#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Requires Host Rab1b for Survival in Macrophages


Yersinia pestis is the bacterial agent that causes the human disease known as plague. While often considered a historic disease, Y. pestis is endemic in rodent populations on several continents and the World Health Organization considers plague to be a reemerging disease. Much of the success of this pathogen comes from its ability to evade clearance by the innate immune system of its host. One weapon in the Y. pestis arsenal is its ability to resist killing when engulfed by macrophages. Upon invasion of macrophages, Y. pestis actively manipulates the cell to generate a protective vacuolar compartment, called the Yersinia containing vacuole (YCV) that allows the bacterium to evade the normal pathogen killing mechanisms of the macrophage. Here we demonstrate that the host protein Rab1b is recruited to the YCV and is required for Y. pestis to inhibit both the acidification and normal maturation of the phagosome to establish a protective niche within the cell. Rab1b is the first protein, either from the host or Y. pestis, shown to contribute to the biogenesis of the YCV. Furthermore, our data suggest a previously unknown impact of Rab1b recruitment in the phagosome maturation pathway.


Vyšlo v časopise: Requires Host Rab1b for Survival in Macrophages. PLoS Pathog 11(10): e32767. doi:10.1371/journal.ppat.1005241
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1005241

Souhrn

Yersinia pestis is the bacterial agent that causes the human disease known as plague. While often considered a historic disease, Y. pestis is endemic in rodent populations on several continents and the World Health Organization considers plague to be a reemerging disease. Much of the success of this pathogen comes from its ability to evade clearance by the innate immune system of its host. One weapon in the Y. pestis arsenal is its ability to resist killing when engulfed by macrophages. Upon invasion of macrophages, Y. pestis actively manipulates the cell to generate a protective vacuolar compartment, called the Yersinia containing vacuole (YCV) that allows the bacterium to evade the normal pathogen killing mechanisms of the macrophage. Here we demonstrate that the host protein Rab1b is recruited to the YCV and is required for Y. pestis to inhibit both the acidification and normal maturation of the phagosome to establish a protective niche within the cell. Rab1b is the first protein, either from the host or Y. pestis, shown to contribute to the biogenesis of the YCV. Furthermore, our data suggest a previously unknown impact of Rab1b recruitment in the phagosome maturation pathway.


Zdroje

1. Perry RD, Fetherston JD. Yersinia pestis—etiologic agent of plague. Clinical microbiology reviews. 1997;10(1):35–66. 8993858

2. Butler T. Plague gives surprises in the first decade of the 21st century in the United States and worldwide. The American journal of tropical medicine and hygiene. 2013;89(4):788–93. Epub 2013/09/18. doi: 10.4269/ajtmh.13-0191 24043686

3. Inglesby TV, Dennis DT, Henderson DA, Bartlett JG, Ascher MS, Eitzen E, et al. Plague as a biological weapon: medical and public health management. Working Group on Civilian Biodefense. Jama. 2000;283(17):2281–90. 10807389

4. Spinner JL, Winfree S, Starr T, Shannon JG, Nair V, Steele-Mortimer O, et al. Yersinia pestis survival and replication within human neutrophil phagosomes and uptake of infected neutrophils by macrophages. Journal of leukocyte biology. 2013. Epub 2013/11/15.

5. Straley SC, Harmon PA. Growth in mouse peritoneal macrophages of Yersinia pestis lacking established virulence determinants. Infection and immunity. 1984;45(3):649–54. Epub 1984/09/01. 6469351

6. Gonzalez RJ, Lane MC, Wagner NJ, Weening EH, Miller VL. Dissemination of a highly virulent pathogen: tracking the early events that define infection. PLoS pathogens. 2015;11(1):e1004587. Epub 2015/01/23. doi: 10.1371/journal.ppat.1004587 25611317

7. Lukaszewski RA, Kenny DJ, Taylor R, Rees DG, Hartley MG, Oyston PC. Pathogenesis of Yersinia pestis infection in BALB/c mice: effects on host macrophages and neutrophils. Infection and immunity. 2005;73(11):7142–50. Epub 2005/10/22. 16239508

8. Janssen WA, Surgalla MJ. Plague bacillus: survival within host phagocytes. Science. 1969;163(3870):950–2. Epub 1969/02/28. 5763880

9. Vagima Y, Zauberman A, Levy Y, Gur D, Tidhar A, Aftalion M, et al. Circumventing Y. pestis Virulence by Early Recruitment of Neutrophils to the Lungs during Pneumonic Plague. PLoS pathogens. 2015;11(5):e1004893. Epub 2015/05/15. doi: 10.1371/journal.ppat.1004893 25974210

10. Burrows TW, Bacon GA. The basis of virulence in Pasteurella pestis: the development of resistance to phagocytosis in vitro. British journal of experimental pathology. 1956;37(3):286–99. Epub 1956/06/01. 13342354

11. Pujol C, Bliska JB. The ability to replicate in macrophages is conserved between Yersinia pestis and Yersinia pseudotuberculosis. Infection and immunity. 2003;71(10):5892–9. 14500510

12. Pujol C, Klein KA, Romanov GA, Palmer LE, Cirota C, Zhao Z, et al. Yersinia pestis can reside in autophagosomes and avoid xenophagy in murine macrophages by preventing vacuole acidification. Infection and immunity. 2009;77(6):2251–61. Epub 2009/03/18. doi: 10.1128/IAI.00068-09 19289509

13. Grabenstein JP, Fukuto HS, Palmer LE, Bliska JB. Characterization of phagosome trafficking and identification of PhoP-regulated genes important for survival of Yersinia pestis in macrophages. Infection and immunity. 2006;74(7):3727–41. Epub 2006/06/23. 16790745

14. Straley SC, Harmon PA. Yersinia pestis grows within phagolysosomes in mouse peritoneal macrophages. Infection and immunity. 1984;45(3):655–9. Epub 1984/09/01. 6469352

15. Grabenstein JP, Marceau M, Pujol C, Simonet M, Bliska JB. The response regulator PhoP of Yersinia pseudotuberculosis is important for replication in macrophages and for virulence. Infection and immunity. 2004;72(9):4973–84. Epub 2004/08/24. 15321989

16. Moreau K, Lacas-Gervais S, Fujita N, Sebbane F, Yoshimori T, Simonet M, et al. Autophagosomes can support Yersinia pseudotuberculosis replication in macrophages. Cellular microbiology. 2010;12(8):1108–23. Epub 2010/02/26. doi: 10.1111/j.1462-5822.2010.01456.x 20180800

17. Ligeon LA, Moreau K, Barois N, Bongiovanni A, Lacorre DA, Werkmeister E, et al. Role of VAMP3 and VAMP7 in the commitment of Yersinia pseudotuberculosis to LC3-associated pathways involving single- or double-membrane vacuoles. Autophagy. 2014;10(9). Epub 2014/07/22.

18. Ponnusamy D, Clinkenbeard KD. Yersinia pestis intracellular parasitism of macrophages from hosts exhibiting high and low severity of plague. PloS one. 2012;7(7):27.

19. Finegold MJ. Pneumonic plague in monkeys. An electron microscopic study. The American journal of pathology. 1969;54(2):167–85. Epub 1969/02/01. 4974722

20. St John AL, Ang WX, Huang MN, Kunder CA, Chan EW, Gunn MD, et al. S1P-Dependent Trafficking of Intracellular Yersinia pestis through Lymph Nodes Establishes Buboes and Systemic Infection. Immunity. 2014;41(3):440–50. Epub 2014/09/23. doi: 10.1016/j.immuni.2014.07.013 25238098

21. Ye Z, Kerschen EJ, Cohen DA, Kaplan AM, van Rooijen N, Straley SC. Gr1+ cells control growth of YopM-negative yersinia pestis during systemic plague. Infection and immunity. 2009;77(9):3791–806. Epub 2009/07/08. doi: 10.1128/IAI.00284-09 19581396

22. Oyston PC, Dorrell N, Williams K, Li SR, Green M, Titball RW, et al. The response regulator PhoP is important for survival under conditions of macrophage-induced stress and virulence in Yersinia pestis. Infection and immunity. 2000;68(6):3419–25. Epub 2000/05/19. 10816493

23. Bozue J, Mou S, Moody KL, Cote CK, Trevino S, Fritz D, et al. The role of the phoPQ operon in the pathogenesis of the fully virulent CO92 strain of Yersinia pestis and the IP32953 strain of Yersinia pseudotuberculosis. Microbial pathogenesis. 2011;50(6):314–21. doi: 10.1016/j.micpath.2011.02.005 21320584

24. Rust JH Jr., Cavanaugh DC, O'Shita R, Marshall JD Jr. The role of domestic animals in the epidemiology of plague. I. Experimental infection of dogs and cats. The Journal of infectious diseases. 1971;124(5):522–6. Epub 1971/11/01. 5115673

25. Stein MP, Muller MP, Wandinger-Ness A. Bacterial pathogens commandeer Rab GTPases to establish intracellular niches. Traffic. 2012;13(12):1565–88. doi: 10.1111/tra.12000 22901006

26. Bhuin T, Roy JK. Rab proteins: the key regulators of intracellular vesicle transport. Experimental cell research. 2014;328(1):1–19. Epub 2014/08/05. doi: 10.1016/j.yexcr.2014.07.027 25088255

27. Kinchen JM, Ravichandran KS. Phagosome maturation: going through the acid test. Nature reviews Molecular cell biology. 2008;9(10):781–95. Epub 2008/09/25. doi: 10.1038/nrm2515 18813294

28. Vieira OV, Botelho RJ, Grinstein S. Phagosome maturation: aging gracefully. The Biochemical journal. 2002;366(Pt 3):689–704. Epub 2002/06/14. 12061891

29. Sarantis H, Balkin DM, De Camilli P, Isberg RR, Brumell JH, Grinstein S. Yersinia entry into host cells requires Rab5-dependent dephosphorylation of PI(4,5)P(2) and membrane scission. Cell host & microbe. 2012;11(2):117–28.

30. Luzio JP, Gray SR, Bright NA. Endosome-lysosome fusion. Biochemical Society transactions. 2010;38(6):1413–6. Epub 2010/12/02. doi: 10.1042/BST0381413 21118098

31. Luzio JP, Pryor PR, Bright NA. Lysosomes: fusion and function. Nature reviews Molecular cell biology. 2007;8(8):622–32. Epub 2007/07/20. 17637737

32. Luzio JP, Pryor PR, Gray SR, Gratian MJ, Piper RC, Bright NA. Membrane traffic to and from lysosomes. Biochemical Society symposium. 2005; (72):77–86. Epub 2005/01/15. 15649132

33. Sturgill-Koszycki S, Schaible UE, Russell DG. Mycobacterium-containing phagosomes are accessible to early endosomes and reflect a transitional state in normal phagosome biogenesis. The EMBO journal. 1996;15(24):6960–8. 9003772

34. Via LE, Deretic D, Ulmer RJ, Hibler NS, Huber LA, Deretic V. Arrest of mycobacterial phagosome maturation is caused by a block in vesicle fusion between stages controlled by rab5 and rab7. The Journal of biological chemistry. 1997;272(20):13326–31. Epub 1997/05/16. 9148954

35. Fratti RA, Backer JM, Gruenberg J, Corvera S, Deretic V. Role of phosphatidylinositol 3-kinase and Rab5 effectors in phagosomal biogenesis and mycobacterial phagosome maturation arrest. The Journal of cell biology. 2001;154(3):631–44. Epub 2001/08/08. 11489920

36. Fratti RA, Chua J, Vergne I, Deretic V. Mycobacterium tuberculosis glycosylated phosphatidylinositol causes phagosome maturation arrest. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(9):5437–42. Epub 2003/04/19. 12702770

37. Seto S, Matsumoto S, Ohta I, Tsujimura K, Koide Y. Dissection of Rab7 localization on Mycobacterium tuberculosis phagosome. Biochemical and biophysical research communications. 2009;387(2):272–7. doi: 10.1016/j.bbrc.2009.06.152 19580780

38. Roberts EA, Chua J, Kyei GB, Deretic V. Higher order Rab programming in phagolysosome biogenesis. The Journal of cell biology. 2006;174(7):923–9. Epub 2006/09/20. 16982798

39. Campoy EM, Zoppino FC, Colombo MI. The early secretory pathway contributes to the growth of the Coxiella-replicative niche. Infection and immunity. 2011;79(1):402–13. doi: 10.1128/IAI.00688-10 20937765

40. Derre I, Isberg RR. Legionella pneumophila replication vacuole formation involves rapid recruitment of proteins of the early secretory system. Infection and immunity. 2004;72(5):3048–53. Epub 2004/04/23. 15102819

41. Dong N, Zhu Y, Lu Q, Hu L, Zheng Y, Shao F. Structurally distinct bacterial TBC-like GAPs link Arf GTPase to Rab1 inactivation to counteract host defenses. Cell. 2012;150(5):1029–41. Epub 2012/09/04. doi: 10.1016/j.cell.2012.06.050 22939626

42. Hardiman CA, Roy CR. AMPylation is critical for Rab1 localization to vacuoles containing Legionella pneumophila. MBio. 2014;5(1):e01035–13. Epub 2014/02/13. doi: 10.1128/mBio.01035-13 24520063

43. Huang B, Hubber A, McDonough JA, Roy CR, Scidmore MA, Carlyon JA. The Anaplasma phagocytophilum-occupied vacuole selectively recruits Rab-GTPases that are predominantly associated with recycling endosomes. Cellular microbiology. 2010;12(9):1292–307. doi: 10.1111/j.1462-5822.2010.01468.x 20345488

44. Huang J, Birmingham CL, Shahnazari S, Shiu J, Zheng YT, Smith AC, et al. Antibacterial autophagy occurs at PI(3)P-enriched domains of the endoplasmic reticulum and requires Rab1 GTPase. Autophagy. 2011;7(1):17–26. Epub 2010/10/29. 20980813

45. Ingmundson A, Delprato A, Lambright DG, Roy CR. Legionella pneumophila proteins that regulate Rab1 membrane cycling. Nature. 2007;450(7168):365–9. 17952054

46. Kagan JC, Stein MP, Pypaert M, Roy CR. Legionella subvert the functions of Rab1 and Sec22b to create a replicative organelle. The Journal of experimental medicine. 2004;199(9):1201–11. Epub 2004/05/01. 15117975

47. Neunuebel MR, Chen Y, Gaspar AH, Backlund PS Jr., Yergey A, Machner MP. De-AMPylation of the small GTPase Rab1 by the pathogen Legionella pneumophila. Science. 2011;333(6041):453–6. Epub 2011/06/18. doi: 10.1126/science.1207193 21680813

48. Rzomp KA, Scholtes LD, Briggs BJ, Whittaker GR, Scidmore MA. Rab GTPases are recruited to chlamydial inclusions in both a species-dependent and species-independent manner. Infection and immunity. 2003;71(10):5855–70. Epub 2003/09/23. 14500507

49. Hardiman CA, McDonough JA, Newton HJ, Roy CR. The role of Rab GTPases in the transport of vacuoles containing Legionella pneumophila and Coxiella burnetii. Biochemical Society transactions. 2012;40(6):1353–9. doi: 10.1042/BST20120167 23176480

50. Tisdale EJ, Bourne JR, Khosravi-Far R, Der CJ, Balch WE. GTP-binding mutants of rab1 and rab2 are potent inhibitors of vesicular transport from the endoplasmic reticulum to the Golgi complex. The Journal of cell biology. 1992;119(4):749–61. Epub 1992/11/01. 1429835

51. Touchot N, Zahraoui A, Vielh E, Tavitian A. Biochemical properties of the YPT-related rab1B protein. Comparison with rab1A. FEBS letters. 1989;256(1–2):79–84. Epub 1989/10/09. 2509243

52. Plutner H, Cox AD, Pind S, Khosravi-Far R, Bourne JR, Schwaninger R, et al. Rab1b regulates vesicular transport between the endoplasmic reticulum and successive Golgi compartments. The Journal of cell biology. 1991;115(1):31–43. Epub 1991/10/01. 1918138

53. Ao X, Zou L, Wu Y. Regulation of autophagy by the Rab GTPase network. Cell Death Differ. 2014;21(3):348–58. Epub 2014/01/21. doi: 10.1038/cdd.2013.187 24440914

54. Huang J, Brumell JH. Bacteria-autophagy interplay: a battle for survival. Nature reviews Microbiology. 2014. Epub 2014/01/05.

55. Mukhopadhyay A, Quiroz JA, Wolkoff AW. Rab1a regulates sorting of early endocytic vesicles. American journal of physiology Gastrointestinal and liver physiology. 2014;306(5):G412–24. Epub 2014/01/11. doi: 10.1152/ajpgi.00118.2013 24407591

56. Mukhopadhyay A, Nieves E, Che FY, Wang J, Jin L, Murray JW, et al. Proteomic analysis of endocytic vesicles: Rab1a regulates motility of early endocytic vesicles. Journal of cell science. 2011;124(Pt 5):765–75. Epub 2011/02/10. doi: 10.1242/jcs.079020 21303926

57. Arasaki K, Toomre DK, Roy CR. The Legionella pneumophila effector DrrA is sufficient to stimulate SNARE-dependent membrane fusion. Cell host & microbe. 2012;11(1):46–57. Epub 2012/01/24.

58. Mihai Gazdag E, Streller A, Haneburger I, Hilbi H, Vetter IR, Goody RS, et al. Mechanism of Rab1b deactivation by the Legionella pneumophila GAP LepB. EMBO reports. 2013;14(2):199–205. Epub 2013/01/05. doi: 10.1038/embor.2012.211 23288104

59. Muller MP, Peters H, Blumer J, Blankenfeldt W, Goody RS, Itzen A. The Legionella effector protein DrrA AMPylates the membrane traffic regulator Rab1b. Science. 2010;329(5994):946–9. Epub 2010/07/24. doi: 10.1126/science.1192276 20651120

60. Schoebel S, Cichy AL, Goody RS, Itzen A. Protein LidA from Legionella is a Rab GTPase supereffector. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(44):17945–50. doi: 10.1073/pnas.1113133108 22011575

61. Arasaki K, Roy CR. Legionella pneumophila promotes functional interactions between plasma membrane syntaxins and Sec22b. Traffic. 2010;11(5):587–600. Epub 2010/02/19. doi: 10.1111/j.1600-0854.2010.01050.x 20163564

62. Mishra AK, Del Campo CM, Collins RE, Roy CR, Lambright DG. The Legionella pneumophila GTPase activating protein LepB accelerates Rab1 deactivation by a non-canonical hydrolytic mechanism. The Journal of biological chemistry. 2013;288(33):24000–11. Epub 2013/07/04. doi: 10.1074/jbc.M113.470625 23821544

63. Sun Y, Connor MG, Pennington JM, Lawrenz MB. Development of bioluminescent bioreporters for in vitro and in vivo tracking of Yersinia pestis. PloS one. 2012;7(10):e47123. doi: 10.1371/journal.pone.0047123 23071730

64. Agaisse H, Burrack LS, Philips JA, Rubin EJ, Perrimon N, Higgins DE. Genome-wide RNAi screen for host factors required for intracellular bacterial infection. Science. 2005;309(5738):1248–51. Epub 2005/07/16. 16020693

65. Misselwitz B, Dilling S, Vonaesch P, Sacher R, Snijder B, Schlumberger M, et al. RNAi screen of Salmonella invasion shows role of COPI in membrane targeting of cholesterol and Cdc42. Molecular systems biology. 2011;7:474. Epub 2011/03/17. doi: 10.1038/msb.2011.7 21407211

66. Galen JE, Nair J, Wang JY, Wasserman SS, Tanner MK, Sztein MB, et al. Optimization of plasmid maintenance in the attenuated live vector vaccine strain Salmonella typhi CVD 908-htrA. Infection and immunity. 1999;67(12):6424–33. Epub 1999/11/24. 10569759

67. Kagan JC, Roy CR. Legionella phagosomes intercept vesicular traffic from endoplasmic reticulum exit sites. Nature cell biology. 2002;4(12):945–54. 12447391

68. Itoh T, Satoh M, Kanno E, Fukuda M. Screening for target Rabs of TBC (Tre-2/Bub2/Cdc16) domain-containing proteins based on their Rab-binding activity. Genes to cells: devoted to molecular & cellular mechanisms. 2006;11(9):1023–37. Epub 2006/08/23.

69. Satoh A, Wang Y, Malsam J, Beard MB, Warren G. Golgin-84 is a rab1 binding partner involved in Golgi structure. Traffic. 2003;4(3):153–61. 12656988

70. Martinez O, Goud B. Rab proteins. Biochimica et biophysica acta. 1998;1404(1–2):101–12. Epub 1998/08/26. 9714762

71. Horwitz MA, Maxfield FR. Legionella pneumophila inhibits acidification of its phagosome in human monocytes. The Journal of cell biology. 1984;99(6):1936–43. Epub 1984/12/01. 6501409

72. Sturgill-Koszycki S, Swanson MS. Legionella pneumophila replication vacuoles mature into acidic, endocytic organelles. The Journal of experimental medicine. 2000;192(9):1261–72. Epub 2000/11/09. 11067875

73. Brumell JH, Scidmore MA. Manipulation of rab GTPase function by intracellular bacterial pathogens. Microbiology and molecular biology reviews: MMBR. 2007;71(4):636–52. Epub 2007/12/08. 18063721

74. Ortiz Sandoval C, Simmen T. Rab proteins of the endoplasmic reticulum: functions and interactors. Biochemical Society transactions. 2012;40(6):1426–32. Epub 2012/11/28. doi: 10.1042/BST20120158 23176493

75. Liu S, Storrie B. Are Rab proteins the link between Golgi organization and membrane trafficking? Cellular and molecular life sciences: CMLS. 2012;69(24):4093–106. Epub 2012/05/15. doi: 10.1007/s00018-012-1021-6 22581368

76. Thornbrough JM, Hundley T, Valdivia R, Worley MJ. Human genome-wide RNAi screen for host factors that modulate intracellular Salmonella growth. PloS one. 2012;7(6):11.

77. Qin QM, Pei J, Ancona V, Shaw BD, Ficht TA, de Figueiredo P. RNAi screen of endoplasmic reticulum-associated host factors reveals a role for IRE1alpha in supporting Brucella replication. PLoS pathogens. 2008;4(7):e1000110. Epub 2008/07/26. doi: 10.1371/journal.ppat.1000110 18654626

78. Gutierrez MG, Vazquez CL, Munafo DB, Zoppino FC, Beron W, Rabinovitch M, et al. Autophagy induction favours the generation and maturation of the Coxiella-replicative vacuoles. Cellular microbiology. 2005;7(7):981–93. Epub 2005/06/15. 15953030

79. Niu H, Yamaguchi M, Rikihisa Y. Subversion of cellular autophagy by Anaplasma phagocytophilum. Cellular microbiology. 2008;10(3):593–605. Epub 2007/11/06. 17979984

80. Newton HJ, McDonough JA, Roy CR. Effector protein translocation by the Coxiella burnetii Dot/Icm type IV secretion system requires endocytic maturation of the pathogen-occupied vacuole. PloS one. 2013;8(1):e54566. Epub 2013/01/26. doi: 10.1371/journal.pone.0054566 23349930

81. Wieland H, Goetz F, Neumeister B. Phagosomal acidification is not a prerequisite for intracellular multiplication of Legionella pneumophila in human monocytes. The Journal of infectious diseases. 2004;189(9):1610–4. Epub 2004/04/30. 15116296

82. Benes P, Vetvicka V, Fusek M. Cathepsin D—many functions of one aspartic protease. Crit Rev Oncol Hematol. 2008;68(1):12–28. doi: 10.1016/j.critrevonc.2008.02.008 18396408

83. Saftig P, Klumperman J. Lysosome biogenesis and lysosomal membrane proteins: trafficking meets function. Nature reviews Molecular cell biology. 2009;10(9):623–35. Epub 2009/08/13. doi: 10.1038/nrm2745 19672277

84. Xu H, Ren D. Lysosomal Physiology. Annual review of physiology. 2015;77:57–80. Epub 2015/02/11. doi: 10.1146/annurev-physiol-021014-071649 25668017

85. Horenkamp FA, Mukherjee S, Alix E, Schauder CM, Hubber AM, Roy CR, et al. Legionella pneumophila subversion of host vesicular transport by SidC effector proteins. Traffic. 2014;15(5):488–99. Epub 2014/02/04. doi: 10.1111/tra.12158 24483784

86. Machner MP, Isberg RR. Targeting of host Rab GTPase function by the intravacuolar pathogen Legionella pneumophila. Developmental cell. 2006;11(1):47–56. Epub 2006/07/11. 16824952

87. Murata T, Delprato A, Ingmundson A, Toomre DK, Lambright DG, Roy CR. The Legionella pneumophila effector protein DrrA is a Rab1 guanine nucleotide-exchange factor. Nature cell biology. 2006;8(9):971–7. Epub 2006/08/15. 16906144

88. Machner MP, Isberg RR. A bifunctional bacterial protein links GDI displacement to Rab1 activation. Science. 2007;318(5852):974–7. Epub 2007/10/20. 17947549

89. Tan Y, Arnold RJ, Luo ZQ. Legionella pneumophila regulates the small GTPase Rab1 activity by reversible phosphorylcholination. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(52):21212–7. Epub 2011/12/14. doi: 10.1073/pnas.1114023109 22158903

90. Tan Y, Luo ZQ. Legionella pneumophila SidD is a deAMPylase that modifies Rab1. Nature. 2011;475(7357):506–9. Epub 2011/07/08. doi: 10.1038/nature10307 21734656

91. Mukherjee S, Liu X, Arasaki K, McDonough J, Galan JE, Roy CR. Modulation of Rab GTPase function by a protein phosphocholine transferase. Nature. 2011;477(7362):103–6. Epub 2011/08/09. doi: 10.1038/nature10335 21822290

92. Chen Y, Tascon I, Neunuebel MR, Pallara C, Brady J, Kinch LN, et al. Structural basis for Rab1 de-AMPylation by the Legionella pneumophila effector SidD. PLoS pathogens. 2013;9(5):e1003382. Epub 2013/05/23. doi: 10.1371/journal.ppat.1003382 23696742

93. Campanacci V, Mukherjee S, Roy CR, Cherfils J. Structure of the Legionella effector AnkX reveals the mechanism of phosphocholine transfer by the FIC domain. The EMBO journal. 2013;32(10):1469–77. Epub 2013/04/11. doi: 10.1038/emboj.2013.82 23572077

94. Doll JM, Zeitz PS, Ettestad P, Bucholtz AL, Davis T, Gage K. Cat-transmitted fatal pneumonic plague in a person who traveled from Colorado to Arizona. The American journal of tropical medicine and hygiene. 1994;51(1):109–14. Epub 1994/07/01. 8059908

95. Abu Kwaik Y, Eisenstein BI, Engleberg NC. Phenotypic modulation by Legionella pneumophila upon infection of macrophages. Infection and immunity. 1993;61(4):1320–9. Epub 1993/04/01. 8454334

96. Al-Khodor S, Price CT, Habyarimana F, Kalia A, Abu Kwaik Y. A Dot/Icm-translocated ankyrin protein of Legionella pneumophila is required for intracellular proliferation within human macrophages and protozoa. Molecular microbiology. 2008;70(4):908–23. Epub 2008/09/25. doi: 10.1111/j.1365-2958.2008.06453.x 18811729

97. Pedersen LL, Radulic M, Doric M, Abu Kwaik Y. HtrA homologue of Legionella pneumophila: an indispensable element for intracellular infection of mammalian but not protozoan cells. Infection and immunity. 2001;69(4):2569–79. Epub 2001/03/20. 11254621

98. Gibson DG, Young L, Chuang RY, Venter JC, Hutchison CA 3rd, Smith HO. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nature methods. 2009;6(5):343–5. doi: 10.1038/nmeth.1318 19363495

99. Seto S, Tsujimura K, Koide Y. Rab GTPases regulating phagosome maturation are differentially recruited to mycobacterial phagosomes. Traffic. 2011;12(4):407–20. doi: 10.1111/j.1600-0854.2011.01165.x 21255211

100. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods (San Diego, Calif). 2001;25(4):402–8. Epub 2002/02/16.

101. Kinder SA, Badger JL, Bryant GO, Pepe JC, Miller VL. Cloning of the YenI restriction endonuclease and methyltransferase from Yersinia enterocolitica serotype O8 and construction of a transformable R-M+ mutant. Gene. 1993;136(1–2):271–5. Epub 1993/12/22. 8294016

102. Chain PS, Carniel E, Larimer FW, Lamerdin J, Stoutland PO, Regala WM, et al. Insights into the evolution of Yersinia pestis through whole-genome comparison with Yersinia pseudotuberculosis. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(38):13826–31. Epub 2004/09/11. 15358858

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 10
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#