#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Plasminogen Controls Inflammation and Pathogenesis of Influenza Virus Infections via Fibrinolysis


Detrimental inflammation of the lungs is a hallmark of severe influenza virus infections. Endothelial cells are the source of cytokine amplification, although mechanisms underlying this process are unknown. Here, using combined pharmacological and gene-deletion approaches, we show that plasminogen controls lung inflammation and pathogenesis of infections with influenza A/PR/8/34, highly pathogenic H5N1 and 2009 pandemic H1N1 viruses. Reduction of virus replication was not responsible for the observed effect. However, pharmacological depletion of fibrinogen, the main target of plasminogen reversed disease resistance of plasminogen-deficient mice or mice treated with an inhibitor of plasminogen-mediated fibrinolysis. Therefore, plasminogen contributes to the deleterious inflammation of the lungs and local fibrin clot formation may be implicated in host defense against influenza virus infections. Our studies suggest that the hemostatic system might be explored for novel treatments against influenza.


Vyšlo v časopise: Plasminogen Controls Inflammation and Pathogenesis of Influenza Virus Infections via Fibrinolysis. PLoS Pathog 9(3): e32767. doi:10.1371/journal.ppat.1003229
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003229

Souhrn

Detrimental inflammation of the lungs is a hallmark of severe influenza virus infections. Endothelial cells are the source of cytokine amplification, although mechanisms underlying this process are unknown. Here, using combined pharmacological and gene-deletion approaches, we show that plasminogen controls lung inflammation and pathogenesis of infections with influenza A/PR/8/34, highly pathogenic H5N1 and 2009 pandemic H1N1 viruses. Reduction of virus replication was not responsible for the observed effect. However, pharmacological depletion of fibrinogen, the main target of plasminogen reversed disease resistance of plasminogen-deficient mice or mice treated with an inhibitor of plasminogen-mediated fibrinolysis. Therefore, plasminogen contributes to the deleterious inflammation of the lungs and local fibrin clot formation may be implicated in host defense against influenza virus infections. Our studies suggest that the hemostatic system might be explored for novel treatments against influenza.


Zdroje

1. Knipe DM, Howley PM, editors (2006) Fields Virology. 5th edition. Philadelphia (Pennsylvania): Lippincott, Williams, & Wilkins.

2. KuikenT, RiteauB, FouchierRA, RimmelzwaanGF (2012) Pathogenesis of influenza virus infections: the good, the bad and the ugly. Curr Opin Virol 2: 276–286.

3. La GrutaNL, KedzierskaK, StambasJ, DohertyPC (2007) A question of self-preservation: immunopathology in influenza virus infection. Immunol Cell Biol 85: 85–92.

4. de JongMD, SimmonsCP, ThanhTT, HienVM, SmithGJ, et al. (2006) Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med 12: 1203–1207.

5. ReedCE, KitaH (2004) The role of protease activation of inflammation in allergic respiratory diseases. J Allergy Clin Immunol 114: 997–1008; quiz 1009.

6. SokolovaE, ReiserG (2007) A novel therapeutic target in various lung diseases: airway proteases and protease-activated receptors. Pharmacol Ther 115: 70–83.

7. SteinhoffM, BuddenkotteJ, ShpacovitchV, RattenhollA, MoormannC, et al. (2005) Proteinase-activated receptors: transducers of proteinase-mediated signaling in inflammation and immune response. Endocr Rev 26: 1–43.

8. BradleyLM, DouglassMF, ChatterjeeD, AkiraS, BaatenBJ (2012) Matrix metalloprotease 9 mediates neutrophil migration into the airways in response to influenza virus-induced toll-like receptor signaling. PLoS Pathog 8: e1002641.

9. KhoufacheK, BerriF, NackenW, VogelAB, DelenneM, et al. (2013) PAR1 contributes to influenza A virus pathogenicity in mice. J Clin Invest 123: 206–214.

10. LeBouderF, MorelloE, RimmelzwaanGF, BosseF, PechouxC, et al. (2008) Annexin II incorporated into influenza virus particles supports virus replication by converting plasminogen into plasmin. J Virol 82: 6820–6828.

11. LeBouderF, LinaB, RimmelzwaanGF, RiteauB (2010) Plasminogen promotes influenza A virus replication through an annexin 2-dependent pathway in the absence of neuraminidase. J Gen Virol 91: 2753–2761.

12. GotoH, KawaokaY (1998) A novel mechanism for the acquisition of virulence by a human influenza A virus. Proc Natl Acad Sci U S A 95: 10224–10228.

13. GotoH, WellsK, TakadaA, KawaokaY (2001) Plasminogen-binding activity of neuraminidase determines the pathogenicity of influenza A virus. J Virol 75: 9297–9301.

14. van HinsberghVW (2012) Endothelium–role in regulation of coagulation and inflammation. Semin Immunopathol 34: 93–106.

15. KellerTT, van der SluijsKF, de KruifMD, GerdesVE, MeijersJC, et al. (2006) Effects on coagulation and fibrinolysis induced by influenza in mice with a reduced capacity to generate activated protein C and a deficiency in plasminogen activator inhibitor type 1. Circ Res 99: 1261–1269.

16. ManzB, BrunotteL, ReutherP, SchwemmleM (2012) Adaptive mutations in NEP compensate for defective H5N1 RNA replication in cultured human cells. Nat Commun 3: 802.

17. PrenticeC (1980) Basis of antifibrinolytic therapy. J Clin Pathol 33: 35–40.

18. GongY, HartE, ShchurinA, Hoover-PlowJ (2008) Inflammatory macrophage migration requires MMP-9 activation by plasminogen in mice. J Clin Invest 118: 3012–3024.

19. WygreckaM, MarshLM, MortyRE, HennekeI, GuentherA, et al. (2009) Enolase-1 promotes plasminogen-mediated recruitment of monocytes to the acutely inflamed lung. Blood 113: 5588–5598.

20. SyrovetsT, TipplerB, RieksM, SimmetT (1997) Plasmin is a potent and specific chemoattractant for human peripheral monocytes acting via a cyclic guanosine monophosphate-dependent pathway. Blood 89: 4574–4583.

21. O'ConnellPA, SuretteAP, LiwskiRS, SvenningssonP, WaismanDM (2010) S100A10 regulates plasminogen-dependent macrophage invasion. Blood 116: 1136–1146.

22. PloplisVA, FrenchEL, CarmelietP, CollenD, PlowEF (1998) Plasminogen deficiency differentially affects recruitment of inflammatory cell populations in mice. Blood 91: 2005–2009.

23. MoonsL, ShiC, PloplisV, PlowE, HaberE, et al. (1998) Reduced transplant arteriosclerosis in plasminogen-deficient mice. J Clin Invest 102: 1788–1797.

24. SoepandiPZ, BurhanE, MangunnegoroH, NawasA, AditamaTY, et al. (2010) Clinical course of avian influenza A(H5N1) in patients at the Persahabatan Hospital, Jakarta, Indonesia, 2005–2008. Chest 138: 665–673.

25. WangZF, SuF, LinXJ, DaiB, KongLF, et al. (2011) Serum D-dimer changes and prognostic implication in 2009 novel influenza A(H1N1). Thromb Res 127: 198–201.

26. UrsoR, BevilacquaN, GentileM, BiagioliD, LauriaFN (2011) Pandemic 2009 H1N1 virus infection associated with purpuric skin lesions: a case report. J Med Case Reports 5: 132.

27. OkayamaS, ArakawaS, OgawaK, MakinoT (2011) A case of hemorrhagic colitis after influenza A infection. J Microbiol Immunol Infect 44(6): 480–483.

28. DegenJL, BuggeTH, GoguenJD (2007) Fibrin and fibrinolysis in infection and host defense. J Thromb Haemost 5(Suppl 1): 24–31.

29. TeijaroJR, WalshKB, CahalanS, FremgenDM, RobertsE, et al. (2011) Endothelial Cells Are Central Orchestrators of Cytokine Amplification during Influenza Virus Infection. Cell 146: 980–991.

30. FislovaT, GocnikM, SladkovaT, DurmanovaV, RajcaniJ, et al. (2009) Multiorgan distribution of human influenza A virus strains observed in a mouse model. Arch Virol 154: 409–419.

31. Sanchez-LanierM, DavisLE, BlisardKS, WoodfinBM, WallaceJM, et al. (1991) Influenza A virus in the mouse: hepatic and cerebral lesions in a Reye's syndrome-like illness. Int J Exp Pathol 72: 489–500.

32. SunX, TseLV, FergusonAD, WhittakerGR (2010) Modifications to the hemagglutinin cleavage site control the virulence of a neurotropic H1N1 influenza virus. J Virol 84: 8683–8690.

33. ZhirnovOP, IkizlerMR, WrightPF (2002) Cleavage of influenza a virus hemagglutinin in human respiratory epithelium is cell associated and sensitive to exogenous antiproteases. J Virol 76: 8682–8689.

34. BertramS, GlowackaI, BlazejewskaP, SoilleuxE, AllenP, et al. (2010) TMPRSS2 and TMPRSS4 facilitate trypsin-independent influenza virus spread in Caco-2 cells. J Virol 84: 10016–10025.

35. BottcherE, MatrosovichT, BeyerleM, KlenkHD, GartenW, et al. (2006) Proteolytic activation of influenza viruses by serine proteases TMPRSS2 and HAT from human airway epithelium. J Virol 80: 9896–9898.

36. ChaipanC, KobasaD, BertramS, GlowackaI, SteffenI, et al. (2009) Proteolytic activation of the 1918 influenza virus hemagglutinin. J Virol 83: 3200–3211.

37. ZhirnovOP, KlenkHD, WrightPF (2011) Aprotinin and similar protease inhibitors as drugs against influenza. Antiviral Res 92: 27–36.

38. Bottcher-FriebertshauserE, FreuerC, SielaffF, SchmidtS, EickmannM, et al. (2010) Cleavage of influenza virus hemagglutinin by airway proteases TMPRSS2 and HAT differs in subcellular localization and susceptibility to protease inhibitors. J Virol 84: 5605–5614.

39. ZhirnovOP, OvcharenkoAV, BukrinskayaAG (1982) Protective effect of protease inhibitors in influenza virus infected animals. Arch Virol 73: 263–272.

40. MunsterVJ, de WitE, van den BrandJM, HerfstS, SchrauwenEJ, et al. (2009) Pathogenesis and transmission of swine-origin 2009 A(H1N1) influenza virus in ferrets. Science 325: 481–483.

41. Couacy-HymannE, DanhoT, KeitaD, BodjoSC, KouakouC, et al. (2009) The first specific detection of a highly pathogenic avian influenza virus (H5N1) in Ivory Coast. Zoonoses Public Health 56: 10–15.

42. HanssMM, FfrenchPO, MornexJF, ChabuetM, BiotF, et al. (2003) Two novel fibrinogen variants found in patients with pulmonary embolism and their families. J Thromb Haemost 1: 1251–1257.

43. LuyendykJP, SullivanBP, GuoGL, WangR (2010) Tissue factor-deficiency and protease activated receptor-1-deficiency reduce inflammation elicited by diet-induced steatohepatitis in mice. Am J Pathol 176: 177–186.

44. BernardD, RiteauB, HansenJD, PhillipsRB, MichelF, et al. (2006) Costimulatory receptors in a teleost fish: typical CD28, elusive CTLA4. J Immunol 176: 4191–4200.

45. PloplisVA, CarmelietP, VazirzadehS, Van VlaenderenI, MoonsL, et al. (1995) Effects of disruption of the plasminogen gene on thrombosis, growth, and health in mice. Circulation 92: 2585–2593.

46. RiteauB, MoreauP, MenierC, Khalil-DaherI, KhosrotehraniK, et al. (2001) Characterization of HLA-G1, -G2, -G3, and -G4 isoforms transfected in a human melanoma cell line. Transplant Proc 33: 2360–2364.

47. KhoufacheK, LeBouderF, MorelloE, LaurentF, RiffaultS, et al. (2009) Protective role for protease-activated receptor-2 against influenza virus pathogenesis via an IFN-gamma-dependent pathway. J Immunol 182: 7795–7802.

48. LeBouderF, KhoufacheK, MenierC, MandouriY, KeffousM, et al. (2009) Immunosuppressive HLA-G molecule is upregulated in alveolar epithelial cells after influenza A virus infection. Hum Immunol 70: 1016–1019.

49. RiteauB, FaureF, MenierC, VielS, CarosellaED, et al. (2003) Exosomes bearing HLA-G are released by melanoma cells. Hum Immunol 64: 1064–1072.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 3
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#