#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Modified Vaccinia Virus Ankara Triggers Type I IFN Production in Murine Conventional Dendritic Cells via a cGAS/STING-Mediated Cytosolic DNA-Sensing Pathway


Modified vaccinia virus Ankara (MVA) is an attenuated vaccinia strain with large deletions of the parental genome that render it non-replicative in mammalian cells. MVA is a safe and effective vaccine against both smallpox and monkeypox. MVA has been investigated as a vaccine vector for infectious diseases and cancers. Dendritic cells (DCs) play important roles in innate and adaptive immunity. A better understanding of how MVA is detected by innate immune sensors in DCs would guide the development of more effective MVA-based vaccines. We report our findings that MVA infection induces the production of type I interferon (IFN) in conventional dendritic cells via a cytosolic DNA-sensing pathway mediated by the newly discovered DNA sensor cGAS, its adaptor STING, and transcription factors IRF3 and IRF7. By contrast, wild-type vaccinia virus fails to activate this pathway. Furthermore, we show that vaccinia virulence factors E3 and N1 play inhibitory roles in the cytosolic DNA-sensing pathway.


Vyšlo v časopise: Modified Vaccinia Virus Ankara Triggers Type I IFN Production in Murine Conventional Dendritic Cells via a cGAS/STING-Mediated Cytosolic DNA-Sensing Pathway. PLoS Pathog 10(4): e32767. doi:10.1371/journal.ppat.1003989
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003989

Souhrn

Modified vaccinia virus Ankara (MVA) is an attenuated vaccinia strain with large deletions of the parental genome that render it non-replicative in mammalian cells. MVA is a safe and effective vaccine against both smallpox and monkeypox. MVA has been investigated as a vaccine vector for infectious diseases and cancers. Dendritic cells (DCs) play important roles in innate and adaptive immunity. A better understanding of how MVA is detected by innate immune sensors in DCs would guide the development of more effective MVA-based vaccines. We report our findings that MVA infection induces the production of type I interferon (IFN) in conventional dendritic cells via a cytosolic DNA-sensing pathway mediated by the newly discovered DNA sensor cGAS, its adaptor STING, and transcription factors IRF3 and IRF7. By contrast, wild-type vaccinia virus fails to activate this pathway. Furthermore, we show that vaccinia virulence factors E3 and N1 play inhibitory roles in the cytosolic DNA-sensing pathway.


Zdroje

1. Fenner F, Henderson DA, Arita I, Jezek Z, Ladnyi ID (1988) Smallpox and its eradication. 1st edn Geneva World Health Organization

2. Moss B (2007) Poxviridae: The viruses and their replication. ; D. M. Knipe e, editor: Lippincott Williams & Wilkins. pp.2905–2946 p.

3. BremanJG, HendersonDA (1998) Poxvirus dilemmas–monkeypox, smallpox, and biologic terrorism. N Engl J Med 339: 556–559.

4. MeyerH, SutterG, MayrA (1991) Mapping of deletions in the genome of the highly attenuated vaccinia virus MVA and their influence on virulence. J Gen Virol 72(Pt 5): 1031–1038.

5. McCurdyLH, LarkinBD, MartinJE, GrahamBS (2004) Modified vaccinia Ankara: potential as an alternative smallpox vaccine. Clin Infect Dis 38: 1749–1753.

6. VollmarJ, ArndtzN, EcklKM, ThomsenT, PetzoldB, et al. (2006) Safety and immunogenicity of IMVAMUNE, a promising candidate as a third generation smallpox vaccine. Vaccine 24: 2065–2070.

7. SutterG, StaibC (2003) Vaccinia vectors as candidate vaccines: the development of modified vaccinia virus Ankara for antigen delivery. Curr Drug Targets Infect Disord 3: 263–271.

8. GomezCE, NajeraJL, KrupaM, EstebanM (2008) The poxvirus vectors MVA and NYVAC as gene delivery systems for vaccination against infectious diseases and cancer. Curr Gene Ther 8: 97–120.

9. GomezCE, NajeraJL, KrupaM, PerdigueroB, EstebanM (2011) MVA and NYVAC as vaccines against emergent infectious diseases and cancer. Curr Gene Ther 11: 189–217.

10. GoepfertPA, ElizagaML, SatoA, QinL, CardinaliM, et al. (2011) Phase 1 safety and immunogenicity testing of DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J Infect Dis 203: 610–619.

11. WyattLS, BelyakovIM, EarlPL, BerzofskyJA, MossB (2008) Enhanced cell surface expression, immunogenicity and genetic stability resulting from a spontaneous truncation of HIV Env expressed by a recombinant MVA. Virology 372: 260–272.

12. GarciaF, Bernaldo de QuirosJC, GomezCE, PerdigueroB, NajeraJL, et al. (2011) Safety and immunogenicity of a modified pox vector-based HIV/AIDS vaccine candidate expressing Env, Gag, Pol and Nef proteins of HIV-1 subtype B (MVA-B) in healthy HIV-1-uninfected volunteers: A phase I clinical trial (RISVAC02). Vaccine 29: 8309–8316.

13. StetsonDB, MedzhitovR (2006) Type I interferons in host defense. Immunity 25: 373–381.

14. KawaiT, AkiraS (2011) Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 34: 637–650.

15. SatpathyAT, WuX, AlbringJC, MurphyKM (2012) Re(de)fining the dendritic cell lineage. Nat Immunol 13: 1145–1154.

16. DengL, DaiP, DingW, GransteinRD, ShumanS (2006) Vaccinia virus infection attenuates innate immune responses and antigen presentation by epidermal dendritic cells. J Virol 80: 9977–9987.

17. DaiP, CaoH, MerghoubT, AvogadriF, WangW, et al. (2011) Myxoma Virus Induces Type I Interferon Production in Murine Plasmacytoid Dendritic Cells via a TLR9/MyD88-, IRF5/IRF7-, and IFNAR-Dependent Pathway. J Virol 85: 10814–10825.

18. CaoH, DaiP, WangW, LiH, YuanJ, et al. (2012) Innate Immune Response of Human Plasmacytoid Dendritic Cells to Poxvirus Infection Is Subverted by Vaccinia E3 via Its Z-DNA/RNA Binding Domain. PLoS One 7: e36823.

19. BowieA, Kiss-TothE, SymonsJA, SmithGL, DowerSK, et al. (2000) A46R and A52R from vaccinia virus are antagonists of host IL-1 and toll-like receptor signaling. Proc Natl Acad Sci U S A 97: 10162–10167.

20. HarteMT, HagaIR, MaloneyG, GrayP, ReadingPC, et al. (2003) The poxvirus protein A52R targets Toll-like receptor signaling complexes to suppress host defense. J Exp Med 197: 343–351.

21. DiPernaG, StackJ, BowieAG, BoydA, KotwalG, et al. (2004) Poxvirus protein N1L targets the I-kappaB kinase complex, inhibits signaling to NF-kappaB by the tumor necrosis factor superfamily of receptors, and inhibits NF-kappaB and IRF3 signaling by toll-like receptors. J Biol Chem 279: 36570–36578.

22. GrahamSC, BaharMW, CoorayS, ChenRA, WhalenDM, et al. (2008) Vaccinia virus proteins A52 and B14 Share a Bcl-2-like fold but have evolved to inhibit NF-kappaB rather than apoptosis. PLoS Pathog 4: e1000128.

23. LynchHE, RayCA, OieKL, PollaraJJ, PettyIT, et al. (2009) Modified vaccinia virus Ankara can activate NF-kappaB transcription factors through a double-stranded RNA-activated protein kinase (PKR)-dependent pathway during the early phase of virus replication. Virology 391: 177–186.

24. WillisKL, PatelS, XiangY, ShislerJL (2009) The effect of the vaccinia K1 protein on the PKR-eIF2alpha pathway in RK13 and HeLa cells. Virology 394: 73–81.

25. KotwalGJ, HuginAW, MossB (1989) Mapping and insertional mutagenesis of a vaccinia virus gene encoding a 13,800-Da secreted protein. Virology 171: 579–587.

26. BartlettN, SymonsJA, TscharkeDC, SmithGL (2002) The vaccinia virus N1L protein is an intracellular homodimer that promotes virulence. J Gen Virol 83: 1965–1976.

27. BrandtT, HeckMC, VijaysriS, JentarraGM, CameronJM, et al. (2005) The N-terminal domain of the vaccinia virus E3L-protein is required for neurovirulence, but not induction of a protective immune response. Virology 333: 263–270.

28. ChangHW, WatsonJC, JacobsBL (1992) The E3L gene of vaccinia virus encodes an inhibitor of the interferon-induced, double-stranded RNA-dependent protein kinase. Proc Natl Acad Sci U S A 89: 4825–4829.

29. XiangY, ConditRC, VijaysriS, JacobsB, WilliamsBR, et al. (2002) Blockade of interferon induction and action by the E3L double-stranded RNA binding proteins of vaccinia virus. J Virol 76: 5251–5259.

30. BeattieE, KauffmanEB, MartinezH, PerkusME, JacobsBL, et al. (1996) Host-range restriction of vaccinia virus E3L-specific deletion mutants. Virus Genes 12: 89–94.

31. BrandtTA, JacobsBL (2001) Both carboxy- and amino-terminal domains of the vaccinia virus interferon resistance gene, E3L, are required for pathogenesis in a mouse model. J Virol 75: 850–856.

32. LanglandJO, KashJC, CarterV, ThomasMJ, KatzeMG, et al. (2006) Suppression of proinflammatory signal transduction and gene expression by the dual nucleic acid binding domains of the vaccinia virus E3L proteins. J Virol 80: 10083–10095.

33. SmithEJ, MarieI, PrakashA, Garcia-SastreA, LevyDE (2001) IRF3 and IRF7 phosphorylation in virus-infected cells does not require double-stranded RNA-dependent protein kinase R or Ikappa B kinase but is blocked by Vaccinia virus E3L protein. J Biol Chem 276: 8951–8957.

34. GuerraS, CaceresA, KnobelochKP, HorakI, EstebanM (2008) Vaccinia virus E3 protein prevents the antiviral action of ISG15. PLoS Pathog 4: e1000096.

35. DengL, DaiP, ParikhT, CaoH, BhojV, et al. (2008) Vaccinia virus subverts a mitochondrial antiviral signaling protein-dependent innate immune response in keratinocytes through its double-stranded RNA binding protein, E3. J Virol 82: 10735–10746.

36. DrillienR, SpehnerD, HanauD (2004) Modified vaccinia virus Ankara induces moderate activation of human dendritic cells. J Gen Virol 85: 2167–2175.

37. WaiblerZ, AnzagheM, LudwigH, AkiraS, WeissS, et al. (2007) Modified vaccinia virus Ankara induces Toll-like receptor-independent type I interferon responses. J Virol 81: 12102–12110.

38. DelaloyeJ, RogerT, Steiner-TardivelQG, Le RoyD, Knaup ReymondM, et al. (2009) Innate immune sensing of modified vaccinia virus Ankara (MVA) is mediated by TLR2-TLR6, MDA-5 and the NALP3 inflammasome. PLoS Pathog 5: e1000480.

39. LaouarY, WelteT, FuXY, FlavellRA (2003) STAT3 is required for Flt3L-dependent dendritic cell differentiation. Immunity 19: 903–912.

40. McKennaHJ (2001) Role of hematopoietic growth factors/flt3 ligand in expansion and regulation of dendritic cells. Curr Opin Hematol 8: 149–154.

41. LinR, HeylbroeckC, PithaPM, HiscottJ (1998) Virus-dependent phosphorylation of the IRF-3 transcription factor regulates nuclear translocation, transactivation potential, and proteasome-mediated degradation. Mol Cell Biol 18: 2986–2996.

42. SatoM, SuemoriH, HataN, AsagiriM, OgasawaraK, et al. (2000) Distinct and essential roles of transcription factors IRF-3 and IRF-7 in response to viruses for IFN-alpha/beta gene induction. Immunity 13: 539–548.

43. HondaK, YanaiH, TakaokaA, TaniguchiT (2005) Regulation of the type I IFN induction: a current view. Int Immunol 17: 1367–1378.

44. KawaiT, TakahashiK, SatoS, CobanC, KumarH, et al. (2005) IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nat Immunol 6: 981–988.

45. AlexopoulouL, HoltAC, MedzhitovR, FlavellRA (2001) Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature 413: 732–738.

46. KawaiT, AkiraS (2006) Innate immune recognition of viral infection. Nat Immunol 7: 131–137.

47. ZhangZ, KimT, BaoM, FacchinettiV, JungSY, et al. (2011) DDX1, DDX21, and DHX36 helicases form a complex with the adaptor molecule TRIF to sense dsRNA in dendritic cells. Immunity 34: 866–878.

48. BarberGN (2011) Innate immune DNA sensing pathways: STING, AIMII and the regulation of interferon production and inflammatory responses. Curr Opin Immunol 23: 10–20.

49. IshikawaH, BarberGN (2008) STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 455: 674–678.

50. IshikawaH, MaZ, BarberGN (2009) STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 461: 788–792.

51. JinL, WatermanPM, JonscherKR, ShortCM, ReisdorphNA, et al. (2008) MPYS, a novel membrane tetraspanner, is associated with major histocompatibility complex class II and mediates transduction of apoptotic signals. Mol Cell Biol 28: 5014–5026.

52. SunW, LiY, ChenL, ChenH, YouF, et al. (2009) ERIS, an endoplasmic reticulum IFN stimulator, activates innate immune signaling through dimerization. Proc Natl Acad Sci U S A 106: 8653–8658.

53. ZhongB, YangY, LiS, WangYY, LiY, et al. (2008) The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation. Immunity 29: 538–550.

54. SauerJD, Sotelo-TrohaK, von MoltkeJ, MonroeKM, RaeCS, et al. (2011) The N-ethyl-N-nitrosourea-induced Goldenticket mouse mutant reveals an essential function of Sting in the in vivo interferon response to Listeria monocytogenes and cyclic dinucleotides. Infect Immun 79: 688–694.

55. WaiblerZ, AnzagheM, FrenzT, SchwantesA, PohlmannC, et al. (2009) Vaccinia virus-mediated inhibition of type I interferon responses is a multifactorial process involving the soluble type I interferon receptor B18 and intracellular components. J Virol 83: 1563–1571.

56. SunL, WuJ, DuF, ChenX, ChenZJ (2013) Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339: 786–791.

57. WuJ, SunL, ChenX, DuF, ShiH, et al. (2013) Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339: 826–830.

58. LiXD, WuJ, GaoD, WangH, SunL, et al. (2013) Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science 341: 1390–1394.

59. MossB, CooperN (1982) Genetic evidence for vaccinia virus-encoded DNA polymerase: isolation of phosphonoacetate-resistant enzyme from the cytoplasm of cells infected with mutant virus. J Virol 43: 673–678.

60. JonesEV, MossB (1984) Mapping of the vaccinia virus DNA polymerase gene by marker rescue and cell-free translation of selected RNA. J Virol 49: 72–77.

61. FredericksenBL, WeiBL, YaoJ, LuoT, GarciaJV (2002) Inhibition of endosomal/lysosomal degradation increases the infectivity of human immunodeficiency virus. J Virol 76: 11440–11446.

62. ButtleDJ, MurataM, KnightCG, BarrettAJ (1992) CA074 methyl ester: a proinhibitor for intracellular cathepsin B. Arch Biochem Biophys 299: 377–380.

63. DeussingJ, RothW, SaftigP, PetersC, PloeghHL, et al. (1998) Cathepsins B and D are dispensable for major histocompatibility complex class II-mediated antigen presentation. Proc Natl Acad Sci U S A 95: 4516–4521.

64. AntoineG, ScheiflingerF, DornerF, FalknerFG (1998) The complete genomic sequence of the modified vaccinia Ankara strain: comparison with other orthopoxviruses. Virology 244: 365–396.

65. Meisinger-HenschelC, SchmidtM, LukassenS, LinkeB, KrauseL, et al. (2007) Genomic sequence of chorioallantois vaccinia virus Ankara, the ancestor of modified vaccinia virus Ankara. J Gen Virol 88: 3249–3259.

66. GaoP, AscanoM, WuY, BarchetW, GaffneyBL, et al. (2013) Cyclic [G(2′,5′)pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell 153: 1094–1107.

67. KranzuschPJ, LeeAS, BergerJM, DoudnaJA (2013) Structure of human cGAS reveals a conserved family of second-messenger enzymes in innate immunity. Cell Rep 3: 1362–1368.

68. CivrilF, DeimlingT, de Oliveira MannCC, AblasserA, MoldtM, et al. (2013) Structural mechanism of cytosolic DNA sensing by cGAS. Nature 498: 332–337.

69. AblasserA, GoldeckM, CavlarT, DeimlingT, WitteG, et al. (2013) cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING. Nature 498: 380–384.

70. GaoP, AscanoM, ZillingerT, WangW, DaiP, et al. (2013) Structure-Function Analysis of STING Activation by c[G(2′,5′)pA(3′,5′)p] and Targeting by Antiviral DMXAA. Cell 154: 748–762.

71. DinerEJ, BurdetteDL, WilsonSC, MonroeKM, KellenbergerCA, et al. (2013) The innate immune DNA sensor cGAS produces a noncanonical cyclic dinucleotide that activates human STING. Cell Rep 3: 1355–1361.

72. ZhangX, ShiH, WuJ, ZhangX, SunL, et al. (2013) Cyclic GMP-AMP containing mixed phosphodiester linkages is an endogenous high-affinity ligand for STING. Mol Cell 51: 226–235.

73. BurdetteDL, MonroeKM, Sotelo-TrohaK, IwigJS, EckertB, et al. (2011) STING is a direct innate immune sensor of cyclic di-GMP. Nature 478: 515–518.

74. RasmussenSB, HoranKA, HolmCK, StranksAJ, MettenleiterTC, et al. (2011) Activation of autophagy by alpha-herpesviruses in myeloid cells is mediated by cytoplasmic viral DNA through a mechanism dependent on stimulator of IFN genes. J Immunol 187: 5268–5276.

75. UnterholznerL, KeatingSE, BaranM, HoranKA, JensenSB, et al. (2010) IFI16 is an innate immune sensor for intracellular DNA. Nat Immunol 11: 997–1004.

76. ZhangZ, YuanB, BaoM, LuN, KimT, et al. (2011) The helicase DDX41 senses intracellular DNA mediated by the adaptor STING in dendritic cells. Nat Immunol 12: 959–965.

77. HondaK, TaniguchiT (2006) IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors. Nat Rev Immunol 6: 644–658.

78. DaffisS, SutharMS, SzretterKJ, GaleMJr, DiamondMS (2009) Induction of IFN-beta and the innate antiviral response in myeloid cells occurs through an IPS-1-dependent signal that does not require IRF-3 and IRF-7. PLoS Pathog 5: e1000607.

79. FischerSF, LudwigH, HolzapfelJ, KvansakulM, ChenL, et al. (2006) Modified vaccinia virus Ankara protein F1L is a novel BH3-domain-binding protein and acts together with the early viral protein E3L to block virus-associated apoptosis. Cell Death Differ 13: 109–118.

80. LudwigH, MagesJ, StaibC, LehmannMH, LangR, et al. (2005) Role of viral factor E3L in modified vaccinia virus ankara infection of human HeLa Cells: regulation of the virus life cycle and identification of differentially expressed host genes. J Virol 79: 2584–2596.

81. LudwigH, SuezerY, WaiblerZ, KalinkeU, SchnierleBS, et al. (2006) Double-stranded RNA-binding protein E3 controls translation of viral intermediate RNA, marking an essential step in the life cycle of modified vaccinia virus Ankara. J Gen Virol 87: 1145–1155.

82. WhiteSD, JacobsBL (2012) The amino terminus of the vaccinia virus E3 protein is necessary to inhibit the interferon response. J Virol 86: 5895–5904.

83. TischerBK, von EinemJ, KauferB, OsterriederN (2006) Two-step red-mediated recombination for versatile high-efficiency markerless DNA manipulation in Escherichia coli. Biotechniques 40: 191–197.

84. WeaverJR, ShamimM, AlexanderE, DaviesDH, FelgnerPL, et al. (2007) The identification and characterization of a monoclonal antibody to the vaccinia virus E3 protein. Virus Res 130: 269–274.

85. LiuL, FuhlbriggeRC, KaribianK, TianT, KupperTS (2006) Dynamic programming of CD8+ T cell trafficking after live viral immunization. Immunity 25: 511–520.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 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#