#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Uracil DNA Glycosylase Counteracts APOBEC3G-Induced Hypermutation of Hepatitis B Viral Genomes: Excision Repair of Covalently Closed Circular DNA


The covalently closed circular DNA (cccDNA) of the hepatitis B virus (HBV) plays an essential role in chronic hepatitis. The cellular repair system is proposed to convert cytoplasmic nucleocapsid (NC) DNA (partially double-stranded DNA) into cccDNA in the nucleus. Recently, antiviral cytidine deaminases, AID/APOBEC proteins, were shown to generate uracil residues in the NC-DNA through deamination, resulting in cytidine-to-uracil (C-to-U) hypermutation of the viral genome. We investigated whether uracil residues in hepadnavirus DNA were excised by uracil-DNA glycosylase (UNG), a host factor for base excision repair (BER). When UNG activity was inhibited by the expression of the UNG inhibitory protein (UGI), hypermutation of NC-DNA induced by either APOBEC3G or interferon treatment was enhanced in a human hepatocyte cell line. To assess the effect of UNG on the cccDNA viral intermediate, we used the duck HBV (DHBV) replication model. Sequence analyses of DHBV DNAs showed that cccDNA accumulated G-to-A or C-to-T mutations in APOBEC3G-expressing cells, and this was extensively enhanced by UNG inhibition. The cccDNA hypermutation generated many premature stop codons in the P gene. UNG inhibition also enhanced the APOBEC3G-mediated suppression of viral replication, including reduction of NC-DNA, pre-C mRNA, and secreted viral particle-associated DNA in prolonged culture. Enhancement of APOBEC3G-mediated suppression by UNG inhibition was not observed when the catalytic site of APOBEC3G was mutated. Transfection experiments of recloned cccDNAs revealed that the combination of UNG inhibition and APOBEC3G expression reduced the replication ability of cccDNA. Taken together, these data indicate that UNG excises uracil residues from the viral genome during or after cccDNA formation in the nucleus and imply that BER pathway activities decrease the antiviral effect of APOBEC3-mediated hypermutation.


Vyšlo v časopise: Uracil DNA Glycosylase Counteracts APOBEC3G-Induced Hypermutation of Hepatitis B Viral Genomes: Excision Repair of Covalently Closed Circular DNA. PLoS Pathog 9(5): e32767. doi:10.1371/journal.ppat.1003361
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003361

Souhrn

The covalently closed circular DNA (cccDNA) of the hepatitis B virus (HBV) plays an essential role in chronic hepatitis. The cellular repair system is proposed to convert cytoplasmic nucleocapsid (NC) DNA (partially double-stranded DNA) into cccDNA in the nucleus. Recently, antiviral cytidine deaminases, AID/APOBEC proteins, were shown to generate uracil residues in the NC-DNA through deamination, resulting in cytidine-to-uracil (C-to-U) hypermutation of the viral genome. We investigated whether uracil residues in hepadnavirus DNA were excised by uracil-DNA glycosylase (UNG), a host factor for base excision repair (BER). When UNG activity was inhibited by the expression of the UNG inhibitory protein (UGI), hypermutation of NC-DNA induced by either APOBEC3G or interferon treatment was enhanced in a human hepatocyte cell line. To assess the effect of UNG on the cccDNA viral intermediate, we used the duck HBV (DHBV) replication model. Sequence analyses of DHBV DNAs showed that cccDNA accumulated G-to-A or C-to-T mutations in APOBEC3G-expressing cells, and this was extensively enhanced by UNG inhibition. The cccDNA hypermutation generated many premature stop codons in the P gene. UNG inhibition also enhanced the APOBEC3G-mediated suppression of viral replication, including reduction of NC-DNA, pre-C mRNA, and secreted viral particle-associated DNA in prolonged culture. Enhancement of APOBEC3G-mediated suppression by UNG inhibition was not observed when the catalytic site of APOBEC3G was mutated. Transfection experiments of recloned cccDNAs revealed that the combination of UNG inhibition and APOBEC3G expression reduced the replication ability of cccDNA. Taken together, these data indicate that UNG excises uracil residues from the viral genome during or after cccDNA formation in the nucleus and imply that BER pathway activities decrease the antiviral effect of APOBEC3-mediated hypermutation.


Zdroje

1. KoikeK, TsutsumiT, FujieH, ShintaniY, KyojiM (2002) Molecular mechanism of viral hepatocarcinogenesis. Oncology 62 Suppl 1: 29–37.

2. TanA, YehSH, LiuCJ, CheungC, ChenPJ (2008) Viral hepatocarcinogenesis: from infection to cancer. Liver Int 28: 175–188.

3. BeckJ, NassalM (2007) Hepatitis B virus replication. World J Gastroenterol 13: 48–64.

4. NguyenDH, LudgateL, HuJ (2008) Hepatitis B virus-cell interactions and pathogenesis. J Cell Physiol 216: 289–294.

5. NassalM (2008) Hepatitis B viruses: reverse transcription a different way. Virus Res 134: 235–249.

6. GhanyM, LiangTJ (2007) Drug targets and molecular mechanisms of drug resistance in chronic hepatitis B. Gastroenterology 132: 1574–1585.

7. LevreroM, PollicinoT, PetersenJ, BelloniL, RaimondoG, et al. (2009) Control of cccDNA function in hepatitis B virus infection. J Hepatol 51: 581–592.

8. SchultzU, GrgacicE, NassalM (2004) Duck hepatitis B virus: an invaluable model system for HBV infection. Adv Virus Res 63: 1–70.

9. FunkA, MhamdiM, WillH, SirmaH (2007) Avian hepatitis B viruses: molecular and cellular biology, phylogenesis, and host tropism. World J Gastroenterol 13: 91–103.

10. KockJ, RoslerC, ZhangJJ, BlumHE, NassalM, et al. (2010) Generation of covalently closed circular DNA of hepatitis B viruses via intracellular recycling is regulated in a virus specific manner. PLoS Pathog 6: e1001082.

11. 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.

12. MuramatsuM, NagaokaH, ShinkuraR, BegumNA, HonjoT (2007) Discovery of activation-induced cytidine deaminase, the engraver of antibody memory. Adv Immunol 94: 1–36.

13. HarrisRS, LiddamentMT (2004) Retroviral restriction by APOBEC proteins. Nat Rev Immunol 4: 868–877.

14. Goila-GaurR, StrebelK (2008) HIV-1 Vif, APOBEC, and intrinsic immunity. Retrovirology 5: 51.

15. MalimMH (2009) APOBEC proteins and intrinsic resistance to HIV-1 infection. Philos Trans R Soc Lond B Biol Sci 364: 675–687.

16. TurelliP, MangeatB, JostS, VianinS, TronoD (2004) Inhibition of hepatitis B virus replication by APOBEC3G. Science 303: 1829.

17. RoslerC, KockJ, KannM, MalimMH, BlumHE, et al. (2005) APOBEC-mediated interference with hepadnavirus production. Hepatology 42: 301–309.

18. BonvinM, AchermannF, GreeveI, StrokaD, KeoghA, et al. (2006) Interferon-inducible expression of APOBEC3 editing enzymes in human hepatocytes and inhibition of hepatitis B virus replication. Hepatology 43: 1364–1374.

19. NoguchiC, HiragaN, MoriN, TsugeM, ImamuraM, et al. (2007) Dual effect of APOBEC3G on Hepatitis B virus. J Gen Virol 88: 432–440.

20. JostS, TurelliP, MangeatB, ProtzerU, TronoD (2007) Induction of antiviral cytidine deaminases does not explain the inhibition of hepatitis B virus replication by interferons. J Virol 81: 10588–10596.

21. NguyenDH, GummuluruS, HuJ (2007) Deamination-independent inhibition of hepatitis B virus reverse transcription by APOBEC3G. J Virol 81: 4465–4472.

22. NguyenDH, HuJ (2008) Reverse transcriptase- and RNA packaging signal-dependent incorporation of APOBEC3G into hepatitis B virus nucleocapsids. J Virol 82: 6852–6861.

23. KockJ, BlumHE (2008) Hypermutation of hepatitis B virus genomes by APOBEC3G, APOBEC3C and APOBEC3H. J Gen Virol 89: 1184–1191.

24. VartanianJP, HenryM, MarchioA, SuspeneR, AynaudMM, et al. (2010) Massive APOBEC3 Editing of Hepatitis B Viral DNA in Cirrhosis. PLoS Pathog 6: e1000928.

25. HarrisRS, SheehyAM, CraigHM, MalimMH, NeubergerMS (2003) DNA deamination: not just a trigger for antibody diversification but also a mechanism for defense against retroviruses. Nat Immunol 4: 641–643.

26. MangeatB, TurelliP, CaronG, FriedliM, PerrinL, et al. (2003) Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts. Nature 424: 99–103.

27. RoslerC, KockJ, MalimMH, BlumHE, von WeizsackerF (2004) Comment on “Inhibition of hepatitis B virus replication by APOBEC3G”. Science 305: 1403; author reply 1403.

28. SuspeneR, GuetardD, HenryM, SommerP, Wain-HobsonS, et al. (2005) Extensive editing of both hepatitis B virus DNA strands by APOBEC3 cytidine deaminases in vitro and in vivo. Proc Natl Acad Sci U S A 102: 8321–8326.

29. SousaMM, KrokanHE, SlupphaugG (2007) DNA-uracil and human pathology. Mol Aspects Med 28: 276–306.

30. Di NoiaJM, NeubergerMS (2007) Molecular mechanisms of antibody somatic hypermutation. Annu Rev Biochem 76: 1–22.

31. ChenR, Le RouzicE, KearneyJA, ManskyLM, BenichouS (2004) Vpr-mediated incorporation of UNG2 into HIV-1 particles is required to modulate the virus mutation rate and for replication in macrophages. J Biol Chem 279: 28419–28425.

32. ManskyLM, PreveralS, SeligL, BenarousR, BenichouS (2000) The interaction of vpr with uracil DNA glycosylase modulates the human immunodeficiency virus type 1 In vivo mutation rate. J Virol 74: 7039–7047.

33. YangB, ChenK, ZhangC, HuangS, ZhangH (2007) Virion-associated uracil DNA glycosylase-2 and apurinic/apyrimidinic endonuclease are involved in the degradation of APOBEC3G-edited nascent HIV-1 DNA. J Biol Chem 282: 11667–11675.

34. KaiserSM, EmermanM (2006) Uracil DNA glycosylase is dispensable for human immunodeficiency virus type 1 replication and does not contribute to the antiviral effects of the cytidine deaminase Apobec3G. J Virol 80: 875–882.

35. MbisaJL, BarrR, ThomasJA, VandegraaffN, DorweilerIJ, et al. (2007) Human immunodeficiency virus type 1 cDNAs produced in the presence of APOBEC3G exhibit defects in plus-strand DNA transfer and integration. J Virol 81: 7099–7110.

36. LangloisMA, NeubergerMS (2008) Human APOBEC3G can restrict retroviral infection in avian cells and acts independently of both UNG and SMUG1. J Virol 82: 4660–4664.

37. SandersonRJ, MosbaughDW (1996) Identification of specific carboxyl groups on uracil-DNA glycosylase inhibitor protein that are required for activity. J Biol Chem 271: 29170–29181.

38. DoiT, KinoshitaK, IkegawaM, MuramatsuM, HonjoT (2003) De novo protein synthesis is required for the activation-induced cytidine deaminase function in class-switch recombination. Proc Natl Acad Sci U S A 100: 2634–2638.

39. BegumNA, KinoshitaK, KakazuN, MuramatsuM, NagaokaH, et al. (2004) Uracil DNA glycosylase activity is dispensable for immunoglobulin class switch. Science 305: 1160–1163.

40. BrussV, GanemD (1991) The role of envelope proteins in hepatitis B virus assembly. Proc Natl Acad Sci U S A 88: 1059–1063.

41. SuzukiT, TakeharaT, OhkawaK, IshidaH, JinushiM, et al. (2003) Intravenous injection of naked plasmid DNA encoding hepatitis B virus (HBV) produces HBV and induces humoral immune response in mice. Biochem Biophys Res Commun 300: 784–788.

42. HenryM, GuetardD, SuspeneR, RusniokC, Wain-HobsonS, et al. (2009) Genetic editing of HBV DNA by monodomain human APOBEC3 cytidine deaminases and the recombinant nature of APOBEC3G. PLoS One 4: e4277.

43. GonzalezMC, SuspeneR, HenryM, GuetardD, Wain-HobsonS, et al. (2009) Human APOBEC1 cytidine deaminase edits HBV DNA. Retrovirology 6: 96.

44. KimHY, ParkGS, KimEG, KangSH, ShinHJ, et al. (2004) Oligomer synthesis by priming deficient polymerase in hepatitis B virus core particle. Virology 322: 22–30.

45. OropezaCE, LiL, McLachlanA (2008) Differential inhibition of nuclear hormone receptor-dependent hepatitis B virus replication by the small heterodimer partner. J Virol 82: 3814–3821.

46. ProtoS, TaylorJA, ChokshiS, NavaratnamN, NaoumovNV (2008) APOBEC and iNOS are not the main intracellular effectors of IFN-gamma-mediated inactivation of Hepatitis B virus replication. Antiviral Res 78: 260–267.

47. KomoharaY, YanoH, ShichijoS, ShimotohnoK, ItohK, et al. (2006) High expression of APOBEC3G in patients infected with hepatitis C virus. J Mol Histol 37: 327–332.

48. YuM, SummersJ (1994) Multiple functions of capsid protein phosphorylation in duck hepatitis B virus replication. J Virol 68: 4341–4348.

49. SummersJ, SmithPM, HuangMJ, YuMS (1991) Morphogenetic and regulatory effects of mutations in the envelope proteins of an avian hepadnavirus. J Virol 65: 1310–1317.

50. Di NoiaJM, RadaC, NeubergerMS (2006) SMUG1 is able to excise uracil from immunoglobulin genes: insight into mutation versus repair. EMBO J 25: 585–595.

51. Di NoiaJ, NeubergerMS (2002) Altering the pathway of immunoglobulin hypermutation by inhibiting uracil-DNA glycosylase. Nature 419: 43–48.

52. Le MireMF, MillerDS, FosterWK, BurrellCJ, JilbertAR (2005) Covalently closed circular DNA is the predominant form of duck hepatitis B virus DNA that persists following transient infection. J Virol 79: 12242–12252.

53. UmedaM, MarusawaH, SenoH, KatsuradaA, NabeshimaM, et al. (2005) Hepatitis B virus infection in lymphatic tissues in inactive hepatitis B carriers. J Hepatol 42: 806–812.

54. SmithHC, BennettRP, KizilyerA, McDougallWM, ProhaskaKM (2011) Functions and regulation of the APOBEC family of proteins. Semin Cell Dev Biol 23: 258–68.

55. StengleinMD, MatsuoH, HarrisRS (2008) Two regions within the amino-terminal half of APOBEC3G cooperate to determine cytoplasmic localization. J Virol 82: 9591–9599.

56. ConticelloSG (2008) The AID/APOBEC family of nucleic acid mutators. Genome Biol 9: 229.

57. MargeridonS, Carrouee-DurantelS, CheminI, BarraudL, ZoulimF, et al. (2008) Rolling circle amplification, a powerful tool for genetic and functional studies of complete hepatitis B virus genomes from low-level infections and for directly probing covalently closed circular DNA. Antimicrob Agents Chemother 52: 3068–3073.

58. RectorA, TachezyR, Van RanstM (2004) A sequence-independent strategy for detection and cloning of circular DNA virus genomes by using multiply primed rolling-circle amplification. J Virol 78: 4993–4998.

59. ChenR, WangH, ManskyLM (2002) Roles of uracil-DNA glycosylase and dUTPase in virus replication. J Gen Virol 83: 2339–2345.

60. StengleinMD, BurnsMB, LiM, LengyelJ, HarrisRS (2010) APOBEC3 proteins mediate the clearance of foreign DNA from human cells. Nat Struct Mol Biol 17: 222–229.

61. 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.

62. RenardM, HenryM, GuetardD, VartanianJP, Wain-HobsonS (2010) APOBEC1 and APOBEC3 cytidine deaminases as restriction factors for hepadnaviral genomes in non-humans in vivo. J Mol Biol 400: 323–334.

63. RogozinIB, BasuMK, JordanIK, PavlovYI, KooninEV (2005) APOBEC4, a new member of the AID/APOBEC family of polynucleotide (deoxy)cytidine deaminases predicted by computational analysis. Cell Cycle 4: 1281–1285.

64. 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.

65. MulderLC, HarariA, SimonV (2008) Cytidine deamination induced HIV-1 drug resistance. Proc Natl Acad Sci U S A 105: 5501–5506.

66. SadlerHA, StengleinMD, HarrisRS, ManskyLM (2010) APOBEC3G contributes to HIV-1 variation through sublethal mutagenesis. J Virol 84: 7396–7404.

67. FagarasanS, KinoshitaK, MuramatsuM, IkutaK, HonjoT (2001) In situ class switching and differentiation to IgA-producing cells in the gut lamina propria. Nature 413: 639–643.

68. GuntherS, LiBC, MiskaS, KrugerDH, MeiselH, et al. (1995) A novel method for efficient amplification of whole hepatitis B virus genomes permits rapid functional analysis and reveals deletion mutants in immunosuppressed patients. J Virol 69: 5437–5444.

69. HirtB (1967) Selective extraction of polyoma DNA from infected mouse cell cultures. J Mol Biol 26: 365–369.

70. HuovinenT, BrockmannEC, AkterS, Perez-GamarraS, Yla-PeltoJ, et al. (2012) Primer extension mutagenesis powered by selective rolling circle amplification. PLoS One 7: e31817.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 5
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#