#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Induces the Premature Death of Human Neutrophils through the Action of Its Lipopolysaccharide


The absence of obvious clinical symptoms during the early stages of brucellosis is linked to the Brucella stealthy strategy and its non-canonical PAMPs, which are low PRRs agonists. Still, there are clinical profiles that require explanation. For instance ‒despite the fact that neutrophils readily ingest Brucella during the onset of infection, brucellosis courses without neutrophilia, and just a low number of infected neutrophils are present in target organs. In the chronic phases, a significant proportion of the patients display absolute neutropenia and bone marrow pancytopenia linked to the myeloid cell linage. Examination of the Brucella infected bone marrow reveals granulomas and phagocytosis of myeloid cells. Based on these observations we explored the fate of native neutrophils during their interaction with Brucella. We found that the bacterium induces the premature cell death of neutrophils without inducing proinflammatory phenotypic changes. This event was reproduced by the lipid A of the Brucella LPS and depends on NADPH-oxidase activation and low ROS formation. We believe that this phenomenon explains ‒at least in part‒ the hematological and histological profiles observed during brucellosis. In addition, it may be that dying Brucella-infected neutrophils serve as “Trojan horse” vehicles for infecting phagocytic cells without promoting activation.


Vyšlo v časopise: Induces the Premature Death of Human Neutrophils through the Action of Its Lipopolysaccharide. PLoS Pathog 11(5): e32767. doi:10.1371/journal.ppat.1004853
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004853

Souhrn

The absence of obvious clinical symptoms during the early stages of brucellosis is linked to the Brucella stealthy strategy and its non-canonical PAMPs, which are low PRRs agonists. Still, there are clinical profiles that require explanation. For instance ‒despite the fact that neutrophils readily ingest Brucella during the onset of infection, brucellosis courses without neutrophilia, and just a low number of infected neutrophils are present in target organs. In the chronic phases, a significant proportion of the patients display absolute neutropenia and bone marrow pancytopenia linked to the myeloid cell linage. Examination of the Brucella infected bone marrow reveals granulomas and phagocytosis of myeloid cells. Based on these observations we explored the fate of native neutrophils during their interaction with Brucella. We found that the bacterium induces the premature cell death of neutrophils without inducing proinflammatory phenotypic changes. This event was reproduced by the lipid A of the Brucella LPS and depends on NADPH-oxidase activation and low ROS formation. We believe that this phenomenon explains ‒at least in part‒ the hematological and histological profiles observed during brucellosis. In addition, it may be that dying Brucella-infected neutrophils serve as “Trojan horse” vehicles for infecting phagocytic cells without promoting activation.


Zdroje

1. Pillay J, den Braber I, Vrisekoop N, Kwast LM, de Boer RJ, Borghans JAM, et al. In vivo labeling with 2H2O reveals a human neutrophil lifespan of 5.4 days. Blood. 2010;116: 625–7. doi: 10.1182/blood-2010-01-259028 20410504

2. Payne CM, Glasser L, Tischler ME, Wyckoff D, Cromey D, Fiederlein R, et al. Programmed cell death of the normal human neutrophil: an in vitro model of senescence. Microsc Res Tech. 1994;28: 327–44. 7919520

3. Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity. 2005;22: 285–94. 15780986

4. Savill JS, Wyllie AH, Henson JE, Walport MJ, Henson PM, Haslett C. Macrophage phagocytosis of aging neutrophils in inflammation. Programmed cell death in the neutrophil leads to its recognition by macrophages. J Clin Invest. 1989;83: 865–75. 2921324

5. Nauseef WM. How human neutrophils kill and degrade microbes: an integrated view. Immunol Rev. 2007;219: 88–102. 17850484

6. Janeway CA. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol. 1989;54 Pt 1: 1–13.

7. McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CCM, et al. Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science. 2010;330: 362–6. doi: 10.1126/science.1195491 20947763

8. Anwar S, Whyte MKB. Neutrophil apoptosis in infectious disease. Exp Lung Res. 2007;33: 519–28. 18075826

9. DeLeo FR. Modulation of phagocyte apoptosis by bacterial pathogens. Apoptosis. 2004;9: 399–413. 15192322

10. Elliott MR, Ravichandran KS. Clearance of apoptotic cells: implications in health and disease. J Cell Biol. 2010;189: 1059–70. doi: 10.1083/jcb.201004096 20584912

11. François M, Le Cabec V, Dupont MA, Sansonetti PJ, Maridonneau-Parini I. Induction of necrosis in human neutrophils by Shigella flexneri requires type III secretion, IpaB and IpaC invasins, and actin polymerization. Infect Immun. 2000;68: 1289–96. 10678940

12. Usher LR, Lawson RA, Geary I, Taylor CJ, Bingle CD, Taylor GW, et al. Induction of neutrophil apoptosis by the Pseudomonas aeruginosa exotoxin pyocyanin: a potential mechanism of persistent infection. J Immunol. 2002;168: 1861–8. 11823520

13. Dacheux D, Toussaint B, Richard M, Brochier G, Croize J, Attree I. Pseudomonas aeruginosa cystic fibrosis isolates induce rapid, type III secretion-dependent, but ExoU-independent, oncosis of macrophages and polymorphonuclear neutrophils. Infect Immun. 2000;68: 2916–24. 10768989

14. Scaife H, Woldehiwet Z, Hart CA, Edwards SW. Anaplasma phagocytophilum reduces neutrophil apoptosis in vivo. Infect Immun. 2003;71: 1995–2001. 12654818

15. Van Zandbergen G, Gieffers J, Kothe H, Rupp J, Bollinger A, Aga E, et al. Chlamydia pneumoniae multiply in neutrophil granulocytes and delay their spontaneous apoptosis. J Immunol. 2004;172: 1768–1776. 14734760

16. Barquero-Calvo E, Chaves-Olarte E, Weiss DS, Guzmán-Verri C, Chacón-Díaz C, Rucavado A, et al. Brucella abortus uses a stealthy strategy to avoid activation of the innate immune system during the onset of infection. PLoS One. 2007;2: e631. 17637846

17. Martirosyan A, Moreno E, Gorvel J-P. An evolutionary strategy for a stealthy intracellular Brucella pathogen. Immunol Rev. 2011;240: 211–34. doi: 10.1111/j.1600-065X.2010.00982.x 21349096

18. Barquero-Calvo E, Conde-Alvarez R, Chacón-Díaz C, Quesada-Lobo L, Martirosyan A, Guzmán-Verri C, et al. The differential interaction of Brucella and Ochrobactrum with innate immunity reveals traits related to the evolution of stealthy pathogens. PLoS One. 2009;4: e5893. doi: 10.1371/journal.pone.0005893 19529776

19. Barquero-Calvo E, Martirosyan A, Ordoñez-Rueda D, Arce-Gorvel V, Alfaro-Alarcón A, Lepidi H, et al. Neutrophils exert a suppressive effect on Th1 responses to intracellular pathogen Brucella abortus. PLoS Pathog. 2013;9: e1003167. doi: 10.1371/journal.ppat.1003167 23458832

20. Gorvel JP, Moreno E. Brucella intracellular life: from invasion to intracellular replication. Vet Microbiol. 2002;90: 281–97. 12414149

21. Roop RM, Gaines JM, Anderson ES, Caswell CC, Martin DW. Survival of the fittest: how Brucella strains adapt to their intracellular niche in the host. Med Microbiol Immunol. 2009;198: 221–38. doi: 10.1007/s00430-009-0123-8 19830453

22. Gross A, Terraza A, Ouahrani-Bettache S, Liautard JP, Dornand J. In vitro Brucella suis infection prevents the programmed cell death of human monocytic cells. Infect Immun. 2000;68: 342–51. 10603407

23. Braude AI. Studies in the pathology and pathogenesis of experimental brucellosis. II. The formation of the hepatic granuloma and its evolution. J Infect Dis. 1951;89: 87–94. 14861465

24. Ackermann MR, Cheville NF, Deyoe BL. Bovine ileal dome lymphoepithelial cells: endocytosis and transport of Brucella abortus strain 19. Vet Pathol. 1988;25: 28–35. 3125659

25. Kreutzer DL, Dreyfus L a, Robertson DC. Interaction of polymorphonuclear leukocytes with smooth and rough strains of Brucella abortus. Infect Immun. 1979;23: 737–42. 110680

26. Martínez de Tejada G, Pizarro-Cerdá J, Moreno E, Moriyón I. The outer membranes of Brucella spp. are resistant to bactericidal cationic peptides. Infect Immun. 1995;63: 3054–61. 7622230

27. Crosby E, Llosa L, Miro Quesada M, Carrillo C, Gotuzzo E. Hematologic changes in brucellosis. J Infect Dis. 1984;150: 419–24. 6481187

28. Ruiz-Castañeda M. Brucelosis. Tercera ed. Ediciones científcas, editor. Mexico, D.F.: La Prensa Médica Mexicana, S.A.; 1986.

29. Copin R, Vitry M-A, Hanot Mambres D, Machelart A, De Trez C, Vanderwinden J-M, et al. In situ microscopy analysis reveals local innate immune response developed around Brucella infected cells in resistant and susceptible mice. PLoS Pathog. 2012;8: e1002575. doi: 10.1371/journal.ppat.1002575 22479178

30. Prouty CC. Studies on the leucocyte content of milk drawn from Brucella abortus infected udders. J Bacteriol. 1934;27: 293–301. 16559701

31. Riley LK, Robertson DC. Ingestion and intracellular survival of Brucella abortus in human and bovine polymorphonuclear leukocytes. Infect Immun. 1984;46: 224–30. 6090315

32. Orduña A, Orduña C, Eiros JM, Bratos MA, Gutiérrez P, Alonso P, et al. Inhibition of the degranulation and myeloperoxidase activity of human polymorphonuclear neutrophils by Brucella melitensis. Microbiología. 1991;7: 113–9. doi: 10.1128/IAI.01162-10 21300774

33. Forestier C, Moreno E, Pizarro-Cerda J, Gorvel J-P. Lysosomal accumulation and recycling of lipopolysaccharide to the cell surface of murine macrophages, an in vitro and in vivo study. J Immunol. 1999;162: 6784–6791. 10352299

34. Moreno E, Gorvel J-P. Invasion, intracellular trafficking and replication of Brucella organisms in professional and non-professional phagocytes. In: López-Goñi I, Moriyón I, editors. Brucella: Molecular and Cellular Biology. United Kingdom: Horizon Scientific Press; 2004. pp. 287–312.

35. Forestier C, Deleuil F, Lapaque N, Moreno E, Gorvel JP. Brucella abortus lipopolysaccharide in murine peritoneal macrophages acts as a down-regulator of T cell activation. J Immunol. 2000;165: 5202–10. 11046053

36. Chaves-Olarte E, Altamirano-Silva P, Guzmán-Verri C, Moreno E. Purification of intracellular bacteria: isolation of viable Brucella abortus from host cells. Host-Bacteria Interactions. Methods Protoc. 2014;1197:245–60 doi: 10.1007/978-1-4939-1261-2_14 25172285

37. Riley LK, Robertson DC. Brucellacidal activity of human and bovine polymorphonuclear leukocyte granule extracts against smooth and rough strains of Brucella abortus. Infect Immun. 1984;46: 231–236. 6090316

38. Salcedo SP, Marchesini MI, Lelouard H, Fugier E, Jolly G, Balor S, et al. Brucella control of dendritic cell maturation is dependent on the TIR-containing protein Btp1. PLoS Pathog. 2008;4: e21. doi: 10.1371/journal.ppat.0040021 18266466

39. Caroff M, Bundle DR, Perry MB. Structure of the O-chain of the phenol-phase soluble cellular lipopolysaccharide of Yersinia enterocolitica serotype O:9. Eur J Biochem. 1984;139: 195–200. 6199199

40. Caroff M, Bundle DR, Perry MB, Cherwonogrodzky JW, Duncan JR. Antigenic S-type lipopolysaccharide of Brucella abortus 1119–3. Infect Immun. 1984;46: 384–8. 6437981

41. Pérez-Gutiérrez C, Llobet E, Llompart CM, Reinés M, Bengoechea JA. Role of lipid A acylation in Yersinia enterocolitica virulence. Infect Immun. 2010;78: 2768–81. doi: 10.1128/IAI.01417-09 20385763

42. Iriarte M, González D, Delrue RM, Monreal D, Conde R, López-Goñi I, et al. Brucella lipopolysaccharide: structure, biosynthesis and genetics. Brucella: Molecular and Cellular Biology. Wymondham, UK: Horizon Bioscience; 2004. pp. 159–191.

43. Conde-Álvarez R, Arce-Gorvel V, Iriarte M, Manček-Keber M, Barquero-Calvo E, Palacios-Chaves L, et al. The lipopolysaccharide core of Brucella abortus acts as a shield against innate immunity recognition. PLoS Pathog. 2012;8: e1002675. doi: 10.1371/journal.ppat.1002675 22589715

44. Kubler-Kielb J, Vinogradov E. The study of the core part and non-repeating elements of the O-antigen of Brucella lipopolysaccharide. Carbohydr Res. 2013;366: 33–7. doi: 10.1016/j.carres.2012.11.004 23261780

45. Velasco J, Bengoechea JA, Brandenburg K, Lindner B, Seydel U, González D, et al. Brucella abortus and its closest phylogenetic relative, Ochrobactrum spp., differ in outer membrane permeability and cationic peptide resistance. Infect Immun. 2000;68: 3210–8. 10816465

46. Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003;21: 335–76. 12524386

47. Sabroe I, Dower SK, Whyte MKB. The role of Toll-like receptors in the regulation of neutrophil migration, activation, and apoptosis. Clin Infect Dis. Oxford University Press; 2005;41 Suppl 7: S421–6. 16237641

48. Zanoni I, Ostuni R, Capuano G, Collini M, Caccia M, Ronchi AE, et al. CD14 regulates the dendritic cell life cycle after LPS exposure through NFAT activation. Nature. 2009;460: 264–8. doi: 10.1038/nature08118 19525933

49. Rasool O, Freer E, Moreno E, Jarstrand C. Effect of Brucella abortus lipopolysaccharide on oxidative metabolism and lysozyme release by human neutrophils. Infect Immun. 1992;60: 1699–702. 1548094

50. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303: 1532–5. 15001782

51. Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, et al. Novel cell death program leads to neutrophil extracellular traps. J Cell Biol. 2007;176: 231–41. 17210947

52. Geering B, Simon H-U. Peculiarities of cell death mechanisms in neutrophils. Cell Death Differ. Nature; 2011;18: 1457–69. doi: 10.1038/cdd.2011.75 21637292

53. Stolk J, Hiltermann TJ, Dijkman JH, Verhoeven AJ. Characteristics of the inhibition of NADPH oxidase activation in neutrophils by apocynin, a methoxy-substituted catechol. Am J Respir Cell Mol Biol. 1994;11: 95–102. 8018341

54. Bayraktutan U, Draper N, Lang D, Shah AM. Expression of functional neutrophil-type NADPH oxidase in cultured rat coronary microvascular endothelial cells. Cardiovasc Res. 1998;38: 256–62. 9683929

55. Roos D, Weening RS, Wyss SR, Aebi HE. Protection of human neutrophils by endogenous catalase: studies with cells from catalase-deficient individuals. J Clin Invest. 1980;65: 1515–22. 7410555

56. Larsen BD, Rampalli S, Burns LE, Brunette S, Dilworth FJ, Megeney LA. Caspase 3/caspase-activated DNase promote cell differentiation by inducing DNA strand breaks. Proc Natl Acad Sci U S A. 2010;107: 4230–5. doi: 10.1073/pnas.0913089107 20160104

57. Kamada S, Funahashi Y, Tsujimoto Y. Caspase-4 and caspase-5, members of the ICE/CED-3 family of cysteine proteases, are CrmA-inhibitable proteases. Cell Death Differ. 1997;4: 473–8. 16465268

58. Ofengeim D, Yuan J. Regulation of RIP1 kinase signalling at the crossroads of inflammation and cell death. Nat Rev Mol Cell Biol. 2013;14: 727–36. doi: 10.1038/nrm3683 24129419

59. Colussi PA, Kumar S. Targeted disruption of caspase genes in mice: what they tell us about the functions of individual caspases in apoptosis. Immunol Cell Biol. 1999;77: 58–63. 10101687

60. Zheng TS, Hunot S, Kuida K, Flavell RA. Caspase knockouts: matters of life and death. Cell Death Differ. 1999;6: 1043–53. 10578172

61. Acorci MJ, Dias-Melicio LA, Golim MA, Bordon-Graciani AP, Peraçoli MTS, Soares AMVC. Inhibition of human neutrophil apoptosis by Paracoccidioides brasiliensis: role of interleukin-8. Scand J Immunol. 2009;69: 73–9. doi: 10.1111/j.1365-3083.2008.02199.x 19144080

62. Afford SC, Pongracz J, Stockley R a, Crocker J, Burnett D. The induction by human interleukin-6 of apoptosis in the promonocytic cell line U937 and human neutrophils. J Biol Chem. 1992;267: 21612–6. 1400472

63. Colotta F, Re F, Polentarutti N, Sozzani S, Mantovani A. Modulation of granulocyte survival and programmed cell death by cytokines and bacterial products. Blood. 1992;80: 2012–2020. 1382715

64. Ocaña MG, Asensi V, Montes AH, Meana A, Celada A, Valle-Garay E. Autoregulation mechanism of human neutrophil apoptosis during bacterial infection. Mol Immunol. 2008;45: 2087–96. 18022234

65. Baran J, Guzik K, Hryniewicz W, Ernst M, Flad HD, Pryjma J. Apoptosis of monocytes and prolonged survival of granulocytes as a result of phagocytosis of bacteria. Infect Immun. 1996;64: 4242–8. 8926095

66. Tumurkhuu G, Koide N, Takahashi K, Hassan F, Islam S, Ito H, et al. Characterization of biological activities of Brucella melitensis lipopolysaccharide. Microbiol Immunol. 2006;50: 421–7. 16785713

67. Weiss DS, Takeda K, Akira S, Zychlinsky A, Moreno E. MyD88, but not toll-like receptors 4 and 2, is required for efficient clearance of Brucella abortus. Infect Immun. 2005;73: 5137–5143. 16041030

68. Goldstein J, Hoffman T, Frasch C, Lizzio EF, Beining PR, Hochstein D, et al. Lipopolysaccharide (LPS) from Brucella abortus is less toxic than that from Escherichia coli, suggesting the possible use of B. abortus or LPS from B. abortus as a carrier in vaccines. Infect Immun. 1992;60: 1385–9. 1548064

69. Dueñas AI, Orduña A, Crespo MS, García-Rodríguez C, Lps B, Ordun A, et al. Interaction of endotoxins with Toll-like receptor 4 correlates with their endotoxic potential and may explain the proinflammatory effect of Brucella spp. LPS. Int Immunol. 2004;16: 1467–75. 15339879

70. Kettritz R, Gaido ML, Haller H, Luft FC, Jennette CJ, Falk RJ. Interleukin-8 delays spontaneous and tumor necrosis factor-alpha-mediated apoptosis of human neutrophils. Kidney Int. 1998;53: 84–91. 9453003

71. Altstaedt J, Kirchner H, Rink L. Cytokine production of neutrophils is limited to interleukin-8. Immunology. 1996;89: 563–8. 9014822

72. Sabroe I, Jones EC, Usher LR, Whyte MKB, Dower SK. Toll-like receptor (TLR)2 and TLR4 in human peripheral blood granulocytes: a critical role for monocytes in leukocyte lipopolysaccharide responses. J Immunol. 2002;168: 4701–10. 11971020

73. Ward C, Chilvers ER, Lawson MF, Pryde JG, Fujihara S, Farrow SN, et al. NF-kappaB activation is a critical regulator of human granulocyte apoptosis in vitro. J Biol Chem. 1999;274: 4309–18. 9933632

74. Fadeel B, Ahlin A, Henter JI, Orrenius S, Hampton MB. Involvement of caspases in neutrophil apoptosis: regulation by reactive oxygen species. Blood. 1998;92: 4808–18. 9845548

75. Gamazo C, Moriyón I. Release of outer membrane fragments by exponentially growing Brucella melitensis cells. Infect Immun. 1987;55: 609–15. 3818086

76. Zwerdling A, Delpino MV, Pasquevich K a, Barrionuevo P, Cassataro J, García Samartino C, et al. Brucella abortus activates human neutrophils. Microbes Infect. 2009;11: 689–97. doi: 10.1016/j.micinf.2009.04.010 19376263

77. Lapaque N, Takeuchi O, Corrales F, Akira S, Moriyon I, Howard JC, et al. Differential inductions of TNF-alpha and IGTP, IIGP by structurally diverse classic and non-classic lipopolysaccharides. Cell Microbiol. 2006;8: 401–13. 16469053

78. Lapaque N, Muller A, Alexopoulou L, Howard JC, Gorvel J-P. Brucella abortus induces Irgm3 and Irga6 expression via type-I IFN by a MyD88-dependent pathway, without the requirement of TLR2, TLR4, TLR5 and TLR9. Microb Pathog. 2009;47: 299–304. doi: 10.1016/j.micpath.2009.09.005 19747534

79. Rodeberg DA, Morris RE, Babcock GF. Azurophilic granules of human neutrophils contain CD14. Infect Immun. 1997;65: 4747–53. 9353060

80. Lapaque N, Forquet F, de Chastellier C, Mishal Z, Jolly G, Moreno E, et al. Characterization of Brucella abortus lipopolysaccharide macrodomains as mega rafts. Cell Microbiol. 2006;8: 197–206. 16441431

81. Lei M, Du L, Jiao H, Cheng Y, Zhang D, Hao Y, et al. Inhibition of mCD14 inhibits TNFα secretion and NO production in RAW264.7 cells stimulated by Brucella melitensis infection. Vet Microbiol. 2012;160: 362–8. doi: 10.1016/j.vetmic.2012.05.039 22770519

82. Lapaque N, Moriyon I, Moreno E, Gorvel J-P. Brucella lipopolysaccharide acts as a virulence factor. Curr Opin Microbiol. 2005;8: 60–6. 15694858

83. Weersink JL, Antal-Szalmas P, Strijp JAG Van, Kessel KPM Van. Quantitation of surface neutrophils CD14 on human monocytes and. J Leukoc Biol. 1997;61: 721–728. 9201263

84. Celli J, de Chastellier C, Franchini D-M, Pizarro-Cerda J, Moreno E, Gorvel J-P. Brucella evades macrophage killing via VirB-dependent sustained interactions with the endoplasmic reticulum. J Exp Med. 2003;198: 545–56. 12925673

85. Simon HU, Haj-Yehia A, Levi-Schaffer F. Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis. 2000;5: 415–8. 11256882

86. Almyroudis NG, Grimm MJ, Davidson BA, Röhm M, Urban CF, Segal BH. NETosis and NADPH oxidase: at the intersection of host defense, inflammation, and injury. Front Immunol. 2013;4: 45. doi: 10.3389/fimmu.2013.00045 23459634

87. Meuth M. Chk1 suppressed cell death. Cell Div. 2010;5: 21. doi: 10.1186/1747-1028-5-21 20813042

88. Festjens N, Vanden Berghe T, Cornelis S, Vandenabeele P. RIP1, a kinase on the crossroads of a cell’s decision to live or die. Cell Death Differ. 2007;14: 400–10. 17301840

89. Olofsson MH, Havelka AM, Brnjic S, Shoshan MC, Linder S. Charting calcium-regulated apoptosis pathways using chemical biology: role of calmodulin kinase II. BMC Chem Biol. 2008;8: 2. doi: 10.1186/1472-6769-8-2 18673549

90. Miao EA, Rajan J V, Aderem A. Caspase-1-induced pyroptotic cell death. Immunol Rev. 2011;243: 206–14. doi: 10.1111/j.1600-065X.2011.01044.x 21884178

91. Bakele M, Joos M, Burdi S, Allgaier N, Pöschel S, Fehrenbacher B, et al. Localization and functionality of the inflammasome in neutrophils. J Biol Chem. 2014;289: 5320–9. doi: 10.1074/jbc.M113.505636 24398679

92. Miao EA, Leaf IA, Treuting PM, Mao DP, Dors M, Sarkar A, et al. Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat Immunol. 2010;11: 1136–42. doi: 10.1038/ni.1960 21057511

93. Brodsky IE, Medzhitov R. Pyroptosis: macrophage suicide exposes hidden invaders. Curr Biol. 2011;21: R72–5. doi: 10.1016/j.cub.2010.12.008 21256438

94. Murray J, Barbara JAJ, Dunkley SA, Lopez AF, Van Ostade X, Condliffe AM, et al. Regulation of neutrophil apoptosis by tumor necrosis factor-alpha: requirement for TNFR55 and TNFR75 for induction of apoptosis in vitro. Blood. 1997;90: 2772–2783. 9326245

95. Aggarwal S, Gollapudi S, Yel L, Gupta AS, Gupta S. TNF-alpha-induced apoptosis in neonatal lymphocytes: TNFRp55 expression and downstream pathways of apoptosis. Genes Immun. 2000;1: 271–9. 11196704

96. Demir C, Karahocagil MK, Esen R, Atmaca M, Gönüllü H, Akdeniz H. Bone marrow biopsy findings in brucellosis patients with hematologic abnormalities. Chin Med J (Engl). 2012;125: 1871–6. 22884045

97. El-Koumi MA, Afify M, Al-Zahrani SH. A prospective study of brucellosis in children: relative frequency of pancytopenia. Mediterr J Hematol Infect Dis. 2013;5: e2013011. doi: 10.4084/MJHID.2013.011 23505599

98. Kokkini G, Giotaki HG, Moutsopoulos HM. Transient hemophagocytosis in Brucella melitensis infection. Arch Pathol Lab Med. 1984;108: 213–6. 6546508

99. Laskay T, van Zandbergen G, Solbach W. Neutrophil granulocytes—Trojan horses for Leishmania major and other intracellular microbes? Trends Microbiol. 2003;11: 210–214. 12781523

100. Chacón-Díaz C, Muñoz-Rodríguez M, Barquero-Calvo E, Guzmán-Verri C, Chaves-Olarte E, Grilló MJ, et al. The use of green fluorescent protein as a marker for Brucella vaccines. Vaccine. 2011;29: 577–82. doi: 10.1016/j.vaccine.2010.09.109 21056079

101. Moreno E, Berman DT, Boettcher L a. Biological activities of Brucella abortus lipopolysaccharides. Infect Immun. 1981;31: 362–70. 6783538

102. Espinoza AM, Pereira R, Macaya-Lizano A V, Hernández M, Goulden M, Rivera C. Comparative light and electron microscopic analyses of tenuivirus major noncapsid protein (NCP) inclusion bodies in infected plants, and of the NCP in vitro. Virology. 1993;195: 156–66. 8317091

103. Rojas N, Freer E, Weintraub A, Ramirez M, Lind S, Moreno E. Immunochemical identification of Brucella abortus lipopolysaccharide epitopes. Clin Diagn Lab Immunol. 1994;1: 206–13. 7496947

104. Reynolds ES. The use of lead citrate at high pH as an electron-opaque stain in electron microscopy. J Cell Biol. 1963;17: 208–12. 13986422

105. Bundle DR, Cherwonogrodzky JW, Gidney MA, Meikle PJ, Perry MB, Peters T. Definition of Brucella A and M epitopes by monoclonal typing reagents and synthetic oligosaccharides. Infect Immun. 1989;57: 2829–36. 2474505

106. Chin AC, Lee WD, Murrin KA, Morck DW, Merrill JK, Dick P, et al. Tilmicosin induces apoptosis in bovine peripheral neutrophils in the presence or in the absence of Pasteurella haemolytica and promotes neutrophil phagocytosis by macrophages. Antimicrob Agents Chemother. 2000;44: 2465–70. 10952596

107. Moreno E, Stackebrandt E, Dorsch M, Wolters J, Busch M, Mayer H. Brucella abortus 16S rRNA and lipid A reveal a phylogenetic relationship with members of the alpha-2 subdivision of the class Proteobacteria. J Bacteriol. 1990;172: 3569–76. 2113907

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#