#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

XRN1 Stalling in the 5’ UTR of Hepatitis C Virus and Bovine Viral Diarrhea Virus Is Associated with Dysregulated Host mRNA Stability


Understanding how a persistent virus like Hepatitis C Virus (HCV) interfaces with the cellular machinery during infection can provide significant insights into mechanisms of pathogenesis. We demonstrate that while trying to degrade HCV transcripts, a major cellular exonuclease called XRN1 stalls and gets repressed in the 5’ noncoding region of the viral mRNA. Interestingly, the region where XRN1 stalls in the 5’ UTR includes the viral IRES that is required for translation initiation, uncovering a novel, unexpected function for this well-studied region. Differential mRNA stability is a major regulator of gene expression in cells. Curiously, repression of the cellular XRN1 exonuclease is associated with a general repression of mRNA decay in general in HCV-infected cells. Thus numerous cellular mRNAs are stabilized and accumulate in a dysregulated fashion during HCV infection. Normally short-lived mRNAs are disproportionately affected—including mRNAs that encode immune regulators and oncogenes. Thus, this study suggests a novel role for the 5’ UTR of HCV in molecular pathogenesis—including hepatocellular carcinoma. Furthermore, the 5’ UTR of Bovine Viral Diarrhea virus also represses the XRN1 enzyme and stabilizes cellular mRNA. Therefore a strategy of 5’ UTR-mediated XRN1 repression appears to be conserved among the vector-independent members of the Flaviviridae.


Vyšlo v časopise: XRN1 Stalling in the 5’ UTR of Hepatitis C Virus and Bovine Viral Diarrhea Virus Is Associated with Dysregulated Host mRNA Stability. PLoS Pathog 11(3): e32767. doi:10.1371/journal.ppat.1004708
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004708

Souhrn

Understanding how a persistent virus like Hepatitis C Virus (HCV) interfaces with the cellular machinery during infection can provide significant insights into mechanisms of pathogenesis. We demonstrate that while trying to degrade HCV transcripts, a major cellular exonuclease called XRN1 stalls and gets repressed in the 5’ noncoding region of the viral mRNA. Interestingly, the region where XRN1 stalls in the 5’ UTR includes the viral IRES that is required for translation initiation, uncovering a novel, unexpected function for this well-studied region. Differential mRNA stability is a major regulator of gene expression in cells. Curiously, repression of the cellular XRN1 exonuclease is associated with a general repression of mRNA decay in general in HCV-infected cells. Thus numerous cellular mRNAs are stabilized and accumulate in a dysregulated fashion during HCV infection. Normally short-lived mRNAs are disproportionately affected—including mRNAs that encode immune regulators and oncogenes. Thus, this study suggests a novel role for the 5’ UTR of HCV in molecular pathogenesis—including hepatocellular carcinoma. Furthermore, the 5’ UTR of Bovine Viral Diarrhea virus also represses the XRN1 enzyme and stabilizes cellular mRNA. Therefore a strategy of 5’ UTR-mediated XRN1 repression appears to be conserved among the vector-independent members of the Flaviviridae.


Zdroje

1. Scheel TK, Rice CM. (2013) Understanding the hepatitis C virus life cycle paves the way for highly effective therapies. Nat Med 19: 837–849 doi: 10.1038/nm.3248 23836234

2. Shlomai A, de Jong YP, Rice CM. (2014). Virus associated malignancies: The role of viral hepatitis in hepatocellular carcinoma. Semin Cancer Biol. 26C:78–88.

3. Bradrick SS, Walters RW, Gromeier M. (2006) The hepatitis C virus 3'-untranslated region or a poly(A) tract promote efficient translation subsequent to the initiation phase. Nucleic Acids Res 34: 1293–1303. 16510853

4. Bung C, Bochkaeva Z, Terenin I, Zinovkin R, Shatsky IN, et al. (2010) Influence of the hepatitis C virus 3'-untranslated region on IRES-dependent and cap-dependent translation initiation. FEBS Lett 584: 837–842. doi: 10.1016/j.febslet.2010.01.015 20079737

5. Bai Y, Zhou K, Doudna JA. (2013) Hepatitis C virus 3'UTR regulates viral translation through direct interactions with the host translation machinery. Nucleic Acids Res 41: 7861–7874. doi: 10.1093/nar/gkt543 23783572

6. Yi M, Lemon SM. (2003) 3’ nontranslated RNA signals required for replication of hepatitis C virus RNA. J Virol 77: 3557–3568. 12610131

7. Kumar A, Ray U, Das S. (2013) Human La protein interaction with GCAC near the initiator AUG enhances hepatitis C Virus RNA replication by promoting linkage between 5' and 3' untranslated regions. J Virol 87: 6713–6726. doi: 10.1128/JVI.00525-13 23552417

8. Friebe P, Lohmann V, Krieger N, Bartenschlager R. (2001) Sequences in the 5' nontranslated region of hepatitis C virus required for RNA replication. J. Virol 75: 12047–12057. 11711595

9. Niepmann M. (2013) Hepatitis C virus RNA translation. Curr Top Microbiol Immunol 369: 143–166. doi: 10.1007/978-3-642-27340-7_6 23463200

10. Jopling CL, Yi M, Lancaster AM, Lemon SM, and Sarnow P. (2005) Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science 309: 1577–1581. 16141076

11. Jopling CL, Schutz S, Sarnow P. (2008) Position-dependent function for a tandem microRNA miR-122-binding site located in the hepatitis C virus RNA genome. Cell Host Microbe 4: 77–85. doi: 10.1016/j.chom.2008.05.013 18621012

12. Cox EM, Sagan SM, Mortimer SA, Doudna JA, Sarnow P. (2013) Enhancement of hepatitis C viral RNA abundance by precursor miR-122 molecules. RNA 19: 1825–1832. doi: 10.1261/rna.040865.113 24106328

13. Wilson JA, Huys A. (2013) miR-122 promotion of the hepatitis C virus life cycle: sound in the silence. Wiley Interdiscip Rev RNA 4: 665–676. doi: 10.1002/wrna.1186 23881584

14. Li Y, Masaki T, Yamane D, McGivern DR, Lemon SM. (2013) Competing and noncompeting activities of miR-122 and the 5' exonuclease Xrn1 in regulation of hepatitis C virus replication. Proc Natl Acad Sci U S A 110: 1881–1886. doi: 10.1073/pnas.1213515110 23248316

15. Thibault PA, Huys A, Dhillon P, Wilson JA. (2013) MicroRNA-122-dependent and -independent replication of Hepatitis C Virus in Hep3B human hepatoma cells. Virology 436: 179–190. doi: 10.1016/j.virol.2012.11.007 23245472

16. Mortimer SA, Doudna JA. (2013) Unconventional miR-122 binding stabilizes the HCV genome by forming a trimolecular RNA structure. Nucleic Acids Res 41: 4230–4240. doi: 10.1093/nar/gkt075 23416544

17. Conrad KD, Giering F, Erfurth C, Neumann A, Fehr C, et al. (2013) MicroRNA-122 dependent binding of Ago2 protein to hepatitis C virus RNA is associated with enhanced RNA stability and translation stimulation. PLoS One. 8(2):e56272. doi: 10.1371/journal.pone.0056272 23405269

18. Li Y, Masaki T, Lemon SM. (2013) miR-122 and the Hepatitis C RNA genome: more than just stability. RNA Biol 10: 919–923. doi: 10.4161/rna.25137 23770926

19. Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, et al. (2013) Treatment of HCV infection by targeting microRNA. N Engl J Med 368: 1685–1694. doi: 10.1056/NEJMoa1209026 23534542

20. Schoenberg DR, Maquat LE. (2012) Regulation of cytoplasmic mRNA decay. Nat Rev Genet 13: 246–259 doi: 10.1038/nrg3160 22392217

21. García-Martínez J, Aranda A, Pérez-Ortín JE. (2004) Genomic run-on evaluates transcription rates for all yeast genes and identifies gene regulatory mechanisms. Mol Cell 15: 303–313. 15260981

22. Cheadle C, Fan J, Cho-Chung YS, Werner T, Ray J, et al. (2005) Stability regulation of mRNA and the control of gene expression. Ann N Y Acad Sci 1058: 196–204. 16394137

23. Elkon R, Zlotorynski E, Zeller KI, Agami R. (2010) Major role for mRNA stability in shaping the kinetics of gene induction. BMC Genomics 11: 259. doi: 10.1186/1471-2164-11-259 20409322

24. Shalem O, Groisman B, Choder M, Dahan O, Pilpel Y. (2011) Transcriptome kinetics is governed by a genome-wide coupling of mRNA production and degradation: a role for RNA Pol II. PLoS Genet. 7(9):e1002273. doi: 10.1371/journal.pgen.1002273 21931566

25. Schwanhäusser B, Busse D, Li N, Dittmar G, Schuchhardt J, et al. (2011) Global quantification of mammalian gene expression control. Nature 473: 337–342. doi: 10.1038/nature10098 21593866

26. Cacace F, Paci P, Cusimano V, Germani A, Farina L. (2012) Stochastic modeling of expression kinetics identifies messenger half-lives and reveals sequential waves of co-ordinated transcription and decay. PLoS Comput Biol 8: e1002772. doi: 10.1371/journal.pcbi.1002772 23144606

27. Mühlemann, O, Jensen, TH. (2012) mRNP quality control goes regulatory. Trends Genet I 70–77.

28. Garneau NL, Wilusz J, Wilusz CJ. (2007) The highways and byways of mRNA decay. Nat Rev Mol Cell Biol 8: 113–126. 17245413

29. Jinek M, Coyle SM, Doudna JA. (2011) Coupled 5' nucleotide recognition and processivity in Xrn1-mediated mRNA decay. Mol Cell 41: 600–608. doi: 10.1016/j.molcel.2011.02.004 21362555

30. Chang JH, Xiang S, Xiang K, Manley JL, Tong L. (2011) Structural and biochemical studies of the 5'→3' exoribonuclease Xrn1. Nat Struct Mol Biol 18: 270–276 doi: 10.1038/nsmb.1984 21297639

31. Reis FP, Pobre V, Silva IJ, Malecki M, Arraiano CM. (2013) The RNase II/RNB family of exoribonucleases: putting the 'Dis' in disease. Wiley Interdiscip Rev RNA 4: 607–615. doi: 10.1002/wrna.1180 23776156

32. Moon SL, Wilusz J. (2013) Cytoplasmic viruses: rage against the (cellular RNA decay) machine. PLoS Pathog. 9(12):e1003762. doi: 10.1371/journal.ppat.1003762 24339774

33. Roby JA, Pijlman GP, Wilusz J, Khromykh AA. (2014) Noncoding subgenomic flavivirus RNA: multiple functions in West Nile virus pathogenesis and modulation of host responses. Viruses 6: 404–427. doi: 10.3390/v6020404 24473339

34. Chapman EG, Moon SL, Wilusz J, Kieft JS (2014) RNA structures that resist degradation by Xrn1 produce a pathogenic Dengue virus RNA. Elife 3: e01892. doi: 10.7554/eLife.01892 24692447

35. Chapman EG, Costantino DA, Rabe JL, Moon SL, Wilusz J, et al. (2014) The structural basis of pathogenic subgenomic flavivirus RNA (sfRNA) production. Science 344: 307–310. doi: 10.1126/science.1250897 24744377

36. Pijlman GP, Funk A, Kondratieva N, Leung J, Torres S, et al. (2008) A highly structured, nuclease-resistant, noncoding RNA produced by flaviviruses is required for pathogenicity. Cell Host Microbe 4: 579–591. doi: 10.1016/j.chom.2008.10.007 19064258

37. Silva PA, Pereira CF, Dalebout TJ, Spaan WJ, Bredenbeek PJ. (2010) An RNA pseudoknot is required for production of yellow fever virus subgenomic RNA by the host nuclease XRN1. J Virol 84: 11395–11406. doi: 10.1128/JVI.01047-10 20739539

38. Funk A, Truong K, Nagasaki T, Torres S, Floden N, et al. (2010) RNA structures required for production of subgenomic flavivirus RNA. J. Virol 84: 11407–11417. doi: 10.1128/JVI.01159-10 20719943

39. Schnettler E, Sterken MG, Leung JY, Metz SW, Geertsema C, et al. (2012) Noncoding flavivirus RNA displays RNA interference suppressor activity in insect and Mammalian cells. J Virol 86: 13486–13500. doi: 10.1128/JVI.01104-12 23035235

40. Pijlman GP. (2014) Flavivirus RNAi suppression: decoding non-coding RNA. Curr Opin Virol 7C: 55–60.

41. Moon SL, Anderson JR, Kumagai Y, Wilusz CJ, Akira S, et al. (2012) A noncoding RNA produced by arthropod-borne flaviviruses inhibits the cellular exoribonuclease XRN1 and alters host mRNA stability. RNA 18: 2029–2040. doi: 10.1261/rna.034330.112 23006624

42. Houe H. (2003) Economic impact of BVDV infection in dairies. Biologicals 31: 137–143. 12770546

43. Walters KA, Syder AJ, Lederer SL, Diamond DL, Paeper B, et al. (2009) Genomic analysis reveals a potential role for cell cycle perturbation in HCV-mediated apoptosis of cultured hepatocytes. PLoS Pathog 5(1):e1000269. doi: 10.1371/journal.ppat.1000269 19148281

44. Pestova TV, Shatsky IN, Fletcher SP, Jackson RJ, Hellen CU. (1998) A prokaryotic-like mode of cytoplasmic eukaryotic ribosome binding to the initiation codon during internal translation initiation of hepatitis C and classical swine fever virus RNAs. Genes Dev 12: 67–83. 9420332

45. Sedano CD, Sarnow P. (2014) Hepatitis C virus subverts liver-specific miR-122 to protect the viral genome from exoribonuclease Xrn2. Cell Host Microbe. 16: 257–264. doi: 10.1016/j.chom.2014.07.006 25121753

46. Grassmann CW, Yu H, Isken O, Behrens SE. (2005) Hepatitis C virus and the related bovine viral diarrhea virus considerably differ in the functional organization of the 5' non-translated region: implications for the viral life cycle. Virology 333: 349–366. 15721367

47. Mukherjee D, Gao M, O'Connor JP, Raijmakers R, Pruijn G, et al. (2002) The mammalian exosome mediates the efficient degradation of mRNAs that contain AU-rich elements. EMBO J 21: 165–174. 11782436

48. Orban TI, Izaurralde E. (2005) Decay of mRNAs targeted by RISC requires XRN1, the Ski complex, and the exosome. RNA 11: 459–469. 15703439

49. Blight KJ, Kolykhalov AA, Rice CM. (2000) Efficient initiation of HCV RNA replication in cell culture. Science 290: 1972–1974. 11110665

50. Pérez-Vilaró G, Scheller N, Saludes V, Díez J. (2012) Hepatitis C virus infection alters P-body composition but is independent of P-body granules. J Virol 86: 8740–8749. doi: 10.1128/JVI.07167-11 22674998

51. Pager CT, Schütz S, Abraham TM, Luo G, Sarnow P. (2013) Modulation of hepatitis C virus RNA abundance and virus release by dispersion of processing bodies and enrichment of stress granules. Virology 435: 472–484. doi: 10.1016/j.virol.2012.10.027 23141719

52. Dölken L, Ruzsics Z, Rädle B, Friedel CC, Zimmer R, et al. (2008) High-resolution gene expression profiling for simultaneous kinetic parameter analysis of RNA synthesis and decay. RNA 14: 1959–1972. doi: 10.1261/rna.1136108 18658122

53. Payne, TL, Blackinton, J, Frisbeem, A, Pickeral, J, Sawant, S, et al. (2014) Transcriptional and Post-transcriptional Regulation of Cytokine Gene Expression in HIV-1 Antigen-specific CD8+ T Cells that Mediate Virus Inhibition. J Virol Jun 4. pii: JVI.00802-14.

54. Breuer K, Foroushani AK, Laird MR, Chen C, Sribnaia A, et al. (2013) InnateDB: systems biology of innate immunity and beyond—recent updates and continuing curation. Nucleic Acids Res 41: D1228–1233. doi: 10.1093/nar/gks1147 23180781

55. Sharova LV, Sharov AA, Nedorezov T, Piao Y, Shaik N, et al. (2009) Database for mRNA half-life of 19,977 genes obtained by DNA microarray analysis of pluripotent and differentiating mouse embryonic stem cells. DNA Res 1: 45–58. doi: 10.1093/dnares/dsn030 19001483

56. Lam LT, Pickeral OK, Peng AC, Rosenwald A, Hurt EM, et al. (2001) Genomic-scale measurement of mRNA turnover and the mechanisms of action of the anti-cancer drug flavopiridol. Genome Biology 2: RESEARCH0041.

57. Shimakami T, Yamane D, Jangra RK, Kempf BJ, Spaniel C, et al. (2012) Stabilization of hepatitis C virus RNA by an Ago2-miR-122 complex. Proc Natl Acad Sci U S A 109: 941–946. doi: 10.1073/pnas.1112263109 22215596

58. Bidet K, Dadlani D, Garcia-Blanco MA. (2014) G3BP1, G3BP2 and CAPRIN1 are required for translation of interferon stimulated mRNAs and are targeted by a Dengue virus non-coding RNA. PLoS Pathog 10(7):e1004242. doi: 10.1371/journal.ppat.1004242 24992036

59. Berry KE, Waghray S, Mortimer SA, Bai Y, Doudna JA. (2011) Crystal structure of the HCV IRES central domain reveals strategy for start-codon positioning. Structure 19: 1456–1466. doi: 10.1016/j.str.2011.08.002 22000514

60. Barnhart MD, Moon SL, Emch AW, Wilusz CJ, et al. (2013) Changes in cellular mRNA stability, splicing, and polyadenylation through HuR protein sequestration by a cytoplasmic RNA virus. Cell Rep 5: 909–917. doi: 10.1016/j.celrep.2013.10.012 24210824

61. Braun JE, Truffault V, Boland A, Huntzinger E, Chang CT, et al. (2012) A direct interaction between DCP1 and XRN1 couples mRNA decapping to 5' exonucleolytic degradation. Nat Struct Mol. Biol 19: 1324–1331. doi: 10.1038/nsmb.2413 23142987

62. Ozgur S, Chekulaeva M, Stoecklin G. (2010) Human Pat1b connects deadenylation with mRNA decapping and controls the assembly of processing bodies. Mol Cell Biol 30: 4308–4323. doi: 10.1128/MCB.00429-10 20584987

63. Sun M, Schwalb B, Pirkl N, Maier KC, Schenk A, et al. (2013) Global analysis of eukaryotic mRNA degradation reveals Xrn1-dependent buffering of transcript levels. Mol Cell 52: 52–62. doi: 10.1016/j.molcel.2013.09.010 24119399

64. Medina DA, Jordán-Pla A, Millán-Zambrano G, Chávez S, Choder M, et al. (2014) Cytoplasmic 5'-3' exonuclease Xrn1p is also a genome-wide transcription factor in yeast. Front Genet 5: 1. doi: 10.3389/fgene.2014.00001 24567736

65. Zhang K, Dion N, Fuchs B, Damron T, Gitelis S, et al. (2002) The human homolog of yeast SEP1 is a novel candidate tumor suppressor gene in osteogenic sarcoma. Gene 298: 121–127. 12426100

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 3
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#