#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Recruitment of RED-SMU1 Complex by Influenza A Virus RNA Polymerase to Control Viral mRNA Splicing


Influenza A viruses are major pathogens which pose continuous animal and public health challenges. Enhancing the knowledge of their life cycle, and especially the understanding of how viral components interact with the host cell, is essential to achieve better prevention and treatment of the disease. The polymerase of influenza A viruses plays a central role in the viral cycle, notably by driving the synthesis of viral messenger RNAs that are translated into viral proteins. Here we identify two human splicing factors, RED and SMU1, that associate with the viral polymerase. We show that these factors jointly regulate the splicing of the NS1 messenger RNA into the shorter NS2 messenger RNA, which encodes a key viral protein named NS2/NEP. We demonstrate that RED and SMU1 are required for efficient expression of NS2/NEP, and for the NS2/NEP-mediated intracellular trafficking of viral components. Overall, our results show that RED and SMU1 are essential for the replication of influenza A viruses. With respect to the need of novel anti-influenza therapies for epidemic and pandemic preparedness, the functional and physical interactions between these cellular splicing factors and the viral transcriptional machinery could be targeted to inhibit viral replication.


Vyšlo v časopise: Recruitment of RED-SMU1 Complex by Influenza A Virus RNA Polymerase to Control Viral mRNA Splicing. PLoS Pathog 10(6): e32767. doi:10.1371/journal.ppat.1004164
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004164

Souhrn

Influenza A viruses are major pathogens which pose continuous animal and public health challenges. Enhancing the knowledge of their life cycle, and especially the understanding of how viral components interact with the host cell, is essential to achieve better prevention and treatment of the disease. The polymerase of influenza A viruses plays a central role in the viral cycle, notably by driving the synthesis of viral messenger RNAs that are translated into viral proteins. Here we identify two human splicing factors, RED and SMU1, that associate with the viral polymerase. We show that these factors jointly regulate the splicing of the NS1 messenger RNA into the shorter NS2 messenger RNA, which encodes a key viral protein named NS2/NEP. We demonstrate that RED and SMU1 are required for efficient expression of NS2/NEP, and for the NS2/NEP-mediated intracellular trafficking of viral components. Overall, our results show that RED and SMU1 are essential for the replication of influenza A viruses. With respect to the need of novel anti-influenza therapies for epidemic and pandemic preparedness, the functional and physical interactions between these cellular splicing factors and the viral transcriptional machinery could be targeted to inhibit viral replication.


Zdroje

1. Resa-InfanteP, JorbaN, ColomaR, OrtinJ (2011) The influenza virus RNA synthesis machine: advances in its structure and function. RNA Biol 8: 207–215.

2. YorkA, FodorE (2013) Biogenesis, assembly and export of viral messenger ribonucleoproteins in the influenza A virus infected cell. RNA biology 10: 1274–1282.

3. ShihSR, SuenPC, ChenYS, ChangSC (1998) A novel spliced transcript of influenza A/WSN/33 virus. Virus genes 17: 179–183.

4. WiseHM, HutchinsonEC, JaggerBW, StuartAD, KangZH, et al. (2012) Identification of a novel splice variant form of the influenza A virus M2 ion channel with an antigenically distinct ectodomain. PLoS pathogens 8: e1002998.

5. HaleBG, RandallRE, OrtinJ, JacksonD (2008) The multifunctional NS1 protein of influenza A viruses. The Journal of general virology 89: 2359–2376.

6. PatersonD, FodorE (2012) Emerging roles for the influenza A virus nuclear export protein (NEP). PLoS pathogens 8: e1003019.

7. SelmanM, DankarSM, ForbesNE, JiaJJ, BrownEG (2012) Adaptive mutation in influenza A virus non-structural gene is linked to host switching and induces a novel protein by alternative splicing. Emerging Microbes & Infections 1: e42.

8. ChiangC, ChenGW, ShihSR (2008) Mutations at alternative 5′ splice sites of M1 mRNA negatively affect influenza A virus viability and growth rate. Journal of virology 82: 10873–10886.

9. ChuaMA, SchmidS, PerezJT, LangloisRA, TenoeverBR (2013) Influenza A virus utilizes suboptimal splicing to coordinate the timing of infection. Cell reports 3: 23–29.

10. PrioreSF, KierzekE, KierzekR, BamanJR, MossWN, et al. (2013) Secondary structure of a conserved domain in the intron of influenza A NS1 mRNA. PLoS One 8: e70615.

11. ShihSR, NemeroffME, KrugRM (1995) The choice of alternative 5′ splice sites in influenza virus M1 mRNA is regulated by the viral polymerase complex. Proceedings of the National Academy of Sciences of the United States of America 92: 6324–6328.

12. RobbNC, FodorE (2012) The accumulation of influenza A virus segment 7 spliced mRNAs is regulated by the NS1 protein. The Journal of general virology 93: 113–118.

13. RobbNC, JacksonD, VreedeFT, FodorE (2010) Splicing of influenza A virus NS1 mRNA is independent of the viral NS1 protein. The Journal of general virology 91: 2331–2340.

14. GaraigortaU, OrtinJ (2007) Mutation analysis of a recombinant NS replicon shows that influenza virus NS1 protein blocks the splicing and nucleo-cytoplasmic transport of its own viral mRNA. Nucleic acids research 35: 4573–4582.

15. StertzS, ShawML (2011) Uncovering the global host cell requirements for influenza virus replication via RNAi screening. Microbes Infect 13: 516–525.

16. WatanabeT, WatanabeS, KawaokaY (2010) Cellular networks involved in the influenza virus life cycle. Cell Host Microbe 7: 427–439.

17. ShihSR, KrugRM (1996) Novel exploitation of a nuclear function by influenza virus: the cellular SF2/ASF splicing factor controls the amount of the essential viral M2 ion channel protein in infected cells. The EMBO journal 15: 5415–5427.

18. TsaiPL, ChiouNT, KussS, Garcia-SastreA, LynchKW, et al. (2013) Cellular RNA binding proteins NS1-BP and hnRNP K regulate influenza A virus RNA splicing. PLoS pathogens 9: e1003460.

19. SpartzAK, HermanRK, ShawJE (2004) SMU-2 and SMU-1, Caenorhabditis elegans homologs of mammalian spliceosome-associated proteins RED and fSAP57, work together to affect splice site choice. Molecular and cellular biology 24: 6811–6823.

20. DassahM, PatzekS, HuntVM, MedinaPE, ZahlerAM (2009) A genetic screen for suppressors of a mutated 5′ splice site identifies factors associated with later steps of spliceosome assembly. Genetics 182: 725–734.

21. ChungT, WangD, KimCS, YadegariR, LarkinsBA (2009) Plant SMU-1 and SMU-2 homologues regulate pre-mRNA splicing and multiple aspects of development. Plant physiology 151: 1498–1512.

22. ArranzR, ColomaR, ChichonFJ, ConesaJJ, CarrascosaJL, et al. (2012) The structure of native influenza virion ribonucleoproteins. Science 338: 1634–1637.

23. ColomaR, ValpuestaJM, ArranzR, CarrascosaJL, OrtínJ, et al. (2009) The structure of a biologically active influenza virus ribonucleoprotein complex. PLoS Pathog 5: e1000491.

24. AssierE, Bouzinba-SegardH, StolzenbergMC, StephensR, BardosJ, et al. (1999) Isolation, sequencing and expression of RED, a novel human gene encoding an acidic-basic dipeptide repeat. Gene 230: 145–154.

25. NeubauerG, KingA, RappsilberJ, CalvioC, WatsonM, et al. (1998) Mass spectrometry and EST-database searching allows characterization of the multi-protein spliceosome complex. Nature genetics 20: 46–50.

26. ZhouZ, LickliderLJ, GygiSP, ReedR (2002) Comprehensive proteomic analysis of the human spliceosome. Nature 419: 182–185.

27. CassonnetP, RolloyC, NeveuG, VidalainPO, ChantierT, et al. (2011) Benchmarking a luciferase complementation assay for detecting protein complexes. Nat Methods 8: 990–992.

28. DengT, EngelhardtOG, ThomasB, AkoulitchevAV, BrownleeGG, et al. (2006) Role of ran binding protein 5 in nuclear import and assembly of the influenza virus RNA polymerase complex. J Virol 80: 11911–11919.

29. GabrielG, HerwigA, KlenkHD (2008) Interaction of polymerase subunit PB2 and NP with importin alpha1 is a determinant of host range of influenza A virus. PLoS Pathog 4: e11.

30. HuarteM, Sanz-EzquerroJJ, RoncalF, OrtínJ, NietoA (2001) PA subunit from influenza virus polymerase complex interacts with a cellular protein with homology to a family of transcriptional activators. J Virol 75: 8597–8604.

31. HegeleA, KamburovA, GrossmannA, SourlisC, WowroS, et al. (2012) Dynamic protein-protein interaction wiring of the human spliceosome. Molecular cell 45: 567–580.

32. ChaseGP, Rameix-WeltiMA, ZvirblieneA, ZvirblisG, GotzV, et al. (2011) Influenza virus ribonucleoprotein complexes gain preferential access to cellular export machinery through chromatin targeting. PLoS Pathog 7: e1002187.

33. GeX, Rameix-WeltiMA, GaultE, ChaseG, dos Santos AfonsoE, et al. (2011) Influenza virus infection induces the nuclear relocalization of the Hsp90 co-chaperone p23 and inhibits the glucocorticoid receptor response. PLoS One 6: e23368.

34. Rameix-WeltiMA, TomoiuA, Dos Santos AfonsoE, van der WerfS, NaffakhN (2009) Avian Influenza A virus polymerase association with nucleoprotein, but not polymerase assembly, is impaired in human cells during the course of infection. Journal of virology 83: 1320–1331.

35. KriefP, Augery-BourgetY, PlaisanceS, MerckMF, AssierE, et al. (1994) A new cytokine (IK) down-regulating HLA class II: monoclonal antibodies, cloning and chromosome localization. Oncogene 9: 3449–3456.

36. CaoLX, Le Bousse-KerdilesMC, ClayD, OshevskiS, JasminC, et al. (1997) Implication of a new molecule IK in CD34+ hematopoietic progenitor cell proliferation and differentiation. Blood 89: 3615–3623.

37. YehPC, YehCC, ChenYC, JuangYL (2012) RED, a spindle pole-associated protein, is required for kinetochore localization of MAD1, mitotic progression, and activation of the spindle assembly checkpoint. The Journal of biological chemistry 287: 11704–11716.

38. AvilovSV, MoisyD, MunierS, SchraidtO, NaffakhN, et al. (2012) Replication-competent influenza A virus that encodes a split-green fluorescent protein-tagged PB2 polymerase subunit allows live-cell imaging of the virus life cycle. J Virol 86: 1433–1448.

39. ValcarcelJ, PortelaA, OrtinJ (1991) Regulated M1 mRNA splicing in influenza virus-infected cells. The Journal of general virology 72 (Pt 6) 1301–1308.

40. SugayaK, HongoE, IshiharaY, TsujiH (2006) The conserved role of Smu1 in splicing is characterized in its mammalian temperature-sensitive mutant. Journal of cell science 119: 4944–4951.

41. CooperTA, WanL, DreyfussG (2009) RNA and disease. Cell 136: 777–793.

42. JagerS, CimermancicP, GulbahceN, JohnsonJR, McGovernKE, et al. (2012) Global landscape of HIV-human protein complexes. Nature 481: 365–370.

43. YorkA, HengrungN, VreedeFT, HuiskonenJT, FodorE (2013) Isolation and characterization of the positive-sense replicative intermediate of a negative-strand RNA virus. Proceedings of the National Academy of Sciences of the United States of America 110: E4238–4245.

44. CloseP, EastP, Dirac-SvejstrupAB, HartmannH, HeronM, et al. (2012) DBIRD complex integrates alternative mRNA splicing with RNA polymerase II transcript elongation. Nature 484: 386–389.

45. MontesM, BecerraS, Sanchez-AlvarezM, SuneC (2012) Functional coupling of transcription and splicing. Gene 501: 104–117.

46. DinkelH, Van RoeyK, MichaelS, DaveyNE, WeatherittRJ, et al. (2013) The eukaryotic linear motif resource ELM: 10 years and counting. Nucleic acids research 42: D259–D266.

47. RobbNC, SmithM, VreedeFT, FodorE (2009) NS2/NEP protein regulates transcription and replication of the influenza virus RNA genome. The Journal of general virology 90: 1398–1407.

48. HeY, XuK, KeinerB, ZhouJ, CzudaiV, et al. (2010) Influenza A virus replication induces cell cycle arrest in G0/G1 phase. Journal of virology 84: 12832–12840.

49. BakkourN, LinYL, MaireS, AyadiL, Mahuteau-BetzerF, et al. (2007) Small-molecule inhibition of HIV pre-mRNA splicing as a novel antiretroviral therapy to overcome drug resistance. PLoS pathogens 3: 1530–1539.

50. MunierS, RollandT, DiotC, JacobY, NaffakhN (2013) Exploration of binary virus-host interactions using an infectious protein complementation assay. Molecular and Cellular Proteomics 12: 2845–2855.

51. KomarovaAV, CombredetC, Meyniel-SchicklinL, ChapelleM, CaignardG, et al. (2011) Proteomic analysis of virus-host interactions in an infectious context using recombinant viruses. Molecular and Cellular Proteomics 10: M110.007443.

52. MatrosovichM, MatrosovichT, GartenW, KlenkHD (2006) New low-viscosity overlay medium for viral plaque assays. Virol J 3: 63.

53. AparicioO, RazquinN, ZaratieguiM, NarvaizaI, FortesP (2006) Adenovirus virus-associated RNA is processed to functional interfering RNAs involved in virus production. Journal of virology 80: 1376–1384.

54. MoisyD, AvilovSV, JacobY, LaoideBM, GeX, et al. (2012) HMGB1 protein binds to influenza virus nucleoprotein and promotes viral replication. Journal of virology 86: 9122–9133.

55. VignuzziM, GerbaudS, van der WerfS, EscriouN (2001) Naked RNA immunization with replicons derived from poliovirus and Semliki Forest virus genomes for the generation of a cytotoxic T cell response against the influenza A virus nucleoprotein. J Gen Virol 82: 1737–1747.

56. LivakKJ, SchmittgenTD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408.

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#