#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Toxin-Induced Necroptosis Is a Major Mechanism of Lung Damage


Staphylococcus aureus (SA) cause a highly inflammatory pneumonia associated with substantial morbidity and mortality. Much of this lung destruction is attributed to toxins that target specific receptors on human and murine cells. We demonstrate that the α-hemolysin (Hla) and other agr-regulated toxins activate RIP1/RIP3/MLKL-mediated necroptosis and IL-1β expression, through a mechanism that involves MLKL pore-formation and inflammasome activation. Cell death can be inhibited by osmoprotectants and K+ repletion. Necroptosis results in alveolar macrophage depletion and loss of anti-inflammatory signaling. Rip3-/- mice maintain significantly greater numbers of alveolar macrophages with anti-inflammatory phenotypes, CD206+ and CD200R+; decreased proinflammatory cytokine production; and improved SA clearance. Necroptosis represents a common mechanism of pulmonary damage activated by multiple SA toxins.


Vyšlo v časopise: Toxin-Induced Necroptosis Is a Major Mechanism of Lung Damage. PLoS Pathog 11(4): e32767. doi:10.1371/journal.ppat.1004820
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004820

Souhrn

Staphylococcus aureus (SA) cause a highly inflammatory pneumonia associated with substantial morbidity and mortality. Much of this lung destruction is attributed to toxins that target specific receptors on human and murine cells. We demonstrate that the α-hemolysin (Hla) and other agr-regulated toxins activate RIP1/RIP3/MLKL-mediated necroptosis and IL-1β expression, through a mechanism that involves MLKL pore-formation and inflammasome activation. Cell death can be inhibited by osmoprotectants and K+ repletion. Necroptosis results in alveolar macrophage depletion and loss of anti-inflammatory signaling. Rip3-/- mice maintain significantly greater numbers of alveolar macrophages with anti-inflammatory phenotypes, CD206+ and CD200R+; decreased proinflammatory cytokine production; and improved SA clearance. Necroptosis represents a common mechanism of pulmonary damage activated by multiple SA toxins.


Zdroje

1. David MZ, Daum RS. Community-associated methicillin-resistant Staphylococcus aureus: epidemiology and clinical consequences of an emerging epidemic. Clin Microbiol Rev. 2010;23(3):616–87. doi: 10.1128/CMR.00081-09 20610826

2. Klein E, Smith DL, Laxminarayan R. Hospitalizations and deaths caused by methicillin-resistant Staphylococcus aureus, United States, 1999–2005. Emerg Infect Dis. 2007;13(12):1840–6. doi: 10.3201/eid1312.070629 18258033

3. Klevens RM, Morrison MA, Nadle J, Petit S, Gershman K, Ray S, et al. Invasive methicillin-resistant Staphylococcus aureus infections in the United States. JAMA. 2007;298(15):1763–71. doi: 10.1001/jama.298.15.1763 17940231

4. Planet PJ, LaRussa SJ, Dana A, Smith H, Xu A, Ryan C, et al. Emergence of the epidemic methicillin-resistant Staphylococcus aureus strain USA300 coincides with horizontal transfer of the arginine catabolic mobile element and speG-mediated adaptations for survival on skin. MBio. 2013;4(6):e00889–13. doi: 10.1128/mBio.00889-13 24345744

5. Inoshima I, Inoshima N, Wilke GA, Powers ME, Frank KM, Wang Y, et al. A Staphylococcus aureus pore-forming toxin subverts the activity of ADAM10 to cause lethal infection in mice. Nat Med. 2011;17(10):1310–4. Epub 2011/09/20. doi: 10.1038/nm.2451 21926978

6. Craven RR, Gao X, Allen IC, Gris D, Bubeck Wardenburg J, McElvania-Tekippe E, et al. Staphylococcus aureus alpha-hemolysin activates the NLRP3-inflammasome in human and mouse monocytic cells. PLoS One. 2009;4(10):e7446. Epub 2009/10/15. doi: 10.1371/journal.pone.0007446 19826485

7. DuMont AL, Yoong P, Day CJ, Alonzo F 3rd, McDonald WH, Jennings MP, et al. Staphylococcus aureus LukAB cytotoxin kills human neutrophils by targeting the CD11b subunit of the integrin Mac-1. Proc Natl Acad Sci U S A. 2013;110(26):10794–9. doi: 10.1073/pnas.1305121110 23754403

8. Ventura CL, Malachowa N, Hammer CH, Nardone GA, Robinson MA, Kobayashi SD, et al. Identification of a novel Staphylococcus aureus two-component leukotoxin using cell surface proteomics. PLoS One. 2010;5(7):e11634. doi: 10.1371/journal.pone.0011634 20661294

9. Chatterjee SS, Joo HS, Duong AC, Dieringer TD, Tan VY, Song Y, et al. Essential Staphylococcus aureus toxin export system. Nat Med. 2013;19(3):364–7. Epub 2013/02/12. doi: 10.1038/nm.3047 23396209

10. Kebaier C, Chamberland RR, Allen IC, Gao X, Broglie PM, Hall JD, et al. Staphylococcus aureus alpha-hemolysin mediates virulence in a murine model of severe pneumonia through activation of the NLRP3 inflammasome. J Infect Dis. 2012;205(5):807–17. doi: 10.1093/infdis/jir846 22279123

11. DuMont AL, Torres VJ. Cell targeting by the Staphylococcus aureus pore-forming toxins: it's not just about lipids. Trends Microbiol. 2014;22(1):21–7. doi: 10.1016/j.tim.2013.10.004 24231517

12. Parker D, Planet PJ, Soong G, Narechania A, Prince A. Induction of type I interferon signaling determines the relative pathogenicity of Staphylococcus aureus strains. PLoS Pathog. 2014;10(2):e1003951. doi: 10.1371/journal.ppat.1003951 24586160

13. Martin FJ, Gomez MI, Wetzel DM, Memmi G, O'Seaghdha M, Soong G, et al. Staphylococcus aureus activates type I IFN signaling in mice and humans through the Xr repeated sequences of protein A. J Clin Invest. 2009;119(7):1931–9. Epub 2009/07/16. 19603548

14. Gomez MI, Lee A, Reddy B, Muir A, Soong G, Pitt A, et al. Staphylococcus aureus protein A induces airway epithelial inflammatory responses by activating TNFR1. Nat Med. 2004;10(8):842–8. doi: 10.1038/nm1079 15247912

15. Cohen TS, Prince AS. Bacterial pathogens activate a common inflammatory pathway through IFNlambda regulation of PDCD4. PLoS Pathog. 2013;9(10):e1003682. Epub 2013/10/08. doi: 10.1371/journal.ppat.1003682 24098127

16. Kapetanovic R, Jouvion G, Fitting C, Parlato M, Blanchet C, Huerre M, et al. Contribution of NOD2 to lung inflammation during Staphylococcus aureus-induced pneumonia. Microbes Infect. 2010;12(10):759–67. doi: 10.1016/j.micinf.2010.05.003 20493961

17. Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157(5):1013–22. doi: 10.1016/j.cell.2014.04.007 24855941

18. Kaczmarek A, Vandenabeele P, Krysko DV. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity. 2013;38(2):209–23. doi: 10.1016/j.immuni.2013.02.003 23438821

19. Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J, et al. Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol. 2014;16(1):55–65. doi: 10.1038/ncb2883 24316671

20. Chen X, Li W, Ren J, Huang D, He WT, Song Y, et al. Translocation of mixed lineage kinase domain-like protein to plasma membrane leads to necrotic cell death. Cell Res. 2014;24(1):105–21. doi: 10.1038/cr.2013.171 24366341

21. Linkermann A, Green DR. Necroptosis. N Engl J Med. 2014;370(5):455–65. doi: 10.1056/NEJMra1310050 24476434

22. Su L, Quade B, Wang H, Sun L, Wang X, Rizo J. A plug release mechanism for membrane permeation by MLKL. Structure. 2014;22(10):1489–500. doi: 10.1016/j.str.2014.07.014 25220470

23. Weng D, Marty-Roix R, Ganesan S, Proulx MK, Vladimer GI, Kaiser WJ, et al. Caspase-8 and RIP kinases regulate bacteria-induced innate immune responses and cell death. Proc Natl Acad Sci U S A. 2014;111(20):7391–6. doi: 10.1073/pnas.1403477111 24799678

24. Robinson N, McComb S, Mulligan R, Dudani R, Krishnan L, Sad S. Type I interferon induces necroptosis in macrophages during infection with Salmonella enterica serovar Typhimurium. Nat Immunol. 2012;13(10):954–62. doi: 10.1038/ni.2397 22922364

25. Degterev A, Hitomi J, Germscheid M, Ch'en IL, Korkina O, Teng X, et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol. 2008;4(5):313–21. Epub 2008/04/15. doi: 10.1038/nchembio.83 18408713

26. Takahashi N, Duprez L, Grootjans S, Cauwels A, Nerinckx W, DuHadaway JB, et al. Necrostatin-1 analogues: critical issues on the specificity, activity and in vivo use in experimental disease models. Cell Death Dis. 2012;3:e437. doi: 10.1038/cddis.2012.176 23190609

27. Sun L, Wang H, Wang Z, He S, Chen S, Liao D, et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell. 2012;148(1–2):213–27. doi: 10.1016/j.cell.2011.11.031 22424234

28. Alonzo F 3rd, Torres VJ. The bicomponent pore-forming leucocidins of Staphylococcus aureus. Microbiol Mol Biol Rev. 2014;78(2):199–230. doi: 10.1128/MMBR.00055-13 24847020

29. Munoz-Planillo R, Kuffa P, Martinez-Colon G, Smith BL, Rajendiran TM, Nunez G. K(+) efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity. 2013;38(6):1142–53. doi: 10.1016/j.immuni.2013.05.016 23809161

30. Kang TB, Yang SH, Toth B, Kovalenko A, Wallach D. Caspase-8 blocks kinase RIPK3-mediated activation of the NLRP3 inflammasome. Immunity. 2013;38(1):27–40. doi: 10.1016/j.immuni.2012.09.015 23260196

31. Vince JE, Wong WW, Gentle I, Lawlor KE, Allam R, O'Reilly L, et al. Inhibitor of apoptosis proteins limit RIP3 kinase-dependent interleukin-1 activation. Immunity. 2012;36(2):215–27. doi: 10.1016/j.immuni.2012.01.012 22365665

32. Yabal M, Muller N, Adler H, Knies N, Gross CJ, Damgaard RB, et al. XIAP restricts TNF- and RIP3-dependent cell death and inflammasome activation. Cell Rep. 2014;7(6):1796–808. doi: 10.1016/j.celrep.2014.05.008 24882010

33. Wang X, Jiang W, Yan Y, Gong T, Han J, Tian Z, et al. RNA viruses promote activation of the NLRP3 inflammasome through a RIP1-RIP3-DRP1 signaling pathway. Nat Immunol. 2014. doi: 10.1038/ni.3015

34. Viboud GI, Bliska JB. Measurement of pore formation by contact-dependent type III protein secretion systems. Methods Enzymol. 2002;358:345–50. 12474398

35. Marriott HM, Jackson LE, Wilkinson TS, Simpson AJ, Mitchell TJ, Buttle DJ, et al. Reactive oxygen species regulate neutrophil recruitment and survival in pneumococcal pneumonia. Am J Respir Crit Care Med. 2008;177(8):887–95. doi: 10.1164/rccm.200707-990OC 18202350

36. Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol. 2014;14(2):81–93. doi: 10.1038/nri3600 24445666

37. Snelgrove RJ, Goulding J, Didierlaurent AM, Lyonga D, Vekaria S, Edwards L, et al. A critical function for CD200 in lung immune homeostasis and the severity of influenza infection. Nat Immunol. 2008;9(9):1074–83. doi: 10.1038/ni.1637 18660812

38. Choi SM, McAleer JP, Zheng M, Pociask DA, Kaplan MH, Qin S, et al. Innate Stat3-mediated induction of the antimicrobial protein Reg3gamma is required for host defense against MRSA pneumonia. J Exp Med. 2013;210(3):551–61. doi: 10.1084/jem.20120260 23401489

39. Powers ME, Bubeck Wardenburg J. Igniting the fire: Staphylococcus aureus virulence factors in the pathogenesis of sepsis. PLoS Pathog. 2014;10(2):e1003871. doi: 10.1371/journal.ppat.1003871 24550724

40. Vandenesch F, Lina G, Henry T. Staphylococcus aureus hemolysins, bi-component leukocidins, and cytolytic peptides: a redundant arsenal of membrane-damaging virulence factors? Front Cell Infect Microbiol. 2012;2:12. doi: 10.3389/fcimb.2012.00012 22919604

41. Weinlich R, Green DR. The Two Faces of Receptor Interacting Protein Kinase-1. Mol Cell. 2014;56(4):469–80. doi: 10.1016/j.molcel.2014.11.001 25459879

42. Wang R, Braughton KR, Kretschmer D, Bach TH, Queck SY, Li M, et al. Identification of novel cytolytic peptides as key virulence determinants for community-associated MRSA. Nat Med. 2007;13(12):1510–4. doi: 10.1038/nm1656 17994102

43. Peschel A, Otto M. Phenol-soluble modulins and staphylococcal infection. Nat Rev Microbiol. 2013;11(10):667–73. doi: 10.1038/nrmicro3110 24018382

44. Laabei M, Jamieson WD, Yang Y, van den Elsen J, Jenkins AT. Investigating the lytic activity and structural properties of Staphylococcus aureus phenol soluble modulin (PSM) peptide toxins. Biochim Biophys Acta. 2014;1838(12):3153–61. doi: 10.1016/j.bbamem.2014.08.026 25194683

45. Berube BJ, Sampedro GR, Otto M, Bubeck Wardenburg J. The psmalpha locus regulates production of Staphylococcus aureus alpha-toxin during infection. Infect Immun. 2014;82(8):3350–8. doi: 10.1128/IAI.00089-14 24866799

46. Dumont AL, Nygaard TK, Watkins RL, Smith A, Kozhaya L, Kreiswirth BN, et al. Characterization of a new cytotoxin that contributes to Staphylococcus aureus pathogenesis. Mol Microbiol. 2011;79(3):814–25. doi: 10.1111/j.1365-2958.2010.07490.x 21255120

47. Greenlee-Wacker MC, Rigby KM, Kobayashi SD, Porter AR, DeLeo FR, Nauseef WM. Phagocytosis of Staphylococcus aureus by human neutrophils prevents macrophage efferocytosis and induces programmed necrosis. J Immunol. 2014;192(10):4709–17. doi: 10.4049/jimmunol.1302692 24729616

48. Robinson KM, Choi SM, McHugh KJ, Mandalapu S, Enelow RI, Kolls JK, et al. Influenza A exacerbates Staphylococcus aureus pneumonia by attenuating IL-1beta production in mice. J Immunol. 2013;191(10):5153–9. doi: 10.4049/jimmunol.1301237 24089191

49. Martin FJ, Parker D, Harfenist BS, Soong G, Prince A. Participation of CD11c(+) leukocytes in methicillin-resistant Staphylococcus aureus clearance from the lung. Infect Immun. 2011;79(5):1898–904. doi: 10.1128/IAI.01299-10 21402768

50. Parker D, Ryan CL, Alonzo F 3rd, Torres VJ, Planet PJ, Prince AS. CD4+ T Cells Promote the Pathogenesis of Staphylococcus aureus Pneumonia. J Infect Dis. 2014. doi: 10.1093/infdis/jiu525

51. Chambers HF, Deleo FR. Waves of resistance: Staphylococcus aureus in the antibiotic era. Nat Rev Microbiol. 2009;7(9):629–41. doi: 10.1038/nrmicro2200 19680247

52. Newton K, Sun X, Dixit VM. Kinase RIP3 is dispensable for normal NF-kappa Bs, signaling by the B-cell and T-cell receptors, tumor necrosis factor receptor 1, and Toll-like receptors 2 and 4. Mol Cell Biol. 2004;24(4):1464–9. Epub 2004/01/30. 14749364

53. Parker D, Prince A. Staphylococcus aureus induces type I IFN signaling in dendritic cells via TLR9. J Immunol. 2012;189(8):4040–6. doi: 10.4049/jimmunol.1201055 22962685

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 4
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#