#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Gammaherpesviral Gene Expression and Virion Composition Are Broadly Controlled by Accelerated mRNA Degradation


Lytic gammaherpesvirus infection restricts host gene expression by promoting widespread degradation of cytoplasmic mRNA through the activity of the viral endonuclease SOX. Though generally assumed to be selective for cellular transcripts, the extent to which SOX impacts viral mRNA stability has remained unknown. We addressed this issue using the model murine gammaherpesvirus MHV68 and, unexpectedly, found that all stages of viral gene expression are controlled through mRNA degradation. Using both comprehensive RNA expression profiling and half-life studies we reveal that the levels of the majority of viral mRNAs but not noncoding RNAs are tempered by MHV68 SOX (muSOX) activity. The targeting of viral mRNA by muSOX is functionally significant, as it impacts intracellular viral protein abundance and progeny virion composition. In the absence of muSOX-imposed gene expression control the viral particles display increased cell surface binding and entry as well as enhanced immediate early gene expression. These phenotypes culminate in a viral replication defect in multiple cell types as well as in vivo, highlighting the importance of maintaining the appropriate balance of viral RNA during gammaherpesviral infection. This is the first example of a virus that fails to broadly discriminate between cellular and viral transcripts during host shutoff and instead uses the targeting of viral messages to fine-tune overall gene expression.


Vyšlo v časopise: Gammaherpesviral Gene Expression and Virion Composition Are Broadly Controlled by Accelerated mRNA Degradation. PLoS Pathog 10(1): e32767. doi:10.1371/journal.ppat.1003882
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003882

Souhrn

Lytic gammaherpesvirus infection restricts host gene expression by promoting widespread degradation of cytoplasmic mRNA through the activity of the viral endonuclease SOX. Though generally assumed to be selective for cellular transcripts, the extent to which SOX impacts viral mRNA stability has remained unknown. We addressed this issue using the model murine gammaherpesvirus MHV68 and, unexpectedly, found that all stages of viral gene expression are controlled through mRNA degradation. Using both comprehensive RNA expression profiling and half-life studies we reveal that the levels of the majority of viral mRNAs but not noncoding RNAs are tempered by MHV68 SOX (muSOX) activity. The targeting of viral mRNA by muSOX is functionally significant, as it impacts intracellular viral protein abundance and progeny virion composition. In the absence of muSOX-imposed gene expression control the viral particles display increased cell surface binding and entry as well as enhanced immediate early gene expression. These phenotypes culminate in a viral replication defect in multiple cell types as well as in vivo, highlighting the importance of maintaining the appropriate balance of viral RNA during gammaherpesviral infection. This is the first example of a virus that fails to broadly discriminate between cellular and viral transcripts during host shutoff and instead uses the targeting of viral messages to fine-tune overall gene expression.


Zdroje

1. RichnerJM, ClydeK, PezdaAC, ChengBY, WangT, et al. (2011) Global mRNA degradation during lytic gammaherpesvirus infection contributes to establishment of viral latency. PLoS Pathog 7: e1002150.

2. KwongAD, FrenkelN (1989) The herpes simplex virus virion host shutoff function. J Virol 63: 4834–4839.

3. StrelowLI, LeibDA (1995) Role of the virion host shutoff (vhs) of herpes simplex virus type 1 in latency and pathogenesis. J Virol 69: 6779–6786.

4. KrausslichHG, NicklinMJ, ToyodaH, EtchisonD, WimmerE (1987) Poliovirus proteinase 2A induces cleavage of eucaryotic initiation factor 4F polypeptide p220. J Virol 61: 2711–2718.

5. ZamoraM, MarissenWE, LloydRE (2002) Multiple eIF4GI-specific protease activities present in uninfected and poliovirus-infected cells. J Virol 76: 165–177.

6. Kuyumcu-MartinezNM, Van EdenME, YounanP, LloydRE (2004) Cleavage of poly(A)-binding protein by poliovirus 3C protease inhibits host cell translation: a novel mechanism for host translation shutoff. Mol Cell Biol 24: 1779–1790.

7. SciabicaKS, DaiQJ, Sandri-GoldinRM (2003) ICP27 interacts with SRPK1 to mediate HSV splicing inhibition by altering SR protein phosphorylation. EMBO J 22: 1608–1619.

8. KoffaMD, ClementsJB, IzaurraldeE, WaddS, WilsonSA, et al. (2001) Herpes simplex virus ICP27 protein provides viral mRNAs with access to the cellular mRNA export pathway. EMBO J 20: 5769–5778.

9. ChenIH, SciabicaKS, Sandri-GoldinRM (2002) ICP27 interacts with the RNA export factor Aly/REF to direct herpes simplex virus type 1 intronless mRNAs to the TAP export pathway. J Virol 76: 12877–12889.

10. KwongAD, KruperJA, FrenkelN (1988) Herpes simplex virus virion host shutoff function. J Virol 62: 912–921.

11. TaddeoB, ZhangW, RoizmanB (2013) The herpes simplex virus host shutoff RNase degrades cellular and viral mRNAs made before infection but not viral mRNA made after infection. J Virol 87: 4516–4522.

12. ShuM, TaddeoB, ZhangW, RoizmanB (2013) Selective degradation of mRNAs by the HSV host shutoff RNase is regulated by the UL47 tegument protein. Proc Natl Acad Sci U S A 110: E1669–1675.

13. HuangC, LokugamageKG, RozovicsJM, NarayananK, SemlerBL, et al. (2011) SARS coronavirus nsp1 protein induces template-dependent endonucleolytic cleavage of mRNAs: viral mRNAs are resistant to nsp1-induced RNA cleavage. PLoS Pathog 7: e1002433.

14. GlaunsingerB, GanemD (2004) Lytic KSHV infection inhibits host gene expression by accelerating global mRNA turnover. Mol Cell 13: 713–723.

15. CovarrubiasS, RichnerJM, ClydeK, LeeYJ, GlaunsingerBA (2009) Host shutoff is a conserved phenotype of gammaherpesvirus infection and is orchestrated exclusively from the cytoplasm. J Virol 83: 9554–9566.

16. CovarrubiasS, GagliaMM, KumarGR, WongW, JacksonAO, et al. (2011) Coordinated destruction of cellular messages in translation complexes by the gammaherpesvirus host shutoff factor and the mammalian exonuclease Xrn1. PLoS Pathog 7: e1002339.

17. LeeYJ, GlaunsingerBA (2009) Aberrant herpesvirus-induced polyadenylation correlates with cellular messenger RNA destruction. PLoS Biol 7: e1000107.

18. KumarGR, GlaunsingerBA (2010) Nuclear import of cytoplasmic poly(A) binding protein restricts gene expression via hyperadenylation and nuclear retention of mRNA. Mol Cell Biol 30: 4996–5008.

19. KumarGR, ShumL, GlaunsingerBA (2011) Importin alpha-mediated nuclear import of cytoplasmic poly(A) binding protein occurs as a direct consequence of cytoplasmic mRNA depletion. Mol Cell Biol 31: 3113–3125.

20. GagliaMM, CovarrubiasS, WongW, GlaunsingerBA (2012) A common strategy for host RNA degradation by divergent viruses. J Virol 86: 9527–9530.

21. RoweM, GlaunsingerB, van LeeuwenD, ZuoJ, SweetmanD, et al. (2007) Host shutoff during productive Epstein-Barr virus infection is mediated by BGLF5 and may contribute to immune evasion. Proc Natl Acad Sci U S A 104: 3366–3371.

22. ZuoJ, ThomasW, van LeeuwenD, MiddeldorpJM, WiertzEJ, et al. (2008) The DNase of gammaherpesviruses impairs recognition by virus-specific CD8+ T cells through an additional host shutoff function. J Virol 82: 2385–2393.

23. JohnsonLS, WillertEK, VirginHW (2010) Redefining the genetics of murine gammaherpesvirus 68 via transcriptome-based annotation. Cell Host Microbe 7: 516–526.

24. MichaelK, KluppBG, MettenleiterTC, KargerA (2006) Composition of pseudorabies virus particles lacking tegument protein US3, UL47, or UL49 or envelope glycoprotein E. J Virol 80: 1332–1339.

25. NohCW, ChoHJ, KangHR, JinHY, LeeS, et al. (2012) The virion-associated open reading frame 49 of murine gammaherpesvirus 68 promotes viral replication both in vitro and in vivo as a derepressor of RTA. J Virol 86: 1109–1118.

26. LingPD, TanJ, SewatanonJ, PengR (2008) Murine gammaherpesvirus 68 open reading frame 75c tegument protein induces the degradation of PML and is essential for production of infectious virus. J Virol 82: 8000–8012.

27. MayJS, ColemanHM, SmillieB, EfstathiouS, StevensonPG (2004) Forced lytic replication impairs host colonization by a latency-deficient mutant of murine gammaherpesvirus-68. J Gen Virol 85: 137–146.

28. HairJR, LyonsPA, SmithKG, EfstathiouS (2007) Control of Rta expression critically determines transcription of viral and cellular genes following gammaherpesvirus infection. J Gen Virol 88: 1689–1697.

29. GasparM, MayJS, SuklaS, FredericoB, GillMB, et al. (2011) Murid herpesvirus-4 exploits dendritic cells to infect B cells. PLoS Pathog 7: e1002346.

30. HwangS, WuTT, TongLM, KimKS, Martinez-GuzmanD, et al. (2008) Persistent gammaherpesvirus replication and dynamic interaction with the host in vivo. J Virol 82: 12498–12509.

31. BartonE, MandalP, SpeckSH (2011) Pathogenesis and host control of gammaherpesviruses: lessons from the mouse. Annu Rev Immunol 29: 351–397.

32. OroskarAA, ReadGS (1989) Control of mRNA stability by the virion host shutoff function of herpes simplex virus. J Virol 63: 1897–1906.

33. LamQ, SmibertCA, KoopKE, LaveryC, CaponeJP, et al. (1996) Herpes simplex virus VP16 rescues viral mRNA from destruction by the virion host shutoff function. EMBO J 15: 2575–2581.

34. SmibertCA, PopovaB, XiaoP, CaponeJP, SmileyJR (1994) Herpes simplex virus VP16 forms a complex with the virion host shutoff protein vhs. J Virol 68: 2339–2346.

35. TaddeoB, SciortinoMT, ZhangW, RoizmanB (2007) Interaction of herpes simplex virus RNase with VP16 and VP22 is required for the accumulation of the protein but not for accumulation of mRNA. Proc Natl Acad Sci U S A 104: 12163–12168.

36. ChengBY, ZhiJ, SantanaA, KhanS, SalinasE, et al. (2012) Tiled microarray identification of novel viral transcript structures and distinct transcriptional profiles during two modes of productive murine gammaherpesvirus 68 infection. J Virol 86: 4340–4357.

37. BechtelJT, WinantRC, GanemD (2005) Host and viral proteins in the virion of Kaposi's sarcoma-associated herpesvirus. J Virol 79: 4952–4964.

38. BortzE, WhiteleggeJP, JiaQ, ZhouZH, StewartJP, et al. (2003) Identification of proteins associated with murine gammaherpesvirus 68 virions. J Virol 77: 13425–13432.

39. ZhuFX, ChongJM, WuL, YuanY (2005) Virion proteins of Kaposi's sarcoma-associated herpesvirus. J Virol 79: 800–811.

40. GlaunsingerB, GanemD (2004) Highly selective escape from KSHV-mediated host mRNA shutoff and its implications for viral pathogenesis. J Exp Med 200: 391–398.

41. ClydeK, GlaunsingerBA (2011) Deep sequencing reveals direct targets of gammaherpesvirus-induced mRNA decay and suggests that multiple mechanisms govern cellular transcript escape. PLoS One 6: e19655.

42. HutinS, LeeY, GlaunsingerBA (2013) An RNA element in human interleukin 6 confers escape from degradation by the gammaherpesvirus SOX protein. J Virol 87: 4672–4682.

43. MettenleiterTC (2004) Budding events in herpesvirus morphogenesis. Virus Res 106: 167–180.

44. GuoH, WangL, PengL, ZhouZH, DengH (2009) Open reading frame 33 of a gammaherpesvirus encodes a tegument protein essential for virion morphogenesis and egress. J Virol 83: 10582–10595.

45. del RioT, DeCosteCJ, EnquistLW (2005) Actin is a component of the compensation mechanism in pseudorabies virus virions lacking the major tegument protein VP22. J Virol 79: 8614–8619.

46. BechtelJ, GrundhoffA, GanemD (2005) RNAs in the virion of Kaposi's sarcoma-associated herpesvirus. J Virol 79: 10138–10146.

47. TerhuneSS, SchroerJ, ShenkT (2004) RNAs are packaged into human cytomegalovirus virions in proportion to their intracellular concentration. J Virol 78: 10390–10398.

48. SathishN, WangX, YuanY (2012) Tegument Proteins of Kaposi's Sarcoma-Associated Herpesvirus and Related Gamma-Herpesviruses. Front Microbiol 3: 98.

49. HwangS, KimKS, FlanoE, WuTT, TongLM, et al. (2009) Conserved herpesviral kinase promotes viral persistence by inhibiting the IRF-3-mediated type I interferon response. Cell Host Microbe 5: 166–178.

50. TrgovcichJ, JohnsonD, RoizmanB (2002) Cell surface major histocompatibility complex class II proteins are regulated by the products of the gamma(1)34.5 and U(L)41 genes of herpes simplex virus 1. J Virol 76: 6974–6986.

51. KellyBJ, FraefelC, CunninghamAL, DiefenbachRJ (2009) Functional roles of the tegument proteins of herpes simplex virus type 1. Virus Res 145: 173–186.

52. BresnahanWA, ShenkTE (2000) UL82 virion protein activates expression of immediate early viral genes in human cytomegalovirus-infected cells. Proc Natl Acad Sci U S A 97: 14506–14511.

53. RochfordR, LutzkeML, AlfinitoRS, ClavoA, CardinRD (2001) Kinetics of murine gammaherpesvirus 68 gene expression following infection of murine cells in culture and in mice. J Virol 75: 4955–4963.

54. GoodwinMM, MollestonJM, CannyS, Abou El HassanM, WillertEK, et al. (2010) Histone deacetylases and the nuclear receptor corepressor regulate lytic-latent switch gene 50 in murine gammaherpesvirus 68-infected macrophages. J Virol 84: 12039–12047.

55. FlanoE, HusainSM, SampleJT, WoodlandDL, BlackmanMA (2000) Latent murine gamma-herpesvirus infection is established in activated B cells, dendritic cells, and macrophages. J Immunol 165: 1074–1081.

56. Sunil-ChandraNP, EfstathiouS, ArnoJ, NashAA (1992) Virological and pathological features of mice infected with murine gamma-herpesvirus 68. J Gen Virol 73 (Pt 9) 2347–2356.

57. MilhoR, SmithCM, MarquesS, AlenquerM, MayJS, et al. (2009) In vivo imaging of murid herpesvirus-4 infection. J Gen Virol 90: 21–32.

58. NekorchukM, HanZ, HsiehTT, SwaminathanS (2007) Kaposi's sarcoma-associated herpesvirus ORF57 protein enhances mRNA accumulation independently of effects on nuclear RNA export. J Virol 81: 9990–9998.

59. SahinBB, PatelD, ConradNK (2010) Kaposi's sarcoma-associated herpesvirus ORF57 protein binds and protects a nuclear noncoding RNA from cellular RNA decay pathways. PLoS Pathog 6: e1000799.

60. AdlerH, MesserleM, WagnerM, KoszinowskiUH (2000) Cloning and mutagenesis of the murine gammaherpesvirus 68 genome as an infectious bacterial artificial chromosome. J Virol 74: 6964–6974.

61. SmythGK (2004) Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3: Article3.

62. WeinbergJB, LutzkeML, AlfinitoR, RochfordR (2004) Mouse strain differences in the chemokine response to acute lung infection with a murine gammaherpesvirus. Viral Immunol 17: 69–77.

63. R Core Team (2013). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. ISBN 3-900051-07-0, URL http://www.R-project.org/.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 1
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#