#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Extreme Genetic Fragility of the HIV-1 Capsid


Genetic robustness, or fragility, is defined as the ability, or lack thereof, of a biological entity to maintain function in the face of mutations. Viruses that replicate via RNA intermediates exhibit high mutation rates, and robustness should be particularly advantageous to them. The capsid (CA) domain of the HIV-1 Gag protein is under strong pressure to conserve functional roles in viral assembly, maturation, uncoating, and nuclear import. However, CA is also under strong immunological pressure to diversify. Therefore, it would be particularly advantageous for CA to evolve genetic robustness. To measure the genetic robustness of HIV-1 CA, we generated a library of single amino acid substitution mutants, encompassing almost half the residues in CA. Strikingly, we found HIV-1 CA to be the most genetically fragile protein that has been analyzed using such an approach, with 70% of mutations yielding replication-defective viruses. Although CA participates in several steps in HIV-1 replication, analysis of conditionally (temperature sensitive) and constitutively non-viable mutants revealed that the biological basis for its genetic fragility was primarily the need to coordinate the accurate and efficient assembly of mature virions. All mutations that exist in naturally occurring HIV-1 subtype B populations at a frequency >3%, and were also present in the mutant library, had fitness levels that were >40% of WT. However, a substantial fraction of mutations with high fitness did not occur in natural populations, suggesting another form of selection pressure limiting variation in vivo. Additionally, known protective CTL epitopes occurred preferentially in domains of the HIV-1 CA that were even more genetically fragile than HIV-1 CA as a whole. The extreme genetic fragility of HIV-1 CA may be one reason why cell-mediated immune responses to Gag correlate with better prognosis in HIV-1 infection, and suggests that CA is a good target for therapy and vaccination strategies.


Vyšlo v časopise: Extreme Genetic Fragility of the HIV-1 Capsid. PLoS Pathog 9(6): e32767. doi:10.1371/journal.ppat.1003461
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003461

Souhrn

Genetic robustness, or fragility, is defined as the ability, or lack thereof, of a biological entity to maintain function in the face of mutations. Viruses that replicate via RNA intermediates exhibit high mutation rates, and robustness should be particularly advantageous to them. The capsid (CA) domain of the HIV-1 Gag protein is under strong pressure to conserve functional roles in viral assembly, maturation, uncoating, and nuclear import. However, CA is also under strong immunological pressure to diversify. Therefore, it would be particularly advantageous for CA to evolve genetic robustness. To measure the genetic robustness of HIV-1 CA, we generated a library of single amino acid substitution mutants, encompassing almost half the residues in CA. Strikingly, we found HIV-1 CA to be the most genetically fragile protein that has been analyzed using such an approach, with 70% of mutations yielding replication-defective viruses. Although CA participates in several steps in HIV-1 replication, analysis of conditionally (temperature sensitive) and constitutively non-viable mutants revealed that the biological basis for its genetic fragility was primarily the need to coordinate the accurate and efficient assembly of mature virions. All mutations that exist in naturally occurring HIV-1 subtype B populations at a frequency >3%, and were also present in the mutant library, had fitness levels that were >40% of WT. However, a substantial fraction of mutations with high fitness did not occur in natural populations, suggesting another form of selection pressure limiting variation in vivo. Additionally, known protective CTL epitopes occurred preferentially in domains of the HIV-1 CA that were even more genetically fragile than HIV-1 CA as a whole. The extreme genetic fragility of HIV-1 CA may be one reason why cell-mediated immune responses to Gag correlate with better prognosis in HIV-1 infection, and suggests that CA is a good target for therapy and vaccination strategies.


Zdroje

1. de VisserJA, HermissonJ, WagnerGP, Ancel MeyersL, Bagheri-ChaichianH, et al. (2003) Perspective: Evolution and detection of genetic robustness. Evolution 57: 1959–1972.

2. WagnerA (2005) Robustness, evolvability, and neutrality. FEBS Lett 579: 1772–1778.

3. ElenaSF (2012) RNA virus genetic robustness: possible causes and some consequences. Curr Opin Virol 2: 525–530.

4. MontvilleR, FroissartR, RemoldSK, TenaillonO, TurnerPE (2005) Evolution of mutational robustness in an RNA virus. PLoS Biol 3: e381.

5. CodonerFM, DarosJA, SoleRV, ElenaSF (2006) The fittest versus the flattest: experimental confirmation of the quasispecies effect with subviral pathogens. PLoS Pathog 2: e136.

6. SanjuanR, CuevasJM, FurioV, HolmesEC, MoyaA (2007) Selection for robustness in mutagenized RNA viruses. PLoS Genet 3: e93.

7. Domingo-CalapP, CuevasJM, SanjuanR (2009) The fitness effects of random mutations in single-stranded DNA and RNA bacteriophages. PLoS Genet 5: e1000742.

8. SanjuanR, MoyaA, ElenaSF (2004) The distribution of fitness effects caused by single-nucleotide substitutions in an RNA virus. Proc Natl Acad Sci U S A 101: 8396–8401.

9. CarrascoP, de la IglesiaF, ElenaSF (2007) Distribution of fitness and virulence effects caused by single-nucleotide substitutions in Tobacco Etch virus. J Virol 81: 12979–12984.

10. BieniaszPD (2009) The cell biology of HIV-1 virion genesis. Cell Host Microbe 5: 550–558.

11. FreedEO (1998) HIV-1 gag proteins: diverse functions in the virus life cycle. Virology 251: 1–15.

12. Ganser-PornillosBK, YeagerM, SundquistWI (2008) The structural biology of HIV assembly. Curr Opin Struct Biol 18: 203–217.

13. SundquistWI, KrausslichHG (2012) HIV-1 Assembly, Budding, and Maturation. Cold Spring Harb Perspect Med 2: a006924.

14. BriggsJA, KrausslichHG (2011) The molecular architecture of HIV. J Mol Biol 410: 491–500.

15. BriggsJA, RichesJD, GlassB, BartonovaV, ZanettiG, et al. (2009) Structure and assembly of immature HIV. Proc Natl Acad Sci U S A 106: 11090–11095.

16. GanserBK, LiS, KlishkoVY, FinchJT, SundquistWI (1999) Assembly and analysis of conical models for the HIV-1 core. Science 283: 80–83.

17. Ganser-PornillosBK, von SchwedlerUK, StrayKM, AikenC, SundquistWI (2004) Assembly properties of the human immunodeficiency virus type 1 CA protein. J Virol 78: 2545–2552.

18. GambleTR, YooS, VajdosFF, von SchwedlerUK, WorthylakeDK, et al. (1997) Structure of the carboxyl-terminal dimerization domain of the HIV-1 capsid protein. Science 278: 849–853.

19. GambleTR, VajdosFF, YooS, WorthylakeDK, HouseweartM, et al. (1996) Crystal structure of human cyclophilin A bound to the amino-terminal domain of HIV-1 capsid. Cell 87: 1285–1294.

20. GittiRK, LeeBM, WalkerJ, SummersMF, YooS, et al. (1996) Structure of the amino-terminal core domain of the HIV-1 capsid protein. Science 273: 231–235.

21. ChangYF, WangSM, HuangKJ, WangCT (2007) Mutations in capsid major homology region affect assembly and membrane affinity of HIV-1 Gag. J Mol Biol 370: 585–597.

22. ZimmermanC, KleinKC, KiserPK, SinghAR, FiresteinBL, et al. (2002) Identification of a host protein essential for assembly of immature HIV-1 capsids. Nature 415: 88–92.

23. LubanJ, BossoltKL, FrankeEK, KalpanaGV, GoffSP (1993) Human immunodeficiency virus type 1 Gag protein binds to cyclophilins A and B. Cell 73: 1067–1078.

24. PornillosO, Ganser-PornillosBK, YeagerM (2011) Atomic-level modelling of the HIV capsid. Nature 469: 424–427.

25. LiS, HillCP, SundquistWI, FinchJT (2000) Image reconstructions of helical assemblies of the HIV-1 CA protein. Nature 407: 409–413.

26. PornillosO, Ganser-PornillosBK, KellyBN, HuaY, WhitbyFG, et al. (2009) X-ray structures of the hexameric building block of the HIV capsid. Cell 137: 1282–1292.

27. LanmanJ, LamTT, BarnesS, SakalianM, EmmettMR, et al. (2003) Identification of novel interactions in HIV-1 capsid protein assembly by high-resolution mass spectrometry. J Mol Biol 325: 759–772.

28. LanmanJ, LamTT, EmmettMR, MarshallAG, SakalianM, et al. (2004) Key interactions in HIV-1 maturation identified by hydrogen-deuterium exchange. Nat Struct Mol Biol 11: 676–677.

29. MateuMG (2009) The capsid protein of human immunodeficiency virus: intersubunit interactions during virus assembly. FEBS J 276: 6098–6109.

30. YamashitaM, PerezO, HopeTJ, EmermanM (2007) Evidence for direct involvement of the capsid protein in HIV infection of nondividing cells. PLoS Pathog 3: 1502–1510.

31. YamashitaM, EmermanM (2004) Capsid is a dominant determinant of retrovirus infectivity in nondividing cells. J Virol 78: 5670–5678.

32. KrishnanL, MatreyekKA, OztopI, LeeK, TipperCH, et al. (2010) The requirement for cellular transportin 3 (TNPO3 or TRN-SR2) during infection maps to human immunodeficiency virus type 1 capsid and not integrase. J Virol 84: 397–406.

33. MatreyekKA, EngelmanA (2011) The requirement for nucleoporin NUP153 during human immunodeficiency virus type 1 infection is determined by the viral capsid. J Virol 85: 7818–7827.

34. LeeK, MulkyA, YuenW, MartinTD, MeyersonNR, et al. (2012) HIV-1 capsid-targeting domain of cleavage and polyadenylation specificity factor 6. J Virol 86: 3851–3860.

35. LeeK, AmbroseZ, MartinTD, OztopI, MulkyA, et al. (2010) Flexible use of nuclear import pathways by HIV-1. Cell Host Microbe 7: 221–233.

36. PriceAJ, FletcherAJ, SchallerT, ElliottT, LeeK, et al. (2012) CPSF6 defines a conserved capsid interface that modulates HIV-1 replication. PLoS Pathog 8: e1002896.

37. SchallerT, OcwiejaKE, RasaiyaahJ, PriceAJ, BradyTL, et al. (2011) HIV-1 capsid-cyclophilin interactions determine nuclear import pathway, integration targeting and replication efficiency. PLoS Pathog 7: e1002439.

38. HatziioannouT, Perez-CaballeroD, CowanS, BieniaszPD (2005) Cyclophilin interactions with incoming human immunodeficiency virus type 1 capsids with opposing effects on infectivity in human cells. J Virol 79: 176–183.

39. SokolskajaE, SayahDM, LubanJ (2004) Target cell cyclophilin A modulates human immunodeficiency virus type 1 infectivity. J Virol 78: 12800–12808.

40. QiM, YangR, AikenC (2008) Cyclophilin A-dependent restriction of human immunodeficiency virus type 1 capsid mutants for infection of nondividing cells. J Virol 82: 12001–12008.

41. StremlauM, OwensCM, PerronMJ, KiesslingM, AutissierP, et al. (2004) The cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World monkeys. Nature 427: 848–853.

42. ManelN, HogstadB, WangY, LevyDE, UnutmazD, et al. (2010) A cryptic sensor for HIV-1 activates antiviral innate immunity in dendritic cells. Nature 467: 214–217.

43. CrawfordH, MatthewsPC, SchaeferM, CarlsonJM, LeslieA, et al. (2011) The hypervariable HIV-1 capsid protein residues comprise HLA-driven CD8+ T-cell escape mutations and covarying HLA-independent polymorphisms. J Virol 85: 1384–1390.

44. DahirelV, ShekharK, PereyraF, MiuraT, ArtyomovM, et al. (2011) Coordinate linkage of HIV evolution reveals regions of immunological vulnerability. Proc Natl Acad Sci U S A 108: 11530–11535.

45. BrockmanMA, BrummeZL, BrummeCJ, MiuraT, SelaJ, et al. (2010) Early selection in Gag by protective HLA alleles contributes to reduced HIV-1 replication capacity that may be largely compensated for in chronic infection. J Virol 84: 11937–11949.

46. CarlsonJM, BrummeCJ, MartinE, ListgartenJ, BrockmanMA, et al. (2012) Correlates of Protective Cellular Immunity Revealed by Analysis of Population-Level Immune Escape Pathways in HIV-1. J Virol 86: 13202–13216.

47. BorrowP, LewickiH, HahnBH, ShawGM, OldstoneMB (1994) Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J Virol 68: 6103–6110.

48. PereyraF, JiaX, McLarenPJ, TelentiA, de BakkerPI, et al. (2010) The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 330: 1551–1557.

49. KiepielaP, NgumbelaK, ThobakgaleC, RamduthD, HoneyborneI, et al. (2007) CD8+ T-cell responses to different HIV proteins have discordant associations with viral load. Nat Med 13: 46–53.

50. KelleherAD, LongC, HolmesEC, AllenRL, WilsonJ, et al. (2001) Clustered mutations in HIV-1 gag are consistently required for escape from HLA-B27-restricted cytotoxic T lymphocyte responses. J Exp Med 193: 375–386.

51. TroyerRM, McNevinJ, LiuY, ZhangSC, KrizanRW, et al. (2009) Variable fitness impact of HIV-1 escape mutations to cytotoxic T lymphocyte (CTL) response. PLoS Pathog 5: e1000365.

52. LeslieAJ, PfafferottKJ, ChettyP, DraenertR, AddoMM, et al. (2004) HIV evolution: CTL escape mutation and reversion after transmission. Nat Med 10: 282–289.

53. ReicinAS, PaikS, BerkowitzRD, LubanJ, LowyI, et al. (1995) Linker insertion mutations in the human immunodeficiency virus type 1 gag gene: effects on virion particle assembly, release, and infectivity. J Virol 69: 642–650.

54. SrinivasakumarN, HammarskjoldML, RekoshD (1995) Characterization of deletion mutations in the capsid region of human immunodeficiency virus type 1 that affect particle formation and Gag-Pol precursor incorporation. J Virol 69: 6106–6114.

55. FitzonT, LeschonskyB, BielerK, PaulusC, SchroderJ, et al. (2000) Proline residues in the HIV-1 NH2-terminal capsid domain: structure determinants for proper core assembly and subsequent steps of early replication. Virology 268: 294–307.

56. von SchwedlerUK, StrayKM, GarrusJE, SundquistWI (2003) Functional surfaces of the human immunodeficiency virus type 1 capsid protein. J Virol 77: 5439–5450.

57. HolmL, KoivulaAK, LehtovaaraPM, HemminkiA, KnowlesJK (1990) Random mutagenesis used to probe the structure and function of Bacillus stearothermophilus alpha-amylase. Protein Eng 3: 181–191.

58. McNattMW, ZangT, HatziioannouT, BartlettM, FofanaIB, et al. (2009) Species-specific activity of HIV-1 Vpu and positive selection of tetherin transmembrane domain variants. PLoS Pathog 5: e1000300.

59. HatziioannouT, CowanS, Von SchwedlerUK, SundquistWI, BieniaszPD (2004) Species-specific tropism determinants in the human immunodeficiency virus type 1 capsid. J Virol 78: 6005–6012.

60. AmbroseZ, LeeK, NdjomouJ, XuH, OztopI, et al. (2012) Human immunodeficiency virus type 1 capsid mutation N74D alters cyclophilin A dependence and impairs macrophage infection. J Virol 86: 4708–4714.

61. SanjuanR, NebotMR, ChiricoN, ManskyLM, BelshawR (2010) Viral mutation rates. J Virol 84: 9733–9748.

62. SanjuanR (2010) Mutational fitness effects in RNA and single-stranded DNA viruses: common patterns revealed by site-directed mutagenesis studies. Philos Trans R Soc Lond B Biol Sci 365: 1975–1982.

63. ForsheyBM, von SchwedlerU, SundquistWI, AikenC (2002) Formation of a human immunodeficiency virus type 1 core of optimal stability is crucial for viral replication. J Virol 76: 5667–5677.

64. JouvenetN, BieniaszPD, SimonSM (2008) Imaging the biogenesis of individual HIV-1 virions in live cells. Nature 454: 236–240.

65. KrausslichHG (1991) Human immunodeficiency virus proteinase dimer as component of the viral polyprotein prevents particle assembly and viral infectivity. Proc Natl Acad Sci U S A 88: 3213–3217.

66. OttDE, CorenLV, ChertovaEN, GagliardiTD, NagashimaK, et al. (2003) Elimination of protease activity restores efficient virion production to a human immunodeficiency virus type 1 nucleocapsid deletion mutant. J Virol 77: 5547–5556.

67. OttDE, CorenLV, ShatzerT (2009) The nucleocapsid region of human immunodeficiency virus type 1 Gag assists in the coordination of assembly and Gag processing: role for RNA-Gag binding in the early stages of assembly. J Virol 83: 7718–7727.

68. BorsettiA, OhagenA, GottlingerHG (1998) The C-terminal half of the human immunodeficiency virus type 1 Gag precursor is sufficient for efficient particle assembly. J Virol 72: 9313–9317.

69. AuerbachMR, ShuC, KaplanA, SinghIR (2003) Functional characterization of a portion of the Moloney murine leukemia virus gag gene by genetic footprinting. Proc Natl Acad Sci U S A 100: 11678–11683.

70. AuerbachMR, BrownKR, SinghIR (2007) Mutational analysis of the N-terminal domain of Moloney murine leukemia virus capsid protein. J Virol 81: 12337–12347.

71. AlinK, GoffSP (1996) Amino acid substitutions in the CA protein of Moloney murine leukemia virus that block early events in infection. Virology 222: 339–351.

72. AllenTM, AltfeldM, GeerSC, KalifeET, MooreC, et al. (2005) Selective escape from CD8+ T-cell responses represents a major driving force of human immunodeficiency virus type 1 (HIV-1) sequence diversity and reveals constraints on HIV-1 evolution. J Virol 79: 13239–13249.

73. Martinez-PicadoJ, PradoJG, FryEE, PfafferottK, LeslieA, et al. (2006) Fitness cost of escape mutations in p24 Gag in association with control of human immunodeficiency virus type 1. J Virol 80: 3617–3623.

74. ManskyLM, TeminHM (1995) Lower in vivo mutation rate of human immunodeficiency virus type 1 than that predicted from the fidelity of purified reverse transcriptase. J Virol 69: 5087–5094.

75. AbramME, FerrisAL, ShaoW, AlvordWG, HughesSH (2010) Nature, position, and frequency of mutations made in a single cycle of HIV-1 replication. J Virol 84: 9864–9878.

76. PerelsonAS, NeumannAU, MarkowitzM, LeonardJM, HoDD (1996) HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span, and viral generation time. Science 271: 1582–1586.

77. RollandM, ManocheewaS, SwainJV, Lanxon-CooksonEC, KimM, et al. (2013) HIV-1 Conserved-Element Vaccines: Relationship between Sequence Conservation and Replicative Capacity. J Virol 87: 5461–5467.

78. TangC, LoeligerE, KindeI, KyereS, MayoK, et al. (2003) Antiviral inhibition of the HIV-1 capsid protein. J Mol Biol 327: 1013–1020.

79. BlairWS, PickfordC, IrvingSL, BrownDG, AndersonM, et al. (2010) HIV capsid is a tractable target for small molecule therapeutic intervention. PLoS Pathog 6: e1001220.

80. StichtJ, HumbertM, FindlowS, BodemJ, MullerB, et al. (2005) A peptide inhibitor of HIV-1 assembly in vitro. Nat Struct Mol Biol 12: 671–677.

81. ZhangH, ZhaoQ, BhattacharyaS, WaheedAA, TongX, et al. (2008) A cell-penetrating helical peptide as a potential HIV-1 inhibitor. J Mol Biol 378: 565–580.

82. LemkeCT, TitoloS, von SchwedlerU, GoudreauN, MercierJF, et al. (2012) Distinct effects of two HIV-1 capsid assembly inhibitor families that bind the same site within the N-terminal domain of the viral CA protein. J Virol 86: 6643–6655.

83. ZhangF, ZangT, WilsonSJ, JohnsonMC, BieniaszPD (2011) Clathrin facilitates the morphogenesis of retrovirus particles. PLoS Pathog 7: e1002119.

84. VarthakaviV, BrowningPJ, SpearmanP (1999) Human immunodeficiency virus replication in a primary effusion lymphoma cell line stimulates lytic-phase replication of Kaposi's sarcoma-associated herpesvirus. J Virol 73: 10329–10338.

85. EdgarRC (2004) MUSCLE: a multiple sequence alignment method with reduced time and space complexity. BMC Bioinformatics 5: 113.

86. ShenkinPS, ErmanB, MastrandreaLD (1991) Information-theoretical entropy as a measure of sequence variability. Proteins 11: 297–313.

87. ShaoW, EverittL, ManchesterM, LoebDD, HutchisonCA3rd, et al. (1997) Sequence requirements of the HIV-1 protease flap region determined by saturation mutagenesis and kinetic analysis of flap mutants. Proc Natl Acad Sci U S A 94: 2243–2248.

88. PareraM, FernandezG, ClotetB, MartinezMA (2007) HIV-1 protease catalytic efficiency effects caused by random single amino acid substitutions. Mol Biol Evol 24: 382–387.

89. LoebDD, SwanstromR, EverittL, ManchesterM, StamperSE, et al. (1989) Complete mutagenesis of the HIV-1 protease. Nature 340: 397–400.

90. ChaoSF, ChanVL, JurankaP, KaplanAH, SwanstromR, et al. (1995) Mutational sensitivity patterns define critical residues in the palm subdomain of the reverse transcriptase of human immunodeficiency virus type 1. Nucleic Acids Res 23: 803–810.

91. van den EntFM, VosA, PlasterkRH (1998) Mutational scan of the human immunodeficiency virus type 2 integrase protein. J Virol 72: 3916–3924.

92. SmithRA, AndersonDJ, PrestonBD (2006) Hypersusceptibility to substrate analogs conferred by mutations in human immunodeficiency virus type 1 reverse transcriptase. J Virol 80: 7169–7178.

93. van OpijnenT, BoerlijstMC, BerkhoutB (2006) Effects of random mutations in the human immunodeficiency virus type 1 transcriptional promoter on viral fitness in different host cell environments. J Virol 80: 6678–6685.

94. KimB, HathawayTR, LoebLA (1996) Human immunodeficiency virus reverse transcriptase. Functional mutants obtained by random mutagenesis coupled with genetic selection in Escherichia coli. J Biol Chem 271: 4872–4878.

95. YasugiT, VidalM, SakaiH, HowleyPM, BensonJD (1997) Two classes of human papillomavirus type 16 E1 mutants suggest pleiotropic conformational constraints affecting E1 multimerization, E2 interaction, and interaction with cellular proteins. J Virol 71: 5942–5951.

96. MorrisonHG, KirchhoffF, DesrosiersRC (1995) Effects of mutations in constant regions 3 and 4 of envelope of simian immunodeficiency virus. Virology 210: 448–455.

97. NakajimaK, NobusawaE, TonegawaK, NakajimaS (2003) Restriction of amino acid change in influenza A virus H3HA: comparison of amino acid changes observed in nature and in vitro. J Virol 77: 10088–10098.

98. YanoT, NobusawaE, NagyA, NakajimaS, NakajimaK (2008) Effects of single-point amino acid substitutions on the structure and function neuraminidase proteins in influenza A virus. Microbiol Immunol 52: 216–223.

99. PakulaAA, YoungVB, SauerRT (1986) Bacteriophage lambda cro mutations: effects on activity and intracellular degradation. Proc Natl Acad Sci U S A 83: 8829–8833.

100. RheeSS, HunterE (1991) Amino acid substitutions within the matrix protein of type D retroviruses affect assembly, transport and membrane association of a capsid. EMBO J 10: 535–546.

101. TerwilligerTC, ZabinHB, HorvathMP, SandbergWS, SchlunkPM (1994) In vivo characterization of mutants of the bacteriophage f1 gene V protein isolated by saturation mutagenesis. J Mol Biol 236: 556–571.

102. MassoM, MatheE, ParvezN, HijaziK, VaismanII (2009) Modeling the functional consequences of single residue replacements in bacteriophage f1 gene V protein. Protein Eng Des Sel 22: 665–671.

103. SuzutaniT, LaceySF, PowellKL, PurifoyDJ, HonessRW (1992) Random mutagenesis of the thymidine kinase gene of varicella-zoster virus. J Virol 66: 2118–2124.

104. EifanSA, ElliottRM (2009) Mutational analysis of the Bunyamwera orthobunyavirus nucleocapsid protein gene. J Virol 83: 11307–11317.

105. StengerDC, YoungBA, FrenchR (2006) Random mutagenesis of wheat streak mosaic virus HC-Pro: non-infectious interfering mutations in a gene dispensable for systemic infection of plants. J Gen Virol 87: 2741–2747.

106. RennellD, BouvierSE, HardyLW, PoteeteAR (1991) Systematic mutation of bacteriophage T4 lysozyme. J Mol Biol 222: 67–88.

107. Strambio-de-CastilliaC, HunterE (1992) Mutational analysis of the major homology region of Mason-Pfizer monkey virus by use of saturation mutagenesis. J Virol 66: 7021–7032.

108. MoyerCL, WiethoffCM, MaierO, SmithJG, NemerowGR (2011) Functional genetic and biophysical analyses of membrane disruption by human adenovirus. J Virol 85: 2631–2641.

109. Van Der VeldenA, KaminskiA, JacksonRJ, BelshamGJ (1995) Defective point mutants of the encephalomyocarditis virus internal ribosome entry site can be complemented in trans. Virology 214: 82–90.

110. PerisJB, DavisP, CuevasJM, NebotMR, SanjuanR (2010) Distribution of fitness effects caused by single-nucleotide substitutions in bacteriophage f1. Genetics 185: 603–609.

111. MarkiewiczP, KleinaLG, CruzC, EhretS, MillerJH (1994) Genetic studies of the lac repressor. XIV. Analysis of 4000 altered Escherichia coli lac repressors reveals essential and non-essential residues, as well as “spacers” which do not require a specific sequence. J Mol Biol 240: 421–433.

112. GuoHH, ChoeJ, LoebLA (2004) Protein tolerance to random amino acid change. Proc Natl Acad Sci U S A 101: 9205–9210.

113. IhssenJ, KowarikM, WiesliL, ReissR, WackerM, et al. (2012) Structural insights from random mutagenesis of Campylobacter jejuni oligosaccharyltransferase PglB. BMC Biotechnol 12: 67.

114. OlinsPO, BauerSC, Braford-GoldbergS, SterbenzK, PolazziJO, et al. (1995) Saturation mutagenesis of human interleukin-3. J Biol Chem 270: 23754–23760.

115. HuangW, PetrosinoJ, HirschM, ShenkinPS, PalzkillT (1996) Amino acid sequence determinants of beta-lactamase structure and activity. J Mol Biol 258: 688–703.

116. ChenC, RobertsVA, RittenbergMB (1992) Generation and analysis of random point mutations in an antibody CDR2 sequence: many mutated antibodies lose their ability to bind antigen. J Exp Med 176: 855–866.

117. AxeDD, FosterNW, FershtAR (1998) A search for single substitutions that eliminate enzymatic function in a bacterial ribonuclease. Biochemistry 37: 7157–7166.

118. KawashimaH, YamagishiJ, YamayoshiM, OhueM, FukuiT, et al. (1992) Structure-activity relationships in human interleukin-1 alpha: identification of key residues for expression of biological activities. Protein Eng 5: 171–176.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 6
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#