#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Molecularly Cloned, Live-Attenuated Japanese Encephalitis Vaccine SA-14-2 Virus: A Conserved Single Amino Acid in the Hairpin of the Viral E Glycoprotein Determines Neurovirulence in Mice


A group of mosquito-borne flaviviruses that cause fatal encephalitis in humans is among the most important of all emerging human pathogens of global significance. This group includes Japanese encephalitis (JE), West Nile, St. Louis encephalitis, and Murray Valley encephalitis viruses. In this work, we have developed a reverse genetics system for SA14-14-2, a live JE vaccine that is most commonly used in JE-endemic areas, by constructing an infectious bacterial artificial chromosome that contains the full-length SA14-14-2 cDNA. Using this infectious SA14-14-2 cDNA, combined with a mouse model for JEV infection, we have identified a key viral neurovirulence factor, a conserved single amino acid in the ij hairpin adjacent to the fusion loop of the viral E glycoprotein, which regulates viral infectivity into neurons within the central nervous system in vivo and neuronal cells of mouse and human in vitro. Thus, our findings elucidate the molecular basis of the neurovirulence caused by JEV and other closely related encephalitic flaviviruses, a major step in understanding their neuropathogenesis. From a clinical perspective, the discovery of the viral neurovirulence factor and its role will have direct application to the design of a novel class of broad-spectrum antivirals to treat and prevent infection of JEV and other taxonomically related neurotropic flaviviruses.


Vyšlo v časopise: A Molecularly Cloned, Live-Attenuated Japanese Encephalitis Vaccine SA-14-2 Virus: A Conserved Single Amino Acid in the Hairpin of the Viral E Glycoprotein Determines Neurovirulence in Mice. PLoS Pathog 10(7): e32767. doi:10.1371/journal.ppat.1004290
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004290

Souhrn

A group of mosquito-borne flaviviruses that cause fatal encephalitis in humans is among the most important of all emerging human pathogens of global significance. This group includes Japanese encephalitis (JE), West Nile, St. Louis encephalitis, and Murray Valley encephalitis viruses. In this work, we have developed a reverse genetics system for SA14-14-2, a live JE vaccine that is most commonly used in JE-endemic areas, by constructing an infectious bacterial artificial chromosome that contains the full-length SA14-14-2 cDNA. Using this infectious SA14-14-2 cDNA, combined with a mouse model for JEV infection, we have identified a key viral neurovirulence factor, a conserved single amino acid in the ij hairpin adjacent to the fusion loop of the viral E glycoprotein, which regulates viral infectivity into neurons within the central nervous system in vivo and neuronal cells of mouse and human in vitro. Thus, our findings elucidate the molecular basis of the neurovirulence caused by JEV and other closely related encephalitic flaviviruses, a major step in understanding their neuropathogenesis. From a clinical perspective, the discovery of the viral neurovirulence factor and its role will have direct application to the design of a novel class of broad-spectrum antivirals to treat and prevent infection of JEV and other taxonomically related neurotropic flaviviruses.


Zdroje

1. EndyTP, NisalakA (2002) Japanese encephalitis virus: ecology and epidemiology. Curr Top Microbiol Immunol 267: 11–48.

2. MackenzieJS, BarrettAD, DeubelV (2002) The Japanese encephalitis serological group of flaviviruses: a brief introduction to the group. Curr Top Microbiol Immunol 267: 1–10.

3. MackenzieJS, GublerDJ, PetersenLR (2004) Emerging flaviviruses: the spread and resurgence of Japanese encephalitis, West Nile and dengue viruses. Nat Med 10: S98–109.

4. MackenzieJS, JohansenCA, RitchieSA, van den HurkAF, HallRA (2002) Japanese encephalitis as an emerging virus: the emergence and spread of Japanese encephalitis virus in Australasia. Curr Top Microbiol Immunol 267: 49–73.

5. GublerDJ (2007) The continuing spread of West Nile virus in the western hemisphere. Clin Infect Dis 45: 1039–1046.

6. NashD, MostashariF, FineA, MillerJ, O'LearyD, et al. (2001) The outbreak of West Nile virus infection in the New York City area in 1999. N Engl J Med 344: 1807–1814.

7. WeaverSC, BarrettAD (2004) Transmission cycles, host range, evolution and emergence of arboviral disease. Nat Rev Microbiol 2: 789–801.

8. NettRJ, CampbellGL, ReisenWK (2009) Potential for the emergence of Japanese encephalitis virus in California. Vector Borne Zoonotic Dis 9: 511–517.

9. WeaverSC, ReisenWK (2010) Present and future arboviral threats. Antiviral Res 85: 328–345.

10. Campbell GL, Hills SL, Fischer M, Jacobson JA, Hoke CH, et al.. (2011) Estimated global incidence of Japanese encephalitis: a systematic review. Bull World Health Organ 89: : 766–774, 774A–774E.

11. MonathTP (2002) Japanese encephalitis vaccines: current vaccines and future prospects. Curr Top Microbiol Immunol 267: 105–138.

12. SolomonT, VaughnDW (2002) Pathogenesis and clinical features of Japanese encephalitis and West Nile virus infections. Curr Top Microbiol Immunol 267: 171–194.

13. SolomonT (2006) Control of Japanese encephalitis-within our grasp? N Engl J Med 355: 869–871.

14. MisraUK, KalitaJ (2010) Overview: Japanese encephalitis. Prog Neurobiol 91: 108–120.

15. DoklandT, WalshM, MackenzieJM, KhromykhAA, EeKH, et al. (2004) West Nile virus core protein: tetramer structure and ribbon formation. Structure 12: 1157–1163.

16. MaL, JonesCT, GroeschTD, KuhnRJ, PostCB (2004) Solution structure of dengue virus capsid protein reveals another fold. Proc Natl Acad Sci U S A 101: 3414–3419.

17. KuhnRJ, ZhangW, RossmannMG, PletnevSV, CorverJ, et al. (2002) Structure of dengue virus: implications for flavivirus organization, maturation, and fusion. Cell 108: 717–725.

18. MukhopadhyayS, KimBS, ChipmanPR, RossmannMG, KuhnRJ (2003) Structure of West Nile virus. Science 302: 248.

19. ZhangW, ChipmanPR, CorverJ, JohnsonPR, ZhangY, et al. (2003) Visualization of membrane protein domains by cryo-electron microscopy of dengue virus. Nat Struct Biol 10: 907–912.

20. ChenY, MaguireT, HilemanRE, FrommJR, EskoJD, et al. (1997) Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Nat Med 3: 866–871.

21. DavisCW, NguyenHY, HannaSL, SanchezMD, DomsRW, et al. (2006) West Nile virus discriminates between DC-SIGN and DC-SIGNR for cellular attachment and infection. J Virol 80: 1290–1301.

22. Navarro-SanchezE, AltmeyerR, AmaraA, SchwartzO, FieschiF, et al. (2003) Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses. EMBO Rep 4: 723–728.

23. PokidyshevaE, ZhangY, BattistiAJ, Bator-KellyCM, ChipmanPR, et al. (2006) Cryo-EM reconstruction of dengue virus in complex with the carbohydrate recognition domain of DC-SIGN. Cell 124: 485–493.

24. TassaneetrithepB, BurgessTH, Granelli-PipernoA, TrumpfhellerC, FinkeJ, et al. (2003) DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med 197: 823–829.

25. ChuJJ, NgML (2004) Infectious entry of West Nile virus occurs through a clathrin-mediated endocytic pathway. J Virol 78: 10543–10555.

26. van der SchaarHM, RustMJ, WaartsBL, van der Ende-MetselaarH, KuhnRJ, et al. (2007) Characterization of the early events in dengue virus cell entry by biochemical assays and single-virus tracking. J Virol 81: 12019–12028.

27. van der SchaarHM, RustMJ, ChenC, van der Ende-MetselaarH, WilschutJ, et al. (2008) Dissecting the cell entry pathway of dengue virus by single-particle tracking in living cells. PLoS Pathog 4: e1000244.

28. BressanelliS, StiasnyK, AllisonSL, SturaEA, DuquerroyS, et al. (2004) Structure of a flavivirus envelope glycoprotein in its low-pH-induced membrane fusion conformation. EMBO J 23: 728–738.

29. LiaoM, Sanchez-San MartinC, ZhengA, KielianM (2010) In vitro reconstitution reveals key intermediate states of trimer formation by the dengue virus membrane fusion protein. J Virol 84: 5730–5740.

30. ModisY, OgataS, ClementsD, HarrisonSC (2004) Structure of the dengue virus envelope protein after membrane fusion. Nature 427: 313–319.

31. NayakV, DessauM, KuceraK, AnthonyK, LedizetM, et al. (2009) Crystal structure of dengue virus type 1 envelope protein in the postfusion conformation and its implications for membrane fusion. J Virol 83: 4338–4344.

32. StiasnyK, KosslC, LepaultJ, ReyFA, HeinzFX (2007) Characterization of a structural intermediate of flavivirus membrane fusion. PLoS Pathog 3: e20.

33. HarrisonSC (2008) Viral membrane fusion. Nat Struct Mol Biol 15: 690–698.

34. Lindenbach BD, Thiel HJ, Rice CM (2007) Flaviviridae: the viruses and their replication. In: Knipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA et al.., editors. Fields virology. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins Publishers. pp. 1101–1152.

35. BrintonMA (2002) The molecular biology of West Nile Virus: a new invader of the western hemisphere. Annu Rev Microbiol 56: 371–402.

36. ParanjapeSM, HarrisE (2010) Control of dengue virus translation and replication. Curr Top Microbiol Immunol 338: 15–34.

37. VillordoSM, GamarnikAV (2009) Genome cyclization as strategy for flavivirus RNA replication. Virus Res 139: 230–239.

38. WestawayEG, MackenzieJM, KhromykhAA (2002) Replication and gene function in Kunjin virus. Curr Top Microbiol Immunol 267: 323–351.

39. GillespieLK, HoenenA, MorganG, MackenzieJM (2010) The endoplasmic reticulum provides the membrane platform for biogenesis of the flavivirus replication complex. J Virol 84: 10438–10447.

40. HsuNY, IlnytskaO, BelovG, SantianaM, ChenYH, et al. (2010) Viral reorganization of the secretory pathway generates distinct organelles for RNA replication. Cell 141: 799–811.

41. WelschS, MillerS, Romero-BreyI, MerzA, BleckCK, et al. (2009) Composition and three-dimensional architecture of the dengue virus replication and assembly sites. Cell Host Microbe 5: 365–375.

42. LiL, LokSM, YuIM, ZhangY, KuhnRJ, et al. (2008) The flavivirus precursor membrane-envelope protein complex: structure and maturation. Science 319: 1830–1834.

43. ZhangY, CorverJ, ChipmanPR, ZhangW, PletnevSV, et al. (2003) Structures of immature flavivirus particles. EMBO J 22: 2604–2613.

44. LorenzIC, AllisonSL, HeinzFX, HeleniusA (2002) Folding and dimerization of tick-borne encephalitis virus envelope proteins prM and E in the endoplasmic reticulum. J Virol 76: 5480–5491.

45. StadlerK, AllisonSL, SchalichJ, HeinzFX (1997) Proteolytic activation of tick-borne encephalitis virus by furin. J Virol 71: 8475–8481.

46. YuIM, ZhangW, HoldawayHA, LiL, KostyuchenkoVA, et al. (2008) Structure of the immature dengue virus at low pH primes proteolytic maturation. Science 319: 1834–1837.

47. ZhangY, ZhangW, OgataS, ClementsD, StraussJH, et al. (2004) Conformational changes of the flavivirus E glycoprotein. Structure 12: 1607–1618.

48. Gubler DJ, Kuno G, Markoff L (2007) Flaviviruses. In: Knipe DM, Howley PM, Griffin DE, Lamb RA, Martin MA et al.., editors. Fields virology. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins Publishers. pp. 1153–1252.

49. BeasleyDW, LewthwaiteP, SolomonT (2008) Current use and development of vaccines for Japanese encephalitis. Expert Opin Biol Ther 8: 95–106.

50. Wilder-SmithA, HalsteadSB (2010) Japanese encephalitis: update on vaccines and vaccine recommendations. Curr Opin Infect Dis 23: 426–431.

51. FischerM, LindseyN, StaplesJE, HillsS (2010) Centers for Disease Control and Prevention (2010) Japanese encephalitis vaccines: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 59: 1–27.

52. PlesnerAM (2003) Allergic reactions to Japanese encephalitis vaccine. Immunol Allergy Clin North Am 23: 665–697.

53. YuY (2010) Phenotypic and genotypic characteristics of Japanese encephalitis attenuated live vaccine virus SA14-14-2 and their stabilities. Vaccine 28: 3635–3641.

54. HalsteadSB, ThomasSJ (2010) Japanese encephalitis: new options for active immunization. Clin Infect Dis 50: 1155–1164.

55. JelinekT (2009) Ixiaro: a new vaccine against Japanese encephalitis. Expert Rev Vaccines 8: 1501–1511.

56. KollaritschH, Paulke-KorinekM, Dubischar-KastnerK (2009) IC51 Japanese encephalitis vaccine. Expert Opin Biol Ther 9: 921–931.

57. Centers for Disease Control and Prevention (2011) Recommendations for use of a booster dose of inactivated vero cell culture-derived Japanese encephalitis vaccine: advisory committee on immunization practices, 2011. MMWR Morb Mortal Wkly Rep 60: 661–663.

58. Centers for Disease Control and Prevention (2011) Update on Japanese encephalitis vaccine for children: United States, May 2011. MMWR Morb Mortal Wkly Rep 60: 664–665.

59. ChambersTJ, NestorowiczA, MasonPW, RiceCM (1999) Yellow fever/Japanese encephalitis chimeric viruses: construction and biological properties. J Virol 73: 3095–3101.

60. MonathTP, LevenbookI, SoikeK, ZhangZX, RatterreeM, et al. (2000) Chimeric yellow fever virus 17D-Japanese encephalitis virus vaccine: dose-response effectiveness and extended safety testing in rhesus monkeys. J Virol 74: 1742–1751.

61. HalsteadSB, ThomasSJ (2011) New Japanese encephalitis vaccines: alternatives to production in mouse brain. Expert Rev Vaccines 10: 355–364.

62. AppaiahgariMB, VratiS (2012) Clinical development of IMOJEV (R)-a recombinant Japanese encephalitis chimeric vaccine (JE-CV). Expert Opin Biol Ther 12: 1251–1263.

63. AiharaS, RaoCM, YuYX, LeeT, WatanabeK, et al. (1991) Identification of mutations that occurred on the genome of Japanese encephalitis virus during the attenuation process. Virus Genes 5: 95–109.

64. NiH, ChangGJ, XieH, TrentDW, BarrettAD (1995) Molecular basis of attenuation of neurovirulence of wild-type Japanese encephalitis virus strain SA14. J Gen Virol 76: 409–413.

65. NitayaphanS, GrantJA, ChangGJ, TrentDW (1990) Nucleotide sequence of the virulent SA-14 strain of Japanese encephalitis virus and its attenuated vaccine derivative, SA-14-14-2. Virology 177: 541–552.

66. CeciliaD, GouldEA (1991) Nucleotide changes responsible for loss of neuroinvasiveness in Japanese encephalitis virus neutralization-resistant mutants. Virology 181: 70–77.

67. ChambersTJ, DrollDA, JiangX, WoldWS, NickellsJA (2007) JE Nakayama/JE SA14-14-2 virus structural region intertypic viruses: biological properties in the mouse model of neuroinvasive disease. Virology 366: 51–61.

68. HasegawaH, YoshidaM, ShiosakaT, FujitaS, KobayashiY (1992) Mutations in the envelope protein of Japanese encephalitis virus affect entry into cultured cells and virulence in mice. Virology 191: 158–165.

69. LeeE, HallRA, LobigsM (2004) Common E protein determinants for attenuation of glycosaminoglycan-binding variants of Japanese encephalitis and West Nile viruses. J Virol 78: 8271–8280.

70. NiH, BarrettAD (1996) Molecular differences between wild-type Japanese encephalitis virus strains of high and low mouse neuroinvasiveness. J Gen Virol 77: 1449–1455.

71. NiH, BarrettAD (1998) Attenuation of Japanese encephalitis virus by selection of its mouse brain membrane receptor preparation escape variants. Virology 241: 30–36.

72. ShahPS, TanakaM, KhanAH, MathengeEG, FukeI, et al. (2006) Molecular characterization of attenuated Japanese encephalitis live vaccine strain ML-17. Vaccine 24: 402–411.

73. SumiyoshiH, TignorGH, ShopeRE (1995) Characterization of a highly attenuated Japanese encephalitis virus generated from molecularly cloned cDNA. J Infect Dis 171: 1144–1151.

74. WuSC, LinCW, LeeSC, LianWC (2003) Phenotypic and genotypic characterization of the neurovirulence and neuroinvasiveness of a large-plaque attenuated Japanese encephalitis virus isolate. Microbes Infect 5: 475–480.

75. KimJM, YunSI, SongBH, HahnYS, LeeCH, et al. (2008) A single N-linked glycosylation site in the Japanese encephalitis virus prM protein is critical for cell type-specific prM protein biogenesis, virus particle release, and pathogenicity in mice. J Virol 82: 7846–7862.

76. YunSI, ChoiYJ, SongBH, LeeYM (2009) 3' cis-acting elements that contribute to the competence and efficiency of Japanese encephalitis virus genome replication: functional importance of sequence duplications, deletions, and substitutions. J Virol 83: 7909–7930.

77. SongBH, YunGN, KimJK, YunSI, LeeYM (2012) Biological and genetic properties of SA14-14-2, a live-attenuated Japanese encephalitis vaccine that is currently available for humans. J Microbiol 50: 698–706.

78. YunSI, KimSY, RiceCM, LeeYM (2003) Development and application of a reverse genetics system for Japanese encephalitis virus. J Virol 77: 6450–6465.

79. NybakkenGE, NelsonCA, ChenBR, DiamondMS, FremontDH (2006) Crystal structure of the West Nile virus envelope glycoprotein. J Virol 80: 11467–11474.

80. LucaVC, AbiMansourJ, NelsonCA, FremontDH (2012) Crystal structure of the Japanese encephalitis virus envelope protein. J Virol 86: 2337–2346.

81. NiH, BurnsNJ, ChangGJ, ZhangMJ, WillsMR, et al. (1994) Comparison of nucleotide and deduced amino acid sequence of the 5' non-coding region and structural protein genes of the wild-type Japanese encephalitis virus strain SA14 and its attenuated vaccine derivatives. J Gen Virol 75: 1505–1510.

82. World Health Organization (2005) Global advisory committee on vaccine safety, 9–10 June 2005. Wkly Epidemiol Rec 80: 242–247.

83. MukhopadhyayS, KuhnRJ, RossmannMG (2005) A structural perspective of the flavivirus life cycle. Nat Rev Microbiol 3: 13–22.

84. StiasnyK, HeinzFX (2006) Flavivirus membrane fusion. J Gen Virol 87: 2755–2766.

85. ReyFA, HeinzFX, MandlC, KunzC, HarrisonSC (1995) The envelope glycoprotein from tick-borne encephalitis virus at 2 A resolution. Nature 375: 291–298.

86. AllisonSL, SchalichJ, StiasnyK, MandlCW, HeinzFX (2001) Mutational evidence for an internal fusion peptide in flavivirus envelope protein E. J Virol 75: 4268–4275.

87. LeeE, LobigsM (2002) Mechanism of virulence attenuation of glycosaminoglycan-binding variants of Japanese encephalitis virus and Murray Valley encephalitis virus. J Virol 76: 4901–4911.

88. LeeJW, ChuJJ, NgML (2006) Quantifying the specific binding between West Nile virus envelope domain III protein and the cellular receptor αVβ3 integrin. J Biol Chem 281: 1352–1360.

89. BeasleyDW, BarrettAD (2002) Identification of neutralizing epitopes within structural domain III of the West Nile virus envelope protein. J Virol 76: 13097–13100.

90. CrillWD, RoehrigJT (2001) Monoclonal antibodies that bind to domain III of dengue virus E glycoprotein are the most efficient blockers of virus adsorption to Vero cells. J Virol 75: 7769–7773.

91. KaufmannB, NybakkenGE, ChipmanPR, ZhangW, DiamondMS, et al. (2006) West Nile virus in complex with the Fab fragment of a neutralizing monoclonal antibody. Proc Natl Acad Sci U S A 103: 12400–12404.

92. WuKP, WuCW, TsaoYP, KuoTW, LouYC, et al. (2003) Structural basis of a flavivirus recognized by its neutralizing antibody: solution structure of the domain III of the Japanese encephalitis virus envelope protein. J Biol Chem 278: 46007–46013.

93. StiasnyK, BressanelliS, LepaultJ, ReyFA, HeinzFX (2004) Characterization of a membrane-associated trimeric low-pH-induced form of the class II viral fusion protein E from tick-borne encephalitis virus and its crystallization. J Virol 78: 3178–3183.

94. ReyFA (2003) Dengue virus envelope glycoprotein structure: new insight into its interactions during viral entry. Proc Natl Acad Sci U S A 100: 6899–6901.

95. KaufmannB, ChipmanPR, HoldawayHA, JohnsonS, FremontDH, et al. (2009) Capturing a flavivirus pre-fusion intermediate. PLoS Pathog 5: e1000672.

96. LinSR, ZouG, HsiehSC, QingM, TsaiWY, et al. (2011) The helical domains of the stem region of dengue virus envelope protein are involved in both virus assembly and entry. J Virol 85: 5159–5171.

97. PangerlK, HeinzFX, StiasnyK (2011) Mutational analysis of the zippering reaction during flavivirus membrane fusion. J Virol 85: 8495–8501.

98. SchmidtAG, YangPL, HarrisonSC (2010) Peptide inhibitors of dengue virus entry target a late-stage fusion intermediate. PLoS Pathog 6: e1000851.

99. ChuJJ, NgML (2004) Interaction of West Nile virus with αvβ3 integrin mediates virus entry into cells. J Biol Chem 279: 54533–54541.

100. HurrelbrinkRJ, McMinnPC (2001) Attenuation of Murray Valley encephalitis virus by site-directed mutagenesis of the hinge and putative receptor-binding regions of the envelope protein. J Virol 75: 7692–7702.

101. LeeE, LobigsM (2000) Substitutions at the putative receptor-binding site of an encephalitic flavivirus alter virulence and host cell tropism and reveal a role for glycosaminoglycans in entry. J Virol 74: 8867–8875.

102. GotoA, YoshiiK, ObaraM, UekiT, MizutaniT, et al. (2005) Role of the N-linked glycans of the prM and E envelope proteins in tick-borne encephalitis virus particle secretion. Vaccine 23: 3043–3052.

103. HannaSL, PiersonTC, SanchezMD, AhmedAA, MurtadhaMM, et al. (2005) N-linked glycosylation of West Nile virus envelope proteins influences particle assembly and infectivity. J Virol 79: 13262–13274.

104. LorenzIC, KartenbeckJ, MezzacasaA, AllisonSL, HeinzFX, et al. (2003) Intracellular assembly and secretion of recombinant subviral particles from tick-borne encephalitis virus. J Virol 77: 4370–4382.

105. TajimaS, NeromeR, NukuiY, KatoF, TakasakiT, et al. (2010) A single mutation in the Japanese encephalitis virus E protein (S123R) increases its growth rate in mouse neuroblastoma cells and its pathogenicity in mice. Virology 396: 298–304.

106. MonathTP, ArroyoJ, LevenbookI, ZhangZX, CatalanJ, et al. (2002) Single mutation in the flavivirus envelope protein hinge region increases neurovirulence for mice and monkeys but decreases viscerotropism for monkeys: relevance to development and safety testing of live, attenuated vaccines. J Virol 76: 1932–1943.

107. ArroyoJ, GuirakhooF, FennerS, ZhangZX, MonathTP, et al. (2001) Molecular basis for attenuation of neurovirulence of a yellow fever virus/Japanese encephalitis virus chimera vaccine (ChimeriVax-JE). J Virol 75: 934–942.

108. ChenLK, LinYL, LiaoCL, LinCG, HuangYL, et al. (1996) Generation and characterization of organ-tropism mutants of Japanese encephalitis virus in vivo and in vitro. Virology 223: 79–88.

109. ZhaoZ, DateT, LiY, KatoT, MiyamotoM, et al. (2005) Characterization of the E-138 (Glu/Lys) mutation in Japanese encephalitis virus by using a stable, full-length, infectious cDNA clone. J Gen Virol 86: 2209–2220.

110. MelianEB, HinzmanE, NagasakiT, FirthAE, WillsNM, et al. (2010) NS1' of flaviviruses in the Japanese encephalitis virus serogroup is a product of ribosomal frameshifting and plays a role in viral neuroinvasiveness. J Virol 84: 1641–1647.

111. FirthAE, AtkinsJF (2009) A conserved predicted pseudoknot in the NS2A-encoding sequence of West Nile and Japanese encephalitis flaviviruses suggests NS1' may derive from ribosomal frameshifting. Virol J 6: 14.

112. YeQ, LiXF, ZhaoH, LiSH, DengYQ, et al. (2012) A single nucleotide mutation in NS2A of Japanese encephalitis-live vaccine virus (SA14-14-2) ablates NS1' formation and contributes to attenuation. J Gen Virol 93: 1959–1964.

113. EckelsKH, YuYX, DuboisDR, MarchetteNJ, TrentDW, et al. (1988) Japanese encephalitis virus live-attenuated vaccine, Chinese strain SA14-14-2; adaptation to primary canine kidney cell cultures and preparation of a vaccine for human use. Vaccine 6: 513–518.

114. SigelMM (1952) Influence of age on susceptibility to virus infections with particular reference to laboratory animals. Annu Rev Microbiol 6: 247–280.

115. Monath TP (1986) Pathobiology of the flaviviruses. In: Schlesinger S, Schlesinger MJ, editors. The Togaviridae and Flaviviridae. New York, NY: Plenum. pp. 375–440.

116. ThompsonJD, GibsonTJ, PlewniakF, JeanmouginF, HigginsDG (1997) The CLUSTAL X windows interface: flexible strategies for multiple sequence alignment aided by quality analysis tools. Nucleic Acids Res 25: 4876–4882.

117. GoujonM, McWilliamH, LiW, ValentinF, SquizzatoS, et al. (2010) A new bioinformatics analysis tools framework at EMBL-EBI. Nucleic Acids Res 38: W695–699.

118. KieferF, ArnoldK, KunzliM, BordoliL, SchwedeT (2009) The SWISS-MODEL Repository and associated resources. Nucleic Acids Res 37: D387–392.

119. ReedLJ, MuenchH (1938) A simple method of estimating fifty percent endpoints. Am J Hyg 27: 493–497.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 7
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#