#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Early Virus-Host Interactions Dictate the Course of a Persistent Infection


Lymphocytic Choriomenengitis Virus (LCMV) is an important model for the investigation of the pathogenesis of persistent viral infections. As with humans infected with hepatitis C and Human Immunodeficiency Virus-1, adult mice persistently infected with immunosuppressive strains of LCMV express high levels of negative immune regulators that suppress the adaptive T cell immune response thereby facilitating viral persistence. Unknown, however, is whether and how very early interactions between the virus and the infected host affect the establishment of a persistent infection. Here, we describe host-virus interactions within the first 8–12 hours of infection are critical for establishing a persistent infection. While early induction of an anti-viral type-I interferons is essential for the subsequent adaptive immune response required to clear the virus, LCMV is able to overcome the programmed innate immune response by over-stimulating this response early. This affects not only the rate of viral growth in the host, but also the ability to infect specific immune cells that help shape an effective adaptive immune response. We further describe how and where LCMV is sensed by this early immune response, identify the critical timing of early virus-host interactions that lead to a persistent infection, and identify an early dysregulated immune signature associated with a persistent viral infection. Altogether, these observations are critical to understanding how early virus-host interactions determines the course of infection.


Vyšlo v časopise: Early Virus-Host Interactions Dictate the Course of a Persistent Infection. PLoS Pathog 11(1): e32767. doi:10.1371/journal.ppat.1004588
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004588

Souhrn

Lymphocytic Choriomenengitis Virus (LCMV) is an important model for the investigation of the pathogenesis of persistent viral infections. As with humans infected with hepatitis C and Human Immunodeficiency Virus-1, adult mice persistently infected with immunosuppressive strains of LCMV express high levels of negative immune regulators that suppress the adaptive T cell immune response thereby facilitating viral persistence. Unknown, however, is whether and how very early interactions between the virus and the infected host affect the establishment of a persistent infection. Here, we describe host-virus interactions within the first 8–12 hours of infection are critical for establishing a persistent infection. While early induction of an anti-viral type-I interferons is essential for the subsequent adaptive immune response required to clear the virus, LCMV is able to overcome the programmed innate immune response by over-stimulating this response early. This affects not only the rate of viral growth in the host, but also the ability to infect specific immune cells that help shape an effective adaptive immune response. We further describe how and where LCMV is sensed by this early immune response, identify the critical timing of early virus-host interactions that lead to a persistent infection, and identify an early dysregulated immune signature associated with a persistent viral infection. Altogether, these observations are critical to understanding how early virus-host interactions determines the course of infection.


Zdroje

1. MarrackP, KapplerJ, MitchellT (1999) Type I interferons keep activated T cells alive. J Exp Med 189: 521–530.

2. CurtsingerJM, ValenzuelaJO, AgarwalP, LinsD, MescherMF (2005) Type I IFNs provide a third signal to CD8 T cells to stimulate clonal expansion and differentiation. J Immunol 174: 4465–4469.

3. AicheleP, UnsoeldH, KoschellaM, SchweierO, KalinkeU, et al. (2006) CD8 T cells specific for lymphocytic choriomeningitis virus require type I IFN receptor for clonal expansion. J Immunol 176: 4525–4529.

4. KolumamGA, ThomasS, ThompsonLJ, SprentJ, Murali-KrishnaK (2005) Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J Exp Med 202: 637–650.

5. BonjardimCA, FerreiraPC, KroonEG (2009) Interferons: signaling, antiviral and viral evasion. Immunol Lett 122: 1–11.

6. ParkSH, RehermannB (2014) Immune responses to HCV and other hepatitis viruses. Immunity 40: 13–24.

7. GoubauD, DeddoucheS, Reis e SousaC (2013) Cytosolic sensing of viruses. Immunity 38: 855–869.

8. HayesM, SalvatoM (2012) Arenavirus evasion of host anti-viral responses. Viruses 4: 2182–2196.

9. TaylorKE, MossmanKL (2013) Recent advances in understanding viral evasion of type I interferon. Immunology 138: 190–197.

10. BowieAG, UnterholznerL (2008) Viral evasion and subversion of pattern-recognition receptor signalling. Nat Rev Immunol 8: 911–922.

11. LooYM, GaleMJr (2007) Viral regulation and evasion of the host response. Curr Top Microbiol Immunol 316: 295–313.

12. RustagiA, GaleMJr (2014) Innate antiviral immune signaling, viral evasion and modulation by HIV-1. J Mol Biol 426: 1161–1177.

13. ZunigaEI, HahmB, OldstoneMB (2007) Type I interferon during viral infections: multiple triggers for a multifunctional mediator. Curr Top Microbiol Immunol 316: 337–357.

14. FanL, BrieseT, LipkinWI (2010) Z proteins of New World arenaviruses bind RIG-I and interfere with type I interferon induction. J Virol 84: 1785–1791.

15. RodrigoWW, Ortiz-RianoE, PythoudC, KunzS, de la TorreJC, et al. (2012) Arenavirus nucleoproteins prevent activation of nuclear factor kappa B. J Virol 86: 8185–8197.

16. PythoudC, RodrigoWW, PasqualG, RothenbergerS, Martinez-SobridoL, et al. (2012) Arenavirus nucleoprotein targets interferon regulatory factor-activating kinase IKKepsilon. J Virol 86: 7728–7738.

17. BorrowP, Martinez-SobridoL, de la TorreJC (2010) Inhibition of the type I interferon antiviral response during arenavirus infection. Viruses 2: 2443–2480.

18. TeijaroJR, NgC, LeeAM, SullivanBM, SheehanKC, et al. (2013) Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 340: 207–211.

19. WilsonEB, YamadaDH, ElsaesserH, HerskovitzJ, DengJ, et al. (2013) Blockade of chronic type I interferon signaling to control persistent LCMV infection. Science 340: 202–207.

20. BlasiusAL, KrebsP, SullivanBM, OldstoneMB, PopkinDL (2012) Slc15a4, a gene required for pDC sensing of TLR ligands, is required to control persistent viral infection. PLoS Pathog 8: e1002915.

21. WangY, SwieckiM, CellaM, AlberG, SchreiberRD, et al. (2012) Timing and magnitude of type I interferon responses by distinct sensors impact CD8 T cell exhaustion and chronic viral infection. Cell Host Microbe 11: 631–642.

22. SouthernPJ, SinghMK, RiviereY, JacobyDR, BuchmeierMJ, et al. (1987) Molecular characterization of the genomic S RNA segment from lymphocytic choriomeningitis virus. Virology 157: 145–155.

23. BuonoP, PaolellaG, ManciniFP, IzzoP, SalvatoreF (1988) The complete nucleotide sequence of the gene coding for the human aldolase C. Nucleic Acids Res 16: 4733.

24. RiviereY, AhmedR, SouthernPJ, BuchmeierMJ, DutkoFJ, et al. (1985) The S RNA segment of lymphocytic choriomeningitis virus codes for the nucleoprotein and glycoproteins 1 and 2. J Virol 53: 966–968.

25. LeeKJ, NovellaIS, TengMN, OldstoneMB, de La TorreJC (2000) NP and L proteins of lymphocytic choriomeningitis virus (LCMV) are sufficient for efficient transcription and replication of LCMV genomic RNA analogs. J Virol 74: 3470–3477.

26. PinschewerDD, PerezM, de la TorreJC (2003) Role of the virus nucleoprotein in the regulation of lymphocytic choriomeningitis virus transcription and RNA replication. J Virol 77: 3882–3887.

27. Martinez-SobridoL, ZunigaEI, RosarioD, Garcia-SastreA, de la TorreJC (2006) Inhibition of the type I interferon response by the nucleoprotein of the prototypic arenavirus lymphocytic choriomeningitis virus. J Virol 80: 9192–9199.

28. BuchmeierMJ, SouthernPJ, ParekhBS, WooddellMK, OldstoneMB (1987) Site-specific antibodies define a cleavage site conserved among arenavirus GP-C glycoproteins. J Virol 61: 982–985.

29. WrightKE, SpiroRC, BurnsJW, BuchmeierMJ (1990) Post-translational processing of the glycoproteins of lymphocytic choriomeningitis virus. Virology 177: 175–183.

30. KunzS, SevillaN, McGavernDB, CampbellKP, OldstoneMB (2001) Molecular analysis of the interaction of LCMV with its cellular receptor [alpha]-dystroglycan. J Cell Biol 155: 301–310.

31. SalvatoMS, ShimomayeEM (1989) The completed sequence of lymphocytic choriomeningitis virus reveals a unique RNA structure and a gene for a zinc finger protein. Virology 173: 1–10.

32. StreckerT, EichlerR, MeulenJ, WeissenhornW, Dieter KlenkH, et al. (2003) Lassa virus Z protein is a matrix protein and sufficient for the release of virus-like particles [corrected]. J Virol 77: 10700–10705.

33. PerezM, CravenRC, de la TorreJC (2003) The small RING finger protein Z drives arenavirus budding: implications for antiviral strategies. Proc Natl Acad Sci U S A 100: 12978–12983.

34. AhmedR, SalmiA, ButlerLD, ChillerJM, OldstoneMB (1984) Selection of genetic variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice. Role in suppression of cytotoxic T lymphocyte response and viral persistence. J Exp Med 160: 521–540.

35. SalvatoM, BorrowP, ShimomayeE, OldstoneMB (1991) Molecular basis of viral persistence: a single amino acid change in the glycoprotein of lymphocytic choriomeningitis virus is associated with suppression of the antiviral cytotoxic T-lymphocyte response and establishment of persistence. J Virol 65: 1863–1869.

36. SullivanBM, EmonetSF, WelchMJ, LeeAM, CampbellKP, et al. (2011) Point mutation in the glycoprotein of lymphocytic choriomeningitis virus is necessary for receptor binding, dendritic cell infection, and long-term persistence. Proc Natl Acad Sci U S A 108: 2969–2974.

37. BergthalerA, FlatzL, HegazyAN, JohnsonS, HorvathE, et al. (2010) Viral replicative capacity is the primary determinant of lymphocytic choriomeningitis virus persistence and immunosuppression. Proc Natl Acad Sci U S A 107: 21641–21646.

38. SmeltSC, BorrowP, KunzS, CaoW, TishonA, et al. (2001) Differences in affinity of binding of lymphocytic choriomeningitis virus strains to the cellular receptor alpha-dystroglycan correlate with viral tropism and disease kinetics. J Virol 75: 448–457.

39. SevillaN, KunzS, HolzA, LewickiH, HomannD, et al. (2000) Immunosuppression and resultant viral persistence by specific viral targeting of dendritic cells. J Exp Med 192: 1249–1260.

40. CaoW, HenryMD, BorrowP, YamadaH, ElderJH, et al. (1998) Identification of alpha-dystroglycan as a receptor for lymphocytic choriomeningitis virus and Lassa fever virus. Science 282: 2079–2081.

41. MatloubianM, KolhekarSR, SomasundaramT, AhmedR (1993) Molecular determinants of macrophage tropism and viral persistence: importance of single amino acid changes in the polymerase and glycoprotein of lymphocytic choriomeningitis virus. J Virol 67: 7340–7349.

42. LeeAM, CruiteJ, WelchMJ, SullivanB, OldstoneMB (2013) Pathogenesis of Lassa fever virus infection: I. Susceptibility of mice to recombinant Lassa Gp/LCMV chimeric virus. Virology 442: 114–121.

43. MacalM, LewisGM, KunzS, FlavellR, HarkerJA, et al. (2012) Plasmacytoid dendritic cells are productively infected and activated through TLR-7 early after arenavirus infection. Cell Host Microbe 11: 617–630.

44. NgCT, OldstoneMB (2012) Infected CD8alpha- dendritic cells are the predominant source of IL-10 during establishment of persistent viral infection. Proc Natl Acad Sci U S A 109: 14116–14121.

45. MuellerSN, MatloubianM, ClemensDM, SharpeAH, FreemanGJ, et al. (2007) Viral targeting of fibroblastic reticular cells contributes to immunosuppression and persistence during chronic infection. Proc Natl Acad Sci U S A 104: 15430–15435.

46. NgCT, NayakBP, SchmedtC, OldstoneMB (2012) Immortalized clones of fibroblastic reticular cells activate virus-specific T cells during virus infection. Proc Natl Acad Sci U S A 109: 7823–7828.

47. BorrowP, EvansCF, OldstoneMB (1995) Virus-induced immunosuppression: immune system-mediated destruction of virus-infected dendritic cells results in generalized immune suppression. J Virol 69: 1059–1070.

48. SevillaN, KunzS, McGavernD, OldstoneMB (2003) Infection of dendritic cells by lymphocytic choriomeningitis virus. Curr Top Microbiol Immunol 276: 125–144.

49. SevillaN, McGavernDB, TengC, KunzS, OldstoneMB (2004) Viral targeting of hematopoietic progenitors and inhibition of DC maturation as a dual strategy for immune subversion. J Clin Invest 113: 737–745.

50. MullerS, HunzikerL, EnzlerS, Buhler-JungoM, Di SantoJP, et al. (2002) Role of an intact splenic microarchitecture in early lymphocytic choriomeningitis virus production. J Virol 76: 2375–2383.

51. OdermattB, EpplerM, LeistTP, HengartnerH, ZinkernagelRM (1991) Virus-triggered acquired immunodeficiency by cytotoxic T-cell-dependent destruction of antigen-presenting cells and lymph follicle structure. Proc Natl Acad Sci U S A 88: 8252–8256.

52. TishonA, BorrowP, EvansC, OldstoneMB (1993) Virus-induced immunosuppression. 1. Age at infection relates to a selective or generalized defect. Virology 195: 397–405.

53. JinHT, AndersonAC, TanWG, WestEE, HaSJ, et al. (2010) Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proc Natl Acad Sci U S A 107: 14733–14738.

54. BrooksDG, TrifiloMJ, EdelmannKH, TeytonL, McGavernDB, et al. (2006) Interleukin-10 determines viral clearance or persistence in vivo. Nat Med 12: 1301–1309.

55. EjrnaesM, FilippiCM, MartinicMM, LingEM, TogherLM, et al. (2006) Resolution of a chronic viral infection after interleukin-10 receptor blockade. J Exp Med 203: 2461–2472.

56. BarberDL, WherryEJ, MasopustD, ZhuB, AllisonJP, et al. (2006) Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439: 682–687.

57. ZajacAJ, BlattmanJN, Murali-KrishnaK, SourdiveDJ, SureshM, et al. (1998) Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med 188: 2205–2213.

58. BrooksDG, TeytonL, OldstoneMB, McGavernDB (2005) Intrinsic functional dysregulation of CD4 T cells occurs rapidly following persistent viral infection. J Virol 79: 10514–10527.

59. BrooksDG, WalshKB, ElsaesserH, OldstoneMB (2010) IL-10 directly suppresses CD4 but not CD8 T cell effector and memory responses following acute viral infection. Proc Natl Acad Sci U S A 107: 3018–3023.

60. MackernessKJ, CoxMA, LillyLM, WeaverCT, HarringtonLE, et al. (2010) Pronounced virus-dependent activation drives exhaustion but sustains IFN-gamma transcript levels. J Immunol 185: 3643–3651.

61. OldstoneMB, BlountP, SouthernPJ, LampertPW (1986) Cytoimmunotherapy for persistent virus infection reveals a unique clearance pattern from the central nervous system. Nature 321: 239–243.

62. WaggonerSN, CornbergM, SelinLK, WelshRM (2012) Natural killer cells act as rheostats modulating antiviral T cells. Nature 481: 394–398.

63. BlasiusAL, ArnoldCN, GeorgelP, RutschmannS, XiaY, et al. (2010) Slc15a4, AP-3, and Hermansky-Pudlak syndrome proteins are required for Toll-like receptor signaling in plasmacytoid dendritic cells. Proc Natl Acad Sci U S A 107: 19973–19978.

64. RichterK, PerriardG, BehrendtR, SchwendenerRA, SexlV, et al. (2013) Macrophage and T cell produced IL-10 promotes viral chronicity. PLoS Pathog 9: e1003735.

65. DieboldSS, KaishoT, HemmiH, AkiraS, Reis e SousaC (2004) Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science 303: 1529–1531.

66. HeilF, HemmiH, HochreinH, AmpenbergerF, KirschningC, et al. (2004) Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 303: 1526–1529.

67. WalshKB, TeijaroJR, ZunigaEI, WelchMJ, FremgenDM, et al. (2012) Toll-like receptor 7 is required for effective adaptive immune responses that prevent persistent virus infection. Cell Host Microbe 11: 643–653.

68. OldstoneMB, CampbellKP (2011) Decoding arenavirus pathogenesis: essential roles for alpha-dystroglycan-virus interactions and the immune response. Virology 411: 170–179.

69. ZhouS, CernyAM, ZachariaA, FitzgeraldKA, Kurt-JonesEA, et al. (2010) Induction and inhibition of type I interferon responses by distinct components of lymphocytic choriomeningitis virus. J Virol 84: 9452–9462.

70. WielandSF, TakahashiK, BoydB, Whitten-BauerC, NgoN, et al. (2014) Human plasmacytoid dendritic cells sense lymphocytic choriomeningitis virus-infected cells in vitro. J Virol 88: 752–757.

71. DreuxM, GaraigortaU, BoydB, DecembreE, ChungJ, et al. (2012) Short-range exosomal transfer of viral RNA from infected cells to plasmacytoid dendritic cells triggers innate immunity. Cell Host Microbe 12: 558–570.

72. SanchezAB, de la TorreJC (2006) Rescue of the prototypic Arenavirus LCMV entirely from plasmid. Virology 350: 370–380.

73. BattegayM, CooperS, AlthageA, BanzigerJ, HengartnerH, et al. (1991) Quantification of lymphocytic choriomeningitis virus with an immunological focus assay in 24- or 96-well plates. J Virol Methods 33: 191–198.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 1
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#