An A14U Substitution in the 3' Noncoding Region of the M Segment of Viral RNA Supports Replication of Influenza Virus with an NS1 Deletion by Modulating Alternative Splicing of M Segment mRNAs

Min Zheng, Pui Wang, Wenjun Song, Siu-Ying Lau, Siwen Liu, Xiaofeng Huang, Bobo Wing-Yee Mok, Yen-Chin Liu, Yixin Chen, Kwok-Yung Yuen, Honglin Chen, Min Zheng, Pui Wang, Wenjun Song, Siu-Ying Lau, Siwen Liu, Xiaofeng Huang, Bobo Wing-Yee Mok, Yen-Chin Liu, Yixin Chen, Kwok-Yung Yuen, Honglin Chen

Abstract

The NS1 protein of influenza virus has multiple functions and is a determinant of virulence. Influenza viruses with NS1 deletions (DelNS1 influenza viruses) are a useful tool for studying virus replication and can serve as effective live attenuated vaccines, but deletion of NS1 severely diminishes virus replication, hampering functional studies and vaccine production. We found that WSN-DelNS1 viruses passaged in cells consistently adapted to gain an A14U substitution in the 3' noncoding region of the M segment of viral RNA (vRNA) which restored replicative ability. DelNS1-M-A14U viruses cannot inhibit interferon expression in virus infected-cells, providing an essential model for studying virus replication in the absence of the NS1 protein. Characterization of DelNS1-M-A14U virus showed that the lack of NS1 has no apparent effect on expression of other viral proteins, with the exception of M mRNAs. Expression of the M transcripts, M1, M2, mRNA3, and mRNA4, is regulated by alternative splicing. The A14U substitution changes the splicing donor site consensus sequence of mRNA3, altering expression of M transcripts, with M2 expression significantly increased and mRNA3 markedly suppressed in DelNS1-M-A14U, but not DelNS1-M-WT, virus-infected cells. Further analysis revealed that the A14U substitution also affects promoter function during replication of the viral genome. The M-A14U mutation increases M vRNA synthesis in DelNS1 virus infection and enhances alternative splicing of M2 mRNA in the absence of other viral proteins. The findings demonstrate that NS1 is directly involved in influenza virus replication through modulation of alternative splicing of M transcripts and provide strategic information important to construction of vaccine strains with NS1 deletions.

Importance: Nonstructural protein (NS1) of influenza virus has multiple functions. Besides its role in antagonizing host antiviral activity, NS1 is also believed to be involved in regulating virus replication, but mechanistic details are not clear. The NS1 protein is a virulence determinant which inhibits both innate and adaptive immunity and live attenuated viruses with NS1 deletions show promise as effective vaccines. However, deletion of NS1 causes severe attenuation of virus replication during infection, impeding functional studies and vaccine development. We characterized a replication-competent DelNS1 virus which carries an A14U substitution in the 3' noncoding region of the vRNA M segment. We found that M-A14U mutation supports virus replication through modulation of alternative splicing of mRNAs transcribed from the M segment. Our findings give insight into the role of NS1 in influenza virus replication and provide an approach for constructing replication-competent strains with NS1 deletions for use in functional and vaccine studies.

Copyright © 2015, Zheng et al.

Figures

FIG 1
FIG 1
Construction and establishment of stabilized WSN-DelNS1 virus. (A) Schematic illustration of the NS segment transcripts and the NS mutant with an NS1 deletion (DelNS1). The DelNS1 plasmid was constructed by deleting the intron region ranging from nt 57 to 528 in the NS segment. NCR, noncoding region. (B) Confirmation of NS1 deletion in rescued DelNS1 viruses. Viral RNA was extracted from P1 virus, and the NS segment was amplified by RT-PCR and analyzed using agarose gel electrophoresis. (C) Plaque sizes of DelNS1 and wild-type A/WSN/33 viruses. (D) Titer (PFU/ml) of DelNS1 virus after each passage. (E) Sequence analysis of the DelNS1 virus genome revealed an A-to-U substitution at nucleotide position 14 in the M segment noncoding region. The noncoding region is marked by a black line, and the A14U mutation is indicated with an arrowhead. (F) Comparison of rescue efficiency for WSN-DelNS1 viruses containing M-WT and M-A14U, M-A14U-CM15, M-A14C, and M-A14G substitutions. NR, not rescued. (G) Rescue efficiency of PR8-DelNS1 viruses containing M-WT or M-A14U. The DelNS1 viruses were rescued with the indicated M-WT or M mutant plasmids in mixed HEK293T and MDCK cell cultures and then titrated by plaque assay. Values plotted are means (± standard deviations [SD]) (n = 3) and are representative of data from at least 5 independent experiments.
FIG 2
FIG 2
Growth kinetics of the M-A14U-DelNS1 virus in MDCK and Vero cells. (A and B) Column-purified reverse genetic WSN-WT, WSN-DelNS1-M-A14U, and WSN-DelNS1-M-WT viruses were used to infect MDCK cells (A) or Vero cells (B) at a multiplicity of infection (MOI) of 0.1. Supernatants were collected at the indicated time points and virus titrated by plaque assay. (C) Column-purified reverse genetic PR8-WT and PR8-DelNS1-M-A14U viruses were used to infect MDCK cells (left panel) and Vero cells (right panel) at an MOI of 0.1. Supernatants were collected at 24 h postinfection and titrated by plaque assay. The values (mean ± SD; n = 3) plotted are representative data from at least 3 independent experiments.
FIG 3
FIG 3
Loss of IFN-β suppression activity in WSN-DelNS1 viruses. (A) HEK293T cells were transfected with an IFN-β reporter plasmid 24 h prior to infection with either WSN-WT or WSN-DelNS1-M-A14U at an MOI of 1 or with the positive control, Sendai virus (SeV), at 50 HA units. After 24 h, cells were harvested and cell lysates prepared for estimation of luciferase activity. The luciferase assays were performed in triplicate, and values were normalized to the Renilla luciferase control. (B) IFN-β and viral NP mRNA levels were quantified by qRT-PCR after MDCK cells were infected with WSN-WT or WSN-DelNS1-M-A14U at an MOI of 0.1 and cultured for 16 h. Values were normalized against canine actin. (C) Suppression of IRF3 dimerization in HEK293T cells infected with WSN-WT, WSN-DelNS1-M-A14U, or Sendai virus was analyzed by native gel electrophoresis. (D) Activity in suppressing IFN-β expression was compared in MDCK cells infected with WSN-DelNS1-M-WT or WSN-DelNS1-M-A14U virus. Similar to the experiment described for panel B, MDCK cells were infected with the indicated viruses at an MOI of 0.1, and at 16 h postinfection, IFN-β and NP mRNA levels were quantified by qPCR. Values were normalized to canine actin. (E) Groups of six BALB/c mice, aged 6 to 8 weeks, were intranasally inoculated with 5 × 104 PFU of WSN-WT or WSN-DelNS1-M-A14U mutant virus, and body weight was monitored daily for 14 days postinfection. (F) Replication efficiency of viruses in lung tissues of infected mice. Groups of three mice were infected with 104 PFU of WSN-WT, WSN-DelNS1-M-WT, or WSN-DelNS1-M-A14U mutant viruses and then euthanized at 72 h postinfection, with lung tissues from each mouse being collected and homogenized for virus titration by plaque assay using MDCK cells. Statistical significance was analyzed by one-way analysis of variance (ANOVA) or Student's t test (**, P < 0.01). The bars plotted show means ± SD (n = 3), and the results represent at least three independent experiments.
FIG 4
FIG 4
Effect of M-A14U mutation on M1 and M2 protein expression. (A) MDCK cells were infected with WSN-WT or WSN-DelNS1-M-A14U virus at an MOI of 2. Cells were harvested at the indicated time points and cell lysates analyzed by Western blotting with specific antibodies, as described in Materials and Methods. (B) MDCK cells were infected with WSN-DelNS1-M-WT or WSN-DelNS1-M-A14U virus at an MOI of 0.1. At 16 h postinfection, cell lysates were collected for Western blotting with specific antibodies. (C) MDCK cells were infected with WSN-WT or WSN-M-A14U virus at an MOI of 5. Cell lysates were prepared at the indicated time points for Western blotting with specific antibodies. β-Tubulin was included as a loading control. All of the results are representative of three independent experiments.
FIG 5
FIG 5
Effect of M-A14U substitution on alternative splicing of M transcripts. (A) Schematic illustration of the M segment transcripts. The sequence of the 3′ noncoding region of the vRNA M segment is shown in red, with the mRNA3 splicing donor (SD) site highlighted in yellow. Splicing consensus sequences for the donor site are indicated in green above the noncoding region sequence (M, A or C; R, A or G). (B) Analysis of levels of different M mRNAs in virus-infected cells. MDCK cells were infected with the indicated viruses at an MOI of 0.1. Total RNA was isolated at 16 h postinfection. The mRNA levels for M1, M2, mRNA3, and M4 were determined by quantitative RT-PCR, as described in Materials and Methods. (C) HEK293T cells were transfected with pHW2000-WSN-M-WT or pHW2000-WSN-M-A14U plasmid with or without cotransfection of pCX-WSN-NS1 for coexpression of NS1. At 48 h posttransfection, total RNAs were isolated. After DNase treatment, the mRNA levels were determined by quantitative RT-PCR. (D) MDCK cells were infected with PR8-WT, PR8-M-A14U, or PR8-DelNS1-M-A14U virus at an MOI of 0.1. At 16 h postinfection, total RNAs were isolated and mRNA levels measured by qPCR. M2/M1 mRNA and mRNA3/M1 mRNA ratios are shown. All the results plotted indicate means ± SD (n = 3) and are representative of three independent experiments.
FIG 6
FIG 6
Effect of M-A14U substitution on M2/M1 mRNA ratio. (A) MDCK cells were infected with WSN-WT, WSN-M-A14U, WSN-M-A14G, or WSN-M-G12C-CM13 virus at an MOI of 5. Total RNAs were extracted at the indicated time points and levels of mRNA3 determined by quantitative RT-PCR. (B) Analysis of growth kinetics of WSN-WT, WSN-M-A14U, WSN-M-A14G, and WSN-M-G12C-CM13 viruses. MDCK cells were infected with these viruses at an MOI of 0.001. Supernatants were collected at the indicated time points and titrated by plaque assay. (C to E) MDCK cells were infected with WSN-WT, WSN-M-A14U, WSN-DelNS1-M-WT, or WSN-DelNS1-M-A14U virus at an MOI of 0.1. At 16 h after infection, total RNAs were isolated, and the M2/M1, mRNA3/M1, and M4/M1 mRNA ratios were determined by quantitative RT-PCR. (F) Effect of mRNA3 on virus replication. HEK293T cells were transfected with pCX-WSN-mRNA3 or a control vector 24 h prior to infection with WSN-DelNS1-M-A14U virus at an MOI of 0.1. Supernatants were collected at the indicated time points and virus titrated by plaque assay. All of the bars and points plotted indicate means ± SD (n = 3) from three independent experiments. **, P = 0.0013 by Student's t test.
FIG 7
FIG 7
Regulation of M mRNA splicing host machinery by viral NS1 and polymerase proteins. (A) HEK293T cells were transfected with increasing amounts of pCX-WSN-NS1. At 24 h after transfection, cells were infected with WSN-DelNS1-M-WT, WSN-DelNS1-M-A14U, or WSN-WT virus at an MOI of 0.5. Total RNAs were isolated at 8 h postinfection and M1 mRNA, M2 mRNA, and mRNA3 levels analyzed by quantitative RT-PCR. (B) HEK293T cells were transfected with WT and various mutants of the M segment cloned into pHW2000 plasmids, which express M vRNA, with or without the pCX-WSN-NS1 plasmid. Cell lysates were prepared at 48 h posttransfection for Western blotting with specific antibodies. β-Tubulin was included as a loading control. (C) Effect of A14U substitution on M vRNA replication. MDCK cells were infected with WSN-WT, WSN-M-A14U, or WSN-M-A14G virus at an MOI of 5. At 4 and 10 h postinfection, total RNAs were isolated for qRT-PCR analysis, and relative amounts of vRNA were determined as described above. (D) The A14U substitution enhances M vRNA replication. MDCK cells were infected with WSN-DelNS1-M-WT or WSN-DelNS1-M-A14U virus at an MOI of 0.1. Total RNAs were isolated at 16 h after infection. M and NP vRNA levels were determined by quantitative RT-PCR, as described above. All bars plotted show means ± SD (n = 3). The results represent three independent experiments. **, P < 0.01; ***, P = 0.0002 (by Student's t test).

References

    1. Taubenberger JK, Kash JC. 2010. Influenza virus evolution, host adaptation, and pandemic formation. Cell Host Microbe 7:440–451. doi:10.1016/j.chom.2010.05.009.
    1. Webster RG, Bean WJ, Gorman OT, Chambers TM, Kawaoka Y. 1992. Evolution and ecology of influenza A viruses. Microbiol Rev 56:152–179.
    1. Palese P, Shaw ML. 2006. Orthmyxoviridae: the viruses and their replication, p 1647–1689. In Knipe DM, Howley P (ed), Fields virology, 5th ed Lippincott Williams & Wilkins, Philadelphia, PA.
    1. Nelson MI, Viboud C, Simonsen L, Bennett RT, Griesemer SB, St George K, Taylor J, Spiro DJ, Sengamalay NA, Ghedin E, Taubenberger JK, Holmes EC. 2008. Multiple reassortment events in the evolutionary history of H1N1 influenza A virus since 1918. PLoS Pathog 4:e1000012. doi:10.1371/journal.ppat.1000012.
    1. Naffakh N, Tomoiu A, Rameix-Welti MA, van der Werf S. 2008. Host restriction of avian influenza viruses at the level of the ribonucleoproteins. Annu Rev Microbiol 62:403–424. doi:10.1146/annurev.micro.62.081307.162746.
    1. Eisfeld AJ, Neumann G, Kawaoka Y. 2015. At the centre: influenza A virus ribonucleoproteins. Nat Rev Microbiol 13:28–41. doi:10.1038/nrmicro3367.
    1. Wise HM, Hutchinson EC, Jagger BW, Stuart AD, Kang ZH, Robb N, Schwartzman LM, Kash JC, Fodor E, Firth AE, Gog JR, Taubenberger JK, Digard P. 2012. Identification of a novel splice variant form of the influenza A virus M2 ion channel with an antigenically distinct ectodomain. PLoS Pathog 8:e1002998. doi:10.1371/journal.ppat.1002998.
    1. Wise HM, Foeglein A, Sun J, Dalton RM, Patel S, Howard W, Anderson EC, Barclay WS, Digard P. 2009. A complicated message: identification of a novel PB1-related protein translated from influenza A virus segment 2 mRNA. J Virol 83:8021–8031. doi:10.1128/JVI.00826-09.
    1. Jagger BW, Wise HM, Kash JC, Walters KA, Wills NM, Xiao YL, Dunfee RL, Schwartzman LM, Ozinsky A, Bell GL, Dalton RM, Lo A, Efstathiou S, Atkins JF, Firth AE, Taubenberger JK, Digard P. 2012. An overlapping protein-coding region in influenza A virus segment 3 modulates the host response. Science 337:199–204. doi:10.1126/science.1222213.
    1. Chen W, Calvo PA, Malide D, Gibbs J, Schubert U, Bacik I, Basta S, O'Neill R, Schickli J, Palese P, Henklein P, Bennink JR, Yewdell JW. 2001. A novel influenza A virus mitochondrial protein that induces cell death. Nat Med 7:1306–1312. doi:10.1038/nm1201-1306.
    1. Wise HM, Barbezange C, Jagger BW, Dalton RM, Gog JR, Curran MD, Taubenberger JK, Anderson EC, Digard P. 2011. Overlapping signals for translational regulation and packaging of influenza A virus segment 2. Nucleic Acids Res 39:7775–7790. doi:10.1093/nar/gkr487.
    1. Shi M, Jagger BW, Wise HM, Digard P, Holmes EC, Taubenberger JK. 2012. Evolutionary conservation of the PA-X open reading frame in segment 3 of influenza A virus. J Virol 86:12411–12413. doi:10.1128/JVI.01677-12.
    1. Brooke CB, Ince WL, Wrammert J, Ahmed R, Wilson PC, Bennink JR, Yewdell JW. 2013. Most influenza a virions fail to express at least one essential viral protein. J Virol 87:3155–3162. doi:10.1128/JVI.02284-12.
    1. Rossman JS, Jing X, Leser GP, Lamb RA. 2010. Influenza virus M2 protein mediates ESCRT-independent membrane scission. Cell 142:902–913. doi:10.1016/j.cell.2010.08.029.
    1. Pinto LH, Dieckmann GR, Gandhi CS, Papworth CG, Braman J, Shaughnessy MA, Lear JD, Lamb RA, DeGrado WF. 1997. A functionally defined model for the M2 proton channel of influenza A virus suggests a mechanism for its ion selectivity. Proc Natl Acad Sci U S A 94:11301–11306. doi:10.1073/pnas.94.21.11301.
    1. Cao S, Liu X, Yu M, Li J, Jia X, Bi Y, Sun L, Gao GF, Liu W. 2012. A nuclear export signal in the matrix protein of influenza A virus is required for efficient virus replication. J Virol 86:4883–4891. doi:10.1128/JVI.06586-11.
    1. Valcarcel J, Portela A, Ortin J. 1991. Regulated M1 mRNA splicing in influenza virus-infected cells. J Gen Virol 72:1301–1308. doi:10.1099/0022-1317-72-6-1301.
    1. Chua MA, Schmid S, Perez JT, Langlois RA, Tenoever BR. 2013. Influenza A virus utilizes suboptimal splicing to coordinate the timing of infection. Cell Rep 3:23–29. doi:10.1016/j.celrep.2012.12.010.
    1. Shih SR, Krug RM. 1996. Novel exploitation of a nuclear function by influenza virus: the cellular SF2/ASF splicing factor controls the amount of the essential viral M2 ion channel protein in infected cells. EMBO J 15:5415–5427.
    1. Shih SR, Nemeroff ME, Krug RM. 1995. The choice of alternative 5′ splice sites in influenza virus M1 mRNA is regulated by the viral polymerase complex. Proc Natl Acad Sci U S A 92:6324–6328. doi:10.1073/pnas.92.14.6324.
    1. Robb NC, Fodor E. 2012. The accumulation of influenza A virus segment 7 spliced mRNAs is regulated by the NS1 protein. J Gen Virol 93:113–118. doi:10.1099/vir.0.035485-0.
    1. van Wielink R, Harmsen MM, Martens DE, Peeters BP, Wijffels RH, Moormann RJ. 2012. Mutations in the M-gene segment can substantially increase replication efficiency of NS1 deletion influenza A virus in MDCK cells. J Virol 86:12341–12350. doi:10.1128/JVI.01725-12.
    1. Hale BG, Randall RE, Ortin J, Jackson D. 2008. The multifunctional NS1 protein of influenza A viruses. J Gen Virol 89:2359–2376. doi:10.1099/vir.0.2008/004606-0.
    1. Krug RM. 2015. Functions of the influenza A virus NS1 protein in antiviral defense. Curr Opin Virol 12C:1–6.
    1. Garcia-Sastre A, Egorov A, Matassov D, Brandt S, Levy DE, Durbin JE, Palese P, Muster T. 1998. Influenza A virus lacking the NS1 gene replicates in interferon-deficient systems. Virology 252:324–330. doi:10.1006/viro.1998.9508.
    1. Egorov A, Brandt S, Sereinig S, Romanova J, Ferko B, Katinger D, Grassauer A, Alexandrova G, Katinger H, Muster T. 1998. Transfectant influenza A viruses with long deletions in the NS1 protein grow efficiently in Vero cells. J Virol 72:6437–6441.
    1. Mok BW, Song W, Wang P, Tai H, Chen Y, Zheng M, Wen X, Lau SY, Wu WL, Matsumoto K, Yuen KY, Chen H. 2012. The NS1 protein of influenza A virus interacts with cellular processing bodies and stress granules through RNA-associated protein 55 (RAP55) during virus infection. J Virol 86:12695–12707. doi:10.1128/JVI.00647-12.
    1. Wen X, Huang X, Mok BW, Chen Y, Zheng M, Lau SY, Wang P, Song W, Jin DY, Yuen KY, Chen H. 2014. NF90 exerts antiviral activity through regulation of PKR phosphorylation and stress granules in infected cells. J Immunol 192:3753–3764. doi:10.4049/jimmunol.1302813.
    1. Song W, Wang P, Mok BW, Lau SY, Huang X, Wu WL, Zheng M, Wen X, Yang S, Chen Y, Li L, Yuen KY, Chen H. 2014. The K526R substitution in viral protein PB2 enhances the effects of E627K on influenza virus replication. Nat Commun 5:5509. doi:10.1038/ncomms6509.
    1. Hoffmann E, Neumann G, Kawaoka Y, Hobom G, Webster RG. 2000. A DNA transfection system for generation of influenza A virus from eight plasmids. Proc Natl Acad Sci U S A 97:6108–6113. doi:10.1073/pnas.100133697.
    1. Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods 25:402–408. doi:10.1006/meth.2001.1262.
    1. Kistner O, Barrett PN, Mundt W, Reiter M, Schober-Bendixen S, Dorner F. 1998. Development of a mammalian cell (Vero) derived candidate influenza virus vaccine. Vaccine 16:960–968. doi:10.1016/S0264-410X(97)00301-0.
    1. van Wielink R, Harmsen MM, Martens DE, Peeters BP, Wijffels RH, Moormann RJ. 2011. MDCK cell line with inducible allele B NS1 expression propagates delNS1 influenza virus to high titres. Vaccine 29:6976–6985. doi:10.1016/j.vaccine.2011.07.037.
    1. Chiang C, Chen GW, Shih SR. 2008. Mutations at alternative 5′ splice sites of M1 mRNA negatively affect influenza A virus viability and growth rate. J Virol 82:10873–10886. doi:10.1128/JVI.00506-08.
    1. Enami M, Enami K. 2000. Characterization of influenza virus NS1 protein by using a novel helper-virus-free reverse genetic system. J Virol 74:5556–5561. doi:10.1128/JVI.74.12.5556-5561.2000.
    1. Ichinohe T, Pang IK, Iwasaki A. 2010. Influenza virus activates inflammasomes via its intracellular M2 ion channel. Nat Immunol 11:404–410. doi:10.1038/ni.1861.
    1. Gannage M, Dormann D, Albrecht R, Dengjel J, Torossi T, Ramer PC, Lee M, Strowig T, Arrey F, Conenello G, Pypaert M, Andersen J, Garcia-Sastre A, Munz C. 2009. Matrix protein 2 of influenza A virus blocks autophagosome fusion with lysosomes. Cell Host Microbe 6:367–380. doi:10.1016/j.chom.2009.09.005.
    1. Paterson D, Fodor E. 2012. Emerging roles for the influenza A virus nuclear export protein (NEP). PLoS Pathog 8:e1003019. doi:10.1371/journal.ppat.1003019.
    1. Dubois J, Terrier O, Rosa-Calatrava M. 2014. Influenza viruses and mRNA splicing: doing more with less. mBio 5(3):e00070-00014. doi:10.1128/mBio.00070-14.
    1. Manz B, Brunotte L, Reuther P, Schwemmle M. 2012. Adaptive mutations in NEP compensate for defective H5N1 RNA replication in cultured human cells. Nat Commun 3:802. doi:10.1038/ncomms1804.
    1. Nemeroff ME, Barabino SM, Li Y, Keller W, Krug RM. 1998. Influenza virus NS1 protein interacts with the cellular 30 kDa subunit of CPSF and inhibits 3′end formation of cellular pre-mRNAs. Mol Cell 1:991–1000. doi:10.1016/S1097-2765(00)80099-4.
    1. Garaigorta U, Ortin J. 2007. Mutation analysis of a recombinant NS replicon shows that influenza virus NS1 protein blocks the splicing and nucleo-cytoplasmic transport of its own viral mRNA. Nucleic Acids Res 35:4573–4582. doi:10.1093/nar/gkm230.
    1. Salvatore M, Basler CF, Parisien JP, Horvath CM, Bourmakina S, Zheng H, Muster T, Palese P, Garcia-Sastre A. 2002. Effects of influenza A virus NS1 protein on protein expression: the NS1 protein enhances translation and is not required for shutoff of host protein synthesis. J Virol 76:1206–1212. doi:10.1128/JVI.76.3.1206-1212.2002.
    1. Fernandez-Sesma A, Marukian S, Ebersole BJ, Kaminski D, Park MS, Yuen T, Sealfon SC, Garcia-Sastre A, Moran TM. 2006. Influenza virus evades innate and adaptive immunity via the NS1 protein. J Virol 80:6295–6304. doi:10.1128/JVI.02381-05.
    1. Pica N, Langlois RA, Krammer F, Margine I, Palese P. 2012. NS1-truncated live attenuated virus vaccine provides robust protection to aged mice from viral challenge. J Virol 86:10293–10301. doi:10.1128/JVI.01131-12.
    1. Mueller SN, Langley WA, Carnero E, Garcia-Sastre A, Ahmed R. 2010. Immunization with live attenuated influenza viruses that express altered NS1 proteins results in potent and protective memory CD8+ T-cell responses. J Virol 84:1847–1855. doi:10.1128/JVI.01317-09.
    1. Talon J, Salvatore M, O'Neill RE, Nakaya Y, Zheng H, Muster T, Garcia-Sastre A, Palese P. 2000. Influenza A and B viruses expressing altered NS1 proteins: a vaccine approach. Proc Natl Acad Sci U S A 97:4309–4314. doi:10.1073/pnas.070525997.
    1. Richt JA, Garcia-Sastre A. 2009. Attenuated influenza virus vaccines with modified NS1 proteins. Curr Top Microbiol Immunol 333:177–195. doi:10.1007/978-3-540-92165-3_9.
    1. Wacheck V, Egorov A, Groiss F, Pfeiffer A, Fuereder T, Hoeflmayer D, Kundi M, Popow-Kraupp T, Redlberger-Fritz M, Mueller CA, Cinatl J, Michaelis M, Geiler J, Bergmann M, Romanova J, Roethl E, Morokutti A, Wolschek M, Ferko B, Seipelt J, Dick-Gudenus R, Muster T. 2010. A novel type of influenza vaccine: safety and immunogenicity of replication-deficient influenza virus created by deletion of the interferon antagonist NS1. J Infect Dis 201:354–362. doi:10.1086/649428.
    1. Wressnigg N, Voss D, Wolff T, Romanova J, Ruthsatz T, Mayerhofer I, Reiter M, Nakowitsch S, Humer J, Morokutti A, Muster T, Egorov A, Kittel C. 2009. Development of a live-attenuated influenza B DeltaNS1 intranasal vaccine candidate. Vaccine 27:2851–2857. doi:10.1016/j.vaccine.2009.02.087.
    1. Kochs G, Martinez-Sobrido L, Lienenklaus S, Weiss S, Garcia-Sastre A, Staeheli P. 2009. Strong interferon-inducing capacity of a highly virulent variant of influenza A virus strain PR8 with deletions in the NS1 gene. J Gen Virol 90:2990–2994. doi:10.1099/vir.0.015727-0.

Source: PubMed

3
S'abonner