NY99, Sarafend, B956, and Koutango viruses) were compared in the periphery (spleen and sera) and in the central nervous system (brain and spinal cord). Each point represents the means ± SEM of the log10 IU per gram of tissue or per ml for sera from three animals. The exception is day 7 postinfection, where only 2 animals were available for titration for WNVNY99- and WNVKOU-infected mice. An asterisk indicates differences that are statistically significant (P < 0.05).
Viral replication in the central nervous system is similar for both WNVNY99 and WNVKOU.
Virus replication in the CNS was investigated to determine whether differences in pathogenesis also extend to this immune-privileged site (Fig. 3B and D). High levels of virus were detected for WNVNY99 on days 5 and 7 postinfection, with little reduction in viral load between these days. While levels for WNVKOU were slightly lower, significant differences were not observed between the two viruses. For WNVSarafend, virus was not detected until day 7 postinfection. The prototype strain, WNVB956, did not have detectable virus in the CNS, suggesting this virus was unable to invade the CNS. Collectively, these data suggest that differences in virulence between WNVNY99 and WNVKOU involve mechanisms that function not only in the CNS but also in the periphery.
WNVKOU induces poor neutralizing antibody responses.
Neutralizing antibody responses play an essential role in the control of severe WNV infection and can be detected as early as day 4 postinfection for WNV (43, 44). Therefore, we investigated differences in the levels of neutralizing antibodies produced following infection with WNVNY99 and WNVKOU at 7, 14, and 21 days postinfection using a sublethal dose of 1 IU to ensure enough survivors at each time point for testing. There were no detectable neutralizing antibodies for mice infected with WNVKOU at 7 or 14 days postinfection, whereas mice infected with WNVNY99 had detectable neutralizing antibody responses of 67 and 533 at days 7 and 14, respectively (Table 4). Low levels of neutralizing antibodies were detected for WNVKOU-infected mice at 21 days postinfection (average, 40) compared to WNVNY99-infected mice, which had relatively high levels of neutralizing antibodies (average, 2,560). Thus, while WNVKOU infection did elicit an antibody response in mice, reaching a titer of 533 by day 21 postinfection, this response was still considerably lower than that in WNVNY99-infected mice at the same time point (average, 5,120). Also interesting was the observation that antibodies produced in WNVNY99-infected mice were able to neutralize WNVKOU, albeit to a lower titer than for its cognate virus, while no WNVNY99-neutralizing activity was found in the sera of the WNVKOU-infected animals (Table 4). Collectively, the data obtained on antibody responses postinfection suggest that WNVKOU is less able to induce critical neutralizing antibodies that are essential for the control of infection.
TABLE 4.
Average antibody responses following viral infection
Infecting virusa | Day p.i. | Ab response tob: |
|||
---|---|---|---|---|---|
NY99 (avg total Ab) | NY99 (avg neutralizing Ab) | Koutango (avg total Ab) | Koutango (avg neutralizing Ab) | ||
NY99 | 7 | 426 | 67 | 10 | 20 |
NY99 | 14 | 960 | 533 | 40 | 40 |
NY99 | 21 | 5,120 | 2,560 | 170 | 160 |
Koutango | 7 | 20 | <10 | 20 | <10 |
Koutango | 14 | 53 | <10 | 87 | <10 |
Koutango | 21 | 266 | <10 | 533 | 40 |
Mice were infected with 1 IU of virus. Three mice were bled per time point.
Total antibody responses were determined by fixed-cell ELISA, and neutralizing antibody responses were determined by virus neutralization assay.
Cytokine responses, histopathology, and viral antigen distribution.
To further investigate differences in pathology between WNVNY99- and WNVKOU-infected mice, tissues were harvested on days 3 and 7 postinfection with 100 IU of virus to study levels of viral antigen and development of pathology. Mice terminated on day 3 p.i. were devoid of gross and histological changes in the CNS. However, by day 7 p.i., meningoencephalitis was evident in both the WNVNY99- and WNVKOU-infected mice, with more severe changes seen in the latter group (Fig. 4 and 5). Both groups of animals exhibited meningeal and perivascular infiltration of lymphocytes, monocytes, some neutrophils, and mild to moderate gliosis, but these features were more exaggerated in the WNVKOU-infected animals than the WNVNY99-infected animals, with the former also featuring marked neuropil infiltration of leukocytes (compare Fig. 4C to D). Moreover, while the spinal cord was histologically unremarkable in the WNVNY99-infected animals, there was marked loss of motor neurons and axonal degeneration in the spinal cord of the WNVKOU-infected mice (Fig. 5). However, apoptosis did not appear to be a feature of this loss, as no cells positive for activated caspase 3 were detected by IHC (data not shown).
FIG 4.
Meningoencephalitis in mice infected with WNVNY99 (A and C) and WNVKOU (B and D). While meningeal infiltration, perivascular cuffing, and neuropil infiltration of lymphocytes and monocytes are present in mice infected with either virus, the inflammatory infiltrates are more severe in the WNVKOU-infected animals. HE staining was used. Original magnifications are ×40 for panels A and B and ×200 for panels C and D.
FIG 5.
WNVKOU causes notable myelitis (B and D) compared to WNVNY99 (A and C), with loss of motor neurons and mild demyelination leaving clear spaces in the neuropil (B and D). Panels A and B show HE stain at an original magnification of ×100. Panels C and D show Luxol fast blue stain at an original magnification of ×100.
Regional distribution of viral antigen was similar in WNVNY99- and WNVKOU-infected mice, but the immunolabeling was notably more intense and widespread in WNVKOU-infected mice than in WNVNY99-infected mice, indicating greater viral protein accumulation (compare Fig. 6A and C to B and D), particularly in the thalamus and basal ganglia (Fig. 6C and D). Since the antibody used for the IHC, 4G4, binds to cultured cells infected at a similar MOI for these two viruses, WNVNY99 and WNVKOU, and with similar affinity (data not shown), it is unlikely that the difference in intensity seen during IHC analysis is due to differences in the affinity of monoclonal antibody binding to a particular virus.
FIG 6.
Immunolabeling for WNV antigen in WNVNY99 (A and C)- and WNVKOU (B and D)-infected mice. Paraformaldehyde-fixed, paraffin-embedded tissues were immunolabeled with the flavivirus-NS1-specific MAb 4G4, and binding was visualized with the Envision reagents, leaving virus-antigen-positive cells with a reddish-brown coloration. While distribution of virus antigen was similar for the two viruses, the labeling intensity and number of positive cells were more pronounced in the WNVKOU-infected mice (B and D) than in the WNVNY99-infected mice (A and C). Images in panels A and B are hippocampus at an original magnification of ×40. Images in panels C and D are thalamus at an original magnification of ×100.
The marked differences in histopathology and viral clearance in extraneural tissues between WNVNY99- and WNVKOU-infected mice prompted us to examine the cytokine profile in serum and brain of a small number of animals infected with 100 IU of either virus (three animals per virus on days 1, 3, and 5 postinfection and two animals per virus on day 7 postinfection), using a multiplex Luminex assay for IL-6, IL-10, IL-12p70, TNF-α, IFN-γ, and MCP-1 (CCL2). Expression of MCP-1 increased steadily to an average of 5.4 × 103 pg/ml of brain homogenate and 1.48 × 103 pg/ml in serum in the WNVNY99-infected mice on day 7 postinfection. In contrast, the MCP-1 levels in WNVKOU-infected animals remained low or undetectable in serum and peaked at 1.27 × 103 pg/ml in brain homogenate on day 5 postinfection. Similar patterns of expression were noted for IL-10, IL-12, and IFN-γ, although overall the levels were lower, while there was no detectable TNF-α in WNVKOU-infected animals at any time point postinfection and only a single WNVNY99-infected animal had TNF-α in brain homogenate (165.3 pg/ml) on day 7 postinfection. In contrast, the WNVKOU-infected animals had a very marked IL-6 response on day 5 postinfection (average, 2,529 pg/ml), exceeding the response in WNVNY99-infected animals (average, 152 pg/ml). These data suggest that mice infected with WNVKOU are less responsive overall with respect to innate defense mechanisms than WNVNY99-infected mice.
Sequence differences between WNVNY99 and WNVKOU genomes.
To identify whether known virulence determinants also were present in the WNVKOU genome that could contribute to the enhanced virulence phenotype, comparisons between published sequences WNVNY99 (accession number HQ596519) and WNVKOU (accession number EU082200) were performed. Sequence comparison between WNVNY99 and WNVKOU revealed 89.2% identity at the amino acid level, with a total of 371 amino acid substitutions with 173 nonconservative changes (Table 5). The structural genes accounted for 103 amino acid changes (54 nonconservative), while the remaining 268 changes (119 nonconservative) were located in the nonstructural region. Interestingly, known virulence determinants for WNVNY99 (19–21) were not shared with WNVKOU, including amino acids at positions 72 in prM (S in WNVNY99 and M in WNVKOU), 154 to 156 in E (NYS in WNVNY99 and NFP in WNVKOU), 249 in NS3 (P in WNVNY99 and T in WNVKOU), and 653 in NS5 (F in WNVNY99 and S in WNVKOU). Particular areas of the genome with accumulated amino acid changes between WNVNY99 and WNVKOU that may represent important virulence motifs include E domain III (10 nonconservative changes) and the prM protein (13 nonconservative changes).
TABLE 5.
Amino acid differences between viruses NY99-4132 and Koutango Dak ArD 5443
Gene and aa position in polyprotein (corresponding protein position) | NY99 4132 virusa amino acid | Koutango Dak ArD 5443 virusb amino acid | No. of substitutions in each gene (no. of nonconservative changes), % total changes |
---|---|---|---|
C | 13 (6), 46 | ||
11 (11) | S | N | |
25 (25) | L | P | |
63 (63) | R | K | |
71 (71) | G | K | |
100 (100) | S | T | |
108 (108) | K | T | |
111 (111) | I | L | |
113 (113) | V | C | |
114 (114) | M | I | |
115 (115) | I | L | |
117 (117) | L | M | |
118 (118) | I | L | |
120 (120) | S | T | |
prM | 27 (15), 56 | ||
126 (3) | L | F | |
128 (5) | N | S | |
132 (9) | K | R | |
141 (18) | D | E | |
143 (20) | T | A | |
147 (24) | T | S | |
150 (27) | T | I | |
152 (29) | A | S | |
156 (33) | L | Q | |
158 (35) | I | T | |
162 (39) | M | I | |
181 (58) | A | P | |
193 (70) | T | D | |
195 (72) | S | M | |
197 (74) | V | I | |
205 (82) | T | I | |
206 (83) | K | R | |
207 (84) | T | G | |
208 (85) | R | K | |
209 (86) | H | N | |
210 (87) | S | P | |
235 (112) | M | L | |
264 (141) | I | L | |
270 (147) | S | G | |
273 (150) | M | T | |
279 (156) | V | A | |
280 (157) | V | I | |
E | 63 (33), 52 | ||
313 (23) | L | I | |
321 (31) | V | I | |
333 (43) | V | I | |
336 (46) | M | L | |
337 (47) | N | S | |
339 (49) | E | S | |
341 (51) | A | T | |
354 (64) | T | A | |
355 (65) | V | I | |
358 (68) | L | P | |
378 (88) | P | S | |
380 (90) | F | Y | |
383 (93) | R | K | |
410 (120) | A | T | |
414 (124) | K | Q | |
416 (126) | I | V | |
418 (128) | R | K | |
419 (129) | T | I | |
421 (131) | L | Q | |
430 (140) | A | S | |
439 (149) | V | L | |
445 (155) | Y | F | |
446 (156) | S | P | |
449 (159) | V | R | |
450 (160) | G | T | |
456 (166) | R | T | |
457 (167) | F | I | |
459 (169) | I | V | |
460 (170) | T | S | |
462 (172) | A | S | |
466 (176) | Y | S | |
472 (182) | E | D | |
478 (188) | V | I | |
486 (196) | I | V | |
489 (199) | N | S | |
498 (208) | T | S | |
500 (210) | T | S | |
516 (226) | S | A | |
518 (228) | A | P | |
520 (230) | S | A | |
521 (231) | T | S | |
522 (232) | V | T | |
530 (240) | M | L | |
543 (253) | I | V | |
566 (276) | S | N | |
568 (278) | T | N | |
600 (310) | K | Q | |
602 (312) | L | H | |
604 (314) | T | H | |
618 (328) | Q | K | |
651 (361) | F | Y | |
653 (363) | S | A | |
654 (364) | V | S | |
655 (365) | A | S | |
656 (366) | T | A | |
660 (370) | K | I | |
680 (390) | E | D | |
700 (410) | T | A | |
702 (412) | L | V | |
724 (434) | V | I | |
728 (438) | V | L | |
757 (467) | A | V | |
769 (479) | R | K | |
NS1 | 43 (21), 49 | ||
800 (9) | S | T | |
820 (29) | M | V | |
825 (34) | Y | F | |
826 (35) | Y | H | |
837 (46) | I | V | |
842 (51) | K | A | |
845 (54) | V | I | |
848 (57) | L | I | |
861 (70) | A | S | |
862 (71) | V | I | |
884 (93) | Q | H | |
896 (105) | T | P | |
897 (106) | A | S | |
904 (113) | I | M | |
914 (123) | L | I | |
937 (146) | Q | S | |
938 (147) | N | R | |
944 (153) | L | M | |
960 (169) | L | M | |
965 (174) | S | I | |
983 (192) | L | Q | |
985 (194) | I | V | |
999 (205) | R | G | |
1010 (219) | E | D | |
1025 (234) | D | E | |
1026 (235) | G | S | |
1027 (236) | I | V | |
1031 (240) | D | E | |
1036 (245) | V | I | |
1042 (251) | R | K | |
1052 (261) | K | M | |
1066 (275) | I | L | |
1075 (284) | T | K | |
1078 (287) | L | V | |
1080 (289) | E | A | |
1081 (290) | S | L | |
1084 (293) | H | R | |
1106 (315) | S | T | |
1117 (326) | D | E | |
1129 (338) | Q | T | |
1134 (343) | K | R | |
1140 (349) | Q | K | |
1142 (351) | N | E | |
NS2A | 32 (14), 44 | ||
1177 (34) | M | V | |
1181 (38) | L | M | |
1182 (39) | I | V | |
1186 (43) | V | T | |
1197 (54) | V | L | |
1198 (55) | L | V | |
1201 (58) | V | I | |
1211 (68) | S | A | |
1239 (96) | K | R | |
1240 (97) | A | S | |
1255 (112) | V | A | |
1262 (119) | H | Q | |
1264 (121) | A | C | |
1265 (122) | R | H | |
1266 (123) | Q | H | |
1267 (124) | I | L | |
1268 (125) | L | I | |
1269 (126) | L | T | |
1290 (147) | T | S | |
1301 (158) | L | M | |
1310 (167) | R | K | |
1311 (168) | R | C | |
1324 (181) | V | I | |
1326 (183) | I | V | |
1330 (187) | I | L | |
1333 (190) | K | R | |
1335 (192) | S | N | |
1336 (193) | A | S | |
1342 (199) | G | S | |
1355 (212) | L | A | |
1359 (216) | M | L | |
1366 (223) | I | L | |
NS2B | 9 (2), 22 | ||
1400 (26) | I | V | |
1433 (59) | D | N | |
1435 (61) | S | T | |
1438 (64) | S | N | |
1449 (75) | V | I | |
1459 (85) | F | Y | |
1473 (99) | M | I | |
1481 (107) | I | V | |
1494 (120) | V | I | |
NS3 | 48 (19), 40 | ||
1538 (33) | S | N | |
1592 (87) | H | Q | |
1597 (92) | Q | V | |
1614 (109) | V | I | |
1615 (110) | R | Q | |
1620 (115) | V | I | |
1660 (155) | I | V | |
1667 (162) | I | V | |
1677 (172) | D | E | |
1680 (175) | I | A | |
1682 (177) | A | V | |
1708 (203) | R | K | |
1720 (215) | R | K | |
1725 (220) | A | V | |
1754 (249) | P | T | |
1758 (253) | N | S | |
1772 (267) | T | H | |
1778 (273) | P | S | |
1811 (306) | E | S | |
1827 (322) | T | S | |
1829 (324) | D | E | |
1836 (331) | S | A | |
1841 (336) | L | I | |
1852 (347) | S | T | |
1861 (356) | T | V | |
1863 (358) | K | R | |
1873 (368) | M | L | |
1887 (382) | K | R | |
1889 (384) | V | I | |
1940 (435) | I | L | |
1951 (446) | E | D | |
1954 (449) | A | S | |
1955 (450) | V | I | |
1972 (467) | S | A | |
1991 (486) | F | S | |
2011 (506) | I | V | |
2014 (509) | F | L | |
2016 (511) | Q | E | |
2023 (518) | Y | F | |
2024 (519) | T | S | |
2045 (540) | T | A | |
2053 (548) | A | S | |
2061 (556) | V | I | |
2067 (562) | R | K | |
2074 (569) | R | K | |
2075 (570) | T | V | |
2078 (573) | I | V | |
2102 (597) | I | T | |
NS4A | 22 (11), 50 | ||
2127 (3) | I | V | |
2129 (5) | L | V | |
2135 (11) | K | R | |
2142 (18) | G | T | |
2145 (21) | W | M | |
2148 (24) | L | F | |
2178 (54) | I | V | |
2179 (55) | A | V | |
2186 (62) | V | I | |
2188 (64) | T | S | |
2189 (65) | M | L | |
2193 (69) | F | L | |
2209 (85) | A | V | |
2213 (89) | V | T | |
2221 (97) | A | S | |
2222 (98) | E | D | |
2224 (100) | P | S | |
2234 (110) | L | M | |
2259 (135) | V | I | |
2264 (140) | V | I | |
2265 (141) | M | L | |
2266 (142) | T | A | |
NS4B | 28 (13), 46 | ||
2276 (3) | M | L | |
2281 (8) | K | R | |
2284 (11) | S | N | |
2288 (15) | S | N | |
2291 (18) | G | A | |
2293 (20) | R | G | |
2294 (21) | I | V | |
2296 (23) | V | K | |
2300 (27) | F | L | |
2301 (28) | S | N | |
2302 (29) | M | I | |
2314 (41) | A | S | |
2321 (48) | T | S | |
2369 (96) | L | F | |
2373 (100) | A | V | |
2387 (114) | A | T | |
2427 (154) | I | M | |
2449 (176) | I | V | |
2450 (177) | M | I | |
2456 (183) | L | I | |
2459 (186) | V | L | |
2460 (187) | V | I | |
2466 (193) | K | R | |
2476 (203) | T | S | |
2477 (204) | A | S | |
2513 (240) | I | V | |
2518 (245) | I | V | |
2521 (248) | M | L | |
NS5 | 86 (39), 45 | ||
2539 (11) | V | F | |
2543 (15) | R | K | |
2548 (21) | K | R | |
2553 (25) | T | N | |
2561 (33) | I | V | |
2565 (38) | S | L | |
2573 (45) | K | R | |
2602 (74) | V | I | |
2657 (129) | G | K | |
2663 (135) | R | K | |
2667 (139) | C | S | |
2682 (154) | A | P | |
2684 (156) | V | I | |
2690 (162) | I | L | |
2691 (163) | R | K | |
2705 (177) | R | K | |
2709 (181) | V | I | |
2722 (194) | K | R | |
2725 (197) | L | V | |
2757 (229) | V | I | |
2760 (232) | S | A | |
2762 (234) | N | S | |
2775 (247) | R | K | |
2802 (274) | S | N | |
2805 (277) | S | G | |
2809 (281) | N | R | |
2815 (287) | R | K | |
2816 (288) | R | Q | |
2824 (296) | H | L | |
2826 (298) | E | P | |
2840 (312) | D | E | |
2901 (373) | K | R | |
2902 (374) | Y | K | |
2905 (377) | N | D | |
2909 (381) | N | D | |
2913 (385) | A | N | |
2916 (388) | A | K | |
2920 (392) | R | K | |
2925 (397) | S | T | |
2931 (403) | R | S | |
2950 (422) | R | S | |
2966 (438) | D | E | |
2967 (439) | E | Q | |
3048 (520) | I | V | |
3054 (526) | T | G | |
3055 (527) | R | K | |
3074 (546) | R | K | |
3081 (553) | A | G | |
3085 (557) | E | R | |
3086 (558) | L | F | |
3088 (560) | D | E | |
3093 (565) | R | L | |
3116 (588) | D | N | |
3118 (590) | R | K | |
3160 (632) | I | V | |
3163 (635) | D | E | |
3169 (641) | T | G | |
3173 (645) | G | E | |
3174 (646) | P | A | |
3175 (647) | K | R | |
3177 (649) | R | K | |
3178 (650) | T | S | |
3181 (653) | F | S | |
3231 (703) | T | V | |
3234 (706) | Y | H | |
3238 (710) | Q | Y | |
3247 (719) | T | A | |
3268 (740) | V | I | |
3282 (754) | R | K | |
3283 (755) | D | E | |
3300 (772) | F | L | |
3318 (790) | V | M | |
3333 (805) | G | K | |
3344 (816) | E | A | |
3353 (825) | E | D | |
3365 (837) | K | R | |
3388 (860) | A | P | |
3397 (869) | Q | H | |
3398 (870) | V | A | |
3413 (885) | V | I | |
3418 (890) | S | T | |
3420 (892) | K | R | |
3425 (897) | T | A | |
3426 (898) | T | S | |
3427 (899) | L | V | |
3441 (903) | T | S | |
Total | 371 (173), 47 |
DISCUSSION
With the increasing global activity of West Nile virus, including the emergence of lineage II strains, causing widespread disease across Europe (8, 9), and the emergence of the first virulent strain of WNV to cause an outbreak in Australia (4), there is a growing need to better understand the ability of different WNV strains to cause disease. This knowledge will lay the foundation for more effective therapeutics or vaccine candidates. While an extensive body of work exists on the pathogenesis of lineage I and II strains in mice, particularly the North American strains, little is known about the capacity for disease induction for WNV strains proposed to branch outside these major lineages. In this study, we have investigated the capacity of a proposed lineage VI strain (WNVSarawak) and a proposed lineage VII strain (WNVKOU) to cause disease in an established murine model of WNV disease.
WNVKOU was first isolated in 1968 from the blood of a gerbil (Tatera kempi) in the Koutango region of Senegal and later from gerbils in Somalia (45). WNVKOU also has been found in Mastomys species and Lemnyscomys species in Senegal and the Central African Republic (46). Aedes aegypti mosquitoes have been confirmed as a competent vector for WNVKOU (47); therefore, it is expected that WNVKOU can be transmitted by vectors similar to those of other WNV strains. However, since the only isolations of WNVKOU have been from gerbils, which are not commonly associated with WNV strains from other lineages, the transmission cycle of WNVKOU may be ecologically distinct. Despite being included in a number of phylogenetic studies of WNV, little is known about this virus, including its ability to cause infection in humans or animals. While a single symptomatic human infection has been reported in a laboratory worker in Senegal, there have been no natural human cases attributed to WNVKOU (48). However, an unusual outbreak of neurological disease occurred in Sudan in 2002 in areas relatively close to the region where the Koutango virus was first isolated. During this outbreak, the causative agent was deemed to be a West Nile-like virus, but no viral isolates were obtained, precluding further genetic analysis (49). This outbreak was atypical in that it occurred solely in children and also was associated with severe neurological involvement, including seizures, a symptom prominent in adult mice infected with WNVKOU in this study.
WNVSarawak was isolated in 1966 from Culex pseudovishnui mosquitoes collected in Sarawak, Borneo, and originally was classified as a strain of Kunjin virus (50). Further characterization of this virus led to its reclassification into lineage VI (51, 52). WNVSarawak is an example of the sporadic activity of WNV in southeast Asia and has not been associated specifically with disease in humans or animals. Results from this study suggest that this virus is attenuated in established murine models; thus, it is less likely to pose a risk to humans. WNVSarawak contains an unglycosylated envelope protein at residues 154 to 156 that has previously been reported to be an important determinant of virulence for lineage I strains (19, 22, 24, 53). Glycosylation of the envelope protein also has been associated with increased particle secretion and infectivity, promoting more efficient packaging and replication (25). Therefore, based on previous studies using lineage I strains, it was expected that WNVSarawak would have replicated at the same efficiency as other unglycosylated strains. However, WNVSarawak replicated as efficiently as other glycosylated WNV strains. Collectively, the in vitro and in vivo data for WNVSarawak question the contribution of the glycosylated envelope protein alone in terms of virulence for viruses branching outside the major lineages.
The argument is further strengthened with survival and virulence data for the unglycosylated lineage VII virus, WNVKOU. While in cell culture this virus was not able to replicate to the same efficiency as its glycosylated counterparts, in vivo data suggest that WNVKOU is more virulent than the North American NY99 strain, both in terms of time to death and overall mortality. A closer investigation of the mechanism behind this enhanced virulence revealed that replication in CNS tissues was similar for both WNVNY99- and WNVKOU-infected animals, indicating that both viruses are able to efficiently enter the CNS and replicate in neurons. However, a significant difference was observed in the ability of these two viruses to replicate and subsequently be cleared from the periphery of the mouse. Consistent with published works (54), WNVNY99 was able to mount significant viremia and viral load in the spleen, which peaked by day 3 and then was cleared subsequently or significantly reduced by the animal's immune response. However, this pattern was not shared by mice infected with WNVKOU, which had a delayed but more prominent viremia that was not cleared before the mice died. Furthermore, no reduction in the viral load in the spleen was observed for the duration of the experiment. The poor viral clearance in WNVKOU-infected animals most likely is due to the reduced induction of neutralizing antibodies, a critical control measure in severe WNV infection (44). In peripheral tissues, WNV infection is thought to be restricted or controlled by innate and adaptive immune responses, including serum IgM (44), IFN-α/β (55), IFN-γ (56), cytolytic CD8+ T cells (54), and cell-intrinsic IRF-3-dependent antiviral responses (57). Further investigation into these aspects of the innate and adaptive immune response comparing WNVNY99- and WNVKOU-infected mice will shed light on their contribution to differences observed in pathogenicity.
While overall the total amount of infectious virus detected in both WNVNY99- and WNVKOU-infected mice was similar in both the brain and spinal cord, there were notable differences in the levels of viral antigen in brain sections, where more widespread viral antigen was detected in WNVKOU-infected animals on day 7 postinfection. Overall, more neuropathology was observed in WNVKOU-infected animals than in WNVNY99-infected animals; however, the mechanism behind neuron loss appears to be caspase 3 independent. A very marked IL-6 response was detected in brain and periphery of WNVKOU-infected mice, and this may have contributed to increased cellular stress. Notably, Cusick et al. showed that IL-6, produced by microglia and infiltrating monocytes, causes seizures following virus infection (58). Seizures were a prominent feature of the clinical presentation of the WNVKOU-infected mice in the terminal phase. Aberrant cellular stress responses, including cyclic AMP (cAMP) response element-binding transcription factor homologous protein (CHOP), may be responsible for the enhanced pathology of WNVKOU-induced neuronal damage, as this protein has been shown to be activated during WNV infection, leading to apoptosis (59). Alternatively, WNV infection is known to trigger apoptosis by activating noncaspase proteases, such as calpains and cathepsins (60, 61). Levels of these enzymes during infection may provide some insight into the differences in pathology seen between WNVKOU- and WNVNY99-infected animals. In addition, further studies into the postneuroinvasion events in disease progression may provide insight into additional virus and host determinants leading to the enhanced virulence of WNVKOU.
Sequence analysis between WNVNY99 and WNVKOU revealed a high level of nonconservative amino acid changes, with nearly 50% of all amino acid changes within each gene segment representing a nonconservative change. In addition, WNVKOU did not contain any known published virulence motifs, including residue 72 of prM and E protein glycosylation, associated with enhanced viral assembly and/or secretion of virus (19, 21), 249 in NS3, which has been associated with enhanced virulence in birds (62), or 653 of NS5, associated with increased resistance to interferon (20), suggesting that additional virulence determinants within the genome are responsible for the enhanced virulence phenotype of WNVKOU. Of particular interest may be the accumulated nonconservative changes in E domain III, which is known to contain potent antibody neutralizing epitopes (29, 43). Amino acid changes in the WNVKOU genome in E domain III may explain the induction of a poor neutralizing antibody response seen in WNVKOU-infected mice. Previous studies have identified key contact residues (amino acids 306, 307, 330, and 332) in E domain III that form the neutralizing epitope of the potent humanized neutralizing monoclonal antibody E16 (63). While amino acid changes were not identified at these particular residues for WNVKOU, nonconservative changes close to these residues at 310, 312, 314, and 328 were identified which may affect neutralizing epitopes for WNVKOU. Furthermore, nonconservative amino acid changes also were identified in close proximity to the furin cleavage site at residues 86 and 87, which may influence virus maturation. This in turn could influence the neutralizing antibody response to WNVKOU infection, as a recent study has identified that the maturation status of WNV virions affects the efficiency of neutralization by antibody (64).
Additional West Nile-like viruses have been isolated from south Asia, the Czech Republic, and Russia and are proposed to form lineages III to V (13, 14, 65). Since cases of WNV encephalitis and febrile disease (66), as well as multifocal retinitis (67), have been reported for some of these new viruses, further analysis for similarities and differences in disease development compared to lineage I and II strains is warranted. Collectively, data generated during this project have identified a virulent West Nile-like virus that branches outside the major lineages and has the potential to cause disease in humans and animals.
ACKNOWLEDGMENTS
We thank Kim Pham for technical assistance during this project, John MacKenzie and Vincent Deubel for provision of viruses used during this study, and Mario Lobigs for critical review of the manuscript.
This work was supported by the Australian Research Council Discovery Grant DP0987010 (N.A.P. and R.A.H.) and The University of Queensland Early Career Researcher Grant (N.A.P.).
Footnotes
Published ahead of print 18 June 2014
REFERENCES
- 1.Murgue B, Zeller H, Deubel V. 2002. The ecology and epidemiology of West Nile virus in Africa, Europe and Asia. Curr. Top. Microbiol. Immunol. 267:195–221 [DOI] [PubMed] [Google Scholar]
- 2.Lanciotti RS, Roehrig JT, Deubel V, Smith J, Parker M, Steele K, Crise B, Volpe KE, Crabtree MB, Scherret JH, Hall RA, MacKenzie JS, Cropp CB, Panigrahy B, Ostlund E, Schmitt B, Malkinson M, Banet C, Weissman J, Komar N, Savage HM, Stone W, McNamara T, Gubler DJ. 1999. Origin of the West Nile virus responsible for an outbreak of encephalitis in the northeastern United States. Science 286:2333–2337. 10.1126/science.286.5448.2333 [DOI] [PubMed] [Google Scholar]
- 3.Murray KO, Mertens E, Despres P. 2010. West Nile virus and its emergence in the United States of America. Vet. Res. 41:67. 10.1051/vetres/2010039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Frost MJ, Zhang J, Edmonds JH, Prow NA, Gu X, Davis R, Hornitzky C, Arzey KE, Finlaison D, Hick P, Read A, Hobson-Peters J, May FJ, Doggett SL, Haniotis J, Russell RC, Hall RA, Khromykh AA, Kirkland PD. 2012. Characterization of virulent West Nile virus Kunjin strain, Australia, 2011. Emerg. Infect. Dis. 18:792–800. 10.3201/eid1805.111720 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Roche SE, Wicks R, Garner MG, East IJ, Paskin R, Moloney BJ, Carr M, Kirkland P. 2013. Descriptive overview of the 2011 epidemic of arboviral disease in horses in Australia. Aust. Vet. J. 91:5–13. 10.1111/avj.12018 [DOI] [PubMed] [Google Scholar]
- 6.Kutasi O, Bakonyi T, Lecollinet S, Biksi I, Ferenczi E, Bahuon C, Sardi S, Zientara S, Szenci O. 2011. Equine encephalomyelitis outbreak caused by a genetic lineage 2 West Nile virus in Hungary. J. Vet. Intern. Med. 25:586–591. 10.1111/j.1939-1676.2011.0715.x [DOI] [PubMed] [Google Scholar]
- 7.Mann RA, Fegan M, O'Riley K, Motha J, Warner S. 2013. Molecular characterization and phylogenetic analysis of Murray Valley encephalitis virus and West Nile virus (Kunjin subtype) from an arbovirus disease outbreak in horses in Victoria, Australia, in 2011. J. Vet. Diagn. Investig. 25:35–44. 10.1177/1040638712467985 [DOI] [PubMed] [Google Scholar]
- 8.Spissu N, Panichi G, Montisci A, Fiore F. 2013. West Nile virus outbreak in Sardinia, Italy, in 2011. J. Infect. Dev. Ctries. 7:6–9. 10.3855/jidc.3305 [DOI] [PubMed] [Google Scholar]
- 9.Garcia-Bocanegra I, Jaen-Tellez JA, Napp S, Arenas-Montes A, Fernandez-Morente M, Fernandez-Molera V, Arenas A. 2011. West Nile fever outbreak in horses and humans, Spain, 2010. Emerg. Infect. Dis. 17:2397–2399. 10.3201/eid1712.110651 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Danis K, Papa A, Theocharopoulos G, Dougas G, Athanasiou M, Detsis M, Baka A, Lytras T, Mellou K, Bonovas S, Panagiotopoulos T. 2011. Outbreak of West Nile virus infection in Greece, 2010. Emerg. Infect. Dis. 17:1868–1872. 10.3201/eid1710.110525 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Arnold C. 2012. West Nile virus bites back. Lancet Neurol. 11:1023–1024. 10.1016/S1474-4422(12)70278-8 [DOI] [PubMed] [Google Scholar]
- 12.Mackenzie JS, Williams DT. 2009. The zoonotic flaviviruses of southern, south-eastern and eastern Asia, and Australasia: the potential for emergent viruses. Zoonoses Public Health 56:338–356. 10.1111/j.1863-2378.2008.01208.x [DOI] [PubMed] [Google Scholar]
- 13.Bondre VP, Jadi RS, Mishra AC, Yergolkar PN, Arankalle VA. 2007. West Nile virus isolates from India: evidence for a distinct genetic lineage. J. Gen. Virol. 88(Part 3):875–884. 10.1099/vir.0.82403-0 [DOI] [PubMed] [Google Scholar]
- 14.May FJ, Davis CT, Tesh RB, Barrett AD. 2011. Phylogeography of West Nile virus: from the cradle of evolution in Africa to Eurasia, Australia, and the Americas. J. Virol. 85:2964–2974. 10.1128/JVI.01963-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bakonyi T, Hubalek Z, Rudolf I, Nowotny N. 2005. Novel flavivirus or new lineage of West Nile virus, central Europe. Emerg. Infect. Dis. 11:225–231. 10.3201/eid1102.041028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.McMullen AR, Albayrak H, May FJ, Davis CT, Beasley DW, Barrett AD. 2013. Molecular evolution of lineage 2 West Nile virus. J. Gen. Virol. 94(Part 2):318–325. 10.1099/vir.0.046888-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Daffis S, Lazear HM, Liu WJ, Audsley M, Engle M, Khromykh AA, Diamond MS. 2011. The naturally attenuated Kunjin strain of West Nile virus shows enhanced sensitivity to the host type I interferon response. J. Virol. 85:5664–5668. 10.1128/JVI.00232-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Keller BC, Fredericksen BL, Samuel MA, Mock RE, Mason PW, Diamond MS, Gale M., Jr 2006. Resistance to alpha/beta interferon is a determinant of West Nile virus replication fitness and virulence. J. Virol. 80:9424–9434. 10.1128/JVI.00768-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Beasley DW, Whiteman MC, Zhang S, Huang CY, Schneider BS, Smith DR, Gromowski GD, Higgs S, Kinney RM, Barrett AD. 2005. Envelope protein glycosylation status influences mouse neuroinvasion phenotype of genetic lineage 1 West Nile virus strains. J. Virol. 79:8339–8347. 10.1128/JVI.79.13.8339-8347.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Laurent-Rolle M, Boer EF, Lubick KJ, Wolfinbarger JB, Carmody AB, Rockx B, Liu W, Ashour J, Shupert WL, Holbrook MR, Barrett AD, Mason PW, Bloom ME, Garcia-Sastre A, Khromykh AA, Best SM. 2010. The NS5 protein of the virulent West Nile virus NY99 strain is a potent antagonist of type I interferon-mediated JAK-STAT signaling. J. Virol. 84:3503–3515. 10.1128/JVI.01161-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Setoh YX, Prow NA, Hobson-Peters J, Lobigs M, Young PR, Khromykh AA, Hall RA. 2012. Identification of residues in West Nile virus pre-membrane protein that influence viral particle secretion and virulence. J. Gen. Virol. 93(Part 9):1965–1975. 10.1099/vir.0.044453-0 [DOI] [PubMed] [Google Scholar]
- 22.Audsley M, Edmonds JH, Liu W, Mokhonov V, Mokhonova E, Melian EB, Prow N, Hall RA, Khromykh AA. 2011. Virulence determinants between New York 99 and Kunjin strains of West Nile virus. Virology 414:63–73. 10.1016/j.virol.2011.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Schuessler A, Funk A, Lazear HM, Cooper DA, Torres S, Daffis S, Jha BK, Kumagai Y, Takeuchi O, Hertzog P, Silverman R, Akira S, Barton DJ, Diamond MS, Khromykh AA. 2012. West Nile virus noncoding subgenomic RNA contributes to viral evasion of the type I interferon-mediated antiviral response. J. Virol. 86:5708–5718. 10.1128/JVI.00207-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Beasley DW, Davis CT, Whiteman M, Granwehr B, Kinney RM, Barrett AD. 2004. Molecular determinants of virulence of West Nile virus in North America. Arch. Virol. Suppl. 18:35–41 [DOI] [PubMed] [Google Scholar]
- 25.Hanna SL, Pierson TC, Sanchez MD, Ahmed AA, Murtadha MM, Doms RW. 2005. N-linked glycosylation of West Nile virus envelope proteins influences particle assembly and infectivity. J. Virol. 79:13262–13274. 10.1128/JVI.79.21.13262-13274.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Davis LE, DeBiasi R, Goade DE, Haaland KY, Harrington JA, Harnar JB, Pergam SA, King MK, DeMasters BK, Tyler KL. 2006. West Nile virus neuroinvasive disease. Ann. Neurol. 60:286–300. 10.1002/ana.20959 [DOI] [PubMed] [Google Scholar]
- 27.Sejvar JJ, Marfin AA. 2006. Manifestations of West Nile neuroinvasive disease. Rev. Med. Virol. 16:209–224. 10.1002/rmv.501 [DOI] [PubMed] [Google Scholar]
- 28.Colpitts TM, Conway MJ, Montgomery RR, Fikrig E. 2012. West Nile Virus: biology, transmission, and human infection. Clin. Microbiol. Rev. 25:635–648. 10.1128/CMR.00045-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Samuel MA, Diamond MS. 2006. Pathogenesis of West Nile virus infection: a balance between virulence, innate and adaptive immunity, and viral evasion. J. Virol. 80:9349–9360. 10.1128/JVI.01122-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Getts DR, Matsumoto I, Muller M, Getts MT, Radford J, Shrestha B, Campbell IL, King NJ. 2007. Role of IFN-gamma in an experimental murine model of West Nile virus-induced seizures. J. Neurochem. 103:1019–1030. 10.1111/j.1471-4159.2007.04798.x [DOI] [PubMed] [Google Scholar]
- 31.Getts DR, Terry RL, Getts MT, Muller M, Rana S, Shrestha B, Radford J, Van Rooijen N, Campbell IL, King NJ. 2008. Ly6c+ “inflammatory monocytes” are microglial precursors recruited in a pathogenic manner in West Nile virus encephalitis. J. Exp. Med. 205:2319–2337. 10.1084/jem.20080421 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Getts DR, Terry RL, Getts MT, Muller M, Rana S, Deffrasnes C, Ashhurst TM, Radford J, Hofer M, Thomas S, Campbell IL, King NJ. 2012. Targeted blockade in lethal West Nile virus encephalitis indicates a crucial role for very late antigen (VLA)-4-dependent recruitment of nitric oxide-producing macrophages. J. Neuroinflammation 9:246. 10.1186/1742-2094-9-246 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang Y, Lobigs M, Lee E, Koskinen A, Mullbacher A. 2006. CD8(+) T cell-mediated immune responses in West Nile virus (Sarafend strain) encephalitis are independent of gamma interferon. J. Gen. Virol. 87(Part 12):3599–3609. 10.1099/vir.0.81306-0 [DOI] [PubMed] [Google Scholar]
- 34.Stins MF, Badger JL, Kim KS. 2001. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb. Pathog. 30:19–28. 10.1006/mpat.2000.0406 [DOI] [PubMed] [Google Scholar]
- 35.Yamshchikov G, Borisevich V, Seregin A, Chaporgina E, Mishina M, Mishin V, Kwok CW, Yamshchikov V. 2004. An attenuated West Nile prototype virus is highly immunogenic and protects against the deadly NY99 strain: a candidate for live WN vaccine development. Virology 330:304–312. 10.1016/j.virol.2004.09.014 [DOI] [PubMed] [Google Scholar]
- 36.Reed LJ, Muench H. 1938. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 27:493–497 [Google Scholar]
- 37.Adams SC, Broom AK, Sammels LM, Hartnett AC, Howard MJ, Coelen RJ, Mackenzie JS, Hall RA. 1995. Glycosylation and antigenic variation among Kunjin virus isolates. Virology 206:49–56. 10.1016/S0042-6822(95)80018-2 [DOI] [PubMed] [Google Scholar]
- 38.Clark DC, Lobigs M, Lee E, Howard MJ, Clark K, Blitvich BJ, Hall RA. 2007. In situ reactions of monoclonal antibodies with a viable mutant of Murray Valley encephalitis virus reveal an absence of dimeric NS1 protein. J. Gen. Virol. 88(Part 4):1175–1183. 10.1099/vir.0.82609-0 [DOI] [PubMed] [Google Scholar]
- 39.Hall RA, Broom AK, Hartnett AC, Howard MJ, Mackenzie JS. 1995. Immunodominant epitopes on the NS1 protein of MVE and KUN viruses serve as targets for a blocking ELISA to detect virus-specific antibodies in sentinel animal serum. J. Virol. Methods 51:201–210. 10.1016/0166-0934(94)00105-P [DOI] [PubMed] [Google Scholar]
- 40.Prince HE, Yeh C. 2013. Reactivity of human IgM binding murine monoclonal 6B6C1 (IgG2a) with other murine monoclonal IgG antibodies. J. Clin. Lab. Anal. 27:27–30. 10.1002/jcla.21557 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bielefeldt-Ohmann H, Tolnay AE, Reisenhauer CE, Hansen TR, Smirnova N, Van Campen H. 2008. Transplacental infection with non-cytopathic bovine viral diarrhoea virus types 1b and 2: viral spread and molecular neuropathology. J. Comp. Pathol. 138:72–85. 10.1016/j.jcpa.2007.10.006 [DOI] [PubMed] [Google Scholar]
- 42.Tolnay AE, Baskin CR, Tumpey TM, Sabourin PJ, Sabourin CL, Long JP, Pyles JA, Albrecht RA, Garcia-Sastre A, Katze MG, Bielefeldt-Ohmann H. 2010. Extrapulmonary tissue responses in cynomolgus macaques (Macaca fascicularis) infected with highly pathogenic avian influenza A (H5N1) virus. Arch. Virol. 155:905–914. 10.1007/s00705-010-0662-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Diamond MS, Shrestha B, Mehlhop E, Sitati E, Engle M. 2003. Innate and adaptive immune responses determine protection against disseminated infection by West Nile encephalitis virus. Viral Immunol. 16:259–278. 10.1089/088282403322396082 [DOI] [PubMed] [Google Scholar]
- 44.Diamond MS, Sitati EM, Friend LD, Higgs S, Shrestha B, Engle M. 2003. A critical role for induced IgM in the protection against West Nile virus infection. J. Exp. Med. 198:1853–1862. 10.1084/jem.20031223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Butenko AM, Semashko IV, Skvortsova TM, Gromashevskii VL, Kondrashina NG. Detection of the Koutango virus (Flavivirus, Togaviridae) in Somalia. Med. Parazitol. 1986:65–68 [PubMed] [Google Scholar]
- 46.Karabatsos N. 1985. International catalogue of arboviruses, including certain other viruses of vertebrates, 3rd ed. American Society of Tropical Medicine and Hygiene for The Subcommittee on Information Exchange of the American Committee on Arthropod-borne Viruses, San Antonio, TX [Google Scholar]
- 47.Coz J, Valade M, Cornet M, Robin Y. 1976. Transovarian transmission of a Flavivirus, the Koutango virus, in Aedes aegypti L. C. R. Acad. Sci. Hebd. Seances Acad. Sci. D 283:109–110 [PubMed] [Google Scholar]
- 48.Shope RE. 2003. Epidemiology of other arthropod-borne flaviviruses infecting humans. Adv. Virus Res. 61:373–391. 10.1016/S0065-3527(03)61009-2 [DOI] [PubMed] [Google Scholar]
- 49.Depoortere E, Kavle J, Keus K, Zeller H, Murri S, Legros D. 2004. Outbreak of West Nile virus causing severe neurological involvement in children, Nuba Mountains, Sudan, 2002. Trop. Med. Int. Health 9:730–736. 10.1111/j.1365-3156.2004.01253.x [DOI] [PubMed] [Google Scholar]
- 50.Ching CY, Casals J, Bowen ET, Simpson DI, Platt GS, Way HJ, Smith CE. 1970. Arbovirus infections in Sarawak: the isolation of Kunjin virus from mosquitoes of the Culex pseudovishnui group. Ann. Trop. Med. Parasitol. 64:263–268 [DOI] [PubMed] [Google Scholar]
- 51.Scherret JH, Poidinger M, Mackenzie JS, Broom AK, Deubel V, Lipkin WI, Briese T, Gould EA, Hall RA. 2001. The relationships between West Nile and Kunjin viruses. Emerg. Infect. Dis. 7:697–705. 10.3201/eid0704.010418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Scherret JH, Mackenzie JS, Hall RA, Deubel V, Gould EA. 2002. Phylogeny and molecular epidemiology of West Nile and Kunjin viruses. Curr. Top. Microbiol. Immunol. 267:373–390 [DOI] [PubMed] [Google Scholar]
- 53.Shirato K, Miyoshi H, Goto A, Ako Y, Ueki T, Kariwa H, Takashima I. 2004. Viral envelope protein glycosylation is a molecular determinant of the neuroinvasiveness of the New York strain of West Nile virus. J. Gen. Virol. 85(Part 12):3637–3645. 10.1099/vir.0.80247-0 [DOI] [PubMed] [Google Scholar]
- 54.Shrestha B, Diamond MS. 2004. Role of CD8+ T cells in control of West Nile virus infection. J. Virol. 78:8312–8321. 10.1128/JVI.78.15.8312-8321.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.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. 10.1128/JVI.79.21.13350-13361.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wang T, Scully E, Yin Z, Kim JH, Wang S, Yan J, Mamula M, Anderson JF, Craft J, Fikrig E. 2003. IFN-gamma-producing gamma delta T cells help control murine West Nile virus infection. J. Immunol. 171:2524–2531. 10.4049/jimmunol.171.5.2524 [DOI] [PubMed] [Google Scholar]
- 57.Bourne N, Scholle F, Silva MC, Rossi SL, Dewsbury N, Judy B, De Aguiar JB, Leon MA, Estes DM, Fayzulin R, Mason PW. 2007. Early production of type I interferon during West Nile virus infection: role for lymphoid tissues in IRF3-independent interferon production. J. Virol. 81:9100–9108. 10.1128/JVI.00316-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Cusick MF, Libbey JE, Patel DC, Doty DJ, Fujinami RS. 2013. Infiltrating macrophages are key to the development of seizures following virus infection. J. Virol. 87:1849–1860. 10.1128/JVI.02747-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Medigeshi GR, Lancaster AM, Hirsch AJ, Briese T, Lipkin WI, Defilippis V, Fruh K, Mason PW, Nikolich-Zugich J, Nelson JA. 2007. West Nile virus infection activates the unfolded protein response, leading to CHOP induction and apoptosis. J. Virol. 81:10849–10860. 10.1128/JVI.01151-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hail N, Jr, Carter BZ, Konopleva M, Andreeff M. 2006. Apoptosis effector mechanisms: a requiem performed in different keys. Apoptosis 11:889–904. 10.1007/s10495-006-6712-8 [DOI] [PubMed] [Google Scholar]
- 61.Kroemer G, Martin SJ. 2005. Caspase-independent cell death. Nat. Med. 11:725–730. 10.1038/nm1263 [DOI] [PubMed] [Google Scholar]
- 62.Brault AC, Huang CY, Langevin SA, Kinney RM, Bowen RA, Ramey WN, Panella NA, Holmes EC, Powers AM, Miller BR. 2007. A single positively selected West Nile viral mutation confers increased virogenesis in American crows. Nat. Genet. 39:1162–1166. 10.1038/ng2097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Oliphant T, Engle M, Nybakken GE, Doane C, Johnson S, Huang L, Gorlatov S, Mehlhop E, Marri A, Chung KM, Ebel GD, Kramer LD, Fremont DH, Diamond MS. 2005. Development of a humanized monoclonal antibody with therapeutic potential against West Nile virus. Nat. Med. 11:522–530. 10.1038/nm1240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lee PD, Mukherjee S, Edeling MA, Dowd KA, Austin SK, Manhart CJ, Diamond MS, Fremont DH, Pierson TC. 2013. The Fc region of an antibody impacts the neutralization of West Nile viruses in different maturation states. J. Virol. 87:13729–13740. 10.1128/JVI.02340-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Balakrishnan A, Butte DK, Jadhav SM. 2013. Complete genome sequence of West Nile virus isolated from Alappuzha district, Kerala, India. Genome Announc. 1:e00230–13. 10.1128/genomeA.00230-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Banerjee K. 1996. Emerging viral infections with special reference to India. Indian J. Med. Res. 103:177–200 [PubMed] [Google Scholar]
- 67.Shukla J, Saxena D, Rathinam S, Lalitha P, Joseph CR, Sharma S, Soni M, Rao PV, Parida M. 2012. Molecular detection and characterization of West Nile virus associated with multifocal retinitis in patients from southern India. Int. J. Infect. Dis. 16:e53–e59. 10.1016/j.ijid.2011.09.020 [DOI] [PubMed] [Google Scholar]