#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Circumventing . Virulence by Early Recruitment of Neutrophils to the Lungs during Pneumonic Plague


The pathogen Yersinia pestis is the causative agent of pneumonic plague, as well as a potential bioweapon. The nature of this disease involves an initial non-inflammatory phase where the influx of neutrophils to the lungs is suppressed, allowing bacterial propagation in this organ. Using the mouse model of pneumonic plague, we demonstrate that the early expression of neutrophil chemoattractants and adhesion molecules in the lungs is delayed concomitant with a delayed recruitment of neutrophils to the lung. We also show that the Y. pestis virulence factor YopJ is involved in the early suppression of chemoattractants mRNA expression in the lung early after infection, but it seems that additional Y. pestis factors interfere with the protein synthesis of these chemoattractants. Indeed, administration of recombinant KC and MIP-2 to the infected lung of G-CSF treated mice restored the early neutrophil influx to the lungs, leading to a significant reduction in bacterial burden. The treatment has also proved efficacious in reducing mortality. This study highlights the complex virulence mechanisms employed by Y. pestis to diminish the early homing of neutrophils to the lungs thereby allowing bacterial propagation and disease progression.


Vyšlo v časopise: Circumventing . Virulence by Early Recruitment of Neutrophils to the Lungs during Pneumonic Plague. PLoS Pathog 11(5): e32767. doi:10.1371/journal.ppat.1004893
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004893

Souhrn

The pathogen Yersinia pestis is the causative agent of pneumonic plague, as well as a potential bioweapon. The nature of this disease involves an initial non-inflammatory phase where the influx of neutrophils to the lungs is suppressed, allowing bacterial propagation in this organ. Using the mouse model of pneumonic plague, we demonstrate that the early expression of neutrophil chemoattractants and adhesion molecules in the lungs is delayed concomitant with a delayed recruitment of neutrophils to the lung. We also show that the Y. pestis virulence factor YopJ is involved in the early suppression of chemoattractants mRNA expression in the lung early after infection, but it seems that additional Y. pestis factors interfere with the protein synthesis of these chemoattractants. Indeed, administration of recombinant KC and MIP-2 to the infected lung of G-CSF treated mice restored the early neutrophil influx to the lungs, leading to a significant reduction in bacterial burden. The treatment has also proved efficacious in reducing mortality. This study highlights the complex virulence mechanisms employed by Y. pestis to diminish the early homing of neutrophils to the lungs thereby allowing bacterial propagation and disease progression.


Zdroje

1. Craig A, Mai J, Cai S, Jeyaseelan S. Neutrophil recruitment to the lungs during bacterial pneumonia. Infect Immun. 2009;77(2):568–75. Epub 2008/11/19. IAI.00832-08 [pii]. 19015252; PubMed Central PMCID: PMC2632043. doi: 10.1128/IAI.00832-08

2. King KY, Goodell MA. Inflammatory modulation of HSCs: viewing the HSC as a foundation for the immune response. Nat Rev Immunol. 2011;11(10):685–92. Epub 2011/09/10. doi: 10.1038/nri3062 21904387

3. Rogers HW, Unanue ER. Neutrophils are involved in acute, nonspecific resistance to Listeria monocytogenes in mice. Infect Immun. 1993;61(12):5090–6. 8225586

4. Borregaard N. Neutrophils, from marrow to microbes. Immunity. 2010;33(5):657–70. doi: 10.1016/j.immuni.2010.11.011 21094463

5. Bozic CR, Kolakowski LF Jr., Gerard NP, Garcia-Rodriguez C, von Uexkull-Guldenband C, Conklyn MJ, et al. Expression and biologic characterization of the murine chemokine KC. J Immunol. 1995;154(11):6048–57. Epub 1995/06/01. 7751647

6. Strieter RM, Kunkel SL. Acute lung injury: the role of cytokines in the elicitation of neutrophils. J Investig Med. 1994;42(4):640–51. Epub 1994/12/01. 8521027

7. Williams MR, Azcutia V, Newton G, Alcaide P, Luscinskas FW. Emerging mechanisms of neutrophil recruitment across endothelium. Trends Immunol. 2011;32(10):461–9. Epub 2011/08/16. doi: 10.1016/j.it.2011.06.009 21839681

8. Mocsai A. Diverse novel functions of neutrophils in immunity, inflammation, and beyond. J Exp Med. 2013;210(7):1283–99. Epub 2013/07/05. doi: 10.1084/jem.20122220 23825232

9. Mizgerd JP. Acute Lower Respiratory Tract Infection. New England Journal of Medicine. 2008;358(7):716–27. doi: 10.1056/NEJMra074111 18272895

10. Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006;6(3):173–82. Epub 2006/02/25. 16498448

11. Tsai WC, Strieter RM, Mehrad B, Newstead MW, Zeng X, Standiford TJ. CXC chemokine receptor CXCR2 is essential for protective innate host response in murine Pseudomonas aeruginosa pneumonia. Infect Immun. 2000;68(7):4289–96. 10858247

12. Tateda K, Moore TA, Newstead MW, Tsai WC, Zeng X, Deng JC, et al. Chemokine-dependent neutrophil recruitment in a murine model of Legionella pneumonia: potential role of neutrophils as immunoregulatory cells. Infect Immun. 2001;69(4):2017–24. Epub 2001/03/20. 11254553

13. Matsuzaki G, Umemura M. Interleukin-17 as an effector molecule of innate and acquired immunity against infections. Microbiol Immunol. 2007;51(12):1139–47. 18094532

14. Eisele NA, Lee-Lewis H, Besch-Williford C, Brown CR, Anderson DM. Chemokine receptor CXCR2 mediates bacterial clearance rather than neutrophil recruitment in a murine model of pneumonic plague. Am J Pathol. 2011;178(3):1190–200. doi: 10.1016/j.ajpath.2010.11.067 21356370

15. Laws TR, Davey MS, Titball RW, Lukaszewski R. Neutrophils are important in early control of lung infection by Yersinia pestis. Microbes Infect. 2010;12(4):331–5. doi: 10.1016/j.micinf.2010.01.007 20114086

16. Perry RD, Fetherston JD. Yersinia pestis—etiologic agent of plague. Clin Microbiol Rev. 1997;10(1):35–66. 8993858

17. Kool JL. Risk of person-to-person transmission of pneumonic plague. Clin Infect Dis. 2005;40(8):1166–72. 15791518

18. Pollitzer R. Plague studies. IX. Epidemiology. Bull World Health Organ. 1954;9(1):131–70.

19. 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

20. Agar SL, Sha J, Foltz SM, Erova TE, Walberg KG, Parham TE, et al. Characterization of a mouse model of plague after aerosolization of Yersinia pestis CO92. Microbiology. 2008;154(Pt 7):1939–48. doi: 10.1099/mic.0.2008/017335-0 18599822

21. Bubeck SS, Cantwell AM, Dube PH. Delayed inflammatory response to primary pneumonic plague occurs in both outbred and inbred mice. Infect Immun. 2007;75(2):697–705. 17101642

22. Lathem WW, Crosby SD, Miller VL, Goldman WE. Progression of primary pneumonic plague: a mouse model of infection, pathology, and bacterial transcriptional activity. Proc Natl Acad Sci U S A. 2005;102(49):17786–91. 16306265

23. Price PA, Jin J, Goldman WE. Pulmonary infection by Yersinia pestis rapidly establishes a permissive environment for microbial proliferation. Proc Natl Acad Sci U S A. 2012;109(8):3083–8. doi: 10.1073/pnas.1112729109 22308352

24. Cornelis GR, Wolf-Watz H. The Yersinia Yop virulon: a bacterial system for subverting eukaryotic cells. Mol Microbiol. 1997;23(5):861–7. Epub 1997/03/01. 9076724

25. Mota LJ, Cornelis GR. The bacterial injection kit: type III secretion systems. Ann Med. 2005;37(4):234–49. Epub 2005/07/16. 16019722

26. Viboud GI, Bliska JB. Yersinia outer proteins: role in modulation of host cell signaling responses and pathogenesis. Annu Rev Microbiol. 2005;59:69–89. Epub 2005/04/26. 15847602

27. Shannon JG, Hasenkrug AM, Dorward DW, Nair V, Carmody AB, Hinnebusch BJ. Yersinia pestis subverts the dermal neutrophil response in a mouse model of bubonic plague. MBio. 2013;4(5):e00170–13. Epub 2013/08/29. doi: 10.1128/mBio.00170-13 23982068

28. Welkos S, Friedlander A, McDowell D, Weeks J, Tobery S. V antigen of Yersinia pestis inhibits neutrophil chemotaxis. Microb Pathog. 1998;24(3):185–96. 9514641

29. Pechous RD, Sivaraman V, Price PA, Stasulli NM, Goldman WE. Early host cell targets of Yersinia pestis during primary pneumonic plague. PLoS Pathog. 2013;9(10):e1003679. Epub 2013/10/08. doi: 10.1371/journal.ppat.1003679 24098126

30. Vagima Y, Levy Y, Gur D, Tidhar A, Aftalion M, Abramovich H, et al. Early sensing of Yersinia pestis airway infection by bone marrow cells. Front Cell Infect Microbiol. 2012;2:143. Epub 2012/11/29. doi: 10.3389/fcimb.2012.00143 23189271

31. Greenlee KJ, Werb Z, Kheradmand F. Matrix metalloproteinases in lung: multiple, multifarious, and multifaceted. Physiol Rev. 2007;87(1):69–98. Epub 2007/01/24. 17237343

32. Furze RC, Rankin SM. Neutrophil mobilization and clearance in the bone marrow. Immunology. 2008;125(3):281–8. Epub 2009/01/09. IMM2950 [pii]. 19128361; PubMed Central PMCID: PMC2669132. doi: 10.1111/j.1365-2567.2008.02950.x

33. Bosio CM, Goodyear AW, Dow SW. Early interaction of Yersinia pestis with APCs in the lung. J Immunol. 2005;175(10):6750–6. Epub 2005/11/08. 16272331

34. Aird WC. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circ Res. 2007;100(2):158–73. Epub 2007/02/03. 17272818

35. Bliska JB, Wang X, Viboud GI, Brodsky IE. Modulation of innate immune responses by Yersinia type III secretion system translocators and effectors. Cell Microbiol. 2013;15(10):1622–31. Epub 2013/07/10. doi: 10.1111/cmi.12164 23834311

36. Navarro L, Alto NM, Dixon JE. Functions of the Yersinia effector proteins in inhibiting host immune responses. Curr Opin Microbiol. 2005;8(1):21–7. Epub 2005/02/08. 15694853

37. Trosky JE, Liverman AD, Orth K. Yersinia outer proteins: Yops. Cell Microbiol. 2008;10(3):557–65. Epub 2007/12/18. doi: 10.1111/j.1462-5822.2007.01109.x 18081726.

38. Phillipson M, Kubes P. The neutrophil in vascular inflammation. Nat Med. 2011;17(11):1381–90. Epub 2011/11/09. doi: 10.1038/nm.2514 22064428

39. Scott DW, Patel RP. Endothelial heterogeneity and adhesion molecules N-glycosylation: implications in leukocyte trafficking in inflammation. Glycobiology. 2013;23(6):622–33. Epub 2013/03/01. doi: 10.1093/glycob/cwt014 23445551

40. Denecker G, Totemeyer S, Mota LJ, Troisfontaines P, Lambermont I, Youta C, et al. Effect of low- and high-virulence Yersinia enterocolitica strains on the inflammatory response of human umbilical vein endothelial cells. Infect Immun. 2002;70(7):3510–20. Epub 2002/06/18. 12065490

41. Cornelis GR. Yersinia type III secretion: send in the effectors. J Cell Biol. 2002;158(3):401–8. 12163464

42. Viboud GI, Mejia E, Bliska JB. Comparison of YopE and YopT activities in counteracting host signalling responses to Yersinia pseudotuberculosis infection. Cell Microbiol. 2006;8(9):1504–15. Epub 2006/08/23. 16922868

43. Viboud GI, So SS, Ryndak MB, Bliska JB. Proinflammatory signalling stimulated by the type III translocation factor YopB is counteracted by multiple effectors in epithelial cells infected with Yersinia pseudotuberculosis. Mol Microbiol. 2003;47(5):1305–15. 12603736

44. Palmer LE, Hobbie S, Galan JE, Bliska JB. YopJ of Yersinia pseudotuberculosis is required for the inhibition of macrophage TNF-alpha production and downregulation of the MAP kinases p38 and JNK. Mol Microbiol. 1998;27(5):953–65. Epub 1998/04/16. 9535085

45. Ruckdeschel K. Immunomodulation of macrophages by pathogenic Yersinia species. Arch Immunol Ther Exp (Warsz). 2002;50(2):131–7. Epub 2002/05/23. 12022702

46. Zauberman A, Cohen S, Mamroud E, Flashner Y, Tidhar A, Ber R, et al. Interaction of Yersinia pestis with macrophages: limitations in YopJ-dependent apoptosis. Infect Immun. 2006;74(6):3239–50. Epub 2006/05/23. 16714551

47. Zhou L, Tan A, Hershenson MB. Yersinia YopJ inhibits pro-inflammatory molecule expression in human bronchial epithelial cells. Respir Physiol Neurobiol. 2004;140(1):89–97. Epub 2004/04/28. 15109931

48. Sauvonnet N, Lambermont I, van der Bruggen P, Cornelis GR. YopH prevents monocyte chemoattractant protein 1 expression in macrophages and T-cell proliferation through inactivation of the phosphatidylinositol 3-kinase pathway. Mol Microbiol. 2002;45(3):805–15. Epub 2002/07/26. 12139625

49. Cantwell AM, Bubeck SS, Dube PH. YopH inhibits early pro-inflammatory cytokine responses during plague pneumonia. BMC Immunol. 2010;11:29. Epub 2010/06/23. doi: 10.1186/1471-2172-11-29 20565713

50. Orth K, Xu Z, Mudgett MB, Bao ZQ, Palmer LE, Bliska JB, et al. Disruption of signaling by Yersinia effector YopJ, a ubiquitin-like protein protease. Science. 2000;290(5496):1594–7. Epub 2000/11/25. 11090361

51. Zhou H, Monack DM, Kayagaki N, Wertz I, Yin J, Wolf B, et al. Yersinia virulence factor YopJ acts as a deubiquitinase to inhibit NF-kappa B activation. J Exp Med. 2005;202(10):1327–32. Epub 2005/11/23. 16301742

52. Mittal R, Peak-Chew SY, McMahon HT. Acetylation of MEK2 and I kappa B kinase (IKK) activation loop residues by YopJ inhibits signaling. Proc Natl Acad Sci U S A. 2006;103(49):18574–9. Epub 2006/11/23. 17116858

53. Mukherjee S, Keitany G, Li Y, Wang Y, Ball HL, Goldsmith EJ, et al. Yersinia YopJ acetylates and inhibits kinase activation by blocking phosphorylation. Science. 2006;312(5777):1211–4. Epub 2006/05/27. 16728640

54. Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 1999;18(49):6853–66. Epub 1999/12/22. 10602461

55. Tebo JM, Datta S, Kishore R, Kolosov M, Major JA, Ohmori Y, et al. Interleukin-1-mediated stabilization of mouse KC mRNA depends on sequences in both 5'- and 3'-untranslated regions. J Biol Chem. 2000;275(17):12987–93. Epub 2000/04/25. 10777600

56. Mizgerd JP, Lupa MM, Spieker MS. NF-kappaB p50 facilitates neutrophil accumulation during LPS-induced pulmonary inflammation. BMC Immunol. 2004;5:10. Epub 2004/06/11. 15189567

57. Lemaitre N, Sebbane F, Long D, Hinnebusch BJ. Yersinia pestis YopJ suppresses tumor necrosis factor alpha induction and contributes to apoptosis of immune cells in the lymph node but is not required for virulence in a rat model of bubonic plague. Infect Immun. 2006;74(9):5126–31. Epub 2006/08/24. 16926404

58. Straley SC, Bowmer WS. Virulence genes regulated at the transcriptional level by Ca2+ in Yersinia pestis include structural genes for outer membrane proteins. Infect Immun. 1986;51(2):445–54. Epub 1986/02/01. 3002984

59. Zauberman A, Velan B, Mamroud E, Flashner Y, Shafferman A, Cohen S. Disparity between Yersinia pestis and Yersinia enterocolitica O:8 in YopJ/YopP-dependent functions. Adv Exp Med Biol. 2007;603:312–20. Epub 2007/10/31. 17966427

60. Lathem WW, Price PA, Miller VL, Goldman WE. A plasminogen-activating protease specifically controls the development of primary pneumonic plague. Science. 2007;315(5811):509–13. Epub 2007/01/27. 17255510

61. Caulfield AJ, Walker ME, Gielda LM, Lathem WW. The Pla protease of Yersinia pestis degrades fas ligand to manipulate host cell death and inflammation. Cell Host Microbe. 2014;15(4):424–34. Epub 2014/04/12. doi: 10.1016/j.chom.2014.03.005 24721571

62. Gorgen I, Hartung T, Leist M, Niehorster M, Tiegs G, Uhlig S, et al. Granulocyte colony-stimulating factor treatment protects rodents against lipopolysaccharide-induced toxicity via suppression of systemic tumor necrosis factor-alpha. J Immunol. 1992;149(3):918–24. Epub 1992/08/01. 1378868

63. Roberts AW. G-CSF: a key regulator of neutrophil production, but that's not all! Growth Factors. 2005;23(1):33–41. Epub 2005/07/16. 16019425

64. Andreasen C, Carbonetti NH. Pertussis toxin inhibits early chemokine production to delay neutrophil recruitment in response to Bordetella pertussis respiratory tract infection in mice. Infect Immun. 2008;76(11):5139–48. Epub 2008/09/04. doi: 10.1128/IAI.00895-08 18765723

65. Sun K, Salmon SL, Lotz SA, Metzger DW. Interleukin-12 promotes gamma interferon-dependent neutrophil recruitment in the lung and improves protection against respiratory Streptococcus pneumoniae infection. Infect Immun. 2007;75(3):1196–202. Epub 2007/01/11. 17210665

66. Bi Y, Zhou J, Yang H, Wang X, Zhang X, Wang Q, et al. IL-17A produced by neutrophils protects against pneumonic plague through orchestrating IFN-gamma-activated macrophage programming. J Immunol. 2014;192(2):704–13. Epub 2013/12/18. doi: 10.4049/jimmunol.1301687 24337746

67. Soehnlein O. Direct and alternative antimicrobial mechanisms of neutrophil-derived granule proteins. J Mol Med (Berl). 2009;87(12):1157–64. Epub 2009/07/31. doi: 10.1007/s00109-009-0508-6 19641860

68. Soehnlein O, Zernecke A, Weber C. Neutrophils launch monocyte extravasation by release of granule proteins. Thromb Haemost. 2009;102(2):198–205. Epub 2009/08/05. doi: 10.1160/TH08-11-0720 19652869

69. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13(3):159–75. Epub 2013/02/26. doi: 10.1038/nri3399 23435331

70. Galimand M, Carniel E, Courvalin P. Resistance of Yersinia pestis to antimicrobial agents. Antimicrob Agents Chemother. 2006;50(10):3233–6. Epub 2006/09/29. 17005799

71. Ben-Gurion R, Shafferman A. Essential virulence determinants of different Yersinia species are carried on a common plasmid. Plasmid. 1981;5(2):183–7. Epub 1981/03/01. 7243971

72. Flashner Y, Mamroud E, Tidhar A, Ber R, Aftalion M, Gur D, et al. Generation of Yersinia pestis attenuated strains by signature-tagged mutagenesis in search of novel vaccine candidates. Infect Immun. 2004;72(2):908–15. Epub 2004/01/27. 14742535

73. Zauberman A, Tidhar A, Levy Y, Bar-Haim E, Halperin G, Flashner Y, et al. Yersinia pestis endowed with increased cytotoxicity is avirulent in a bubonic plague model and induces rapid protection against pneumonic plague. PLoS One. 2009;4(6):e5938. doi: 10.1371/journal.pone.0005938 19529770

74. Flashner Y, Mamroud E, Tidhar A, Ber R, Aftalion M, Gur D, et al. Identification of genes involved in Yersinia pestis virulence by signature-tagged mutagenesis. Adv Exp Med Biol. 2003;529:31–3. Epub 2003/05/22. 12756723

75. Datsenko KA, Wanner BL. One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A. 2000;97(12):6640–5. Epub 2000/06/01. 10829079

76. Derbise A, Lesic B, Dacheux D, Ghigo JM, Carniel E. A rapid and simple method for inactivating chromosomal genes in Yersinia. FEMS Immunol Med Microbiol. 2003;38(2):113–6. Epub 2003/09/18. 13129645

77. Tidhar A, Flashner Y, Cohen S, Levi Y, Zauberman A, Gur D, et al. The NlpD lipoprotein is a novel Yersinia pestis virulence factor essential for the development of plague. PLoS One. 2009;4(9):e7023. doi: 10.1371/journal.pone.0007023 19759820

78. Reed LJ MH. A simple method of estimating fifty percent endpoints. TheAmerican Journal of Hygiene. 1938;27:493–7.

79. Lin KY, Guarnieri FG, Staveley-O'Carroll KF, Levitsky HI, August JT, Pardoll DM, et al. Treatment of established tumors with a novel vaccine that enhances major histocompatibility class II presentation of tumor antigen. Cancer Res. 1996;56(1):21–6. Epub 1996/01/01. 8548765

80. Vagima Y, Avigdor A, Goichberg P, Shivtiel S, Tesio M, Kalinkovich A, et al. MT1-MMP and RECK are involved in human CD34+ progenitor cell retention, egress, and mobilization. J Clin Invest. 2009;119(3):492–503. doi: 10.1172/JCI36541 19197139

81. Vagima Y, Lapid K, Kollet O, Goichberg P, Alon R, Lapidot T. Pathways implicated in stem cell migration: the SDF-1/CXCR4 axis. Methods Mol Biol. 2011;750:277–89. Epub 2011/05/28. doi: 10.1007/978-1-61779-145-1_19 21618098

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 5
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#