Skip to main content
Erschienen in: Virology Journal 1/2016

Open Access 01.12.2016 | Research

Newcastle disease virus-vectored West Nile fever vaccine is immunogenic in mammals and poultry

verfasst von: Jinliang Wang, Jie Yang, Jinying Ge, Ronghong Hua, Renqiang Liu, Xiaofeng Li, Xijun Wang, Yu Shao, Encheng Sun, Donglai Wu, Chengfeng Qin, Zhiyuan Wen, Zhigao Bu

Erschienen in: Virology Journal | Ausgabe 1/2016

Abstract

Background

West Nile virus (WNV) is an emerging zoonotic pathogen which is harmful to human and animal health. Effective vaccination in susceptible hosts should protect against WNV infection and significantly reduce viral transmission between animals and from animals to humans. A versatile vaccine suitable for different species that can be delivered via flexible routes remains an essential unmet medical need. In this study, we developed a recombinant avirulent Newcastle disease virus (NDV) LaSota strain expressing WNV premembrane/envelope (PrM/E) proteins (designated rLa-WNV-PrM/E) and evaluated its immunogenicity in mice, horses, chickens, ducks and geese.

Results

Mouse immunization experiments disclosed that rLa-WNV-PrM/E induces significant levels of WNV-neutralizing antibodies and E protein-specific CD4+ and CD8+ T-cell responses. Moreover, recombinant rLa-WNV-PrM/E elicited significant levels of WNV-specific IgG in horses upon delivery via intramuscular immunization, and in chickens, ducks and geese via intramuscular, oral or intranasal immunization.

Conclusions

Our results collectively support the utility of rLa-WNV-PrM/E as a promising WNV veterinary vaccine candidate for mammals and poultry.

Background

West Nile virus (WNV) is the causative agent of West Nile fever (WNF), a major emerging zoonotic disease shown to have a significant negative impact on both human and animal health since the first recorded case in Uganda in 1937. WNV is a member of the genus Flavivirus belonging to the family Flaviviridae. The virus is one of the most widespread arthropod-transmitted pathogens, and is extensively distributed worldwide throughout Africa, Europe, Asia and North America. WNV has a broad host spectrum comprising several species of birds (including poultry), mammals, amphibians and reptiles. Culex mosquitoes play an important role as the primary global WNV transmission vector, and are responsible for the incidental infection of humans and horses, which are considered dead-end hosts of WNV [14].
Vaccination in sensitive host animals, especially those abundant in number and closely associated with humans, such as horses, poultry and other bird species, should protect against WNV infection and significantly reduce transmission between animals and from animals to humans. Currently, several injection-delivered vaccines [58] are licensed for horses, but not other sensitive host animals. A versatile vaccine suitable for different species that can be delivered via flexible administration routes therefore remains an unmet medical requirement.
Newcastle disease virus (NDV) has been actively developed and evaluated as a vaccine vector for the control of human and animal diseases [916]. NDV vector vaccines can be effectively delivered via intramuscular or intratracheal inoculation in mammals and intramuscular, intranasal or oral (through water or feed) inoculation in poultry [11, 12, 1721]. In the current study, we generated a recombinant nonvirulent NDV LaSota virus strain expressing WNV pre-membrane (PrM) and envelope protein (E), two surface glycoproteins that form a heterodimer on the viral surface [22] and are responsible for eliciting the majority of protective immune responses [23]. Immunogenicity of the recombinant NDV in mammals and poultry delivered via different immunization routes was further evaluated.

Methods

Construction of recombinant NDV LaSota virus

The chemically synthesized mammalian codon-optimized WNV PrM/E gene (strain NY99, GenBank No. DQ211652.1) was cloned and inserted into the Pme I site between the P and M genes of full-length genomic cDNA of NDV LaSota [11]. The resultant plasmid was co-transfected with eukaryotic plasmids expressing NDV nucleoprotein (NP), phosphate protein (P) and large polymerase protein (L), following an established protocol [11]. The rescued recombinant virus was designated rLa-WNV-PrM/E. Expression of WNV PrM and E proteins was confirmed via indirect immunofluorescence and western blot assays. Mouse anti-WNV E monoclonal antibody (developed in our laboratory), mouse anti-PrM monoclonal antibody [24] and chicken anti-NDV serum [11] was used as primary antibodies. Fluorescein isothiocyanate (FITC)-conjugated goat anti-mouse antibody (Sigma, St. Louis, MO) and Tetramethylrhodamine (TRITC)-conjugated rabbit anti-chicken antibody (Sigma, St. Louis, MO) was used as secondary antibodies for immunofluorescence assay. Chicken anti-NDV serum and mouse anti-WNV serum (developed in our laboratory) were used as primary antibodies, horseradish-peroxidase (HRP)-conjugated goat anti-chicken IgG and goat anti-mouse IgG (SouthernBiotech, Birmingham, AL) were used as secondary antibodies for western blot assay.
To determine the pathogenicity of rLa-WNV-PrM/E in poultry, mean death time, intracerebral pathogenicity index, and intravenous pathogenicity index were determined in embryonated specific pathogen-free (SPF) chickens or eggs according to the OIE Manual [25]. To assess pathogenicity in mouse, ten 6-week-old female C57BL/6 mice (Vital River, Beijing, China) were inoculated intramuscularly with 0.1 ml diluted allantoic fluid containing 1 × 108 EID50 (50 % Embryo Infectious Dose) rLa-WNV-PrM/E and intranasally with 0.03 ml diluted allantoic fluid containing 3 × 107 EID50 rLa-WNV-PrM/E. Mice were examined daily for 3 weeks for signs of illness, weight loss or death.

Animal immunization studies

For mouse immunization, ten 6-week-old female C57BL/6 mice (Vital River, Beijing, China) were intramuscularly vaccinated with 0.1 ml diluted allantoic fluid containing 1 × 108 EID50 rLa-WNV-PrM/E twice with a 3-week interval. Splenocytes for assay of E protein-specific CD4+ and CD8+ T-cell responses were harvested 10 days after the first or second dose. Serum samples for the serological assay were prepared 2 weeks after each dose.
For horse immunization, five adult horses were intramuscularly inoculated with 2 ml diluted allantoic fluid containing 2 × 109 EID50 rLa-WNV-PrM/E, and five administered with 2 ml phosphate-buffered saline (PBS) as the control group. Three weeks after the first dose, a booster with the same vaccine was delivered using the same dosage and route. Serum samples were collected for serological assay 2 weeks after each immunization.
For poultry immunization, three groups (ten per group) of 4-week-old SPF chickens were assessed: intramuscular inoculation with 0.1 ml diluted allantoic fluid containing 1 × 108 EID50 rLa-WNV-PrM/E (Group One), oral inoculation with 10 ml diluted allantoic fluid containing 1 × 1010 EID50 rLa-WNV-PrM/E mixed with 500 g chicken feed and 300 ml water (Group Two), whereby feeding was stopped 5 h before inoculation, and intramuscular and oral inoculation with PBS (Group Three). Three groups (ten per group) of 4-week-old SPF ducks were immunized following the above procedure. For immunization of geese, four groups (15 per group) of 4-week-old birds were examined: intramuscular inoculation with 0.5 ml diluted allantoic fluid containing 5 × 108 EID50 rLa-WNV-PrM/E (Group One), intranasal inoculation with 0.5 ml diluted allantoic fluid containing 5 × 108 EID50 rLa-WNV-PrM/E via eye drops and nostril instillation (Group Two), oral inoculation with 0.5 ml diluted allantoic fluid containing 5 × 108 EID50 rLa-WNV-PrM/E via buccal cavity instillation (Group Three), and intramuscular inoculation with 0.5 ml PBS (Group Four). Three weeks after the first dose, chickens, ducks and geese were boosted with the vaccine using the same doses and routes. Serum samples were collected for serological assay 2 weeks after each immunization. All poultry were housed in the Experimental Animal Center of Harbin Veterinary Research Institute.

Analysis of WNV-specific IgG, neutralizing and NDV HI antibodies

Enzyme-linked immunosorbent assay (ELISA) for determining antigen-specific IgG in mouse serum was performed as described previously [26]. Briefly, purified mammalian cells producing WNV virus-like particles (4 μg/ml, containing PrM and E proteins, unpublished) were used as coating antigen. Antibodies were detected using HRP-labeled goat anti-mouse IgG (SouthernBiotech, Birmingham, AL) secondary antibody. A standard curve was generated by coating with serially diluted mouse IgG (Southern Biotech, Birmingham, AL) at known concentrations. A linear equation was obtained based on the standard IgG concentration and their O.D values, thus the concentration of WNV-specific IgG was calculated according to the linear equation based on their O.D values and expressed as the amount of IgG per ml of serum (ng/ml). The above coating antigen was also used for ELISA detection of WNV-specific IgG in horse and poultry sera. HRP-labeled goat anti-horse IgG (SouthernBiotech, Birmingham, AL) and goat anti-chicken IgG (SouthernBiotech, Birmingham, AL) were used as secondary antibodies for horse and chicken serum detection, and mouse anti-duck IgG (AbD Serotec, Oxford, UK) and HRP-labeled goat anti-mouse IgG (SouthernBiotech, Birmingham, AL) for duck and goose serum detection. Due to the lack of purified IgG for these animals, results were expressed as O.D. values relative to negative controls.
Mouse serum neutralizing antibody levels were determined using the WNV plaque reduction neutralization test (PRNT) in the Biosafety Level 3 facility of Beijing Institute of Microbiology and Epidemiology. Briefly, 320 μl of 10-fold serially diluted mouse serum (heat-inactivated at 56 °C for 30 min before use) was mixed with 320 μl medium containing 150 plaque-forming units (PFU) of WNV (strain NY99) and incubated at 37 °C for 1 h. Next, the mixture was added to BHK-21 cells in the wells of a six-well plate and incubated at 37 °C for 1 h. Following removal of the mixture, cells washed three times with PBS. Cells were overlaid with 2 ml DMEM-agarose, and incubation continued at 37 °C. After 72 h, cells were fixed with 4 % paraformaldehyde and subsequently stained with 1.5 % crystal violet to visualize plaques. Neutralization titers were expressed as the reciprocal of the highest dilution of serum showing at least 50 % reduction in number of plaques, compared with the negative control. Neutralizing antibodies of chicken, duck, goose and horse sera were not assessed due to unavailability of the BSL-3 facility during the experimental period. NDV hemagglutinin inhibition (HI) antibodies of immunized animals were determined following a previously described protocol [11].

Flow cytometric analysis of the mouse CD4+ and CD8+ T-cell response

The WNV E protein-specific CD4+ and CD8+ T-cell response in C57BL/6 mice was determined via flow cytometry using established protocols [27]. rLa-WNV-PrM/E-immunized mice were sacrificed on day 10 after the first and second immunizations. Mouse splenocytes were prepared as documented by Ye et al. [28]. Briefly, spleens were removed from euthanized mice, cut into small sections, and homogenized by gentle rubbing. After low-speed centrifugation, the supernatant was removed, cells gently re-suspended in red blood cell lysis buffer (Sigma), and incubated on ice for 1 min. Splenocytes (1 × 106) were stimulated with 20 μg/ml WNV E-specific CD4 peptide (PVGRLVTVNPFVSVA, H-2 b , [29]) or CD8 peptide (LGMSNRDFL, H-2D b , [30] for 6 h in presence of 10 ng/ml Brefeldin A (eBioscience, San Diego, CA) to assess the CD4+ and CD8+ T-cell response, respectively. Cells were washed twice with PBS containing 3 % fetal calf serum and subsequently stained with Peridinin-Chlorophyll-Protein-Complex (PerCP)-conjugated rat anti-mouse CD4 (or CD8) and phycoerythrin (PE)-conjugated rat anti-mouse CD3 antibody (BD Pharmingen, San Diego, CA). Next, cells were fixed and permeabilized with Fix&Perm Buffer (eBioscience, San Diego, CA) and stained for intracellular interferon-gamma (IFN-γ) with an allophycocyanin (APC)-conjugated rat anti-mouse IFN-γ antibody (BD Pharmingen). The levels of CD4+ or CD8+ T-cell responses were determined using flow cytometry on a BD FACSAria Station (BD Immunocytometry Systems, San Jose, CA). Data were analyzed with FlowJo software (Treestar Inc, Ashland, OR).

Statistical analysis

Data on virus titers, antibody titers and mouse T cell responses were analyzed using two-tailed Student’s t test with the Excel program (Microsoft, Redmond, WA). To describe the p value significance, the following convention was used: not significant, p > 0.05; significant, p ≤ 0.05; highly significant, p ≤ 0.01.

Results

Generation of rLa-WNV-PrM/E virus and in vitro characterization

Recombinant NDV expressing WNV PrM/E proteins was generated by inserting the PrM/E gene between the P and M genes in NDV genome cDNA (Fig. 1a). The presence of PrM/E was confirmed via RT-PCR. PrM/E protein expression was confirmed via western blot and indirect immunofluorescence staining of rLa-WNV-PrM/E-infected BHK-21 cells. Western blot detected the presence of both E (~45 kDa) and PrM (~25 kDa) proteins (Fig. 1b), which were further confirmed via indirect immunofluorescence with specific monoclonal antibodies against each protein. As expected, rLa-infected BHK-21 cells were not stained (Fig. 1c).
The growth titer of rLa-WNV-PrM/E in embryonated chicken eggs was comparable to that of parental rLaSota. Genetic stability of rLa-WNV-PrM/E was assessed by serial passage of the virus in SPF chicken eggs, and confirmed with RT-PCR and immunofluorescence (data not shown). Mean death time (>120 h), intracerebral pathogenicity index (=0), and intravenous pathogenicity index (=0) results demonstrated the lentogenic nature of rLa-WNV-PrM/E in poultry (data not shown). The genetic stability of WNV PrM/E gene within rLa-WNV-PrM/E was assessed by serially passage (at least 10 passages) of the virus in embryonated SPF chicken eggs, the presence and expression of PrM/E was confirmed by RT-PCR and indirect immunofluorescence assay. The results demonstrated the PrM/E gene can be stably maintained and expressed. Ten mice receiving intramuscular inoculation at a dose of 1 × 108 EID50 and intranasal inoculation of 3 × 107 EID50 rLa-WNV-PrM/E survived with no abnormalities during the 2-week observation period. No significant differences in weight gain were observed after inoculation. Our results indicate rLa-WNV-PrM/E is safe for mice (data not shown).

The recombinant virus induces significant WNV-specific humoral and T-cell responses in mice

WNV-specific IgG (Fig. 2a) was detected using ELISA. Notably, the IgG antibody level was significantly boosted after the second dose (p < 0.01). Serum neutralizing antibodies were analyzed with a WNV plaque reduction assay. As shown in Fig. 2b, WNV-neutralizing antibodies were detected after the first dose, and significantly boosted after the second dose (p < 0.01). NDV neutralizing antibodies were also detected after the first dose, and significantly boosted after the second dose (p < 0.01) (Fig. 2c).
The WNV E protein-specific CD4 epitope (PVGRLVTVNPFVSVA, H-2 b , [29] and CD8 epitope (LGMSNRDFL, H-2D b , [30] were used to assess rLa-WNV-PrM/E-induced specific T-cell responses in mice. As shown in Fig. 3, rLa-WNV-PrM/E immunization induced significant epitope-specific IFN-γ-producing CD4+ and CD8+ T-cell responses after the first dose, which were significantly boosted after the second dose (p < 0.05).

rLa-WNV-PrM/E administered via different immunization routes induces significant WNV IgG antibody production in horses and poultry

Given that rLa-WNV-PrM/E induces good humoral responses in mice, we performed horse immunization with the vaccine. Horses received two doses of the vaccine via the intramuscular route with a 3-week interval. WNV-specific IgG was detected after the first dose, and significantly boosted after the second dose (p < 0.01) (Fig. 4a). HI antibodies against NDV were also detected in horse after the first dose, and significantly boosted after the second dose (p < 0.01) (Fig. 4b). To determine whether rLa-WNV-PrM/E induces an immune response in poultry, SPF chickens were intramuscularly or orally inoculated with the recombinant virus twice with a 3-week interval. In intramuscularly immunized chickens, the WNV-specific IgG was detected after the first dose, and significantly boosted after the second dose (p < 0.05) (Fig. 5a I). In orally immunized chickens, IgG was also detected after the first dose, but only slightly boosted after the second dose (Fig. 5a II). NDV HI antibodies was detected after the first dose, and significantly boosted after the second dose (p < 0.05) in intramuscularly immunized chickens (Fig. 5b I). In orally immunized chickens, NDV HI antibody was also detected after the first dose, but only slightly boosted after the second dose with no statistical significance (Fig. 5b II). The same immunization procedure was performed for ducks. WNV IgG in intramuscularly (Fig. 6a I) and orally (Fig. 6a II) immunized duck sera was induced after the first dose, and boosted significantly after the second dose (p < 0.05). NDV HI antibodies were detected after the first dose, and significantly boosted after the second dose in intramuscularly (Fig. 6b I) and orally immunized ducks (p < 0.01) (Fig. 6b II). Groups of outbred geese were either intramuscularly, intranasally or orally inoculated with rLa-WNV-PrM/E. Intramuscularly immunized geese produced detectable WNV IgG after the first dose, which was significantly boosted after the second dose (p < 0.05) (Fig. 7a I). Intranasally (Fig. 7a II) and orally (Fig. 7a III) immunized geese also produced a detectable level of IgG after the first dose, which was only slightly boosted after the second dose (p > 0.05). NDV HI antibodies were detected after the first dose, and significantly boosted after the second dose in intramuscularly (Fig. 7b I), intranasally (Fig. 7b II) and orally (Fig. 7b III) immunized geese (p < 0.01).

Discussion

WNV is an important zoonotic pathogen widely distributed geographically, with emergence of increasingly neuroevasive strains. Here, a recombinant NDV LaSota virus expressing WNV PrM and E proteins, rLa-WNV-PrM/E, was constructed as a candidate veterinary vaccine for WNV prevention and control. rLa-WNV-PrM/E elicited significant levels of neutralizing antibodies and WNV-specific T-cell responses in mice, as well as WNV-specific IgG in horses, chickens, ducks, and geese, support the immunogenicity of the newly generated recombinant virus in mammals and poultry.
Mice are sensitive to WNV infection, and thus commonly used as the model animal for WNV vaccine evaluation and other related studies. In our experiments, mice intramuscularly inoculated with rLa-WNV-PrM/E produced significant WNV-neutralizing antibodies and specific IgG. Neutralizing antibodies play a crucial role in WNV control and clearance [31]. We used the 50 % plaque reduction assay for determining the levels of neutralizing antibody against WNV NY99 in mice sera. This method is recommended by WHO for testing the potency of Japanese encephalitis vaccine. The cut-off value for testing serum seroprotection is 1 log10 (a ten-fold dilution of serum that reduces plaque formation by 50 % is sufficient for protection against viral challenge) [32]. In an earlier study, mice immunized actively or passively that possessed WNV-neutralizing antibodies higher than 1 log10 were protected against the lethal WNV challenge [33]. In our experiments, the neutralizing antibody titer of rLa-WNV-PrM/E-immunized mice reached up to 1.3 log10 after the first dose. After administration of the second dose, the neutralizing antibody titer was significantly boosted (~2.2 log10), implying that rLa-WNV-PrM/E confers robust protection against lethal WNV infection in mice. In sera of mice, high levels of anti-WNV E IgG were elicited after the first dose, which were significantly boosted after the second dose. IgG findings were in accordance with the neutralizing antibody pattern, indicating a linear correlation in rLa-WNV-PrM/E-immunized animals. T-cell responses are additionally critical in controlling WNV infection. CD4+ T-cells play a dominant protective role in viral clearance via facilitating antibody responses and sustaining WNV-specific CD8+ T-cell responses in the central nervous system (CNS) [34]. The efficacy of CD8+ T cells in controlling WNV infection has also been characterized. An earlier study showed that while neutralizing antibodies play a central role in terminating WNV viremia, CD8+ T-cells are essential for preventing sustained WNV infection in peripheral and CNS compartments [35]. Live vector vaccines have a significant advantage in that they effectively elicit cellular responses [3638]. In our experiments, rLa-WNV-PrM/E induced high levels of WNV-specific CD4+ and CD8+ T-cell responses. Although a challenge study was not conducted at this time due to the unavailability of the BSL-3 laboratory, given the neutralizing antibody and T-cell response results, we presume that the rLa-WNV-PrM/E could confer protection in mice. Intramuscularly immunized horses produced WNV specific IgG following the first dose, which was significantly boosted after the second dose. Considering the close association between IgG and neutralizing antibody levels, we propose that horses acquire protective immunity after vaccination. Further neutralization assays and challenge studies are required to confirm the efficacy of the vaccine in horses.
Vaccination of sensitive hosts not only protects the animal itself but also prevents transmission of WNV from animals to humans. Several veterinary WNV vaccines are currently available, including inactivated whole virus vaccine [5, 39, 40], DNA vaccines [4143], recombinant canarypox-vectored vaccine [6, 44] and recombinant Yellow Fever 17D vaccine [45, 46]. Notably, canarypox-vectored WNV vaccine has been shown to effectively elicit WNV-specific neutralizing antibodies and confer protection in horses, geese, cats and dogs against lethal WNV challenge [6, 44, 47, 48]. These vaccines require delivery via intramuscular inoculation. The current study demonstrated that rLa-WNV-PrM/E is immunogenic in not only mice, horses and poultry when administered intramuscularly, but also in poultry upon delivery via intranasal or oral inoculation. Based on the collective findings, we propose that rLa-WNV-PrM/E is a promising veterinary candidate vaccine for multiple mammalian and avian species that can be delivered via flexible inoculation routes.
Domestic poultry, such as chickens, ducks and geese, are susceptible to WNV and develop clinical signs and viremia (usually sufficient to infect mosquitoes), thus contributing to WNV transmission. Chickens are widely used as sentinel animals in the early warning of WNV prevalence [2, 4952]. Several studies have additionally provided evidence of the susceptibility of domestic or captive ducks to WNV [3, 53, 54]. Ducks develop high-titer viremia in blood and are capable of shedding virus orally [55]. Geese are also susceptible to WNV, especially young geese, and develop a variety of neurological signs, often resulting in a significant number of deaths [4, 56, 57]. Geese represent another experimental animal model for WNV vaccine evaluation [5860]. Notably, WNV human infection and isolation of the virus has recently been reported in mosquitoes in the Xinjiang Uygur Autonomous Region in Northwest China [61, 62]. China has an estimated 12 billion or more poultry, including four billion ducks and geese. Many domestic birds, especially ducks and geese, are raised in backyards and open ranges under poor biosecurity conditions and live in high-density groups close to large human populations. Since these birds may serve as important amplifying hosts of WNV, control of WNV circulation in birds is important for public health. For economic reasons, poultry farmers are generally unwilling to pay additional vaccine and labor costs for vaccination solely against WNV. As NDV is one of the most lethal and economically important pathogens for poultry (at least chickens and geese), farmers use live vaccines, such as LaSota strain, to protect against infection. In our study, most domestic poultry showed NDV HI antibody titters higher than 4 log2 after vaccination, irrespective of the delivery route (Figs. 5b, 6b and 7b). A HI antibody titer higher than 3 log2 is usually sufficient to protect poultry from lethal challenge of NDV. In this scenario, farmers will not need to pay additional vaccine and labor costs to protect against WNV infection by using rLa-WNV-PrM/E instead of NDV live vaccine for routine vaccination. Moreover, oral and intranasal immunization routes are more convenient than intramuscular immunization for poultry as well as water fowl and migratory and resident wild birds.

Conclusions

In summary, rLa-WNV-PrM/E vaccination in susceptible animals is important for protection against WNV infection. Our findings demonstrate that rLa-WNV-PrM/E delivered via multiple immunization routes is immunogenic in both mammals and poultry.

Acknowledgements

This study was supported by The National Key Technology R&D Program (2013BAD12B05) and National High-Tech Research and Development Program of China (2011AA10A212).

Authors’ contributions

ZB and ZW designed and oversaw the experiments and wrote the manuscript. JY and JW rescued and characterized the recombinant virus. JW and JY determined the mice IgG and HI antibodies, ZW carried out T cell response assays. JW, JG, XW, RH, RL, YS, ES and DW carried out the horse and poultry immunization assay. XL and CQ determined the mice neutralizing antibodies. All authors have read and approved the submitted manuscript.

Competing interests

The authors declare that they have no competing interests.
The present study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the Ministry of Science and Technology of the People’s Republic of China. The protocol was approved by the Animal Research Ethics Committee of Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Langevin SA, Bunning M, Davis B, Komar N. Experimental infection of chickens as candidate sentinels for West Nile virus. Emerg Infect Dis. 2001;7:726–9.CrossRefPubMedPubMedCentral Langevin SA, Bunning M, Davis B, Komar N. Experimental infection of chickens as candidate sentinels for West Nile virus. Emerg Infect Dis. 2001;7:726–9.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Himsworth CG, Gurney KE, Neimanis AS, Wobeser GA, Leighton FA. An outbreak of West Nile virus infection in captive lesser scaup (Aythya affinis) ducklings. Avian Dis. 2009;53:129–34.CrossRefPubMed Himsworth CG, Gurney KE, Neimanis AS, Wobeser GA, Leighton FA. An outbreak of West Nile virus infection in captive lesser scaup (Aythya affinis) ducklings. Avian Dis. 2009;53:129–34.CrossRefPubMed
4.
Zurück zum Zitat Banet-Noach C, Simanov L, Malkinson M. Direct (non-vector) transmission of West Nile virus in geese. Avian Pathol. 2003;32:489–94.CrossRefPubMed Banet-Noach C, Simanov L, Malkinson M. Direct (non-vector) transmission of West Nile virus in geese. Avian Pathol. 2003;32:489–94.CrossRefPubMed
5.
Zurück zum Zitat Ng T, Hathaway D, Jennings N, Champ D, Chiang YW, Chu HJ. Equine vaccine for West Nile virus. Dev Biol (Basel). 2003;114:221–7. Ng T, Hathaway D, Jennings N, Champ D, Chiang YW, Chu HJ. Equine vaccine for West Nile virus. Dev Biol (Basel). 2003;114:221–7.
6.
Zurück zum Zitat El Garch H, Minke JM, Rehder J, Richard S, Edlund Toulemonde C, Dinic S, Andreoni C, Audonnet JC, Nordgren R, Juillard V. A West Nile virus (WNV) recombinant canarypox virus vaccine elicits WNV-specific neutralizing antibodies and cell-mediated immune responses in the horse. Vet Immunol Immunopathol. 2008;123:230–9.CrossRefPubMed El Garch H, Minke JM, Rehder J, Richard S, Edlund Toulemonde C, Dinic S, Andreoni C, Audonnet JC, Nordgren R, Juillard V. A West Nile virus (WNV) recombinant canarypox virus vaccine elicits WNV-specific neutralizing antibodies and cell-mediated immune responses in the horse. Vet Immunol Immunopathol. 2008;123:230–9.CrossRefPubMed
7.
Zurück zum Zitat Bowen RA, Bosco-Lauth A, Syvrud K, Thomas A, Meinert TR, Ludlow DR, Cook C, Salt J, Ons E. Protection of horses from West Nile virus Lineage 2 challenge following immunization with a whole, inactivated WNV lineage 1 vaccine. Vaccine. 2014;32:5455–9.CrossRefPubMed Bowen RA, Bosco-Lauth A, Syvrud K, Thomas A, Meinert TR, Ludlow DR, Cook C, Salt J, Ons E. Protection of horses from West Nile virus Lineage 2 challenge following immunization with a whole, inactivated WNV lineage 1 vaccine. Vaccine. 2014;32:5455–9.CrossRefPubMed
8.
Zurück zum Zitat Minke JM, Siger L, Cupillard L, Powers B, Bakonyi T, Boyum S, Nowotny N, Bowen R. Protection provided by a recombinant ALVAC((R))-WNV vaccine expressing the prM/E genes of a lineage 1 strain of WNV against a virulent challenge with a lineage 2 strain. Vaccine. 2011;29:4608–12.CrossRefPubMed Minke JM, Siger L, Cupillard L, Powers B, Bakonyi T, Boyum S, Nowotny N, Bowen R. Protection provided by a recombinant ALVAC((R))-WNV vaccine expressing the prM/E genes of a lineage 1 strain of WNV against a virulent challenge with a lineage 2 strain. Vaccine. 2011;29:4608–12.CrossRefPubMed
9.
10.
Zurück zum Zitat Bukreyev A, Collins PL. Newcastle disease virus as a vaccine vector for humans. Curr Opin Mol Ther. 2008;10:46–55.PubMed Bukreyev A, Collins PL. Newcastle disease virus as a vaccine vector for humans. Curr Opin Mol Ther. 2008;10:46–55.PubMed
11.
Zurück zum Zitat Ge J, Deng G, Wen Z, Tian G, Wang Y, Shi J, Wang X, Li Y, Hu S, Jiang Y, et al. Newcastle disease virus-based live attenuated vaccine completely protects chickens and mice from lethal challenge of homologous and heterologous H5N1 avian influenza viruses. J Virol. 2007;81:150–8.CrossRefPubMed Ge J, Deng G, Wen Z, Tian G, Wang Y, Shi J, Wang X, Li Y, Hu S, Jiang Y, et al. Newcastle disease virus-based live attenuated vaccine completely protects chickens and mice from lethal challenge of homologous and heterologous H5N1 avian influenza viruses. J Virol. 2007;81:150–8.CrossRefPubMed
12.
Zurück zum Zitat Ge J, Tian G, Zeng X, Jiang Y, Chen H, Bua Z. Generation and evaluation of a Newcastle disease virus-based H9 avian influenza live vaccine. Avian Dis. 2010;54:294–6.CrossRefPubMed Ge J, Tian G, Zeng X, Jiang Y, Chen H, Bua Z. Generation and evaluation of a Newcastle disease virus-based H9 avian influenza live vaccine. Avian Dis. 2010;54:294–6.CrossRefPubMed
13.
Zurück zum Zitat DiNapoli JM, Yang L, Samal SK, Murphy BR, Collins PL, Bukreyev A. Respiratory tract immunization of non-human primates with a Newcastle disease virus-vectored vaccine candidate against Ebola virus elicits a neutralizing antibody response. Vaccine. 2010;29:17–25.CrossRefPubMedPubMedCentral DiNapoli JM, Yang L, Samal SK, Murphy BR, Collins PL, Bukreyev A. Respiratory tract immunization of non-human primates with a Newcastle disease virus-vectored vaccine candidate against Ebola virus elicits a neutralizing antibody response. Vaccine. 2010;29:17–25.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat DiNapoli JM, Nayak B, Yang L, Finneyfrock BW, Cook A, Andersen H, Torres-Velez F, Murphy BR, Samal SK, Collins PL, Bukreyev A. Newcastle disease virus-vectored vaccines expressing the hemagglutinin or neuraminidase protein of H5N1 highly pathogenic avian influenza virus protect against virus challenge in monkeys. J Virol. 2010;84:1489–503.CrossRefPubMed DiNapoli JM, Nayak B, Yang L, Finneyfrock BW, Cook A, Andersen H, Torres-Velez F, Murphy BR, Samal SK, Collins PL, Bukreyev A. Newcastle disease virus-vectored vaccines expressing the hemagglutinin or neuraminidase protein of H5N1 highly pathogenic avian influenza virus protect against virus challenge in monkeys. J Virol. 2010;84:1489–503.CrossRefPubMed
15.
Zurück zum Zitat Kortekaas J, de Boer SM, Kant J, Vloet RP, Antonis AF, Moormann RJ. Rift Valley fever virus immunity provided by a paramyxovirus vaccine vector. Vaccine. 2010;28:4394–401.CrossRefPubMed Kortekaas J, de Boer SM, Kant J, Vloet RP, Antonis AF, Moormann RJ. Rift Valley fever virus immunity provided by a paramyxovirus vaccine vector. Vaccine. 2010;28:4394–401.CrossRefPubMed
16.
Zurück zum Zitat Khattar SK, Collins PL, Samal SK. Immunization of cattle with recombinant Newcastle disease virus expressing bovine herpesvirus-1 (BHV-1) glycoprotein D induces mucosal and serum antibody responses and provides partial protection against BHV-1. Vaccine. 2010;28:3159–70.CrossRefPubMedPubMedCentral Khattar SK, Collins PL, Samal SK. Immunization of cattle with recombinant Newcastle disease virus expressing bovine herpesvirus-1 (BHV-1) glycoprotein D induces mucosal and serum antibody responses and provides partial protection against BHV-1. Vaccine. 2010;28:3159–70.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Liu Q, Mena I, Ma J, Bawa B, Krammer F, Lyoo YS, Lang Y, Morozov I, Mahardika GN, Ma W, et al. Newcastle Disease Virus-Vectored H7 and H5 Live Vaccines Protect Chickens from Challenge with H7N9 or H5N1 Avian Influenza Viruses. J Virol. 2015;89:7401–8.CrossRefPubMedPubMedCentral Liu Q, Mena I, Ma J, Bawa B, Krammer F, Lyoo YS, Lang Y, Morozov I, Mahardika GN, Ma W, et al. Newcastle Disease Virus-Vectored H7 and H5 Live Vaccines Protect Chickens from Challenge with H7N9 or H5N1 Avian Influenza Viruses. J Virol. 2015;89:7401–8.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Zhao W, Spatz S, Zhang Z, Wen G, Garcia M, Zsak L, Zsak L, Yu Q. Newcastle disease virus (NDV) recombinants expressing infectious laryngotracheitis virus (ILTV) glycoproteins gB and gD protect chickens against ILTV and NDV challenges. J Virol. 2014;88:8397–406. Zhao W, Spatz S, Zhang Z, Wen G, Garcia M, Zsak L, Zsak L, Yu Q. Newcastle disease virus (NDV) recombinants expressing infectious laryngotracheitis virus (ILTV) glycoproteins gB and gD protect chickens against ILTV and NDV challenges. J Virol. 2014;88:8397–406.
19.
Zurück zum Zitat Marquardt WW, Snyder DB, Savage PK, Kadavil SK, Yancey FS. Antibody response to Newcastle disease virus given by two different routes as measured by ELISA and hemagglutination-inhibition test and associated tracheal immunity. Avian Dis. 1985;29:71–9.CrossRefPubMed Marquardt WW, Snyder DB, Savage PK, Kadavil SK, Yancey FS. Antibody response to Newcastle disease virus given by two different routes as measured by ELISA and hemagglutination-inhibition test and associated tracheal immunity. Avian Dis. 1985;29:71–9.CrossRefPubMed
20.
Zurück zum Zitat Jayawardane GW, Spradbrow PB. Mucosal immunity in chickens vaccinated with the V4 strain of Newcastle disease virus. Vet Microbiol. 1995;46:69–77.CrossRefPubMed Jayawardane GW, Spradbrow PB. Mucosal immunity in chickens vaccinated with the V4 strain of Newcastle disease virus. Vet Microbiol. 1995;46:69–77.CrossRefPubMed
21.
Zurück zum Zitat Wambura PN, Wilson C. Protective antibody response following oral vaccination of feral pigeons (Columba livia) with Newcastle disease vaccine (strain I-2) coated on oiled rice. Vet Res Commun. 2009;33:921–6.CrossRefPubMed Wambura PN, Wilson C. Protective antibody response following oral vaccination of feral pigeons (Columba livia) with Newcastle disease vaccine (strain I-2) coated on oiled rice. Vet Res Commun. 2009;33:921–6.CrossRefPubMed
22.
Zurück zum Zitat Wengler G, Wengler G. Cell-associated West Nile flavivirus is covered with E + pre-M protein heterodimers which are destroyed and reorganized by proteolytic cleavage during virus release. J Virol. 1989;63:2521–6.PubMedPubMedCentral Wengler G, Wengler G. Cell-associated West Nile flavivirus is covered with E + pre-M protein heterodimers which are destroyed and reorganized by proteolytic cleavage during virus release. J Virol. 1989;63:2521–6.PubMedPubMedCentral
23.
Zurück zum Zitat Sanchez MD, Pierson TC, McAllister D, Hanna SL, Puffer BA, Valentine LE, Murtadha MM, Hoxie JA, Doms RW. Characterization of neutralizing antibodies to West Nile virus. Virology. 2005;336:70–82.CrossRefPubMed Sanchez MD, Pierson TC, McAllister D, Hanna SL, Puffer BA, Valentine LE, Murtadha MM, Hoxie JA, Doms RW. Characterization of neutralizing antibodies to West Nile virus. Virology. 2005;336:70–82.CrossRefPubMed
24.
Zurück zum Zitat Guo LP, Huo H, Wang XL, Bu ZG, Hua RH. Generation and characterization of a monoclonal antibody against prM protein of West Nile virus. Monoclon Antib Immunodiagn Immunother. 2014;33:438–43.CrossRefPubMedPubMedCentral Guo LP, Huo H, Wang XL, Bu ZG, Hua RH. Generation and characterization of a monoclonal antibody against prM protein of West Nile virus. Monoclon Antib Immunodiagn Immunother. 2014;33:438–43.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat OIE. Manual of Diagnostic Tests and Vaccines for Terrestrial Animals 2011. Paris: Office International des Epizooties; 2011. OIE. Manual of Diagnostic Tests and Vaccines for Terrestrial Animals 2011. Paris: Office International des Epizooties; 2011.
26.
Zurück zum Zitat Kong D, Wen Z, Su H, Ge J, Chen W, Wang X, Wu C, Yang C, Chen H, Bu Z. Newcastle disease virus-vectored Nipah encephalitis vaccines induce B and T cell responses in mice and long-lasting neutralizing antibodies in pigs. Virol. 2012;432:327–35.CrossRef Kong D, Wen Z, Su H, Ge J, Chen W, Wang X, Wu C, Yang C, Chen H, Bu Z. Newcastle disease virus-vectored Nipah encephalitis vaccines induce B and T cell responses in mice and long-lasting neutralizing antibodies in pigs. Virol. 2012;432:327–35.CrossRef
27.
Zurück zum Zitat Wen Z, Ye L, Gao Y, Pan L, Dong K, Bu Z, Compans RW, Yang C. Immunization by influenza virus-like particles protects aged mice against lethal influenza virus challenge. Antiviral Res. 2009;84:215–24.CrossRefPubMedPubMedCentral Wen Z, Ye L, Gao Y, Pan L, Dong K, Bu Z, Compans RW, Yang C. Immunization by influenza virus-like particles protects aged mice against lethal influenza virus challenge. Antiviral Res. 2009;84:215–24.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Ye L, Bu Z, Vzorov A, Taylor D, Compans RW, Yang C. Surface stability and immunogenicity of the human immunodeficiency virus envelope glycoprotein: role of the cytoplasmic domain. J Virol. 2004;78:13409–19.CrossRefPubMedPubMedCentral Ye L, Bu Z, Vzorov A, Taylor D, Compans RW, Yang C. Surface stability and immunogenicity of the human immunodeficiency virus envelope glycoprotein: role of the cytoplasmic domain. J Virol. 2004;78:13409–19.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Brien JD, Uhrlaub JL, Nikolich-Zugich J. West Nile virus-specific CD4 T cells exhibit direct antiviral cytokine secretion and cytotoxicity and are sufficient for antiviral protection. J Immunol. 2008;181:8568–75.CrossRefPubMedPubMedCentral Brien JD, Uhrlaub JL, Nikolich-Zugich J. West Nile virus-specific CD4 T cells exhibit direct antiviral cytokine secretion and cytotoxicity and are sufficient for antiviral protection. J Immunol. 2008;181:8568–75.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Brien JD, Uhrlaub JL, Nikolich-Zugich J. Protective capacity and epitope specificity of CD8(+) T cells responding to lethal West Nile virus infection. Eur J Immunol. 2007;37:1855–63.CrossRefPubMed Brien JD, Uhrlaub JL, Nikolich-Zugich J. Protective capacity and epitope specificity of CD8(+) T cells responding to lethal West Nile virus infection. Eur J Immunol. 2007;37:1855–63.CrossRefPubMed
31.
Zurück zum Zitat Diamond MS, Shrestha B, Marri A, Mahan D, Engle M. B cells and antibody play critical roles in the immediate defense of disseminated infection by West Nile encephalitis virus. J Virol. 2003;77:2578–86.CrossRefPubMedPubMedCentral Diamond MS, Shrestha B, Marri A, Mahan D, Engle M. B cells and antibody play critical roles in the immediate defense of disseminated infection by West Nile encephalitis virus. J Virol. 2003;77:2578–86.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Ferguson M, Kurane I, Wimalaratne O, Shin J, Wood D, group WHOic. WHO informal consultation on the scientific basis of specifications for production and control of inactivated Japanese encephalitis vaccines for human use, Geneva, Switzerland, 1–2 June 2006. Vaccine. 2007;25:5233–43.CrossRefPubMed Ferguson M, Kurane I, Wimalaratne O, Shin J, Wood D, group WHOic. WHO informal consultation on the scientific basis of specifications for production and control of inactivated Japanese encephalitis vaccines for human use, Geneva, Switzerland, 1–2 June 2006. Vaccine. 2007;25:5233–43.CrossRefPubMed
33.
Zurück zum Zitat Muraki Y, Fujita T, Matsuura M, Fuke I, Manabe S, Ishikawa T, Okuno Y, Morita K. The efficacy of inactivated West Nile vaccine (WN-VAX) in mice and monkeys. J Virol. 2015;12:54.CrossRef Muraki Y, Fujita T, Matsuura M, Fuke I, Manabe S, Ishikawa T, Okuno Y, Morita K. The efficacy of inactivated West Nile vaccine (WN-VAX) in mice and monkeys. J Virol. 2015;12:54.CrossRef
34.
Zurück zum Zitat Sitati EM, Diamond MS. CD4+ T-cell responses are required for clearance of West Nile virus from the central nervous system. J Virol. 2006;80:12060–9.CrossRefPubMedPubMedCentral Sitati EM, Diamond MS. CD4+ T-cell responses are required for clearance of West Nile virus from the central nervous system. J Virol. 2006;80:12060–9.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Mathew A, Terajima M, West K, Green S, Rothman AL, Ennis FA, Kennedy JS. Identification of murine poxvirus-specific CD8+ CTL epitopes with distinct functional profiles. J Immunol. 2005;174:2212–9.CrossRefPubMed Mathew A, Terajima M, West K, Green S, Rothman AL, Ennis FA, Kennedy JS. Identification of murine poxvirus-specific CD8+ CTL epitopes with distinct functional profiles. J Immunol. 2005;174:2212–9.CrossRefPubMed
37.
Zurück zum Zitat Song GY, Srivastava T, Ishizaki H, Lacey SF, Diamond DJ, Ellenhorn JD. Recombinant modified vaccinia virus ankara (MVA) expressing wild-type human p53 induces specific antitumor CTL expansion. Cancer Invest. 2011;29:501–10.CrossRefPubMedPubMedCentral Song GY, Srivastava T, Ishizaki H, Lacey SF, Diamond DJ, Ellenhorn JD. Recombinant modified vaccinia virus ankara (MVA) expressing wild-type human p53 induces specific antitumor CTL expansion. Cancer Invest. 2011;29:501–10.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Wang Z, La Rosa C, Lacey SF, Maas R, Mekhoubad S, Britt WJ, Diamond DJ. Attenuated poxvirus expressing three immunodominant CMV antigens as a vaccine strategy for CMV infection. J Clin Virol. 2006;35:324–31.CrossRefPubMed Wang Z, La Rosa C, Lacey SF, Maas R, Mekhoubad S, Britt WJ, Diamond DJ. Attenuated poxvirus expressing three immunodominant CMV antigens as a vaccine strategy for CMV infection. J Clin Virol. 2006;35:324–31.CrossRefPubMed
39.
Zurück zum Zitat Samina I, Havenga M, Koudstaal W, Khinich Y, Koldijk M, Malkinson M, Simanov M, Perl S, Gijsbers L, Weverling GJ et al. Safety and efficacy in geese of a PER.C6-based inactivated West Nile virus vaccine. Vaccine. 2007;25:8338–45.CrossRefPubMed Samina I, Havenga M, Koudstaal W, Khinich Y, Koldijk M, Malkinson M, Simanov M, Perl S, Gijsbers L, Weverling GJ et al. Safety and efficacy in geese of a PER.C6-based inactivated West Nile virus vaccine. Vaccine. 2007;25:8338–45.CrossRefPubMed
40.
Zurück zum Zitat Lim CK, Takasaki T, Kotaki A, Kurane I. Vero cell-derived inactivated West Nile (WN) vaccine induces protective immunity against lethal WN virus infection in mice and shows a facilitated neutralizing antibody response in mice previously immunized with Japanese encephalitis vaccine. Virol. 2008;374:60–70.CrossRef Lim CK, Takasaki T, Kotaki A, Kurane I. Vero cell-derived inactivated West Nile (WN) vaccine induces protective immunity against lethal WN virus infection in mice and shows a facilitated neutralizing antibody response in mice previously immunized with Japanese encephalitis vaccine. Virol. 2008;374:60–70.CrossRef
41.
Zurück zum Zitat Davis BS, Chang GJ, Cropp B, Roehrig JT, Martin DA, Mitchell CJ, Bowen R, Bunning ML. West Nile virus recombinant DNA vaccine protects mouse and horse from virus challenge and expresses in vitro a noninfectious recombinant antigen that can be used in enzyme-linked immunosorbent assays. J Virol. 2001;75:4040–7.CrossRefPubMedPubMedCentral Davis BS, Chang GJ, Cropp B, Roehrig JT, Martin DA, Mitchell CJ, Bowen R, Bunning ML. West Nile virus recombinant DNA vaccine protects mouse and horse from virus challenge and expresses in vitro a noninfectious recombinant antigen that can be used in enzyme-linked immunosorbent assays. J Virol. 2001;75:4040–7.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Seregin A, Nistler R, Borisevich V, Yamshchikov G, Chaporgina E, Kwok CW, Yamshchikov V. Immunogenicity of West Nile virus infectious DNA and its noninfectious derivatives. Virol. 2006;356:115–25.CrossRef Seregin A, Nistler R, Borisevich V, Yamshchikov G, Chaporgina E, Kwok CW, Yamshchikov V. Immunogenicity of West Nile virus infectious DNA and its noninfectious derivatives. Virol. 2006;356:115–25.CrossRef
43.
Zurück zum Zitat Chang DC, Liu WJ, Anraku I, Clark DC, Pollitt CC, Suhrbier A, Hall RA, Khromykh AA. Single-round infectious particles enhance immunogenicity of a DNA vaccine against West Nile virus. Nat Biotechnol. 2008;26:571–7.CrossRefPubMed Chang DC, Liu WJ, Anraku I, Clark DC, Pollitt CC, Suhrbier A, Hall RA, Khromykh AA. Single-round infectious particles enhance immunogenicity of a DNA vaccine against West Nile virus. Nat Biotechnol. 2008;26:571–7.CrossRefPubMed
44.
Zurück zum Zitat Karaca K, Bowen R, Austgen LE, Teehee M, Siger L, Grosenbaugh D, Loosemore L, Audonnet JC, Nordgren R, Minke JM. Recombinant canarypox vectored West Nile virus (WNV) vaccine protects dogs and cats against a mosquito WNV challenge. Vaccine. 2005;23:3808–13.CrossRefPubMed Karaca K, Bowen R, Austgen LE, Teehee M, Siger L, Grosenbaugh D, Loosemore L, Audonnet JC, Nordgren R, Minke JM. Recombinant canarypox vectored West Nile virus (WNV) vaccine protects dogs and cats against a mosquito WNV challenge. Vaccine. 2005;23:3808–13.CrossRefPubMed
45.
Zurück zum Zitat Monath TP, Liu J, Kanesa-Thasan N, Myers GA, Nichols R, Deary A, McCarthy K, Johnson C, Ermak T, Shin S, et al. A live, attenuated recombinant West Nile virus vaccine. Proc Natl Acad Sci U S A. 2006;103:6694–9.CrossRefPubMedPubMedCentral Monath TP, Liu J, Kanesa-Thasan N, Myers GA, Nichols R, Deary A, McCarthy K, Johnson C, Ermak T, Shin S, et al. A live, attenuated recombinant West Nile virus vaccine. Proc Natl Acad Sci U S A. 2006;103:6694–9.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Long MT, Gibbs EP, Mellencamp MW, Bowen RA, Seino KK, Zhang S, Beachboard SE, Humphrey PP. Efficacy, duration, and onset of immunogenicity of a West Nile virus vaccine, live Flavivirus chimera, in horses with a clinical disease challenge model. Equine Vet J. 2007;39:491–7.CrossRefPubMed Long MT, Gibbs EP, Mellencamp MW, Bowen RA, Seino KK, Zhang S, Beachboard SE, Humphrey PP. Efficacy, duration, and onset of immunogenicity of a West Nile virus vaccine, live Flavivirus chimera, in horses with a clinical disease challenge model. Equine Vet J. 2007;39:491–7.CrossRefPubMed
47.
Zurück zum Zitat Seino KK, Long MT, Gibbs EP, Bowen RA, Beachboard SE, Humphrey PP, Dixon MA, Bourgeois MA. Comparative efficacies of three commercially available vaccines against West Nile Virus (WNV) in a short-duration challenge trial involving an equine WNV encephalitis model. Clin Vaccine Immunol. 2007;14:1465–71.CrossRefPubMedPubMedCentral Seino KK, Long MT, Gibbs EP, Bowen RA, Beachboard SE, Humphrey PP, Dixon MA, Bourgeois MA. Comparative efficacies of three commercially available vaccines against West Nile Virus (WNV) in a short-duration challenge trial involving an equine WNV encephalitis model. Clin Vaccine Immunol. 2007;14:1465–71.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Minke JM, Siger L, Karaca K, Austgen L, Gordy P, Bowen R, Renshaw RW, Loosmore S, Audonnet JC, Nordgren B. Recombinant canarypoxvirus vaccine carrying the prM/E genes of West Nile virus protects horses against a West Nile virus-mosquito challenge. Arch Virol Suppl. 2004;18:221–30.PubMed Minke JM, Siger L, Karaca K, Austgen L, Gordy P, Bowen R, Renshaw RW, Loosmore S, Audonnet JC, Nordgren B. Recombinant canarypoxvirus vaccine carrying the prM/E genes of West Nile virus protects horses against a West Nile virus-mosquito challenge. Arch Virol Suppl. 2004;18:221–30.PubMed
49.
Zurück zum Zitat Blackmore CG, Stark LM, Jeter WC, Oliveri RL, Brooks RG, Conti LA, Wiersma ST. Surveillance results from the first West Nile virus transmission season in Florida, 2001. Am J Trop Med Hyg. 2003;69:141–50.PubMed Blackmore CG, Stark LM, Jeter WC, Oliveri RL, Brooks RG, Conti LA, Wiersma ST. Surveillance results from the first West Nile virus transmission season in Florida, 2001. Am J Trop Med Hyg. 2003;69:141–50.PubMed
50.
Zurück zum Zitat Chaskopoulou A, Dovas C, Chaintoutis S, Bouzalas I, Ara G, Papanastassopoulou M: Evidence of enzootic circulation of West Nile virus (Nea Santa-Greece-2010, lineage 2), Greece, May to July 2011. Euro Surveill. 2011: 16. Chaskopoulou A, Dovas C, Chaintoutis S, Bouzalas I, Ara G, Papanastassopoulou M: Evidence of enzootic circulation of West Nile virus (Nea Santa-Greece-2010, lineage 2), Greece, May to July 2011. Euro Surveill. 2011: 16.
51.
Zurück zum Zitat Fall AG, Diaite A, Seck MT, Bouyer J, Lefrancois T, Vachiery N, Aprelon R, Faye O, Konate L, Lancelot R. West Nile virus transmission in sentinel chickens and potential mosquito vectors, Senegal River Delta, 2008–2009. Int J Environ Res Public Health. 2013;10:4718–27.CrossRefPubMedPubMedCentral Fall AG, Diaite A, Seck MT, Bouyer J, Lefrancois T, Vachiery N, Aprelon R, Faye O, Konate L, Lancelot R. West Nile virus transmission in sentinel chickens and potential mosquito vectors, Senegal River Delta, 2008–2009. Int J Environ Res Public Health. 2013;10:4718–27.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Komar N. West Nile virus surveillance using sentinel birds. Ann N Y Acad Sci. 2001;951:58–73.CrossRefPubMed Komar N. West Nile virus surveillance using sentinel birds. Ann N Y Acad Sci. 2001;951:58–73.CrossRefPubMed
53.
Zurück zum Zitat Meece JK, Kronenwetter-Koepel TA, Vandermause MF, Reed KD. West Nile virus infection in commercial waterfowl operation, Wisconsin. Emerg Infect Dis. 2006;12:1451–3.CrossRefPubMedPubMedCentral Meece JK, Kronenwetter-Koepel TA, Vandermause MF, Reed KD. West Nile virus infection in commercial waterfowl operation, Wisconsin. Emerg Infect Dis. 2006;12:1451–3.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Wojnarowicz C, Olkowski A, Schwean-Lardner K. First Canadian outbreak of West Nile virus disease in farmed domestic ducks in Saskatchewan. Can Vet J. 2007;48:1270–1.PubMedPubMedCentral Wojnarowicz C, Olkowski A, Schwean-Lardner K. First Canadian outbreak of West Nile virus disease in farmed domestic ducks in Saskatchewan. Can Vet J. 2007;48:1270–1.PubMedPubMedCentral
55.
Zurück zum Zitat Hofmeister E, Porter RE, Franson JC. Experimental susceptibility of wood ducks (Aix sponsa) for West Nile virus. J Wildl Dis. 2015;51:411–8.CrossRefPubMed Hofmeister E, Porter RE, Franson JC. Experimental susceptibility of wood ducks (Aix sponsa) for West Nile virus. J Wildl Dis. 2015;51:411–8.CrossRefPubMed
56.
Zurück zum Zitat Guy JS, Malkinson M. Arbovirus infections. In: Saif YM, editor. Diseases of Poultry. 11th ed. Ames: Iowa State University Press; 2003. p. 388–98. Guy JS, Malkinson M. Arbovirus infections. In: Saif YM, editor. Diseases of Poultry. 11th ed. Ames: Iowa State University Press; 2003. p. 388–98.
57.
Zurück zum Zitat Austin RJ, Whiting TL, Anderson RA, Drebot MA. An outbreak of West Nile virus-associated disease in domestic geese (Anser anser domesticus) upon initial introduction to a geographic region, with evidence of bird to bird transmission. Can Vet J. 2004;45:117–23.PubMedPubMedCentral Austin RJ, Whiting TL, Anderson RA, Drebot MA. An outbreak of West Nile virus-associated disease in domestic geese (Anser anser domesticus) upon initial introduction to a geographic region, with evidence of bird to bird transmission. Can Vet J. 2004;45:117–23.PubMedPubMedCentral
58.
Zurück zum Zitat Samina I, Khinich Y, Simanov M, Malkinson M. An inactivated West Nile virus vaccine for domestic geese-efficacy study and a summary of 4 years of field application. Vaccine. 2005;23:4955–8.CrossRefPubMed Samina I, Khinich Y, Simanov M, Malkinson M. An inactivated West Nile virus vaccine for domestic geese-efficacy study and a summary of 4 years of field application. Vaccine. 2005;23:4955–8.CrossRefPubMed
59.
Zurück zum Zitat Sa ESM, Ellis A, Karaca K, Minke J, Nordgren R, Wu S, Swayne DE. Domestic goose as a model for West Nile virus vaccine efficacy. Vaccine. 2013;31:1045–50.CrossRef Sa ESM, Ellis A, Karaca K, Minke J, Nordgren R, Wu S, Swayne DE. Domestic goose as a model for West Nile virus vaccine efficacy. Vaccine. 2013;31:1045–50.CrossRef
60.
Zurück zum Zitat Jarvi SI, Lieberman MM, Hofmeister E, Nerurkar VR, Wong T, Weeks-Levy C. Protective efficacy of a recombinant subunit West Nile virus vaccine in domestic geese (Anser anser). Vaccine. 2008;26:5338–44.CrossRefPubMed Jarvi SI, Lieberman MM, Hofmeister E, Nerurkar VR, Wong T, Weeks-Levy C. Protective efficacy of a recombinant subunit West Nile virus vaccine in domestic geese (Anser anser). Vaccine. 2008;26:5338–44.CrossRefPubMed
61.
Zurück zum Zitat Lu Z, Fu SH, Cao L, Tang CJ, Zhang S, Li ZX, Tusong M, Yao XH, Zhang HL, Wang PY, et al. Human infection with West Nile Virus, Xinjiang, China, 2011. Emerg Infect Dis. 2014;20:1421–3.CrossRefPubMedPubMedCentral Lu Z, Fu SH, Cao L, Tang CJ, Zhang S, Li ZX, Tusong M, Yao XH, Zhang HL, Wang PY, et al. Human infection with West Nile Virus, Xinjiang, China, 2011. Emerg Infect Dis. 2014;20:1421–3.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Li XL, Fu SH, Liu WB, Wang HY, Lu Z, Tong SX, Li ZX, Nasci RS, Kosoy O, Cui Y, Liang GD. West nile virus infection in Xinjiang, China. Vector Borne Zoonotic Dis. 2013;13:131–3.CrossRefPubMed Li XL, Fu SH, Liu WB, Wang HY, Lu Z, Tong SX, Li ZX, Nasci RS, Kosoy O, Cui Y, Liang GD. West nile virus infection in Xinjiang, China. Vector Borne Zoonotic Dis. 2013;13:131–3.CrossRefPubMed
Metadaten
Titel
Newcastle disease virus-vectored West Nile fever vaccine is immunogenic in mammals and poultry
verfasst von
Jinliang Wang
Jie Yang
Jinying Ge
Ronghong Hua
Renqiang Liu
Xiaofeng Li
Xijun Wang
Yu Shao
Encheng Sun
Donglai Wu
Chengfeng Qin
Zhiyuan Wen
Zhigao Bu
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Virology Journal / Ausgabe 1/2016
Elektronische ISSN: 1743-422X
DOI
https://doi.org/10.1186/s12985-016-0568-5

Weitere Artikel der Ausgabe 1/2016

Virology Journal 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.