#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Role for Host Activation-Induced Cytidine Deaminase in Innate Immune Defense against KSHV


Activation-induced cytidine deaminase (AID) is specifically induced in germinal center B cells to carry out somatic hypermutation and class-switch recombination, two processes responsible for antibody diversification. Because of its mutagenic potential, AID expression and activity are tightly regulated to minimize unwanted DNA damage. Surprisingly, AID expression has been observed ectopically during pathogenic infections. However, the function of AID outside of the germinal centers remains largely uncharacterized. In this study, we demonstrate that infection of human primary naïve B cells with Kaposi's sarcoma-associated herpesvirus (KSHV) rapidly induces AID expression in a cell intrinsic manner. We find that infected cells are marked for elimination by Natural Killer cells through upregulation of NKG2D ligands via the DNA damage pathway, a pathway triggered by AID. Moreover, without having a measurable effect on KSHV latency, AID impinges directly on the viral fitness by inhibiting lytic reactivation and reducing infectivity of KSHV virions. Importantly, we uncover two KSHV-encoded microRNAs that directly regulate AID abundance, further reinforcing the role for AID in the antiviral response. Together our findings reveal additional functions for AID in innate immune defense against KSHV with implications for a broader involvement in innate immunity to other pathogens.


Vyšlo v časopise: A Role for Host Activation-Induced Cytidine Deaminase in Innate Immune Defense against KSHV. PLoS Pathog 9(11): e32767. doi:10.1371/journal.ppat.1003748
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003748

Souhrn

Activation-induced cytidine deaminase (AID) is specifically induced in germinal center B cells to carry out somatic hypermutation and class-switch recombination, two processes responsible for antibody diversification. Because of its mutagenic potential, AID expression and activity are tightly regulated to minimize unwanted DNA damage. Surprisingly, AID expression has been observed ectopically during pathogenic infections. However, the function of AID outside of the germinal centers remains largely uncharacterized. In this study, we demonstrate that infection of human primary naïve B cells with Kaposi's sarcoma-associated herpesvirus (KSHV) rapidly induces AID expression in a cell intrinsic manner. We find that infected cells are marked for elimination by Natural Killer cells through upregulation of NKG2D ligands via the DNA damage pathway, a pathway triggered by AID. Moreover, without having a measurable effect on KSHV latency, AID impinges directly on the viral fitness by inhibiting lytic reactivation and reducing infectivity of KSHV virions. Importantly, we uncover two KSHV-encoded microRNAs that directly regulate AID abundance, further reinforcing the role for AID in the antiviral response. Together our findings reveal additional functions for AID in innate immune defense against KSHV with implications for a broader involvement in innate immunity to other pathogens.


Zdroje

1. DavisonAJ (2002) Evolution of the herpesviruses. Vet Microbiol 86: 69–88.

2. ChangY, CesarmanE, PessinMS, LeeF, CulpepperJ, et al. (1994) Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma. Science 266: 1865–1869.

3. SoulierJ, GrolletL, OksenhendlerE, CacoubP, Cazals-HatemD, et al. (1995) Kaposi's sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman's disease. Blood 86: 1276–1280.

4. CesarmanE, ChangY, MoorePS, SaidJW, KnowlesDM (1995) Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas. N Engl J Med 332: 1186–1191.

5. AmbroziakJA, BlackbournDJ, HerndierBG, GlogauRG, GullettJH, et al. (1995) Herpes-like sequences in HIV-infected and uninfected Kaposi's sarcoma patients. Science 268: 582–583.

6. MesriEA, CesarmanE, ArvanitakisL, RafiiS, MooreMA, et al. (1996) Human herpesvirus-8/Kaposi's sarcoma-associated herpesvirus is a new transmissible virus that infects B cells. J Exp Med 183: 2385–2390.

7. DittmerD, LagunoffM, RenneR, StaskusK, HaaseA, et al. (1998) A cluster of latently expressed genes in Kaposi's sarcoma-associated herpesvirus. J Virol 72: 8309–8315.

8. SaridR, FloreO, BohenzkyRA, ChangY, MoorePS (1998) Transcription mapping of the Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) genome in a body cavity-based lymphoma cell line (BC-1). J Virol 72: 1005–1012.

9. SamolsMA, HuJ, SkalskyRL, RenneR (2005) Cloning and identification of a microRNA cluster within the latency-associated region of Kaposi's sarcoma-associated herpesvirus. J Virol 79: 9301–9305.

10. CaiX, LuS, ZhangZ, GonzalezCM, DamaniaB, et al. (2005) Kaposi's sarcoma-associated herpesvirus expresses an array of viral microRNAs in latently infected cells. Proc Natl Acad Sci U S A 102: 5570–5575.

11. GrundhoffA, SullivanCS, GanemD (2006) A combined computational and microarray-based approach identifies novel microRNAs encoded by human gamma-herpesviruses. RNA 12: 733–750.

12. PfefferS, SewerA, Lagos-QuintanaM, SheridanR, SanderC, et al. (2005) Identification of microRNAs of the herpesvirus family. Nat Methods 2: 269–276.

13. SullivanSG, HirschHH, FranceschiS, SteffenI, AmariEB, et al. (2010) Kaposi sarcoma herpes virus antibody response and viremia following highly active antiretroviral therapy in the Swiss HIV Cohort study. AIDS 24: 2245–2252.

14. GaoSJ, KingsleyL, LiM, ZhengW, ParraviciniC, et al. (1996) KSHV antibodies among Americans, Italians and Ugandans with and without Kaposi's sarcoma. Nat Med 2: 925–928.

15. DialynaIA, GrahamD, RezaeeR, BlueCE, StavrianeasNG, et al. (2004) Anti-HHV-8/KSHV antibodies in infected individuals inhibit infection in vitro. AIDS 18: 1263–1270.

16. KimballLE, CasperC, KoelleDM, MorrowR, CoreyL, et al. (2004) Reduced levels of neutralizing antibodies to Kaposi sarcoma-associated herpesvirus in persons with a history of Kaposi sarcoma. J Infect Dis 189: 2016–2022.

17. LehrnbecherTL, FosterCB, ZhuS, VenzonD, SteinbergSM, et al. (2000) Variant genotypes of FcgammaRIIIA influence the development of Kaposi's sarcoma in HIV-infected men. Blood 95: 2386–2390.

18. MuramatsuM, KinoshitaK, FagarasanS, YamadaS, ShinkaiY, et al. (2000) Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell 102: 553–563.

19. WablM, BurrowsPD, von GabainA, SteinbergC (1985) Hypermutation at the immunoglobulin heavy chain locus in a pre-B-cell line. Proc Natl Acad Sci U S A 82: 479–482.

20. de YebenesVG, RamiroAR (2006) Activation-induced deaminase: light and dark sides. Trends Mol Med 12: 432–439.

21. RamiroAR, JankovicM, EisenreichT, DifilippantonioS, Chen-KiangS, et al. (2004) AID is required for c-myc/IgH chromosome translocations in vivo. Cell 118: 431–438.

22. ShenHM, PetersA, BaronB, ZhuX, StorbU (1998) Mutation of BCL-6 gene in normal B cells by the process of somatic hypermutation of Ig genes. Science 280: 1750–1752.

23. PasqualucciL, MigliazzaA, FracchiollaN, WilliamC, NeriA, et al. (1998) BCL-6 mutations in normal germinal center B cells: evidence of somatic hypermutation acting outside Ig loci. Proc Natl Acad Sci U S A 95: 11816–11821.

24. ConticelloSG, ThomasCJ, Petersen-MahrtSK, NeubergerMS (2005) Evolution of the AID/APOBEC family of polynucleotide (deoxy)cytidine deaminases. Mol Biol Evol 22: 367–377.

25. ChiuYL, GreeneWC (2008) The APOBEC3 cytidine deaminases: an innate defensive network opposing exogenous retroviruses and endogenous retroelements. Annu Rev Immunol 26: 317–353.

26. GourziP, LeonovaT, PapavasiliouFN (2006) A role for activation-induced cytidine deaminase in the host response against a transforming retrovirus. Immunity 24: 779–786.

27. EpeldeguiM, ThapaDR, De la CruzJ, KitchenS, ZackJA, et al. (2010) CD40 ligand (CD154) incorporated into HIV virions induces activation-induced cytidine deaminase (AID) expression in human B lymphocytes. PLoS One 5: e11448.

28. IshikawaC, NakachiS, SenbaM, SugaiM, MoriN (2011) Activation of AID by human T-cell leukemia virus Tax oncoprotein and the possible role of its constitutive expression in ATL genesis. Carcinogenesis 32: 110–119.

29. MachidaK, ChengKT, SungVM, ShimodairaS, LindsayKL, et al. (2004) Hepatitis C virus induces a mutator phenotype: enhanced mutations of immunoglobulin and protooncogenes. Proc Natl Acad Sci U S A 101: 4262–4267.

30. MatsumotoY, MarusawaH, KinoshitaK, EndoY, KouT, et al. (2007) Helicobacter pylori infection triggers aberrant expression of activation-induced cytidine deaminase in gastric epithelium. Nat Med 13: 470–476.

31. MyoungJ, GanemD (2011) Infection of lymphoblastoid cell lines by Kaposi's sarcoma-associated herpesvirus: critical role of cell-associated virus. J Virol 85: 9767–9777.

32. MyoungJ, GanemD (2011) Infection of primary human tonsillar lymphoid cells by KSHV reveals frequent but abortive infection of T cells. Virology 413: 1–11.

33. TranTH, NakataM, SuzukiK, BegumNA, ShinkuraR, et al. (2010) B cell-specific and stimulation-responsive enhancers derepress Aicda by overcoming the effects of silencers. Nat Immunol 11: 148–154.

34. DedeogluF, HorwitzB, ChaudhuriJ, AltFW, GehaRS (2004) Induction of activation-induced cytidine deaminase gene expression by IL-4 and CD40 ligation is dependent on STAT6 and NFkappaB. Int Immunol 16: 395–404.

35. ZhouC, SaxonA, ZhangK (2003) Human activation-induced cytidine deaminase is induced by IL-4 and negatively regulated by CD45: implication of CD45 as a Janus kinase phosphatase in antibody diversification. J Immunol 170: 1887–1893.

36. MattaH, ChaudharyPM (2004) Activation of alternative NF-kappa B pathway by human herpes virus 8-encoded Fas-associated death domain-like IL-1 beta-converting enzyme inhibitory protein (vFLIP). Proc Natl Acad Sci U S A 101: 9399–9404.

37. RauletDH (2003) Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 3: 781–790.

38. GasserS, OrsulicS, BrownEJ, RauletDH (2005) The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436: 1186–1190.

39. NormanJM, MashibaM, McNamaraLA, Onafuwa-NugaA, Chiari-FortE, et al. (2011) The antiviral factor APOBEC3G enhances the recognition of HIV-infected primary T cells by natural killer cells. Nat Immunol 12: 975–983.

40. Stern-GinossarN, MandelboimO (2009) An integrated view of the regulation of NKG2D ligands. Immunology 128: 1–6.

41. JostS, AltfeldM (2013) Control of human viral infections by natural killer cells. Annu Rev Immunol 31: 163–194.

42. EspinozaJL, TakamiA, YoshiokaK, NakataK, SatoT, et al. (2012) Human microRNA-1245 down-regulates the NKG2D receptor in natural killer cells and impairs NKG2D-mediated functions. Haematologica 97: 1295–1303.

43. GiurisatoE, CellaM, TakaiT, KurosakiT, FengY, et al. (2007) Phosphatidylinositol 3-kinase activation is required to form the NKG2D immunological synapse. Mol Cell Biol 27: 8583–8599.

44. EpeldeguiM, HungYP, McQuayA, AmbinderRF, Martinez-MazaO (2007) Infection of human B cells with Epstein-Barr virus results in the expression of somatic hypermutation-inducing molecules and in the accrual of oncogene mutations. Mol Immunol 44: 934–942.

45. MillerG, HestonL, GroganE, GradovilleL, RigsbyM, et al. (1997) Selective switch between latency and lytic replication of Kaposi's sarcoma herpesvirus and Epstein-Barr virus in dually infected body cavity lymphoma cells. J Virol 71: 314–324.

46. VermaSC, BajajBG, CaiQ, SiH, SeelhammerT, et al. (2006) Latency-associated nuclear antigen of Kaposi's sarcoma-associated herpesvirus recruits uracil DNA glycosylase 2 at the terminal repeats and is important for latent persistence of the virus. J Virol 80: 11178–11190.

47. KavliB, SundheimO, AkbariM, OtterleiM, NilsenH, et al. (2002) hUNG2 is the major repair enzyme for removal of uracil from U:A matches, U:G mismatches, and U in single-stranded DNA, with hSMUG1 as a broad specificity backup. J Biol Chem 277: 39926–39936.

48. NilsenH, RosewellI, RobinsP, SkjelbredCF, AndersenS, et al. (2000) Uracil-DNA glycosylase (UNG)-deficient mice reveal a primary role of the enzyme during DNA replication. Mol Cell 5: 1059–1065.

49. CoscoyL (2007) Immune evasion by Kaposi's sarcoma-associated herpesvirus. Nat Rev Immunol 7: 391–401.

50. DorsettY, McBrideKM, JankovicM, GazumyanA, ThaiTH, et al. (2008) MicroRNA-155 suppresses activation-induced cytidine deaminase-mediated Myc-Igh translocation. Immunity 28: 630–638.

51. TengG, HakimpourP, LandgrafP, RiceA, TuschlT, et al. (2008) MicroRNA-155 is a negative regulator of activation-induced cytidine deaminase. Immunity 28: 621–629.

52. de YebenesVG, BelverL, PisanoDG, GonzalezS, VillasanteA, et al. (2008) miR-181b negatively regulates activation-induced cytidine deaminase in B cells. J Exp Med 205: 2199–2206.

53. BorchertGM, HoltonNW, LarsonED (2011) Repression of human activation induced cytidine deaminase by miR-93 and miR-155. BMC Cancer 11: 347.

54. YinQ, McBrideJ, FewellC, LaceyM, WangX, et al. (2008) MicroRNA-155 is an Epstein-Barr virus-induced gene that modulates Epstein-Barr virus-regulated gene expression pathways. J Virol 82: 5295–5306.

55. LinnstaedtSD, GottweinE, SkalskyRL, LuftigMA, CullenBR (2010) Virally induced cellular microRNA miR-155 plays a key role in B-cell immortalization by Epstein-Barr virus. J Virol 84: 11670–11678.

56. RahadianiN, TakakuwaT, TresnasariK, MoriiE, AozasaK (2008) Latent membrane protein-1 of Epstein-Barr virus induces the expression of B-cell integration cluster, a precursor form of microRNA-155, in B lymphoma cell lines. Biochem Biophys Res Commun 377: 579–583.

57. GottweinE, MukherjeeN, SachseC, FrenzelC, MajorosWH, et al. (2007) A viral microRNA functions as an orthologue of cellular miR-155. Nature 450: 1096–1099.

58. SkalskyRL, SamolsMA, PlaisanceKB, BossIW, RivaA, et al. (2007) Kaposi's sarcoma-associated herpesvirus encodes an ortholog of miR-155. J Virol 81: 12836–12845.

59. BartelDP (2009) MicroRNAs: target recognition and regulatory functions. Cell 136: 215–233.

60. GottweinE, CorcoranDL, MukherjeeN, SkalskyRL, HafnerM, et al. (2011) Viral microRNA targetome of KSHV-infected primary effusion lymphoma cell lines. Cell Host Microbe 10: 515–526.

61. LeiX, BaiZ, YeF, XieJ, KimCG, et al. (2010) Regulation of NF-kappaB inhibitor IkappaBalpha and viral replication by a KSHV microRNA. Nat Cell Biol 12: 193–199.

62. ZhouFC, ZhangYJ, DengJH, WangXP, PanHY, et al. (2002) Efficient infection by a recombinant Kaposi's sarcoma-associated herpesvirus cloned in a bacterial artificial chromosome: application for genetic analysis. J Virol 76: 6185–6196.

63. EndoY, MarusawaH, KinoshitaK, MorisawaT, SakuraiT, et al. (2007) Expression of activation-induced cytidine deaminase in human hepatocytes via NF-kappaB signaling. Oncogene 26: 5587–5595.

64. SirianniMC, VincenziL, TopinoS, GiovannettiA, MazzettaF, et al. (2002) NK cell activity controls human herpesvirus 8 latent infection and is restored upon highly active antiretroviral therapy in AIDS patients with regressing Kaposi's sarcoma. Eur J Immunol 32: 2711–2720.

65. MatthewsNC, GoodierMR, RobeyRC, BowerM, GotchFM (2011) Killing of Kaposi's sarcoma-associated herpesvirus-infected fibroblasts during latent infection by activated natural killer cells. Eur J Immunol 41: 1958–1968.

66. MadridAS, GanemD (2012) Kaposi's sarcoma-associated herpesvirus ORF54/dUTPase downregulates a ligand for the NK activating receptor NKp44. J Virol 86: 8693–8704.

67. ThomasM, BonameJM, FieldS, NejentsevS, SalioM, et al. (2008) Down-regulation of NKG2D and NKp80 ligands by Kaposi's sarcoma-associated herpesvirus K5 protects against NK cell cytotoxicity. Proc Natl Acad Sci U S A 105: 1656–1661.

68. NachmaniD, Stern-GinossarN, SaridR, MandelboimO (2009) Diverse herpesvirus microRNAs target the stress-induced immune ligand MICB to escape recognition by natural killer cells. Cell Host Microbe 5: 376–385.

69. WuX, GeraldesP, PlattJL, CascalhoM (2005) The double-edged sword of activation-induced cytidine deaminase. J Immunol 174: 934–941.

70. LiangG, KitamuraK, WangZ, LiuG, ChowdhuryS, et al. (2013) RNA editing of hepatitis B virus transcripts by activation-induced cytidine deaminase. Proc Natl Acad Sci U S A 110: 2246–2251.

71. AriasC, WalshD, HarbellJ, WilsonAC, MohrI (2009) Activation of host translational control pathways by a viral developmental switch. PLoS Pathog 5: e1000334.

72. Lukac DM, Yuan Y (2007) Reactivation and lytic replication of KSHV. In: Arvin A, Campadelli-Fiume G, Mocarski E, Moore PS, Roizman B et al.., editors. Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis. Cambridge.

73. BransteitterR, PhamP, ScharffMD, GoodmanMF (2003) Activation-induced cytidine deaminase deaminates deoxycytidine on single-stranded DNA but requires the action of RNase. Proc Natl Acad Sci U S A 100: 4102–4107.

74. WilsonSJ, TsaoEH, WebbBL, YeH, Dalton-GriffinL, et al. (2007) X box binding protein XBP-1s transactivates the Kaposi's sarcoma-associated herpesvirus (KSHV) ORF50 promoter, linking plasma cell differentiation to KSHV reactivation from latency. J Virol 81: 13578–13586.

75. SiegelAM, RangaswamyUS, NapierRJ, SpeckSH (2010) Blimp-1-dependent plasma cell differentiation is required for efficient maintenance of murine gammaherpesvirus latency and antiviral antibody responses. J Virol 84: 674–685.

76. TobollikS, MeyerL, BuettnerM, KlemmerS, KempkesB, et al. (2006) Epstein-Barr virus nuclear antigen 2 inhibits AID expression during EBV-driven B-cell growth. Blood 108: 3859–3864.

77. SernandezIV, de YebenesVG, DorsettY, RamiroAR (2008) Haploinsufficiency of activation-induced deaminase for antibody diversification and chromosome translocations both in vitro and in vivo. PLoS One 3: e3927.

78. ZhangW, BardwellPD, WooCJ, PoltoratskyV, ScharffMD, et al. (2001) Clonal instability of V region hypermutation in the Ramos Burkitt's lymphoma cell line. Int Immunol 13: 1175–1184.

79. LuCC, LiZ, ChuCY, FengJ, FengJ, et al. (2010) MicroRNAs encoded by Kaposi's sarcoma-associated herpesvirus regulate viral life cycle. EMBO Rep 11: 784–790.

80. LuF, StedmanW, YousefM, RenneR, LiebermanPM (2010) Epigenetic regulation of Kaposi's sarcoma-associated herpesvirus latency by virus-encoded microRNAs that target Rta and the cellular Rbl2-DNMT pathway. J Virol 84: 2697–2706.

81. BellareP, GanemD (2009) Regulation of KSHV lytic switch protein expression by a virus-encoded microRNA: an evolutionary adaptation that fine-tunes lytic reactivation. Cell Host Microbe 6: 570–575.

82. LinX, LiangD, HeZ, DengQ, RobertsonES, et al. (2011) miR-K12-7-5p encoded by Kaposi's sarcoma-associated herpesvirus stabilizes the latent state by targeting viral ORF50/RTA. PLoS One 6: e16224.

83. ZiegelbauerJM, SullivanCS, GanemD (2009) Tandem array-based expression screens identify host mRNA targets of virus-encoded microRNAs. Nat Genet 41: 130–134.

84. OkazakiIM, KotaniA, HonjoT (2007) Role of AID in tumorigenesis. Adv Immunol 94: 245–273.

85. SilvaJM, LiMZ, ChangK, GeW, GoldingMC, et al. (2005) Second-generation shRNA libraries covering the mouse and human genomes. Nat Genet 37: 1281–1288.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 11
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#