#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Epstein-Barr Virus Large Tegument Protein BPLF1 Contributes to Innate Immune Evasion through Interference with Toll-Like Receptor Signaling


Viral infection triggers an early host response through activation of pattern recognition receptors, including Toll-like receptors (TLR). TLR signaling cascades induce production of type I interferons and proinflammatory cytokines involved in establishing an anti-viral state as well as in orchestrating ensuing adaptive immunity. To allow infection, replication, and persistence, (herpes)viruses employ ingenious strategies to evade host immunity. The human gamma-herpesvirus Epstein-Barr virus (EBV) is a large, enveloped DNA virus persistently carried by more than 90% of adults worldwide. It is the causative agent of infectious mononucleosis and is associated with several malignant tumors. EBV activates TLRs, including TLR2, TLR3, and TLR9. Interestingly, both the expression of and signaling by TLRs is attenuated during productive EBV infection. Ubiquitination plays an important role in regulating TLR signaling and is controlled by ubiquitin ligases and deubiquitinases (DUBs). The EBV genome encodes three proteins reported to exert in vitro deubiquitinase activity. Using active site-directed probes, we show that one of these putative DUBs, the conserved herpesvirus large tegument protein BPLF1, acts as a functional DUB in EBV-producing B cells. The BPLF1 enzyme is expressed during the late phase of lytic EBV infection and is incorporated into viral particles. The N-terminal part of the large BPLF1 protein contains the catalytic site for DUB activity and suppresses TLR-mediated activation of NF-κB at, or downstream of, the TRAF6 signaling intermediate. A catalytically inactive mutant of this EBV protein did not reduce NF-κB activation, indicating that DUB activity is essential for attenuating TLR signal transduction. Our combined results show that EBV employs deubiquitination of signaling intermediates in the TLR cascade as a mechanism to counteract innate anti-viral immunity of infected hosts.


Vyšlo v časopise: Epstein-Barr Virus Large Tegument Protein BPLF1 Contributes to Innate Immune Evasion through Interference with Toll-Like Receptor Signaling. PLoS Pathog 10(2): e32767. doi:10.1371/journal.ppat.1003960
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003960

Souhrn

Viral infection triggers an early host response through activation of pattern recognition receptors, including Toll-like receptors (TLR). TLR signaling cascades induce production of type I interferons and proinflammatory cytokines involved in establishing an anti-viral state as well as in orchestrating ensuing adaptive immunity. To allow infection, replication, and persistence, (herpes)viruses employ ingenious strategies to evade host immunity. The human gamma-herpesvirus Epstein-Barr virus (EBV) is a large, enveloped DNA virus persistently carried by more than 90% of adults worldwide. It is the causative agent of infectious mononucleosis and is associated with several malignant tumors. EBV activates TLRs, including TLR2, TLR3, and TLR9. Interestingly, both the expression of and signaling by TLRs is attenuated during productive EBV infection. Ubiquitination plays an important role in regulating TLR signaling and is controlled by ubiquitin ligases and deubiquitinases (DUBs). The EBV genome encodes three proteins reported to exert in vitro deubiquitinase activity. Using active site-directed probes, we show that one of these putative DUBs, the conserved herpesvirus large tegument protein BPLF1, acts as a functional DUB in EBV-producing B cells. The BPLF1 enzyme is expressed during the late phase of lytic EBV infection and is incorporated into viral particles. The N-terminal part of the large BPLF1 protein contains the catalytic site for DUB activity and suppresses TLR-mediated activation of NF-κB at, or downstream of, the TRAF6 signaling intermediate. A catalytically inactive mutant of this EBV protein did not reduce NF-κB activation, indicating that DUB activity is essential for attenuating TLR signal transduction. Our combined results show that EBV employs deubiquitination of signaling intermediates in the TLR cascade as a mechanism to counteract innate anti-viral immunity of infected hosts.


Zdroje

1. Rickinson AB, Kieff E (2007) Epstein-Barr Virus. In: Knipe DM, Howley PM, editors. Field's Virology. Philadelphia: Lippincott Williams & Wilkins. pp. 2655–2700.

2. KutokJL, WangF (2006) Spectrum of Epstein-Barr Virus-Associated Diseases. Annu Rev Pathol Mech Dis 1: 375–404.

3. IwasakiA, MedzhitovR (2010) Regulation of Adaptive Immunity by the Innate Immune System. Science 327: 291–295.

4. Szomolanyi-TsudaE, LiangX, WelshRM, Kurt-JonesEA, FinbergRW (2006) Role for TLR2 in NK Cell-Mediated Control of Murine Cytomegalovirus In Vivo. Journal of Virology 80: 4286–4291.

5. TabetaK, GeorgelP, JanssenE, DuX, HoebeK, et al. (2004) Toll-like receptors 9 and 3 as essential components of innate immune defense against mouse cytomegalovirus infection. Proceedings of the National Academy of Sciences of the United States of America 101: 3516–3521.

6. ZucchiniN, BessouG, TraubS, RobbinsSH, UematsuS, et al. (2008) Cutting Edge: Overlapping Functions of TLR7 and TLR9 for Innate Defense against a Herpesvirus Infection. The Journal of Immunology 180: 5799–5803.

7. DaveyGM, WojtasiakM, ProiettoAI, CarboneFR, HeathWR, et al. (2010) Cutting Edge: Priming of CD8 T Cell Immunity to Herpes Simplex Virus Type 1 Requires Cognate TLR3 Expression In Vivo. The Journal of Immunology 184: 2243–2246.

8. SorensenLN, ReinertLS, MalmgaardL, BartholdyC, ThomsenAR, et al. (2008) TLR2 and TLR9 Synergistically Control Herpes Simplex Virus Infection in the Brain. The Journal of Immunology 181: 8604–8612.

9. CasrougeA, ZhangSY, EidenschenkC, JouanguyE, PuelA, et al. (2006) Herpes Simplex Virus Encephalitis in Human UNC-93B Deficiency. Science 314: 308–312.

10. GuoY, AudryM, CiancanelliM, AlsinaL, AzevedoJ, et al. (2011) Herpes simplex virus encephalitis in a patient with complete TLR3 deficiency: TLR3 is otherwise redundant in protective immunity. The Journal of Experimental Medicine 208: 2083–2098.

11. Perez de DiegoR, Sancho-ShimizuV, LorenzoL, PuelA, PlancoulaineS, et al. (2010) Human TRAF3 Adaptor Molecule Deficiency Leads to Impaired Toll-like Receptor 3 Response and Susceptibility to Herpes Simplex Encephalitis. Immunity 33: 400–411.

12. Sancho-ShimizuV, rez de DiegoR, LorenzoL, HalwaniR, AlangariA, et al. (2011) Herpes simplex encephalitis in children with autosomal recessive and dominant TRIF deficiency. J Clin Invest 121: 4889–4902.

13. ZhangSY, JouanguyE, UgoliniS, SmahiA, ElainG, et al. (2007) TLR3 Deficiency in Patients with Herpes Simplex Encephalitis. Science 317: 1522–1527.

14. PaludanSR, BowieAG, HoranKA, FitzgeraldKA (2011) Recognition of herpesviruses by the innate immune system. Nat Rev Immunol 11: 143–154.

15. ArizaME, GlaserR, KaumayaPTP, JonesC, WilliamsMV (2009) The EBV-Encoded dUTPase Activates NF-kB through the TLR2 and MyD88-Dependent Signaling Pathway. The Journal of Immunology 182: 851–859.

16. GaudreaultE, FiolaS, OlivierM, GosselinJ (2007) Epstein-Barr Virus Induces MCP-1 Secretion by Human Monocytes via TLR2. J Virol 81: 8016–8024.

17. FiolaS, GosselinD, TakadaK, GosselinJ (2010) TLR9 Contributes to the Recognition of EBV by Primary Monocytes and Plasmacytoid Dendritic Cells. The Journal of Immunology 185: 3620–3631.

18. IwakiriD, ZhouL, SamantaM, MatsumotoM, EbiharaT, et al. (2009) Epstein-Barr virus (EBV)-encoded small RNA is released from EBV-infected cells and activates signaling from Toll-like receptor 3. The Journal of Experimental Medicine 206: 2091–2099.

19. LimWH, KiretaS, RussGR, CoatesPTH (2007) Human plasmacytoid dendritic cells regulate immune responses to Epstein-Barr virus (EBV) infection and delay EBV-related mortality in humanized NOD-SCID mice. Blood 109: 1043–1050.

20. QuanTE, RomanRM, RudengaBJ, HolersVM, CraftJE (2010) Epstein-Barr virus promotes interferon-alpha production by plasmacytoid dendritic cells. Arthritis & Rheumatism 62: 1693–1701.

21. MogensenTH (2009) Pathogen Recognition and Inflammatory Signaling in Innate Immune Defenses. Clinical Microbiology Reviews 22: 240–273.

22. TakeuchiO, AkiraS (2009) Innate immunity to virus infection. Immunological Reviews 227: 75–86.

23. van GentM, GriffinBD, BerkhoffEG, van LeeuwenD, BoerIGJ, et al. (2011) EBV Lytic-Phase Protein BGLF5 Contributes to TLR9 Downregulation during Productive Infection. The Journal of Immunology 186: 1694–1702.

24. WertzIE, DixitVM (2010) Signaling to NF-kB: Regulation by Ubiquitination. Cold Spring Harbor Perspectives in Biology 2: 1–19.

25. BooneDL, TurerEE, LeeEG, AhmadRC, WheelerMT, et al. (2004) The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol 5: 1052–1060.

26. BrummelkampTR, NijmanSMB, DiracAMG, BernardsR (2003) Loss of the cylindromatosis tumour suppressor inhibits apoptosis by activating NF-[kappa]B. Nature 424: 797–801.

27. IsaacsonMK, PloeghHL (2009) Ubiquitination, Ubiquitin-like Modifiers, and Deubiquitination in Viral Infection. Cell Host Microbe 5: 559–570.

28. RandowF, LehnerPJ (2009) Viral avoidance and exploitation of the ubiquitin system. Nat Cell Biol 11: 527–534.

29. GredmarkS, SchliekerC, QuesadaV, SpoonerE, PloeghHL (2007) A Functional Ubiquitin-Specific Protease Embedded in the Large Tegument Protein (ORF64) of Murine Gammaherpesvirus 68 Is Active during the Course of Infection. Journal of Virology 81: 10300–10309.

30. KattenhornLM, KorbelGA, KesslerBM, SpoonerE, PloeghHL (2005) A Deubiquitinating Enzyme Encoded by HSV-1 Belongs to a Family of Cysteine Proteases that Is Conserved across the Family Herpesviridae. Mol Cell 19: 547–557.

31. SchliekerC, KorbelGA, KattenhornLM, PloeghHL (2005) A Deubiquitinating Activity Is Conserved in the Large Tegument Protein of the Herpesviridae. Journal of Virology 79: 15582–15585.

32. SompallaeR, GastaldelloS, HildebrandS, ZininN, HassinkG, et al. (2008) Epstein-Barr Virus Encodes Three Bona Fide Ubiquitin-Specific Proteases. Journal of Virology 82: 10477–10486.

33. RessingME, KeatingSE, van LeeuwenD, Koppers-LalicD, PappworthIY, et al. (2005) Impaired Transporter Associated with Antigen Processing-Dependent Peptide Transport during Productive EBV Infection. The Journal of Immunology 174: 6829–6838.

34. BornkammGW, HudewentzJ, FreeseUK, ZimberU (1982) Deletion of the Nontransforming Epstein-Barr Virus Strain P3HR-1 Causes Fusion of the Large Internal Repeat to the DSL Region. Journal of Virology 43: 952–968.

35. Kurt-JonesEA, MandellL, WhitneyC, PadgettA, GosselinK, et al. (2002) Role of Toll-like receptor 2 (TLR2) in neutrophil activation: GM-CSF enhances TLR2 expression and TLR2-mediated interleukin 8 responses in neutrophils. Blood 100: 1860–1868.

36. de JongA, MerkxR, BerlinI, RodenkoB, WijdevenRHM, et al. (2012) Ubiquitin-Based Probes Prepared by Total Synthesis To Profile the Activity of Deubiquitinating Enzymes. ChemBioChem 13: 2251–2258.

37. TsaiCH, LiuMT, ChenMR, LuJ, YangHL, et al. (1997) Characterization of Monoclonal Antibodies to the Zta and DNase proteins of Epstein-Barr Virus. J Biomed Sci 4: 69–77.

38. GastaldelloS, HildebrandS, FaridaniO, CallegariS, PalmkvistM, et al. (2010) A deneddylase encoded by Epstein-Barr virus promotes viral DNA replication by regulating the activity of cullin-RING ligases. Nat Cell Biol 12: 351–361.

39. GastaldelloS, CallegariS, CoppotelliG, HildebrandS, SongM, et al. (2012) Herpes virus deneddylases interrupt the cullin-RING ligase neddylation cycle by inhibiting the binding of CAND1. Journal of Molecular Cell Biology 4: 242–251.

40. WhitehurstCB, NingS, BentzGL, DufourF, GershburgE, et al. (2009) The Epstein-Barr Virus (EBV) Deubiquitinating Enzyme BPLF1 Reduces EBV Ribonucleotide Reductase Activity. Journal of Virology 83: 4345–4353.

41. WhitehurstCB, VaziriC, ShackelfordJ, PaganoJS (2012) Epstein-Barr Virus BPLF1 Deubiquitinates PCNA and Attenuates Polymerase eta Recruitment to DNA Damage Sites. Journal of Virology 86: 8097–8106.

42. GastaldelloS, ChenX, CallegariS, MasucciMG (2013) Caspase-1 Promotes Epstein-Barr Virus Replication by Targeting the Large Tegument Protein Deneddylase to the Nucleus of Productively Infected Cells. PLoS Pathog 9: e1003664.

43. SachdevS, HoffmannA, HanninkM (1998) Nuclear Localization of IkBa Is Mediated by the Second Ankyrin Repeat: the IkBa Ankyrin Repeats Define a Novel Class of cis-Acting Nuclear Import Sequences. Molecular and Cellular Biology 18: 2524–2534.

44. SchmausS, WolfH, SchwarzmannF (2004) The Reading Frame BPLF1 of Epstein-Barr Virus: A Homologue of Herpes Simplex Virus Protein VP16. Virus Genes 29: 267–277.

45. RoweM, GlaunsingerB, van LeeuwenD, ZuoJ, SweetmanD, et al. (2007) Host shutoff during productive Epstein-Barr virus infection is mediated by BGLF5 and may contribute to immune evasion. Proceedings of the National Academy of Sciences 104: 3366–3371.

46. SchliekerC, WeihofenWA, FrijnsE, KattenhornLM, GaudetR, et al. (2007) Structure of a Herpesvirus-Encoded Cysteine Protease Reveals a Unique Class of Deubiquitinating Enzymes. Mol Cell 25: 677–687.

47. KimET, OhSE, LeeYO, GibsonW, AhnJH (2009) Cleavage Specificity of the UL48 Deubiquitinating Protease Activity of Human Cytomegalovirus and the Growth of an Active-Site Mutant Virus in Cultured Cells. Journal of Virology 83: 12046–12056.

48. RobertsAPE, AbaituaF, O'HareP, McNabD, RixonFJ, et al. (2009) Differing Roles of Inner Tegument Proteins pUL36 and pUL37 during Entry of Herpes Simplex Virus Type 1. Journal of Virology 83: 105–116.

49. GredmarkR, IsaacsonMK, KattenhornL, CheungEJ, WatsonN, et al. (2009) A Gammaherpesvirus Ubiquitin-Specific Protease Is Involved in the Establishment of Murine Gammaherpesvirus 68 Infection. Journal of Virology 83: 10644–10652.

50. SaitoS, MurataT, KandaT, IsomuraH, NaritaY, et al. (2013) Epstein-Barr Virus Deubiquitinase Downregulates TRAF6-Mediated NF-kB Signaling during Productive Replication. Journal of Virology 87: 4060–4070.

51. ErnstR, ClaessenJHL, MuellerB, SanyalS, SpoonerE, et al. (2011) Enzymatic Blockade of the Ubiquitin-Proteasome Pathway. PLoS Biol 8: e1000605.

52. OvaaH, KesslerBM, RolénU, GalardyPJ, PloeghHL, et al. (2004) Activity-based ubiquitin-specific protease (USP) profiling of virus-infected and malignant human cells. Proceedings of the National Academy of Sciences of the United States of America 101: 2253–2258.

53. JohannsenE, LuftigM, ChaseMR, WeickselS, Cahir-McFarlandE, et al. (2004) Proteins of purified Epstein-Barr virus. Proceedings of the National Academy of Sciences of the United States of America 101: 16286–16291.

54. AbaituaF, HollinsheadM, BolstadM, CrumpCM, O'HareP (2012) A Nuclear Localization Signal in Herpesvirus Protein VP1-2 Is Essential for Infection via Capsid Routing to the Nuclear Pore. Journal of Virology 86: 8998–9014.

55. JovasevicV, LiangL, RoizmanB (2008) Proteolytic Cleavage of VP1-2 Is Required for Release of Herpes Simplex Virus 1 DNA into the Nucleus. Journal of Virology 82: 3311–3319.

56. SchipkeJ, PohlmannA, DiestelR, BinzA, RudolphK, et al. (2012) The C Terminus of the Large Tegument Protein pUL36 Contains Multiple Capsid Binding Sites That Function Differently during Assembly and Cell Entry of Herpes Simplex Virus. Journal of Virology 86: 3682–3700.

57. WangJ, LovelandAN, KattenhornLM, PloeghHL, GibsonW (2006) High-Molecular-Weight Protein (pUL48) of Human Cytomegalovirus Is a Competent Deubiquitinating Protease: Mutant Viruses Altered in Its Active-Site Cysteine or Histidine Are Viable. Journal of Virology 80: 6003–6012.

58. VazirabadiG, GeigerTR, CoffinIII, MartinJM (2003) Epstein-Barr virus latent membrane protein-1 (LMP-1) and lytic LMP-1 localization in plasma membrane-derived extracellular vesicles and intracellular virions. Journal of General Virology 84: 1997–2008.

59. PandyaJ, WallingDM (2006) Oncogenic Activity of Epstein-Barr Virus Latent Membrane Protein 1 (LMP-1) Is Down-Regulated by Lytic LMP-1. Journal of Virology 80: 8038–8046.

60. EricksonKD, MartinJM (2000) The Late Lytic LMP-1 Protein of Epstein-Barr Virus Can Negatively Regulate LMP-1 Signaling. Journal of Virology 74: 1057–1060.

61. MansurDS, Maluquer de MotesC, UnterholznerL, SumnerRP, FergusonBJ, et al. (2013) Poxvirus Targeting of E3 Ligase ßTrCP by Molecular Mimicry: A Mechanism to Inhibit NF-kB Activation and Promote Immune Evasion and Virulence. PLoS Pathog 9: e1003183.

62. YuY, WangSE, HaywardGS (2005) The KSHV Immediate-Early Transcription Factor RTA Encodes Ubiquitin E3 Ligase Activity that Targets IRF7 for Proteosome-Mediated Degradation. Immunity 22: 59–70.

63. KarimR, TummersB, MeyersC, BiryukovJL, AlamS, et al. (2013) Human Papillomavirus (HPV) Upregulates the Cellular Deubiquitinase UCHL1 to Suppress the Keratinocyte's Innate Immune Response. PLoS Pathog 9: e1003384.

64. DaubeufS, SinghD, TanY, LiuH, FederoffHJ, et al. (2009) HSV ICP0 recruits USP7 to modulate TLR-mediated innate response. Blood 113: 3264–3275.

65. van KasterenPB, BeugelingC, NinaberDK, Frias-StaheliN, van BoheemenS, et al. (2012) Arterivirus and Nairovirus Ovarian Tumor Domain-Containing Deubiquitinases Target Activated RIG-I To Control Innate Immune Signaling. Journal of Virology 86: 773–785.

66. van KasterenPB, Bailey-ElkinBA, JamesTW, NinaberDK, BeugelingC, et al. (2013) Deubiquitinase function of arterivirus papain-like protease 2 suppresses the innate immune response in infected host cells. Proceedings of the National Academy of Sciences 110: E838–E847.

67. BalakirevMY, JaquinodM, HaasAL, ChroboczekJ (2002) Deubiquitinating Function of Adenovirus Proteinase. Journal of Virology 76: 6323–6331.

68. InnKS, LeeSH, RathbunJY, WongLY, TothZ, et al. (2011) Inhibition of RIG-I mediated signaling by Kaposi's sarcoma-associated herpesvirus-encoded deubiquitinase ORF64. Journal of Virology 85: 10899–10904.

69. RessingME, WiertzEJHJ (2008) Manipulation of the immune response by Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus: Consequences for tumor development. Seminars in Cancer Biology 18: 379–380.

70. MohamedMR, McFaddenG (2009) NF-kB inhibitors: Strategies from poxviruses. Cell Cycle 8: 3125–3132.

71. WuL, FossumE, JooCH, InnKS, ShinYC, et al. (2009) Epstein-Barr Virus LF2: an Antagonist to Type I Interferon. Journal of Virology 83: 1140–1146.

72. HahnAM, HuyeLE, NingS, Webster-CyriaqueJ, PaganoJS (2005) Interferon Regulatory Factor 7 Is Negatively Regulated by the Epstein-Barr Virus Immediate-Early Gene, BZLF-1. Journal of Virology 79: 10040–10052.

73. WangJT, DoongSL, TengSC, LeeCP, TsaiCH, et al. (2009) Epstein-Barr Virus BGLF4 Kinase Suppresses the Interferon Regulatory Factor 3 Signaling Pathway. Journal of Virology 83: 1856–1869.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 2
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#