#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Cytoplasmic Viral RNA-Dependent RNA Polymerase Disrupts the Intracellular Splicing Machinery by Entering the Nucleus and Interfering with Prp8


RNA-dependent RNA polymerase (RdRp) is an enzyme that catalyzes the replication from an RNA template and is encoded in the genomes of all RNA viruses. RNA viruses in general replicate in cytoplasm and interfere host cellular gene expression by utilizing proteolytic destruction of cellular targets as the primary mechanism. However, several cytoplasmic RNA viral proteins have been found in the nucleus. What do they do in the nucleus? This study utilized picornaviral polymerase to probe the function of RdRp in the nucleus. Our findings reveal a novel mechanism of viruses attacking hosts whereby picornaviral 3D polymerase (3Dpol) enters the nucleus and targets the central pre-mRNA processing factor 8 (Prp8) to block pre-mRNA splicing and mRNA synthesis. The 3Dpol inhibits the second catalytic step of the splicing process, resulting in the accumulation of the lariat-form and the reduction of the mRNA. These results provide new insights into the strategy of a cytoplasmic RNA virus attacking host cell, that differs from viral shutting off cellular transcription and translation which contributes to the viral pathogenesis. To our knowledge, this study shows for the first time that a cytoplasmic RNA virus uses its polymerase to alter cellular gene expression by hijacking the splicing machinery.


Vyšlo v časopise: Cytoplasmic Viral RNA-Dependent RNA Polymerase Disrupts the Intracellular Splicing Machinery by Entering the Nucleus and Interfering with Prp8. PLoS Pathog 10(6): e32767. doi:10.1371/journal.ppat.1004199
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004199

Souhrn

RNA-dependent RNA polymerase (RdRp) is an enzyme that catalyzes the replication from an RNA template and is encoded in the genomes of all RNA viruses. RNA viruses in general replicate in cytoplasm and interfere host cellular gene expression by utilizing proteolytic destruction of cellular targets as the primary mechanism. However, several cytoplasmic RNA viral proteins have been found in the nucleus. What do they do in the nucleus? This study utilized picornaviral polymerase to probe the function of RdRp in the nucleus. Our findings reveal a novel mechanism of viruses attacking hosts whereby picornaviral 3D polymerase (3Dpol) enters the nucleus and targets the central pre-mRNA processing factor 8 (Prp8) to block pre-mRNA splicing and mRNA synthesis. The 3Dpol inhibits the second catalytic step of the splicing process, resulting in the accumulation of the lariat-form and the reduction of the mRNA. These results provide new insights into the strategy of a cytoplasmic RNA virus attacking host cell, that differs from viral shutting off cellular transcription and translation which contributes to the viral pathogenesis. To our knowledge, this study shows for the first time that a cytoplasmic RNA virus uses its polymerase to alter cellular gene expression by hijacking the splicing machinery.


Zdroje

1. BedardKM, SemlerBL (2004) Regulation of picornavirus gene expression. Microbes and infection/Institut Pasteur 6: 702–713.

2. WeidmanMK, SharmaR, RaychaudhuriS, KunduP, TsaiW, et al. (2003) The interaction of cytoplasmic RNA viruses with the nucleus. Virus research 95: 75–85.

3. WengKF, LiML, HungCT, ShihSR (2009) Enterovirus 71 3C protease cleaves a novel target CstF-64 and inhibits cellular polyadenylation. PLoS pathogens 5: e1000593.

4. ChangLY, HuangLM, GauSS, WuYY, HsiaSH, et al. (2007) Neurodevelopment and cognition in children after enterovirus 71 infection. The New England journal of medicine 356: 1226–1234.

5. WhittonJL, CornellCT, FeuerR (2005) Host and virus determinants of picornavirus pathogenesis and tropism. Nature reviews Microbiology 3: 765–776.

6. LinKH, HwangKP, KeGM, WangCF, KeLY, et al. (2006) Evolution of EV71 genogroup in Taiwan from 1998 to 2005: an emerging of subgenogroup C4 of EV71. Journal of medical virology 78: 254–262.

7. DingNZ, WangXM, SunSW, SongQ, LiSN, et al. (2009) Appearance of mosaic enterovirus 71 in the 2008 outbreak of China. Virus research 145: 157–161.

8. SeiffA (2012) Cambodia unravels cause of mystery illness. Lancet 380: 206.

9. MarcotteLL, WassAB, GoharaDW, PathakHB, ArnoldJJ, et al. (2007) Crystal structure of poliovirus 3CD protein: virally encoded protease and precursor to the RNA-dependent RNA polymerase. Journal of virology 81: 3583–3596.

10. ThompsonAA, PeersenOB (2004) Structural basis for proteolysis-dependent activation of the poliovirus RNA-dependent RNA polymerase. The EMBO journal 23: 3462–3471.

11. PaulAV, van BoomJH, FilippovD, WimmerE (1998) Protein-primed RNA synthesis by purified poliovirus RNA polymerase. Nature 393: 280–284.

12. PaulAV, PetersJ, MugaveroJ, YinJ, van BoomJH, et al. (2003) Biochemical and genetic studies of the VPg uridylylation reaction catalyzed by the RNA polymerase of poliovirus. Journal of virology 77: 891–904.

13. PaulAV, YinJ, MugaveroJ, RiederE, LiuY, et al. (2003) A “slide-back” mechanism for the initiation of protein-primed RNA synthesis by the RNA polymerase of poliovirus. The Journal of biological chemistry 278: 43951–43960.

14. HsuNY, IlnytskaO, BelovG, SantianaM, ChenYH, et al. (2010) Viral reorganization of the secretory pathway generates distinct organelles for RNA replication. Cell 141: 799–811.

15. NgKK, ArnoldJJ, CameronCE (2008) Structure-function relationships among RNA-dependent RNA polymerases. Current topics in microbiology and immunology 320: 137–156.

16. WuY, LouZ, MiaoY, YuY, DongH, et al. (2010) Structures of EV71 RNA-dependent RNA polymerase in complex with substrate and analogue provide a drug target against the hand-foot-and-mouth disease pandemic in China. Protein & cell 1: 491–500.

17. CampagnolaG, WeygandtM, ScogginK, PeersenO (2008) Crystal structure of coxsackievirus B3 3Dpol highlights the functional importance of residue 5 in picornavirus polymerases. Journal of virology 82: 9458–9464.

18. ChenTC, ChangHY, LinPF, ChernJH, HsuJT, et al. (2009) Novel antiviral agent DTriP-22 targets RNA-dependent RNA polymerase of enterovirus 71. Antimicrobial agents and chemotherapy 53: 2740–2747.

19. AminevAG, AminevaSP, PalmenbergAC (2003) Encephalomyocarditis virus (EMCV) proteins 2A and 3BCD localize to nuclei and inhibit cellular mRNA transcription but not rRNA transcription. Virus research 95: 59–73.

20. AminevaSP, AminevAG, PalmenbergAC, GernJE (2004) Rhinovirus 3C protease precursors 3CD and 3CD′ localize to the nuclei of infected cells. The Journal of general virology 85: 2969–2979.

21. KrogerusC, SamuilovaO, PoyryT, JokitaloE, HyypiaT (2007) Intracellular localization and effects of individually expressed human parechovirus 1 non-structural proteins. The Journal of general virology 88: 831–841.

22. SharmaR, RaychaudhuriS, DasguptaA (2004) Nuclear entry of poliovirus protease-polymerase precursor 3CD: implications for host cell transcription shut-off. Virology 320: 195–205.

23. ClarkME, HammerleT, WimmerE, DasguptaA (1991) Poliovirus proteinase 3C converts an active form of transcription factor IIIC to an inactive form: a mechanism for inhibition of host cell polymerase III transcription by poliovirus. The EMBO journal 10: 2941–2947.

24. ClarkME, LiebermanPM, BerkAJ, DasguptaA (1993) Direct cleavage of human TATA-binding protein by poliovirus protease 3C in vivo and in vitro. Molecular and cellular biology 13: 1232–1237.

25. YalamanchiliP, DattaU, DasguptaA (1997) Inhibition of host cell transcription by poliovirus: cleavage of transcription factor CREB by poliovirus-encoded protease 3Cpro. Journal of virology 71: 1220–1226.

26. YalamanchiliP, WeidmanK, DasguptaA (1997) Cleavage of transcriptional activator Oct-1 by poliovirus encoded protease 3Cpro. Virology 239: 176–185.

27. FalkMM, GrigeraPR, BergmannIE, ZibertA, MulthaupG, et al. (1990) Foot-and-mouth disease virus protease 3C induces specific proteolytic cleavage of host cell histone H3. Journal of virology 64: 748–756.

28. WeidmanMK, YalamanchiliP, NgB, TsaiW, DasguptaA (2001) Poliovirus 3C protease-mediated degradation of transcriptional activator p53 requires a cellular activity. Virology 291: 260–271.

29. JuricaMS, MooreMJ (2003) Pre-mRNA splicing: awash in a sea of proteins. Molecular cell 12: 5–14.

30. TurnerIA, NormanCM, ChurcherMJ, NewmanAJ (2004) Roles of the U5 snRNP in spliceosome dynamics and catalysis. Biochemical Society transactions 32: 928–931.

31. ValadkhanS, JaladatY (2010) The spliceosomal proteome: at the heart of the largest cellular ribonucleoprotein machine. Proteomics 10: 4128–4141.

32. PenaV, LiuS, BujnickiJM, LuhrmannR, WahlMC (2007) Structure of a multipartite protein-protein interaction domain in splicing factor prp8 and its link to retinitis pigmentosa. Molecular cell 25: 615–624.

33. GalejWP, OubridgeC, NewmanAJ, NagaiK (2013) Crystal structure of Prp8 reveals active site cavity of the spliceosome. Nature 493: 638–643.

34. ButlerMI, PoulterRT (2005) The PRP8 inteins in Cryptococcus are a source of phylogenetic and epidemiological information. Fungal genetics and biology: FG & B 42: 452–463.

35. WahlMC, WillCL, LuhrmannR (2009) The spliceosome: design principles of a dynamic RNP machine. Cell 136: 701–718.

36. GraingerRJ, BeggsJD (2005) Prp8 protein: at the heart of the spliceosome. Rna 11: 533–557.

37. LiuS, RauhutR, VornlocherHP, LuhrmannR (2006) The network of protein-protein interactions within the human U4/U6.U5 tri-snRNP. Rna 12: 1418–1430.

38. LiX, ZhangW, XuT, RamseyJ, ZhangL, et al. (2013) Comprehensive in vivo RNA-binding site analyses reveal a role of Prp8 in spliceosomal assembly. Nucleic acids research 41: 3805–3818.

39. UmenJG, GuthrieC (1995) The second catalytic step of pre-mRNA splicing. Rna 1: 869–885.

40. KramerA (1996) The structure and function of proteins involved in mammalian pre-mRNA splicing. Annual review of biochemistry 65: 367–409.

41. WachtelC, ManleyJL (2009) Splicing of mRNA precursors: the role of RNAs and proteins in catalysis. Molecular bioSystems 5: 311–316.

42. AronovaA, BacikovaD, CrottiLB, HorowitzDS, SchwerB (2007) Functional interactions between Prp8, Prp18, Slu7, and U5 snRNA during the second step of pre-mRNA splicing. Rna 13: 1437–1444.

43. LinKT, LuRM, TarnWY (2004) The WW domain-containing proteins interact with the early spliceosome and participate in pre-mRNA splicing in vivo. Molecular and cellular biology 24: 9176–9185.

44. LeeKM, Hsu IaW, TarnWY (2010) TRAP150 activates pre-mRNA splicing and promotes nuclear mRNA degradation. Nucleic acids research 38: 3340–3350.

45. Huang daW, ShermanBT, LempickiRA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols 4: 44–57.

46. McBrideAE, SchlegelA, KirkegaardK (1996) Human protein Sam68 relocalization and interaction with poliovirus RNA polymerase in infected cells. Proceedings of the National Academy of Sciences of the United States of America 93: 2296–2301.

47. LinJY, ChenTC, WengKF, ChangSC, ChenLL, et al. (2009) Viral and host proteins involved in picornavirus life cycle. Journal of biomedical science 16: 103.

48. BelshamGJ (2009) Divergent picornavirus IRES elements. Virus research 139: 183–192.

49. HoBC, YuSL, ChenJJ, ChangSY, YanBS, et al. (2011) Enterovirus-induced miR-141 contributes to shutoff of host protein translation by targeting the translation initiation factor eIF4E. Cell host & microbe 9: 58–69.

50. AlvarezE, CastelloA, CarrascoL, IzquierdoJM (2011) Alternative splicing, a new target to block cellular gene expression by poliovirus 2A protease. Biochemical and biophysical research communications 414: 142–147.

51. ShihSR, StollarV, LinJY, ChangSC, ChenGW, et al. (2004) Identification of genes involved in the host response to enterovirus 71 infection. Journal of neurovirology 10: 293–304.

52. MacejakDG, SarnowP (1991) Internal initiation of translation mediated by the 5′ leader of a cellular mRNA. Nature 353: 90–94.

53. JohannesG, SarnowP (1998) Cap-independent polysomal association of natural mRNAs encoding c-myc, BiP, and eIF4G conferred by internal ribosome entry sites. Rna 4: 1500–1513.

54. ThomaC, BergaminiG, GalyB, HundsdoerferP, HentzeMW (2004) Enhancement of IRES-mediated translation of the c-myc and BiP mRNAs by the poly(A) tail is independent of intact eIF4G and PABP. Molecular cell 15: 925–935.

55. GoharaDW, HaCS, KumarS, GhoshB, ArnoldJJ, et al. (1999) Production of “authentic” poliovirus RNA-dependent RNA polymerase (3D(pol)) by ubiquitin-protease-mediated cleavage in Escherichia coli. Protein expression and purification 17: 128–138.

56. van OoijMJ, VogtDA, PaulA, CastroC, KuijpersJ, et al. (2006) Structural and functional characterization of the coxsackievirus B3 CRE(2C): role of CRE(2C) in negative- and positive-strand RNA synthesis. The Journal of general virology 87: 103–113.

57. PathakHB, ArnoldJJ, WiegandPN, HargittaiMR, CameronCE (2007) Picornavirus genome replication: assembly and organization of the VPg uridylylation ribonucleoprotein (initiation) complex. The Journal of biological chemistry 282: 16202–16213.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 6
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#