#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

An -Methyltransferase Is Required for Infection of Tick Cells by


Since its discovery in 1994, Human Granulocytic Anaplasmosis (HGA) has become the second most commonly diagnosed tick-borne disease in the US, and it is gaining importance in several countries in Europe. HGA is caused by Anaplasma phagocytophilum, a bacterium transmitted by black-legged ticks and their relatives. Whereas several of the molecules and processes leading to infection of human cells have been identified, little is known about their counterparts in the tick. We analyzed the effects of a mutation in a gene encoding an o-methyltransferase that is involved in methylation of an outer membrane protein. The mutation of the OMT appears to be important for the ability of A. phagocytophilum to adhere to, invade, and replicate in tick cells. Several tests including binding assays, microscopic analysis of the infection cycle within tick cells, gene expression assays, and biochemical assays using recombinant OMT strongly suggested that the mutation of the o-methyltransferase gene arrested the growth and development of this bacterium within tick cells. Proteomic analyses identified several possible OMT substrates, and in vitro methylation assays using recombinant o-methyltransferase identified an outer membrane protein, Msp4, as a specifically methyl-modified target. Our results indicated that methylation was important for infection of tick cells by A. phagocytophilum, and suggested possible strategies to block transmission of this emerging pathogen. The solved crystal structure of the o-methyltransferase will further stimulate the search for small molecule inhibitors that could break the tick transmission cycle of A. phagocytophilum in nature.


Vyšlo v časopise: An -Methyltransferase Is Required for Infection of Tick Cells by. PLoS Pathog 11(11): e32767. doi:10.1371/journal.ppat.1005248
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1005248

Souhrn

Since its discovery in 1994, Human Granulocytic Anaplasmosis (HGA) has become the second most commonly diagnosed tick-borne disease in the US, and it is gaining importance in several countries in Europe. HGA is caused by Anaplasma phagocytophilum, a bacterium transmitted by black-legged ticks and their relatives. Whereas several of the molecules and processes leading to infection of human cells have been identified, little is known about their counterparts in the tick. We analyzed the effects of a mutation in a gene encoding an o-methyltransferase that is involved in methylation of an outer membrane protein. The mutation of the OMT appears to be important for the ability of A. phagocytophilum to adhere to, invade, and replicate in tick cells. Several tests including binding assays, microscopic analysis of the infection cycle within tick cells, gene expression assays, and biochemical assays using recombinant OMT strongly suggested that the mutation of the o-methyltransferase gene arrested the growth and development of this bacterium within tick cells. Proteomic analyses identified several possible OMT substrates, and in vitro methylation assays using recombinant o-methyltransferase identified an outer membrane protein, Msp4, as a specifically methyl-modified target. Our results indicated that methylation was important for infection of tick cells by A. phagocytophilum, and suggested possible strategies to block transmission of this emerging pathogen. The solved crystal structure of the o-methyltransferase will further stimulate the search for small molecule inhibitors that could break the tick transmission cycle of A. phagocytophilum in nature.


Zdroje

1. Bakken JS, Dumler JS. Clinical diagnosis and treatment of human granulocytotropic anaplasmosis. Ann N Y Acad Sci. 2006;1078:236–47. 17114714.

2. Dumler JS. The biological basis of severe outcomes in Anaplasma phagocytophilum infection. FEMS Immunol Med Microbiol. 2012;64(1):13–20. 22098465. doi: 10.1111/j.1574-695X.2011.00909.x

3. CDC. Anaplasmosis: Statistics and Epidemiology. 2012.

4. CDC. Notice to readers: final 2013 reports of nationally notifiable infectious diseases. MMWR Morb Mortal Wkly Rep. 2014;63(32):702. 25272402.

5. Jin H, Wei F, Liu Q, Qian J. Epidemiology and Control of Human Granulocytic Anaplasmosis: A Systematic Review. Vector Borne Zoonotic Dis. 2012;2012:4. 22217177.

6. Rejmanek D, Bradburd G, Foley J. Molecular characterization reveals distinct genospecies of Anaplasma phagocytophilum from diverse North American hosts. J Med Microbiol. 2012;61(Pt 2):204–12. 21921109. doi: 10.1099/jmm.0.034702-0

7. Bakken JS, Dumler S. Human granulocytic anaplasmosis. Infect Dis Clin North Am. 2008;22(3):433–48, viii. 18755383. doi: 10.1016/j.idc.2008.03.011

8. Baldridge GD, Scoles GA, Burkhardt NY, Schloeder B, Kurtti TJ, Munderloh UG. Transovarial transmission of Francisella-like endosymbionts and Anaplasma phagocytophilum variants in Dermacentor albipictus (Acari: Ixodidae). J Med Entomol. 2009;46(3):625–32. 19496436.

9. Munderloh UG, Jauron SD, Fingerle V, Leitritz L, Hayes SF, Hautman JM, et al. Invasion and intracellular development of the human granulocytic ehrlichiosis agent in tick cell culture. J Clin Microbiol. 1999;37(8):2518–24. 10405394.

10. Troese MJ, Carlyon JA. Anaplasma phagocytophilum dense-cored organisms mediate cellular adherence through recognition of human P-selectin glycoprotein ligand 1. Infect Immun. 2009;77(9):4018–27. 19596771. doi: 10.1128/IAI.00527-09

11. Rikihisa Y. Molecular events involved in cellular invasion by Ehrlichia chaffeensis and Anaplasma phagocytophilum. Vet Parasitol. 2010;167(2–4):155–66. 19836896. doi: 10.1016/j.vetpar.2009.09.017

12. Severo MS, Stephens KD, Kotsyfakis M, Pedra JH. Anaplasma phagocytophilum: deceptively simple or simply deceptive? Future Microbiol. 2012;7(6):719–31. 22702526. doi: 10.2217/fmb.12.45

13. Pedra JH, Narasimhan S, Rendic D, DePonte K, Bell-Sakyi L, Wilson IB, et al. Fucosylation enhances colonization of ticks by Anaplasma phagocytophilum. Cell Microbiol. 2010;12(9):1222–34. 20331643. doi: 10.1111/j.1462-5822.2010.01464.x

14. Sukumaran B, Narasimhan S, Anderson JF, DePonte K, Marcantonio N, Krishnan MN, et al. An Ixodes scapularis protein required for survival of Anaplasma phagocytophilum in tick salivary glands. J Exp Med. 2006;203(6):1507–17. 16717118.

15. Zivkovic Z, Blouin EF, Manzano-Roman R, Almazan C, Naranjo V, Massung RF, et al. Anaplasma phagocytophilum and Anaplasma marginale elicit different gene expression responses in cultured tick cells. Comp Funct Genomics. 2009;705034(10):705034. 19636428.

16. Mastronunzio JE, Kurscheid S, Fikrig E. Postgenomic analyses reveal development of infectious Anaplasma phagocytophilum during transmission from ticks to mice. J Bacteriol. 2012;194(9):2238–47. 22389475. doi: 10.1128/JB.06791-11

17. Nelson CM, Herron MJ, Felsheim RF, Schloeder BR, Grindle SM, Chavez AO, et al. Whole genome transcription profiling of Anaplasma phagocytophilum in human and tick host cells by tiling array analysis. BMC Genomics. 2008;9(364):364. 18671858.

18. Felsheim RF, Herron MJ, Nelson CM, Burkhardt NY, Barbet AF, Kurtti TJ, et al. Transformation of Anaplasma phagocytophilum. BMC Biotechnol. 2006;6(42):42. 17076894.

19. Chen G, Severo MS, Sakhon OS, Choy A, Herron MJ, Felsheim RF, et al. Anaplasma phagocytophilum dihydrolipoamide dehydrogenase 1 affects host-derived immunopathology during microbial colonization. Infect Immun. 2012;80(9):3194–205. 22753375. doi: 10.1128/IAI.00532-12

20. Crosby FL, Brayton KA, Magunda F, Munderloh UG, Kelley KL, Barbet AF. Reduced Infectivity in cattle for an outer membrane protein mutant of Anaplasma marginale. Appl Environ Microbiol. 2015;81(6):2206–14. Epub 2015/01/18 06:00. 25595772. doi: 10.1128/AEM.03241-14

21. Crosby FL, Wamsley HL, Pate MG, Lundgren AM, Noh SM, Munderloh UG, et al. Knockout of an outer membrane protein operon of Anaplasma marginale by transposon mutagenesis. BMC Genomics. 2014;15(278):278. Epub 2014/04/15 06:00. 24725301.

22. Champion MD. Host-pathogen o-methyltransferase similarity and its specific presence in highly virulent strains of Francisella tularensis suggests molecular mimicry. PLoS One. 2011;6(5):e20295. 21637805. doi: 10.1371/journal.pone.0020295

23. Liscombe DK, Louie GV, Noel JP. Architectures, mechanisms and molecular evolution of natural product methyltransferases. Nat Prod Rep. 2012;29(10):1238–50. 22850796. doi: 10.1039/c2np20029e

24. Pustelny C, Brouwer S, Musken M, Bielecka A, Dotsch A, Nimtz M, et al. The peptide chain release factor methyltransferase PrmC is essential for pathogenicity and environmental adaptation of Pseudomonas aeruginosa PA14. Environ Microbiol. 2013;15(2):597–609. 23278968. doi: 10.1111/1462-2920.12040

25. Chao CC, Chelius D, Zhang T, Mutumanje E, Ching WM. Insight into the virulence of Rickettsia prowazekii by proteomic analysis and comparison with an avirulent strain. Biochim Biophys Acta. 2007;1774(3):373–81. 17301007.

26. Paranjpye RN, Lara JC, Pepe JC, Pepe CM, Strom MS. The type IV leader peptidase/N-methyltransferase of Vibrio vulnificus controls factors required for adherence to HEp-2 cells and virulence in iron-overloaded mice. Infect Immun. 1998;66(12):5659–68. 9826339.

27. Viswanathan VK, Edelstein PH, Pope CD, Cianciotto NP. The Legionella pneumophila iraAB locus is required for iron assimilation, intracellular infection, and virulence. Infect Immun. 2000;68(3):1069–79. 10678909.

28. Dekkers KL, You BJ, Gowda VS, Liao HL, Lee MH, Bau HJ, et al. The Cercospora nicotianae gene encoding dual O-methyltransferase and FAD-dependent monooxygenase domains mediates cercosporin toxin biosynthesis. Fungal Genet Biol. 2007;44(5):444–54. 17074519.

29. Willemsen NM, Hitchen EM, Bodetti TJ, Apolloni A, Warrilow D, Piller SC, et al. Protein methylation is required to maintain optimal HIV-1 infectivity. Retrovirology. 2006;3(92):92. 17169163.

30. Abeykoon A, Wang G, Chao CC, Chock PB, Gucek M, Ching WM, et al. Multi-methylation in Rickettsia OmpB Catalyzed by Lysine Methyltransferases. J Biol Chem. 2014:4. 24497633.

31. Chan YG, Cardwell MM, Hermanas TM, Uchiyama T, Martinez JJ. Rickettsial outer-membrane protein B (rOmpB) mediates bacterial invasion through Ku70 in an actin, c-Cbl, clathrin and caveolin 2-dependent manner. Cell Microbiol. 2009;11(4):629–44. 19134120. doi: 10.1111/j.1462-5822.2008.01279.x

32. Eshghi A, Pinne M, Haake DA, Zuerner RL, Frank A, Cameron CE. Methylation and in vivo expression of the surface-exposed Leptospira interrogans outer-membrane protein OmpL32. Microbiology. 2012;158(Pt 3):622–35. 22174381. doi: 10.1099/mic.0.054767-0

33. Dunning Hotopp JC, Lin M, Madupu R, Crabtree J, Angiuoli SV, Eisen JA, et al. Comparative genomics of emerging human ehrlichiosis agents. PLoS Genet. 2006;2(2):e21. 16482227.

34. Mohan Kumar D, Yamaguchi M, Miura K, Lin M, Los M, Coy JF, et al. Ehrlichia chaffeensis uses its surface protein EtpE to bind GPI-anchored protein DNase X and trigger entry into mammalian cells. PLoS Pathog. 2013;9(10):e1003666. Epub 2013/10/08 06:00. 24098122. doi: 10.1371/journal.ppat.1003666

35. Seidman D, Hebert KS, Truchan HK, Miller DP, Tegels BK, Marconi RT, et al. Essential domains of Anaplasma phagocytophilum invasins utilized to infect mammalian host cells. PLoS Pathog. 2015;11(2):e1004669. Epub 2015/02/07 06:00. 25658707. doi: 10.1371/journal.ppat.1004669

36. Williams JC, McInnis KA, Testerman TL. Adherence of Helicobacter pylori to abiotic surfaces is influenced by serum. Appl Environ Microbiol. 2008;74(4):1255–8. Epub 2007/12/25 09:00. 18156334.

37. Felsheim RF, Chavez AS, Palmer GH, Crosby L, Barbet AF, Kurtti TJ, et al. Transformation of Anaplasma marginale. Vet Parasitol. 2010;167(2–4):167–74. 19837516. doi: 10.1016/j.vetpar.2009.09.018

38. Ge Y, Rikihisa Y. Identification of novel surface proteins of Anaplasma phagocytophilum by affinity purification and proteomics. J Bacteriol. 2007;189(21):7819–28. 17766422.

39. Troese MJ, Kahlon A, Ragland SA, Ottens AK, Ojogun N, Nelson KT, et al. Proteomic analysis of Anaplasma phagocytophilum during infection of human myeloid cells identifies a protein that is pronouncedly upregulated on the infectious dense-cored cell. Infect Immun. 2011;79(11):4696–707. 21844238. doi: 10.1128/IAI.05658-11

40. Garcia IG, Stevenson CEM, Uson I, Meyers CLF, Walsh CT, Lawson DM. The Crystal Structure of the Novobiocin Biosynthetic Enzyme NovP: The First Representative Structure for the TylF O-Methyltransferase Superfamily. J Mol Biol. 2009;395(2):390. 19857499. doi: 10.1016/j.jmb.2009.10.045

41. Abendroth J, Gardberg AS, Robinson JI, Christensen JS, Staker BL, Myler PJ, et al. SAD phasing using iodide ions in a high-throughput structural genomics environment. J Struct Funct Genomics. 2011;12(2):83–95. 21359836. doi: 10.1007/s10969-011-9101-7

42. Kelley LA, Sternberg MJ. Protein structure prediction on the Web: a case study using the Phyre server. Nat Protoc. 2009;4(3):363–71. 19247286. doi: 10.1038/nprot.2009.2

43. Cheng C, Nair AD, Indukuri VV, Gong S, Felsheim RF, Jaworski D, et al. Targeted and random mutagenesis of Ehrlichia chaffeensis for the identification of genes required for in vivo infection. PLoS Pathog. 1371;9(2):e1003171. 23459099. doi: 10.1371/journal.ppat.1003171

44. Noriea NF, Clark TR, Hackstadt T. Targeted knockout of the Rickettsia rickettsii OmpA surface antigen does not diminish virulence in a mammalian model system. MBio. 2015;6(2):00323–15. Epub 2015/04/02 06:00. 25827414.

45. Chen G, Severo MS, Sakhon OS, Choy A, Herron MJ, Felsheim RF, et al. Anaplasma phagocytophilum dihydrolipoamide dehydrogenase 1 affects host-derived immunopathology during microbial colonization. Infect Immun. 2012;80(9):3194–205. 22753375. doi: 10.1128/IAI.00532-12

46. Montagna M, Sassera D, Epis S, Bazzocchi C, Vannini C, Lo N, et al. "Candidatus Midichloriaceae" fam. nov. (Rickettsiales), an Ecologically Widespread Clade of Intracellular Alphaproteobacteria. Appl Environ Microbiol. 2013;79(10):3241–8. 23503305. doi: 10.1128/AEM.03971-12

47. Sockett RE. Predatory lifestyle of Bdellovibrio bacteriovorus. Annu Rev Microbiol. 2009;63:523–39. 19575566. doi: 10.1146/annurev.micro.091208.073346

48. Lambert C, Chang CY, Capeness MJ, Sockett RE. The first bite—profiling the predatosome in the bacterial pathogen Bdellovibrio. PLoS One. 2010;5(1):e8599. 20062540. doi: 10.1371/journal.pone.0008599

49. Lan G, Schulmeister S, Sourjik V, Tu Y. Adapt locally and act globally: strategy to maintain high chemoreceptor sensitivity in complex environments. Mol Syst Biol. 2011;7(475):475. 21407212.

50. Levit MN, Stock JB. Receptor methylation controls the magnitude of stimulus-response coupling in bacterial chemotaxis. J Biol Chem. 2002;277(39):36760–5. 12119291.

51. Terwilliger TC, Wang JY, Koshland DE Jr. Kinetics of receptor modification. The multiply methylated aspartate receptors involved in bacterial chemotaxis. J Biol Chem. 1986;261(23):10814–20. 3015942.

52. Cheng Z, Kumagai Y, Lin M, Zhang C, Rikihisa Y. Intra-leukocyte expression of two-component systems in Ehrlichia chaffeensis and Anaplasma phagocytophilum and effects of the histidine kinase inhibitor closantel. Cell Microbiol. 2006;8(8):1241–52. 16882029.

53. Low JK, Hart-Smith G, Erce MA, Wilkins MR. The Saccharomyces cerevisiae poly(A)-binding protein is subject to multiple post-translational modifications, including the methylation of glutamic acid. Biochem Biophys Res Commun. 2014;443(2):543–8. 24326073. doi: 10.1016/j.bbrc.2013.12.009

54. Abeykoon AH, Chao CC, Wang G, Gucek M, Yang DC, Ching WM. Two protein lysine methyltransferases methylate outer membrane protein B from Rickettsia. J Bacteriol. 2012;194(23):6410–8. 23002218. doi: 10.1128/JB.01379-12

55. Bechah Y, El Karkouri K, Mediannikov O, Leroy Q, Pelletier N, Robert C, et al. Genomic, proteomic, and transcriptomic analysis of virulent and avirulent Rickettsia prowazekii reveals its adaptive mutation capabilities. Genome Res. 2010;20(5):655–63. 20368341. doi: 10.1101/gr.103564.109

56. Chamot-Rooke J, Mikaty G, Malosse C, Soyer M, Dumont A, Gault J, et al. Posttranslational modification of pili upon cell contact triggers N. meningitidis dissemination. Science. 2011;331(6018):778–82. 21311024. doi: 10.1126/science.1200729

57. Krzywinska E, Bhatnagar S, Sweet L, Chatterjee D, Schorey JS. Mycobacterium avium 104 deleted of the methyltransferase D gene by allelic replacement lacks serotype-specific glycopeptidolipids and shows attenuated virulence in mice. Mol Microbiol. 2005;56(5):1262–73. 15882419.

58. Narimatsu M, Noiri Y, Itoh S, Noguchi N, Kawahara T, Ebisu S. Essential role for the gtfA gene encoding a putative glycosyltransferase in the adherence of Porphyromonas gingivalis. Infect Immun. 2004;72(5):2698–702. 15102778.

59. Rodionov AV, Eremeeva ME, Balayeva NM. Isolation and partial characterization of the M(r) 100 kD protein from Rickettsia prowazekii strains of different virulence. Acta Virol. 1991;35(6):557–65. 1687639.

60. de la Fuente J, Massung RF, Wong SJ, Chu FK, Lutz H, Meli M, et al. Sequence analysis of the msp4 gene of Anaplasma phagocytophilum strains. J Clin Microbiol. 2005;43(3):1309–17. 15750101.

61. Oberle SM, Palmer GH, Barbet AF. Expression and immune recognition of the conserved MSP4 outer membrane protein of Anaplasma marginale. Infect Immun. 1993;61(12):5245–51. 7693596.

62. Palmer GH, Eid G, Barbet AF, McGuire TC, McElwain TF. The immunoprotective Anaplasma marginale major surface protein 2 is encoded by a polymorphic multigene family. Infect Immun. 1994;62(9):3808–16. 8063397.

63. Galdiero S, Falanga A, Cantisani M, Tarallo R, Della Pepa ME, D'Oriano V, et al. Microbe-host interactions: structure and role of Gram-negative bacterial porins. Curr Protein Pept Sci. 2012;13(8):843–54. 23305369.

64. Nikaido H. Molecular basis of bacterial outer membrane permeability revisited. Microbiol Mol Biol Rev. 2003;67(4):593–656. 14665678.

65. Vidotto MC, McGuire TC, McElwain TF, Palmer GH, Knowles DP Jr. Intermolecular relationships of major surface proteins of Anaplasma marginale. Infect Immun. 1994;62(7):2940–6. 8005681.

66. Kahlon A, Ojogun N, Ragland SA, Seidman D, Troese MJ, Ottens AK, et al. Anaplasma phagocytophilum Asp14 is an invasin that interacts with mammalian host cells via its C terminus to facilitate infection. Infect Immun. 2013;81(1):65–79. 23071137. doi: 10.1128/IAI.00932-12

67. Ojogun N, Kahlon A, Ragland SA, Troese MJ, Mastronunzio JE, Walker NJ, et al. Anaplasma phagocytophilum Outer Membrane Protein A Interacts with Sialylated Glycoproteins To Promote Infection of Mammalian Host Cells. Infect Immun. 2012;80(11):3748–60. 22907813. doi: 10.1128/IAI.00654-12

68. Seidman D, Ojogun N, Walker NJ, Mastronunzio J, Kahlon A, Hebert KS, et al. Anaplasma phagocytophilum surface protein AipA mediates invasion of mammalian host cells. Cell Microbiol. 2014;16(8):1133–45. 24612118. doi: 10.1111/cmi.12286

69. Vellaiswamy M, Kowalczewska M, Merhej V, Nappez C, Vincentelli R, Renesto P, et al. Characterization of rickettsial adhesin Adr2 belonging to a new group of adhesins in alpha-proteobacteria. Microb Pathog. 2011;50(5):233–42. 21288480. doi: 10.1016/j.micpath.2011.01.009

70. Rikihisa Y, Zhi N, Wormser GP, Wen B, Horowitz HW, Hechemy KE. Ultrastructural and antigenic characterization of a granulocytic ehrlichiosis agent directly isolated and stably cultivated from a patient in New York state. J Infect Dis. 1997;175(1):210–3. 8985223.

71. Goodman JL, Nelson C, Vitale B, Madigan JE, Dumler JS, Kurtti TJ, et al. Direct cultivation of the causative agent of human granulocytic ehrlichiosis. N Engl J Med. 1996;334(4):209–15. 8531996.

72. Oliver JD, Burkhardt NY, Felsheim RF, Kurtti TJ, Munderloh UG. Motility Characteristics Are Altered for Rickettsia bellii Transformed To Overexpress a Heterologous rickA Gene. Appl Environ Microbiol. 2014;80(3):1170–6. 24296498. doi: 10.1128/AEM.03352-13

73. Baldridge GD, Burkhardt N, Herron MJ, Kurtti TJ, Munderloh UG. Analysis of fluorescent protein expression in transformants of Rickettsia monacensis, an obligate intracellular tick symbiont. Appl Environ Microbiol. 2005;71(4):2095–105. 15812043.

74. Riedl J, Crevenna AH, Kessenbrock K, Yu JH, Neukirchen D, Bista M, et al. Lifeact: a versatile marker to visualize F-actin. Nat Methods. 2008;5(7):605–7. 18536722. doi: 10.1038/nmeth.1220

75. Yang F, Shen Y, Camp DG 2nd, Smith RD. High-pH reversed-phase chromatography with fraction concatenation for 2D proteomic analysis. Expert Rev Proteomics. 2012;9(2):129–34. 22462785. doi: 10.1586/epr.12.15

76. Lin-Moshier Y, Sebastian PJ, Higgins L, Sampson ND, Hewitt JE, Marchant JS. Re-evaluation of the role of calcium homeostasis endoplasmic reticulum protein (CHERP) in cellular calcium signaling. J Biol Chem. 2013;288(1):355–67. 23148228. doi: 10.1074/jbc.M112.405761

77. Kabsch W. Integration, scaling, space-group assignment and post-refinement. Acta Crystallogr D Biol Crystallogr. 1107;66(Pt 2):133–44. 20124693. doi: 10.1107/S0907444909047374

78. Grosse-Kunstleve RW, Adams PD. Substructure search procedures for macromolecular structures. Acta Crystallogr D Biol Crystallogr. 2003;59(Pt 11):1966–73. 14573951.

79. McCoy AJ, Grosse-Kunstleve RW, Adams PD, Winn MD, Storoni LC, Read RJ. Phaser crystallographic software. J Appl Crystallogr. 2007;40(Pt 4):658–74. 19461840.

80. Terwilliger TC. Maximum-likelihood density modification. Acta Crystallogr D Biol Crystallogr. 2000;56(Pt 8):965–72. 10944333.

81. Langer G, Cohen SX, Lamzin VS, Perrakis A. Automated macromolecular model building for X-ray crystallography using ARP/wARP version 7. Nat Protoc. 2008;3(7):1171–9. 18600222. doi: 10.1038/nprot.2008.91

82. Adams PD, Afonine PV, Bunkoczi G, Chen VB, Davis IW, Echols N, et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr D Biol Crystallogr. 2010;66(Pt 2):213–21. 20124702. doi: 10.1107/S0907444909052925

83. Murshudov GN, Skubak P, Lebedev AA, Pannu NS, Steiner RA, Nicholls RA, et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr D Biol Crystallogr. 2011;67(Pt 4):355–67. 21460454. doi: 10.1107/S0907444911001314

84. Emsley P, Lohkamp B, Scott WG, Cowtan K. Features and development of Coot. Acta Crystallogr D Biol Crystallogr. 2010;66(Pt 4):486–501. 20383002. doi: 10.1107/S0907444910007493

85. Bailey TL, Elkan C. Fitting a mixture model by expectation maximization to discover motifs in biopolymers. Proc Int Conf Intell Syst Mol Biol. 1994;2:28–36. 7584402.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 11
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#